Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nanomedicine ; 12(4): 1023-1031, 2016 May.
Article in English | MEDLINE | ID: mdl-26733258

ABSTRACT

Hypersensitivity reactions to particulate drugs can partly be caused by complement activation and represent a major complication during intravenous application of nanomedicines. Several liposomal and micellar drugs and carriers, and therapeutic antibodies, were shown to activate complement and induce complement activation-related pseudoallergy (CARPA) in model animals. To explore the possible use of the natural complement inhibitor factor H (FH) against CARPA, we examined the effect of FH on complement activation induced by CARPAgenic drugs. Exogenous FH inhibited complement activation induced by the antifungal liposomal Amphotericin-B (AmBisome), the widely used solvent of anticancer drugs Cremophor EL, and the anticancer monoclonal antibody rituximab in vitro. An engineered form of FH (mini-FH) was more potent inhibitor of Ambisome-, Cremophor EL- and rituximab-induced complement activation than FH. The FH-related protein CFHR1 had no inhibitory effect. Our data suggest that FH or its derivatives may be considered in the pharmacological prevention of CARPA. FROM THE CLINICAL EDITOR: Although liposomes and micelles are already in use in the clinical setting as drug carriers, there remains the potential problem of hypersensitivity due to complement activation. In this article, the authors investigated the use of complement inhibitor factor H (FH) on complement activation and showed good efficacy. The results would therefore suggest the potential application of complement inhibitor in the future.


Subject(s)
Complement Activation/drug effects , Complement Factor H/administration & dosage , Drug Hypersensitivity/drug therapy , Liposomes/adverse effects , Complement Activation/immunology , Complement Factor H/immunology , Drug Carriers/adverse effects , Drug Hypersensitivity/immunology , Drug Hypersensitivity/pathology , Healthy Volunteers , Humans , Micelles , Nanomedicine , Neoplasms/drug therapy , Neoplasms/immunology , Rituximab/adverse effects
2.
J Gene Med ; 17(10-12): 229-43, 2015.
Article in English | MEDLINE | ID: mdl-26369397

ABSTRACT

BACKGROUND: Age-related macular degeneration (AMD) is the most common cause of blindness in the elderly, with no therapy available for 90% of patients. Recent genetic evidence implicates activation of complement in the pathogenesis of AMD. We have recently discovered that adenovirus (Ad)-mediated expression of complement component C3 (AdCMVC3) in the murine retina recapitulates many of the pathological features found in human AMD. In the present study, utilizing a gene therapy approach, we examine whether Ad-mediated expression of complement Factor H (AdCAGfH) attenuates AdCMVC3-mediated retinal pathology. METHODS: AdCMVC3 was co-injected with either AdCAGfH or a negative control virus expressing green fluorescent protein (AdCMVGFP) into the subretinal space of adult mice. The resulting retinal pathology was analyzed by histology and immunocytochemistry and retinal function was quantified by electroretinography. RESULTS: Morphological and functional analyses indicated that AdCMVC3-mediated retinal pathology could be attenuated by AdCAGfH. Specifically, endothelial cell proliferation was reduced by 91% and atrophy of retinal pigment epithelium (RPE) could be attenuated by 69%. AdCAGfH injected eyes exhibited 90-150% greater A-wave and 120-180% greater B-wave amplitudes relative to control eyes. Immunocytochemical analysis of rhodopsin and RPE65 was consistent with the rescue of photoreceptors and RPE in AdCAGfH injected eyes. CONCLUSIONS: C3-induced pathology in murine retina can be attenuated by Ad-mediated expression of Factor H. Expression of Factor H is worthy of further study as a potential gene therapy for AMD.


Subject(s)
Complement C3/metabolism , Complement Factor H/therapeutic use , Adenoviridae , Animals , Complement C3/adverse effects , Complement Factor H/administration & dosage , Genetic Therapy , Genetic Vectors , Humans , Macular Degeneration/therapy , Mice , Retina/drug effects , Retina/pathology
3.
Clin Vaccine Immunol ; 21(10): 1452-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25143339

ABSTRACT

The emergence of antimicrobial resistance among several medically important pathogens represents a serious threat to human health globally and necessitates the development of novel therapeutics. Complement forms a key arm of innate immune defenses against invading pathogens. A mechanism of complement evasion employed by many pathogens is binding of complement inhibitors, including factor H (FH), a key downregulator of the alternative pathway. Most FH-binding bacteria engage FH through regions in FH spanned by domains 6 and 7 and/or 18 through 20. We created a chimeric protein that comprised human FH domains 6 and 7 fused to human IgG1 Fc (FH6,7/HuFc) and tested its activity as an immunotherapeutic against Neisseria meningitidis, which binds FH through domains 6 and 7. FH6,7/HuFc bound to meningococci and effectively blocked FH binding to bacteria. FH6,7/HuFc enhanced human C3 and C4 deposition and facilitated complement-mediated killing in a dose-responsive manner; complement activation and killing were classical pathway dependent. To investigate in vivo efficacy, infant Wistar rats were treated intraperitoneally (IP) with different doses of FH6,7/HuFc and challenged 2 h later with serogroup C strain 4243 given IP. At 8 to 9 h after the challenge, the FH6,7/HuFc-treated rats had >100-fold fewer CFU per ml of blood than control animals pretreated with phosphate-buffered saline (PBS) or FH18-20/HuFc, which does not bind to meningococci (P < 0.0001). These data provide proof of concept of the utility of FH/Fc fusion proteins as anti-infective immunotherapeutics. Because many microbes share a common binding region(s) in FH, FH/Fc chimeric proteins may be a promising candidate for adjunctive therapy against drug-resistant pathogens.


Subject(s)
Anti-Bacterial Agents/pharmacology , Complement Factor H/pharmacology , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulin G/pharmacology , Immunotherapy/methods , Meningococcal Infections/therapy , Neisseria meningitidis, Serogroup C/drug effects , Animals , Anti-Bacterial Agents/administration & dosage , Blood/microbiology , Colony Count, Microbial , Complement Factor H/administration & dosage , Complement Factor H/genetics , Complement System Proteins/immunology , Complement System Proteins/metabolism , Disease Models, Animal , Immunoglobulin Fc Fragments/administration & dosage , Immunoglobulin Fc Fragments/genetics , Immunoglobulin G/administration & dosage , Immunoglobulin G/genetics , Microbial Viability , Neisseria meningitidis, Serogroup C/physiology , Rats, Wistar , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Treatment Outcome
4.
Br J Ophthalmol ; 97(3): 367-70, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23258212

ABSTRACT

PURPOSE: To investigate the inhibitory effect of intravitreally administered human complement factor H (CFH) in a rat model of laser-induced choroidal neovascularisation (CNV). METHODS: Analysis of alternative pathway inhibition by human plasma-purified CFH was conducted by measuring C3 deposition on zymosan particles using rat serum. CNV was induced by laser photocoagulation on Day 0 in the eyes of Brown Norway rats. Human plasma-purified CFH (50 µg/2 µl) or phosphate buffered saline was injected intravitreally on Day 0 (prevention arm) or Day 7 (treatment arm). Seven days after injection, eyes were enucleated and retinal pigment epithelium-choroid-sclera flat mounts were prepared. Areas of CNV were determined in flat mounts and quantified using an image analysis programme. Flat mounts were also stained for membrane attack complex. RESULTS: In rat serum, human CFH inhibited activity of alternative pathway in a dose-dependent manner. On Day 3, mean membrane attack complex deposition in laser-treated retina significantly decreased in CFH-treated eyes (p<0.001). In the prevention arm, the mean CNV area in CFH-treated eyes decreased by 27.0% compared with phosphate buffered saline-treated control eyes on Day 7 (p=0.011). In the treatment arm, the mean CNV area in CFH-treated eyes decreased by 38.3% compared with control eyes on Day 14 (p=0.001). CONCLUSIONS: Intravitreal injection of human CFH resulted in the suppression of formation of new, and regression of preformed laser-induced CNV in the rat model. Human CFH may be a feasible treatment for CNV associated with age-related macular degeneration or other causes.


Subject(s)
Choroidal Neovascularization/drug therapy , Complement Factor H/administration & dosage , Lasers/adverse effects , Animals , Choroidal Neovascularization/etiology , Choroidal Neovascularization/pathology , Complement Inactivating Agents/administration & dosage , Disease Models, Animal , Humans , Intravitreal Injections , Rats , Rats, Inbred BN , Treatment Outcome
5.
Blood ; 118(17): 4705-13, 2011 Oct 27.
Article in English | MEDLINE | ID: mdl-21860027

ABSTRACT

To selectively modulate human complement alternative pathway (CAP) activity implicated in a wide range of acute and chronic inflammatory conditions and to provide local cell surface and tissue-based inhibition of complement-induced damage, we developed TT30, a novel therapeutic fusion protein linking the human complement receptor type 2 (CR2/CD21) C3 fragment (C3frag = iC3b, C3dg, C3d)-binding domain with the CAP inhibitory domain of human factor H (fH). TT30 efficiently blocks ex vivo CAP-dependent C3frag accumulation on activated surfaces, membrane attack complex (MAC) formation and hemolysis of RBCs in a CR2-dependent manner, and with a ∼ 150-fold potency gain over fH, without interference of C3 activation or MAC formation through the classic and lectin pathways. TT30 protects RBCs from hemolysis and remains bound and detectable for at least 24 hours. TT30 selectively inhibits CAP in cynomolgus monkeys and is bioavailable after subcutaneous injection. Using a unique combination of targeting and effector domains, TT30 controls cell surface CAP activation and has substantial potential utility for the treatment of human CAP-mediated diseases.


Subject(s)
Complement C3-C5 Convertases/antagonists & inhibitors , Complement C3d/metabolism , Complement Factor H/therapeutic use , Complement Pathway, Alternative/immunology , Drug Discovery , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/therapeutic use , Immune System Diseases/drug therapy , Immune System Diseases/immunology , Receptors, Complement 3d/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Animals , Complement C3-C5 Convertases/metabolism , Complement Factor H/administration & dosage , Drug Design , Drug Evaluation, Preclinical , Female , Humans , Immune System Diseases/metabolism , Macaca fascicularis , Male , Models, Immunological , Molecular Targeted Therapy/methods , Rabbits , Receptors, Complement 3d/administration & dosage , Recombinant Fusion Proteins/administration & dosage
6.
Peptides ; 31(5): 872-7, 2010 May.
Article in English | MEDLINE | ID: mdl-20132852

ABSTRACT

Sepsis is a serious complication for patients with obstructive jaundice. Although administration of adrenomedullin (AM) in combination with its binding protein (AMBP-1) is protective after injury, it remains unknown whether AM/AMBP-1 ameliorates sepsis-induced organ injury and mortality in the setting of biliary obstruction. The aim of this study is, therefore, to test the efficacy of human AM/AMBP-1 in a rat model of obstructive jaundice and polymicrobial sepsis. To study this, obstructive jaundice was induced in male adult rats (275-325g) by common bile duct ligation (BDL). One week after BDL, the rats were subjected to sepsis by cecal ligation and puncture (CLP). Plasma levels of AM and AMBP-1 were measured at 20h after CLP. In additional groups of BDL+CLP rats, human AM/AMBP-1 (24/80microg/kg body weight (BW)) or vehicle (i.e., human albumin) was administered intravenously at 5h after CLP. Blood and tissue samples were collected at 20h after CLP for various measurements. To determine the long-term effect of human AM/AMBP-1 after BDL+CLP, the gangrenous cecum was removed at 20h after CLP and 7-day survival was recorded. Our results showed that plasma levels of AM were significantly increased while AMBP-1 levels were markedly decreased after BDL+CLP (n=8, P<0.05). Administration of human AM/AMBP-1 attenuated tissue injury and inflammatory responses after BDL+CLP. Moreover, human AM/AMBP-1 significantly increased the survival rate from 21% (n=14) to 53% (n=15). Thus, human AM/AMBP-1 ameliorates sepsis-induced organ injury and mortality in jaundiced rats. Human AM/AMBP-1 can be further developed as a novel treatment for sepsis in jaundiced patients.


Subject(s)
Adrenomedullin/blood , Adrenomedullin/therapeutic use , Complement Factor H/metabolism , Complement Factor H/therapeutic use , Jaundice, Obstructive/blood , Sepsis/drug therapy , Sepsis/physiopathology , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Cecum/surgery , Complement Factor H/administration & dosage , Creatinine/blood , Humans , Interleukin-6/blood , Lactic Acid/blood , Ligation , Male , Rats , Rats, Sprague-Dawley , Sepsis/blood , Tumor Necrosis Factor-alpha/blood
7.
J Immunol ; 183(9): 5928-37, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19828624

ABSTRACT

The alternative pathway (AP) of complement is required for the induction of collagen Ab-induced arthritis (CAIA) in mice. The objective of this study was to examine the effect of a recombinant AP inhibitor containing complement receptor 2 and factor H (CR2-fH) on CAIA in mice. CR2 binds to tissue-fixed activation fragments of C3, and the linked fH is a potent local inhibitor of the AP. CAIA was induced in C57BL/6 mice by i.p. injections of 4 mAb to type II collagen (CII) on day 0 and LPS on day 3. PBS or CR2-fH (250 or 500 microg) were injected i.p. 15 min after the mAb to CII on day 0 and 15 min after LPS on day 3; the mice were sacrificed on day 10. The disease activity score (DAS) was decreased significantly (p < 0.001) in both groups receiving CR2-fH compared with the PBS. Histology scores for inflammation, pannus, bone damage, and cartilage damage decreased in parallel with the DAS. C3 deposition in the synovium and cartilage was significantly reduced (p < 0.0001) in the mice treated with CR2-fH. In vitro studies with immune complexes containing type II collagen and mAb to CII showed that CR2-fH specifically inhibited the AP with minimal effect on the classical pathway (CP) and no effect on the lectin pathway (LP). The relative potency of CR2-fH in vitro was superior to mAbs to factor B and C5. Thus, CR2-fH specifically targets and inhibits the AP of complement in vitro and is effective in CAIA in vivo.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Arthritis, Experimental/immunology , Collagen Type II/immunology , Complement Factor H/physiology , Complement Inactivator Proteins/physiology , Complement Pathway, Alternative/immunology , Receptors, Complement 3d/physiology , Recombinant Fusion Proteins/physiology , Animals , Arthritis, Experimental/pathology , Arthritis, Experimental/therapy , Cattle , Complement Factor H/administration & dosage , Complement Inactivator Proteins/administration & dosage , Drug Combinations , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Complement 3d/administration & dosage , Recombinant Fusion Proteins/administration & dosage
8.
Mol Immunol ; 46(10): 1942-50, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19411110

ABSTRACT

Dense deposit disease (DDD) is strongly associated with the uncontrolled activation of the complement alternative pathway. Factor H (CFH)-deficient (Cfh(-/-)) mice spontaneously develop C3 deposition along the glomerular basement membrane (GBM) with subsequent development of glomerulonephritis with features of DDD, a lesion dependent on C3 activation. In order to understand the role of CFH in preventing renal damage associated with the dysregulation of the alternative pathway we administered purified mouse CFH (mCFH) to Cfh(-/-) mice. 24h following the administration of mCFH we observed an increase in plasma C3 levels with presence of intact C3 in circulation showing that mCFH restored control of C3 activation in fluid phase. mCFH resulted in the reduction of iC3b deposition along the GBM. The exogenous mCFH was readily detectable in plasma but critically not in association with C3 along the GBM. Thus, the reduction in GBM C3 was dependent on the ability of mCFH to regulate C3 activation in plasma. Western blot analysis of glomeruli from Cfh(-/-) mice demonstrated the presence of iC3b. Our data show that the C3 along the GBM in Cfh(-/-) mice is the C3 fragment iC3b and that this is derived from plasma C3 activation. The implication is that successful therapy of DDD is likely to be achieved by therapies that inhibit C3 turnover in plasma.


Subject(s)
Complement Activation/immunology , Complement C3b/immunology , Complement Factor H/immunology , Glomerular Basement Membrane/immunology , Animals , CD11b Antigen/immunology , Complement Activation/drug effects , Complement C3/biosynthesis , Complement Factor H/administration & dosage , Complement Factor H/deficiency , Complement Factor H/pharmacology , Glomerular Basement Membrane/drug effects , Humans , Kidney/drug effects , Kidney/immunology , Mice , Neutrophils/cytology , Neutrophils/drug effects , Neutrophils/immunology , Protein Binding/drug effects , Protein Transport/drug effects
9.
Ann Surg ; 249(2): 310-7, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19212187

ABSTRACT

OBJECTIVE: To determine whether administration of a vasoactive peptide, human adrenomedullin (AM), in combination with its binding protein (ie, AMBP-1), prevents or minimizes hepatic ischemia-reperfusion (I/R) injury. SUMMARY BACKGROUND DATA: Hepatic I/R injury results from tissue hypoxia and subsequent inflammatory responses. Even though numerous pharmacological modalities and substances have been studied to reduce I/R-induced mortality, none have been entirely successful. We have shown that administration of AM/AMBP-1 produces significant beneficial effects under various pathophysiological conditions. However, it remains unknown if human AM/AMBP-1 has any protective effects on hepatic I/R-induced tissue damage and mortality. METHODS: Seventy percent hepatic ischemia was induced in male adult rats by placing a microvascular clip across the hilum of the left and median lobes for 90 minutes. After removing the clip, human AM alone, human AMBP-1 alone, human AM in combination with human AMBP-1 or vehicle was administered intravenously over a period of 30 minutes. Blood and tissue samples were collected 4 hours after reperfusion for various measurements. In additional groups of animals, the nonischemic liver lobes were resected at the end of 90-minute ischemia. The animals were monitored for 7 days and survival was recorded. RESULTS: After hepatic I/R, plasma levels of AM were significantly increased, whereas AMBP-1 levels were markedly decreased. Likewise, gene expression of AM in the liver was increased significantly, whereas AMBP-1 expression was markedly decreased. Administration of AM in combination with AMBP-1 immediately after the onset of reperfusion down-regulated inflammatory cytokines, decreased hepatic neutrophil infiltration, inhibited liver cell apoptosis and necrosis, and reduced liver injury and mortality in a rat model of hepatic I/R. On the other hand, administration of human AM alone or human AMBP-1 alone after hepatic I/R failed to produce significant protection. CONCLUSIONS: Human AM/AMBP-1 may be a novel treatment to attenuate tissue injury after an episode of hepatic ischemia.


Subject(s)
Adrenomedullin/administration & dosage , Complement Factor H/administration & dosage , Liver Diseases/prevention & control , Liver/blood supply , Reperfusion Injury/prevention & control , Vasodilator Agents/administration & dosage , Animals , Disease Models, Animal , Liver Diseases/mortality , Male , Rats , Rats, Sprague-Dawley , Reperfusion Injury/mortality
10.
Regul Pept ; 152(1-3): 82-7, 2009 Jan 08.
Article in English | MEDLINE | ID: mdl-18948146

ABSTRACT

Previous studies have demonstrated that co-administration of rat adrenomedullin (AM) and human AM binding protein-1 (AMBP-1) has various beneficial effects following adverse circulatory conditions. In order to reduce rat proteins to elicit possible immune responses in humans, we determined the effect of human AM combined with human AMBP-1 after intestinal ischemia and reperfusion (I/R). Intestinal ischemia was induced in the rat by occluding the superior mesenteric artery for 90 min. At 60 min after the beginning of reperfusion, human AM/AMBP-1 at 3 different dosages was administered intravenously over 30 min. At 240 min after the treatment, blood and tissue samples were harvested and measured for pro-inflammatory cytokines (i.e., TNF-alpha and IL-6), myeloperoxidase activities in the gut and lungs, and cleaved caspase-3 expression in the lungs, as well as serum levels of hepatic enzymes and lactate. In additional groups of animals, a 10-day survival study was conducted. Results showed that administration of human AM/AMBP-1 reduced pro-inflammatory cytokines, attenuated organ injury, and improved the survival rate in a seemingly dose-response fashion. Co-administration of the highest dose of human AM/AMBP-1 in this study had the optimal therapeutic effect in the rat model of intestinal I/R.


Subject(s)
Adrenomedullin/pharmacology , Complement Factor H/pharmacology , Intestines/blood supply , Ischemia/metabolism , Reperfusion Injury/metabolism , Adrenomedullin/administration & dosage , Adrenomedullin/metabolism , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Complement Factor H/administration & dosage , Complement Factor H/metabolism , Cytokines/metabolism , Humans , Lactic Acid/blood , Rats , Rats, Sprague-Dawley , Survival Rate , Tumor Necrosis Factor-alpha/metabolism
11.
Peptides ; 29(7): 1223-30, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18403050

ABSTRACT

We recently discovered that vascular responsiveness to adrenomedullin (AM), a vasoactive hormone, decreases after hemorrhage, which is markedly improved by the addition of its binding protein AMBP-1. One obstacle hampering the development of AM/AMBP-1 as resuscitation agents in trauma victims is the potential immunogenicity of rat proteins in humans. Although less potent than rat AM, human AM has been shown to increase organ perfusion in rats. We therefore hypothesized that administration of human AM/AMBP-1 improves organ function and survival after severe blood loss in rats. To test this, male Sprague-Dawley rats were bled to and maintained at an MAP of 40 mmHg for 90 min. They were then resuscitated with an equal volume of shed blood in the form of Ringer's lactate (i.e., low-volume resuscitation) over 60 min. At 15 min after the beginning of resuscitation, human AM/AMBP-1 (12/40 or 48/160 microg/kg BW) were administered intravenously over 45 min. Various pathophysiological parameters were measured 4h after resuscitation. In additional groups of animals, a 12-day survival study was conducted. Our result showed that tissue injury as evidenced by increased levels of transaminases, lactate, and creatinine, was present at 4h after hemorrhage and resuscitation. Moreover, pro-inflammatory cytokines TNF-alpha and IL-6 were also significantly elevated. Administration of AM/AMBP-1 markedly attenuated tissue injury, reduced cytokine levels, and improved the survival rate from 29% (vehicle) to 62% (low-dose) or 70% (high-dose). However, neither human AM alone nor human AMBP-1 alone prevented the significant increase in ALT, AST, lactate and creatinine at 4h after the completion of hemorrhage and resuscitation. Moreover, the half-life of human AM and human AMBP-1 in rats was 35.8 min and 1.68 h, respectively. Thus, administration of human AM/AMBP-1 may be a useful approach for attenuating organ injury, and reducing mortality after hemorrhagic shock.


Subject(s)
Adrenomedullin/pharmacology , Complement Factor H/pharmacology , Shock, Hemorrhagic/therapy , Vasodilator Agents/pharmacology , Adrenomedullin/administration & dosage , Adrenomedullin/metabolism , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Complement Factor H/administration & dosage , Creatinine/blood , Dose-Response Relationship, Drug , Half-Life , Humans , Lactic Acid/blood , Male , Models, Animal , Rats , Rats, Sprague-Dawley , Resuscitation/methods , Shock, Hemorrhagic/metabolism , Survival Analysis , Time Factors , Vasodilator Agents/administration & dosage , Vasodilator Agents/metabolism
12.
J Clin Invest ; 118(2): 608-18, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18202746

ABSTRACT

The inflammatory kidney disease membranoproliferative glomerulonephritis type II (MPGN2) is associated with dysregulation of the alternative pathway of complement activation. MPGN2 is characterized by the presence of complement C3 along the glomerular basement membrane (GBM). Spontaneous activation of C3 through the alternative pathway is regulated by 2 plasma proteins, factor H and factor I. Deficiency of either of these regulators results in uncontrolled C3 activation, although the breakdown of activated C3 is dependent on factor I. Deficiency of factor H, but not factor I, is associated with MPGN2 in humans, pigs, and mice. To explain this discordance, mice with single or combined deficiencies of these factors were studied. MPGN2 did not develop in mice with combined factor H and I deficiency or in mice deficient in factor I alone. However, administration of a source of factor I to mice with combined factor H and factor I deficiency triggered both activated C3 fragments in plasma and GBM C3 deposition. Mouse renal transplant studies demonstrated that C3 deposited along the GBM was derived from plasma. Together, these findings provide what we believe to be the first evidence that factor I-mediated generation of activated C3 fragments in the circulation is a critical determinant for the development of MPGN2 associated with factor H deficiency.


Subject(s)
Complement C3/metabolism , Complement Factor H/deficiency , Complement Factor I/physiology , Glomerulonephritis, Membranoproliferative/genetics , Animals , Complement C3/analysis , Complement Factor H/administration & dosage , Complement Factor H/genetics , Complement Factor I/administration & dosage , Complement Factor I/genetics , Glomerulonephritis, Membranoproliferative/blood , Glomerulonephritis, Membranoproliferative/immunology , Mice , Mice, Mutant Strains
13.
J Biomed Mater Res A ; 76(1): 25-34, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16250010

ABSTRACT

In the present work we have bound Pluronic, a class of triblock copolymers consisting of a block of polypropylene oxide (PPO) surrounded on each side by polyethylene oxide (PEO) blocks, to polystyrene surfaces and investigated the thrombogenicity and complement activation of this construct upon exposure to whole blood. The surface was highly inert towards coagulation, unfortunately at the expense of increased complement activation. We, therefore, as an alternative approach, used End-Group Activated Pluronic to conjugate factor H, a regulator of complement activation (RCA), to the surface. The bound factor H did not detach from the surface upon incubation with human serum. Furthermore, factor H bound in a physiological conformation could to a significant degree attenuate complement activation at the Pluronic surface. Thus, we have created a hybrid surface in which the coagulation-inert properties of the original Pluronic are supplemented with a specific complement-inhibitory effect. Medical device technology includes numerous potential applications for crosslinkers that are capable of specifically binding biomolecules to surfaces with retained activity. These applications include coupling of functional biomolecules to biomedical devices such as stents and grafts. The biomolecule may be an RCA, antibody, or other beneficial ligand.


Subject(s)
Biocompatible Materials , Blood Coagulation/drug effects , Complement Activation/drug effects , Complement Factor H/administration & dosage , Poloxamer , Polyethylene Glycols , Complement Factor H/isolation & purification , Cross-Linking Reagents , Drug Stability , Humans , In Vitro Techniques , Materials Testing , Succinimides , Surface Properties , Surface-Active Agents
SELECTION OF CITATIONS
SEARCH DETAIL