Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
J Immunol ; 174(9): 5750-7, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15843577

ABSTRACT

In glomerular diseases of diverse etiologies, dysfunction of the glomerular barrier to protein passage results in proteinuria, and proteinuria is considered an independent risk factor that plays a direct role in inflammation, interstitial fibrosis, and renal failure. The mechanism by which proteinuria leads to nephrotoxic injury is unclear, but a role for complement in mediating interstitial damage appears likely. We describe a strategy for Ag-specific targeting of complement inhibitors using a single chain Ab fragment and show that complement inhibitors targeted to the tubular epithelium protect against tubulointerstitial injury and renal dysfunction in a rat model of puromycin-induced nephrosis. The targeting of systemically administered complement inhibitors markedly enhanced their efficacy and obviated the need to systemically inhibit complement, thus reducing the risk of compromising host defense and immune homeostasis. Targeted inhibition of complement activation by Crry, and of membrane attack complex (MAC) formation by CD59 was equally therapeutic, demonstrating that the MAC plays a key role in proteinuria-induced tubulointerstitial injury. CD59 activity was dependent on its being targeted to the site of complement activation, and this is the first report of specific inhibition of the MAC in vivo after systemic administration of inhibitor. The data establish the MAC is a valid target for pharmaceutical intervention in proteinuric disorders and provide an approach to investigate the role of the MAC in complement-dependent disease under clinically relevant conditions.


Subject(s)
CD59 Antigens/metabolism , Complement Inactivator Proteins/metabolism , Complement Membrane Attack Complex/physiology , Kidney Tubules, Proximal/immunology , Kidney Tubules, Proximal/pathology , Nephrotic Syndrome/immunology , Nephrotic Syndrome/prevention & control , Receptors, Complement/metabolism , Animals , Antigens, Surface , CD59 Antigens/administration & dosage , Complement Inactivator Proteins/chemistry , Complement Inactivator Proteins/pharmacokinetics , Disease Models, Animal , Drug Delivery Systems/methods , Female , Kidney Tubules, Proximal/metabolism , Nephritis, Interstitial/immunology , Nephritis, Interstitial/pathology , Nephritis, Interstitial/prevention & control , Nephrotic Syndrome/pathology , Organ Specificity/immunology , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface , Receptors, Complement/administration & dosage , Receptors, Complement/blood , Receptors, Complement/chemistry , Recombinant Proteins/administration & dosage , Recombinant Proteins/blood , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacokinetics
2.
Am Heart J ; 147(1): 173-80, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14691437

ABSTRACT

BACKGROUND: Increase in vascular permeability and multiorgan dysfunction after cardiopulmonary bypass (CPB) are barriers to successful cardiac surgery in infants. Complement inhibition with TP10, a C3/C5 convertase inhibitor (AVANT Immunotherapeutics, Needham, Mass), blunts post-CPB organ dysfunction in the neonatal pig. Methods and results The pharmacokinetics and safety of TP10 in infants (age <1 year, n = 15) undergoing CPB were examined in a phase I/II open-label prospective trial. TP10 (10 mg/kg) was given intravenously before CPB and also added (10 mg/100 mL prime volume) to the CPB circuit. TP10 plasma levels correlated with C3a levels and measures of clinical course. All infants survived. No adverse events were attributed to TP10. TP10 plasma concentration fell to < or =60 microg/mL 12 hours after CPB. A 2-compartment model was fit to the TP10 blood levels as a function of time. Based on this model, an initial dose of 10 mg/kg over 0.5 hours followed by 10 mg/kg over 23.5 hours is the most appropriate for maintaining TP10 concentration between 100 microg/mL and 160 microg/mL for 24 hours after CPB. C3a was lower 12 hours after CPB than before CPB and still lower 24 hours after CPB. TP10 concentration was inversely correlated with the 12-hour post-CPB to pre-CPB ratio of C3a (Spearman rho -0.76, P = -.016), and with total (rho -0.56, P =.047) and net (rho -0.85, P =.0016) fluid and blood product administration/kg >24 hours after CPB. CONCLUSIONS: TP10 administration to infants appears safe. Pharmacokinetic analysis generated an optimal dosing strategy to achieve effective TP10 levels for 24 hours after CPB. In the infant, TP10 appears to decrease CPB-induced complement activation and protect vascular function. These results support a phase III trial of TP10 in infants requiring CPB.


Subject(s)
Cardiopulmonary Bypass/adverse effects , Complement Inactivator Proteins/pharmacokinetics , Heart Defects, Congenital/blood , Receptors, Complement/metabolism , Capillary Permeability , Complement Inactivator Proteins/adverse effects , Female , Heart Defects, Congenital/surgery , Humans , Infant , Male , Prospective Studies , Statistics, Nonparametric , Syndrome
3.
Br J Pharmacol ; 134(8): 1778-86, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11739255

ABSTRACT

1. Initiation of a peritoneal Arthus reaction by deposition of immune-complexes results in vascular leakage, polymorphonuclear leukocyte (PMN) infiltration, and tumour necrosis factor alpha (TNFalpha) and interleukin-6 (IL-6) production. We now demonstrate in rats that oral administration of the C5a receptor antagonist AcPhe[Orn-Pro-D-Cyclohexylalanine-Trp-Arg] (AcF-[OPdChaWR]; 1 - 10 mg kg(-1) 30 min prior to immune-complex deposition) inhibits these inflammatory markers in the peritoneal Arthus reaction. 2. Initiation of a dermal Arthus reaction resulted in a significant increase in vascular leakage, PMN infiltration, systemic production of TNFalpha and pathological changes in the dermis. 3. Pretreatment of rats with AcF-[OPdChaWR] either intravenously (1 mg kg(-1) 10 min prior to immune-complex deposition) or orally (1 - 10 mg kg(-1) 30 min prior to immune-complex deposition) significantly inhibited immune-complex mediated dermal vascular leakage and systemic cytokine production. Topical pretreatment with AcF-[OPdChaWR] (400 microg site(-1) in 10% dimethyl sulphoxide 10 min prior to immune-complex deposition) also inhibited vascular leakage, as well as histopathological changes associated with a dermal Arthus reaction. 4. Oral administration of 3 mg kg(-1) AcF-[OPdChaWR] resulted in the appearance of the drug in plasma within 5 min, with peak blood levels approximately 0.3 microM reached within 20 min. The plasma elimination half-life was approximately 70 min. The oral activity and bioavailability of AcF-[OPdChaWR], its activity when applied topically to the skin, suggest that small molecule C5a receptor antagonists may have therapeutic utility in dermal inflammatory disorders involving complement activation. 5. This is the first demonstration for either an orally or topically active C5a receptor antagonist, and suggests that small molecule C5a antagonists may have therapeutic utility when given by multiple routes of application.


Subject(s)
Arthus Reaction/drug therapy , Immunosuppressive Agents/pharmacology , Peptides, Cyclic/pharmacology , Receptors, Complement/antagonists & inhibitors , Administration, Oral , Administration, Topical , Animals , Antigens, CD , Arthus Reaction/immunology , Biological Availability , Biomarkers/analysis , Complement C5a/antagonists & inhibitors , Complement C5a/metabolism , Complement Inactivator Proteins/administration & dosage , Complement Inactivator Proteins/pharmacokinetics , Complement Inactivator Proteins/pharmacology , Cytokines/blood , Female , Half-Life , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/pharmacokinetics , Infusions, Intravenous , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/pharmacokinetics , Rats , Rats, Wistar , Receptor, Anaphylatoxin C5a , Time Factors
4.
J Immunol ; 166(10): 6341-8, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11342658

ABSTRACT

The anaphylatoxin C3a is a potent chemotactic peptide and inflammatory mediator released during complement activation which binds to and activates a G-protein-coupled receptor. Molecular cloning of the C3aR has facilitated studies to identify nonpeptide antagonists of the C3aR. A chemical lead that selectively inhibited the C3aR in a high throughput screen was identified and chemically optimized. The resulting antagonist, N(2)-[(2,2-diphenylethoxy)acetyl]-L-arginine (SB 290157), functioned as a competitive antagonist of (125)I-C3a radioligand binding to rat basophilic leukemia (RBL)-2H3 cells expressing the human C3aR (RBL-C3aR), with an IC(50) of 200 nM. SB 290157 was a functional antagonist, blocking C3a-induced C3aR internalization in a concentration-dependent manner and C3a-induced Ca(2+) mobilization in RBL-C3aR cells and human neutrophils with IC(50)s of 27.7 and 28 nM, respectively. SB 290157 was selective for the C3aR in that it did not antagonize the C5aR or six other chemotactic G protein-coupled receptors. Functional antagonism was not solely limited to the human C3aR; SB 290157 also inhibited C3a-induced Ca(2+) mobilization of RBL-2H3 cells expressing the mouse and guinea pig C3aRS: It potently inhibited C3a-mediated ATP release from guinea pig platelets and inhibited C3a-induced potentiation of the contractile response to field stimulation of perfused rat caudal artery. Furthermore, in animal models, SB 290157, inhibited neutrophil recruitment in a guinea pig LPS-induced airway neutrophilia model and decreased paw edema in a rat adjuvant-induced arthritis model. This selective antagonist may be useful to define the physiological and pathophysiological roles of the C3aR.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arginine/pharmacology , Benzhydryl Compounds/pharmacology , Complement C3a/metabolism , Complement Inactivator Proteins/pharmacology , Membrane Proteins , Receptors, Complement/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Arginine/analogs & derivatives , Arginine/metabolism , Arginine/pharmacokinetics , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Benzhydryl Compounds/metabolism , Benzhydryl Compounds/pharmacokinetics , Binding, Competitive , Cell Line , Complement Inactivator Proteins/metabolism , Complement Inactivator Proteins/pharmacokinetics , Disease Models, Animal , Edema/pathology , Edema/prevention & control , Guinea Pigs , Hindlimb , Humans , Injections, Intraperitoneal , Leukocytosis/immunology , Leukocytosis/pathology , Male , Mice , Muscle Contraction/drug effects , Neutrophil Infiltration/drug effects , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Receptors, Complement/metabolism , Tumor Cells, Cultured
5.
J Immunol ; 158(6): 2872-81, 1997 Mar 15.
Article in English | MEDLINE | ID: mdl-9058824

ABSTRACT

A chimeric gene was constructed from the genes coding for the human complement regulatory proteins, membrane cofactor protein (CD46) and decay-accelerating factor (CD55). The recombinant chimeric gene was transfected into Chinese hamster ovary cells. The gene product is a soluble, glycosylated, 110-kDa protein named complement activation blocker-2 (CAB-2). This protein possesses both factor I cofactor activity and decay-accelerating activity, and inactivates classical and alternative C3/C5 convertases in vitro. The specific activity of CAB-2 against cell-associated convertases is greater than that of soluble forms of either membrane cofactor protein or decay-accelerating factor or of both factors combined. CAB-2 also blocks the activation of complement in vivo, inhibiting both the Arthus reaction and Forssman shock in guinea pigs. Studies in rats demonstrate CAB-2 to exhibit favorable biphasic pharmacokinetics with a t1/2 alpha of 10 min and a t1/2 beta of 8 h; the beta phase accounts for 93% of the administered dose. CAB-2 may be an effective therapeutic treatment of acute human diseases in which excessive complement activation causes damage to normal tissues.


Subject(s)
Antigens, CD/genetics , CD55 Antigens/genetics , Complement Inactivator Proteins/genetics , Membrane Glycoproteins/genetics , Recombinant Fusion Proteins/pharmacology , Anaphylaxis/prevention & control , Animals , Antigens, CD/physiology , CD55 Antigens/physiology , Complement Activation/drug effects , Complement Inactivator Proteins/biosynthesis , Complement Inactivator Proteins/isolation & purification , Complement Inactivator Proteins/pharmacokinetics , Female , Guinea Pigs , Membrane Cofactor Protein , Membrane Glycoproteins/physiology , Protein Engineering , Rats , Rats, Inbred Strains , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/pharmacokinetics , Solubility
6.
Clin Exp Immunol ; 94(2): 348-53, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8222327

ABSTRACT

In this study the effect of K-76, a sesquiterpene compound with anticomplementary activity isolated from a fungus culture, on carrageenan-induced colitis was studied from biochemical, histological and immunohistopathological aspects. K-76 suppressed epithelial cell loss, crypt abscess formation, inflammatory cell infiltration, mucosal atrophy, and ulceration. Immunohistochemical examination of the colonic mucosa showed that the number of IgG- and IgM-positive plasma cells and the staining intensity for IgG and C3 were increased in carrageenan-induced colitis, but these changes were inhibited by K-76. Besides, serum mucoprotein concentrations and CH50 levels were lower in the animals treated with carrageenan alone. K-76 exerted multifunctional activity, although its mechanisms of action remain obscure.


Subject(s)
Colitis, Ulcerative/drug therapy , Complement Inactivator Proteins/pharmacology , Sesquiterpenes/pharmacology , Animals , Carrageenan/toxicity , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/immunology , Complement C3/metabolism , Complement Hemolytic Activity Assay , Complement Inactivator Proteins/pharmacokinetics , Disease Models, Animal , Immunoglobulin G/metabolism , Immunoglobulin M/metabolism , Immunohistochemistry , Male , Mucoproteins/blood , Rabbits , Sesquiterpenes/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...