Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Neurosci Res ; 87: 26-32, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25011063

ABSTRACT

Peripheral nerve injury leads to Wallerian degeneration, followed by regeneration, in which functionality and morphology of the nerve are restored. We previously described that deficiency for complement component C6, which prevents formation of the membrane attack complex, slows down degeneration and results in an earlier recovery of sensory function after sciatic nerve injury compared to WT animals. In this study, we determine whether C6(-/-) rats have an intrinsic trait that affects sciatic nerve regeneration after injury. To study the contribution of complement activation on degeneration and regeneration with only minimal effect of complement activation, a crush injury model with only modest complement deposition was used. We compared the morphological and functional aspects of crushed nerves during degeneration and regeneration in C6(-/-) and WT animals. Morphological changes of myelin and axons showed similar degeneration and regeneration patterns in WT and C6(-/-) injured nerves. Functional degeneration and regeneration, recorded by ex vivo electrophysiology and in vivo foot flick test, showed that the timeline of the restoration of nerve conduction and sensory recovery also followed similar patterns in WT and C6(-/-) animals. Our findings suggest that C6 deficiency by itself does not alter the regrowth capacity of the peripheral nerve after crush injury.


Subject(s)
Complement C6/deficiency , Nerve Regeneration , Peripheral Nerve Injuries/physiopathology , Sciatic Nerve/injuries , Sciatic Nerve/physiology , Animals , Axons/physiology , Complement C6/physiology , Male , Myelin Sheath/physiology , Rats , Wallerian Degeneration/physiopathology
2.
J Immunol ; 188(3): 1469-78, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22205026

ABSTRACT

The complement system, especially the alternative pathway, plays essential roles in the induction of injury in collagen Ab-induced arthritis (CAIA) in mice. The goal of the current study was to directly compare the roles of receptors for C3a and C5a, as well as the membrane attack complex, as effector mechanisms in the pathogenesis of CAIA. Clinical disease activity in C3aR(-/-), C5aR(-/-), and C6-deficient (C6-def) mice was decreased by 52, 94, and 65%, respectively, as compared with wild-type mice. Decreases in histopathologic injury as well as in IgG and C3 deposition paralleled the clinical disease activity. A decrease in the percentage of synovial neutrophils was observed in C3aR(-/-), C5aR(-/-), and C6-def mice, and a decrease in macrophages was observed in C3aR(-/-) and C5aR(-/-), but not in C6-def, mice. Synovial mRNA obtained by laser capture microdissection exhibited a decrease in TNF-α in C5aR(-/-) mice and in IL-1ß in both C5aR(-/-) and C6-def mice, whereas C3aR(-/-) mice demonstrated no change in either cytokine. Our findings show that absent C3aR-, C5aR-, or membrane attack complex-initiated effector mechanisms each decrease susceptibility to CAIA, with clinical effects most pronounced in C5aR-deficient mice. Although the absence of C3aR, C5aR, or C6 led to differential deficiencies in effector mechanisms, decreased proximal joint IgG and C3 deposition was common to all three genotypes in comparison with wild-type mice. These data suggest the existence of positive-feedback amplification pathways downstream of all three effectors that promote additional IgG deposition and C3 activation in the joint.


Subject(s)
Arthritis, Experimental/etiology , Complement C6/deficiency , Receptor, Anaphylatoxin C5a/physiology , Receptors, Complement/physiology , Animals , Complement Activation , Complement C3a/immunology , Complement C6/physiology , Cytokines , Disease Susceptibility , Immunoglobulin G , Macrophages/pathology , Mice , Mice, Knockout , Neutrophils/pathology , Synovial Fluid/immunology
4.
Transplantation ; 82(7): 857-64, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17038898

ABSTRACT

BACKGROUND: Many patients on the waiting list for transplants are sensitized from previous blood transfusions, pregnancy, or transplants. We investigated the role of complement in acute and chronic pathology in hearts transplanted to sensitized rats. METHODS: Blood was transfused from allogeneic PVG.R8 rats or control isogeneic PVG.1U rats to C6-sufficient and -deficient PVG.1U rats. Three weeks later hearts were transplanted from PVG.R8 donors and low-dose cyclosporin A was initiated. RESULTS: Allogeneic but not isogeneic blood transfusion elicited strong immunoglobulin (Ig) M, IgG1 and IgG2b alloantibody responses. Sensitization caused accelerated acute rejection of cardiac allografts by C6-sufficient recipients (4 days). In contrast, allografts functioned over 40 days in all C6-deficient recipients, but sensitization caused increased interstitial fibrosis and chronic vasculopathy. Circulating alloantibodies were associated with deposits of C4d on the vascular endothelium together with pericapillary accumulation of neutrophils and macrophages in the grafts. In contrast, T cells accumulated in periarterial lymphatics that did not have C4d deposits. CONCLUSIONS: Presensitization by allogeneic blood transfusion causes accelerated acute graft rejection in the presence of the complete complement cascade. In the absence of C6, macrophages colocalized with deposits of C4d and T cells accumulated in the periarterial lymphatics.


Subject(s)
Antibody Formation , Complement C6/deficiency , Complement C6/physiology , Heart Transplantation/immunology , Isoantibodies/blood , Transfusion Reaction , Animals , Gene Deletion , Graft Survival , Immunoglobulin G/blood , Immunoglobulin G/classification , Immunoglobulin M/blood , Macrophages/immunology , Major Histocompatibility Complex , Models, Animal , Rats , Rats, Inbred Strains , Transplantation, Homologous , Transplantation, Isogeneic
5.
Infect Immun ; 74(5): 2803-8, 2006 May.
Article in English | MEDLINE | ID: mdl-16622217

ABSTRACT

The relative contributions of antibody-induced complement-mediated bacterial lysis and antibody/complement-mediated phagocytosis to host immunity against meningococcal infections are currently unclear. Further, the in vivo effector functions of antibodies may vary depending on their specificity and Fc heavy-chain isotype. In this study, a mouse immunoglobulin G2a (mIgG2a) monoclonal antibody (MN12H2) to meningococcal outer membrane protein PorA (P1.16), its human IgG subclass derivatives (hIgG1 to hIgG4), and an mIgG2a monoclonal antibody (Nmb735) to serogroup B capsular polysaccharide (B-PS) were evaluated for passive protection against meningococcal serogroup B strain 44/76-SL (B:15:P1.7,16) in an infant rat infection model. Complement component C6-deficient (PVG/c-) rats were used to assess the importance of complement-mediated bacterial lysis for protection. The PorA-specific parental mIgG2a and the hIgG1 to hIgG3 derivatives all induced efficient bactericidal activity in vitro in the presence of human or infant rat complement and augmented bacterial clearance in complement-sufficient HsdBrlHan:WIST rats, while the hIgG4 was unable to do so. In C6-deficient PVG/c- rats, lacking complement-mediated bacterial lysis, the augmentation of bacterial clearance by PorA-specific mIgG2a and hIgG1 antibodies was impaired compared to that in the syngeneic complement-sufficient PVG/c+ rat strain. This was in contrast to the case for B-PS-specific mIgG2a, which conferred similar protective activity in both rat strains. These data suggest that while anti-B-PS antibody can provide protection in the infant rats without membrane attack complex formation, the protection afforded by anti-PorA antibody is more dependent on the activation of the whole complement pathway and subsequent bacterial lysis.


Subject(s)
Antibodies, Bacterial/immunology , Bacterial Capsules/immunology , Complement C6/physiology , Neisseria meningitidis, Serogroup B/immunology , Porins/immunology , Animals , Blood Bactericidal Activity , Immunoglobulin G/immunology , Immunoglobulin Isotypes/immunology , Mice , Rats
6.
J Virol ; 80(8): 4038-46, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16571820

ABSTRACT

Glycoprotein C (gC) of herpes simplex virus type 1 (HSV-1) and type 2 (HSV-2) binds complement component C3b and protects virus from complement-mediated neutralization. Differences in complement interacting domains exist between gC of HSV-1 (gC1) and HSV-2 (gC2), since the amino terminus of gC1 blocks complement C5 from binding to C3b, while gC2 fails to interfere with this activity. We previously reported that neutralization of HSV-1 gC-null virus by HSV antibody-negative human serum requires activation of C5 but not of downstream components of the classical complement pathway. In this report, we evaluated whether activation of C5 is sufficient to neutralize HSV-2 gC-null virus, or whether formation of the membrane attack complex by C6 to C9 is required for neutralization. We found that activation of the classical complement pathway up to C5 was sufficient to neutralize HSV-2 gC-null virus by HSV antibody-negative human serum. We evaluated the mechanisms by which complement activation occurred in seronegative human serum. Interestingly, natural immunoglobulin M antibodies bound to virus, which triggered activation of C1q and the classical complement pathway. HSV antibody-negative sera obtained from four individuals differed over an approximately 10-fold range in their potency for complement-mediated virus neutralization. These findings indicate that humans differ in the ability of their innate immune systems to neutralize HSV-1 or HSV-2 gC-null virus and that a critical function of gC1 and gC2 is to prevent C5 activation.


Subject(s)
Antibodies, Viral/immunology , Complement System Proteins/physiology , Immunoglobulin M/immunology , Viral Envelope Proteins/physiology , Animals , Chlorocebus aethiops , Complement C1q/physiology , Complement C3/physiology , Complement C5/physiology , Complement C6/physiology , Complement Pathway, Alternative , Complement Pathway, Mannose-Binding Lectin , Neutralization Tests , Vero Cells
7.
Arthritis Rheum ; 50(8): 2633-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15334478

ABSTRACT

OBJECTIVE: Monosodium urate monohydrate (MSU) crystals promote gouty inflammation that is critically mediated by neutrophil recruitment and activation. Interleukin-8 (IL-8) and closely related chemokines are major neutrophil chemotaxins in experimental gout. But MSU crystals also activate the classical and alternative pathways of complement, and MSU crystals directly cleave C5 on the crystal surface. Unlike IL-8, the roles in acute gout of individual complement-derived peptides and of the terminal C5b-9 complement components that comprise the membrane attack complex (MAC) are unclear. Hence, we studied rabbits deficient in the MAC component C6 to determine if MAC mediated urate crystal-induced arthritis. METHODS: We injected C6-deficient and C6-sufficient rabbit knee joints with 10 mg of pyrogen-free urate crystals and analyzed IL-8 levels, leukocyte influx, and joint inflammation 24 hours later. RESULTS: There was a significant decrease (>60%) in swelling in MSU crystal-injected knees of C6-deficient animals as compared with C6-sufficient animals (P < 0.05). An attenuated rise in MSU crystal-induced joint effusion levels of IL-8 also was observed, which was concordant with diminished numbers of neutrophils (P < 0.05) but not monocytes in MSU crystal-induced knee synovial fluid from C6-deficient animals. Synovial tissue analysis confirmed mononuclear leukocyte infiltration in response to MSU crystal injection in all animals, but substantial neutrophil infiltration only in C6-sufficient animals. CONCLUSION: MAC activation appears to play a major role in intraarticular IL-8 generation and in neutrophil recruitment in experimental acute gouty arthritis of the rabbit knee. C6 and MAC activation may represent novel therapeutic targets for suppression of neutrophil-mediated joint inflammation in gout.


Subject(s)
Complement Membrane Attack Complex/physiology , Gout/etiology , Synovitis/chemically induced , Uric Acid , Animals , Complement C6/physiology , Crystallization , Interleukin-8/analysis , Knee Joint , Neutrophils/pathology , Rabbits
8.
J Immunol ; 169(8): 4620-7, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12370401

ABSTRACT

The lung is known to be particularly susceptible to complement-mediated injury. Both C5a and the membrane attack complex (MAC), which is formed by the terminal components of complement (C5b-C9), can cause acute pulmonary distress in nontransplanted lungs. We used C6-deficient rats to investigate whether MAC causes injury to lung allografts. PVG.R8 lungs were transplanted orthotopically to MHC class I-incompatible PVG.1U recipients. Allografts from C6-sufficient (C6(+)) donors to C6(+) recipients were rejected with an intense vascular infiltration and diffuse alveolar hemorrhage 7 days after transplantation (n = 5). Ab and complement (C3d) deposition was accompanied by extensive vascular endothelial injury and intravascular release of von Willebrand factor. In contrast, lung allografts from C6-deficient (C6(-)) donors to C6(-) recipients survived 13-17 days (n = 5). In the absence of C6, perivascular mononuclear infiltrates of ED1(+) macrophages and CD8(+) T lymphocytes were present 7 days after transplantation, but vascular endothelial cells were quiescent, with minimal von Willebrand factor release and no evidence of alveolar hemorrhage or edema. Lung allografts were performed from C6(-) donors to C6(+) recipients (n = 5) and from C6(+) donors to C6(-) recipients (n = 5) to separate the effects of systemic and local C6 production. Lungs transplanted from C6(+) donors to C6(-) recipients had increased alveolar macrophages and capillary injury. C6 production by lung allografts was demonstrated at the mRNA and protein levels. These results demonstrate that MAC causes vascular injury in lung allografts and that the location of injury is dependent on the source of C6.


Subject(s)
Complement C6/physiology , Complement Membrane Attack Complex/physiology , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Graft Rejection/immunology , Graft Rejection/pathology , Lung Transplantation/immunology , Lung Transplantation/pathology , Acute Disease , Animals , Animals, Congenic , Cell Movement/genetics , Cell Movement/immunology , Complement C3d/metabolism , Complement C6/biosynthesis , Complement C6/deficiency , Complement C6/genetics , Complement Membrane Attack Complex/deficiency , Complement Membrane Attack Complex/genetics , Female , Graft Rejection/genetics , Graft Survival/genetics , Graft Survival/immunology , Histocompatibility Antigens/genetics , Histocompatibility Antigens Class I/genetics , Immunoglobulin G/metabolism , Immunoglobulin M/metabolism , Isoantibodies/biosynthesis , Male , Postoperative Hemorrhage/genetics , Postoperative Hemorrhage/immunology , Postoperative Hemorrhage/pathology , Postoperative Hemorrhage/prevention & control , Pulmonary Alveoli/immunology , Pulmonary Alveoli/pathology , Rats , Rats, Inbred Strains
9.
J Am Soc Nephrol ; 13(4): 928-936, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11912252

ABSTRACT

Although it was once considered only a marker of glomerular damage, accumulating evidence indicates that proteinuria per se is nephrotoxic and contributes to the progression of renal injury. Several studies have demonstrated that activation of complement in proteinuric urine results in tubular and interstitial damage. It was previously demonstrated that acute complement-mediated interstitial disease is induced by C5b-9. Here the role of C5b-9 in the progression of chronic proteinuric renal disease was investigated in a nonimmunologic remnant kidney model. Five-sixths nephrectomies were performed for normocomplementemic control and C6-deficient PVG rats. Tubulointerstitial injury was assessed by measurement of two independent markers of tubular injury (i.e., vimentin and osteopontin), interstitial accumulation of the extracellular matrix components collagen type I, collagen type IV, and laminin, interstitial macrophage infiltration, and renal function. The two groups developed similar levels of proteinuria and BP. Whereas C3 deposition on the brush border was equivalent for rats in the two groups, C5b-9 deposition was observed only for normocomplementemic rats. At day 35, the degrees of both tubulointerstitial injury and renal failure were the same for the two groups. Tubulointerstitial injury in normocomplementemic rats was still severe at day 70. In contrast, interstitial injury in C6-deficient rats had improved markedly at day 70, with improvements in renal function. In a rat model of chronic progressive renal disease secondary to nephron loss, the initial interstitial changes are complement-independent and largely reversible, whereas progressive interstitial fibrosis is mediated predominantly by C5b-9. Treatment to reduce C5b-9 attack in tubular cells may slow progression and facilitate recovery.


Subject(s)
Complement C6/physiology , Kidney/physiopathology , Proteinuria/physiopathology , Animals , Blood Pressure , Chronic Disease , Complement C3/metabolism , Complement Membrane Attack Complex/metabolism , Disease Progression , Extracellular Matrix/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Kidney/pathology , Kidney Glomerulus/pathology , Macrophages/pathology , Male , Osteopontin , Periodic Acid-Schiff Reaction , Proliferating Cell Nuclear Antigen/metabolism , Proteinuria/pathology , Rats , Rats, Inbred Strains , Sialoglycoproteins/metabolism , Staining and Labeling , Vimentin/metabolism
10.
J Immunol ; 168(1): 458-65, 2002 Jan 01.
Article in English | MEDLINE | ID: mdl-11751993

ABSTRACT

Complement is implicated in pathology in the human demyelinating disease multiple sclerosis and in animal models that mimic the demyelination seen in multiple sclerosis. However, the components of the complement system responsible for demyelination in vivo remain unidentified. In this study, we show that C6-deficient (C6-) PVG/c rats, unable to form the membrane attack complex (MAC), exhibit no demyelination and significantly reduced clinical score in the Ab-mediated experimental autoimmune encephalomyelitis model when compared with matched C6-sufficient (C6+) rats. In C6+ rats, perivenous demyelination appeared, accompanied by abundant mononuclear cell infiltration and axonal injury. Neither demyelination nor axonal damage was seen in C6- rats, whereas levels of mononuclear cell infiltration were equivalent to those seen in C6+ rats. Reconstitution of C6 to C6- rats yielded pathology and clinical disease indistinguishable from that in C6+ rats. We conclude that demyelination and axonal damage occur in the presence of Ab and require activation of the entire complement cascade, including MAC deposition. In the absence of MAC deposition, complement activation leading to opsonization and generation of the anaphylatoxins C5a and C3a is insufficient to initiate demyelination.


Subject(s)
Axons/pathology , Complement Activation , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Animals , Complement C6/genetics , Complement C6/pharmacology , Complement C6/physiology , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Deletion , Macromolecular Substances , Multiple Sclerosis/pathology , Myelin Sheath/pathology , Rats , Spinal Cord/pathology
11.
Xenotransplantation ; 8(4): 303-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11737856

ABSTRACT

The present study aimed to investigate the role of cellular immune response in the absence of membrane attack complex (MAC) formation in the concordant mouse-to-rat heart xenografting. Hearts from BALB/c mice were transplanted into the neck vessels of C6-competent (C6(+)) and C6-deficient (C6(-)) PVG rats. Liposome-encapsulated dichloro-methylene diphosphonate (Lip-Cl2MDP) was administered at a dose of 10 ml/kg 2 days before transplantation and every 5 days thereafter. Cyclosporine (CsA) was administered intramuscularly (i.m.) at a dose of 15 mg/kg per day. The heart xenografts were harvested for immuno-histological analysis at the time of rejection and the functioning grafts were removed at 70 days after transplantation. In untreated C6(+) rats, xeno-grafts survived for 2.3 +/- 0.5 days. Treatment with CsA or Lip-Cl(2)MDP in C6(+) rats did not significantly affect graft survival (2.5 +/- 0.6 and 2.3 +/- 0.4 days, respectively). In untreated C6(-) rats, xenografts survived for 5.0 +/- 0.6 days. However, Lip-Cl(2)MDP in C6(-) rats resulted in a prolongation of graft survival to 11 +/- 2.3 days (P < 0.05 vs. untreated C6(-) rats), while treatment with CsA alone in these rats led to more than 70 days' survival in four out of six grafts (61 +/- 16 days). In untreated C6(+) rats, immunohistology showed a severe myocardial necrosis and thrombosis with a scarce cellular infiltrate in the rejected xenografts. By contrast, in untreated C6(-) rats, xenografts were heavily infiltrated by macrophages and T cells. The number of macrophages, but not T cells, was markedly reduced in Lip-Cl(2)MDP-treated rats. In CsA-treated C6(-) rats, the grafts harvested at 70 days after transplantation had a normal morphology, with a minimal cellular infiltrate. Our data indicate that MAC-mediated injury plays an essential role in concordant xenograft rejection. Once this mechanism has been prevented, suppression of T cells allows for long-term xenograft survival.


Subject(s)
Complement C6/deficiency , Graft Rejection/prevention & control , Graft Survival/immunology , Heart Transplantation/immunology , Immunosuppression Therapy , T-Lymphocyte Subsets/immunology , Transplantation, Heterologous/immunology , Animals , Animals, Genetically Modified , Clodronic Acid/pharmacology , Complement C3/analysis , Complement C6/physiology , Cyclosporine/pharmacology , Graft Rejection/immunology , Graft Rejection/pathology , Immunity, Cellular , Immunoglobulin G/analysis , Immunoglobulin M/analysis , Immunosuppressive Agents/pharmacology , Killer Cells, Natural/immunology , Liposomes , Macrophages/immunology , Mice , Mice, Inbred BALB C , Myocardial Contraction , Myocardium/immunology , Myocardium/pathology , Rats , Transplantation, Heterotopic
14.
Infect Immun ; 68(3): 1626-32, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10678982

ABSTRACT

Several products of the activated complement system are known to modulate endothelial cell function in vitro. It has been shown that the membrane attack complex (MAC) (C5b-C9) can enhance tumor necrosis factor alpha (TNF-alpha)-induced expression of P- and E-selectin and intercellular adhesion molecule type 1 in cell cultures of human umbilical vein endothelial cells. In the present study the potential role of this synegism for lung injury during endotoxin-mediated septic shock in vivo was examined using a model of C6-deficient PVG (C-) (RT1(C)) rats and the congenic PVG (C+) (RT1(C)) strain. Following administration of a high (5 mg/kg) or low (0.5 mg/kg) dose of lipopolysaccharide (LPS) (Escherichia coli O55:B5), we determined the expression of cytokines, chemokines, and adhesion molecules as well as the recruitment of leukocytes in the lung. Challenge with intraperitoneal i.p. injections of LPS resulted in a strong induction of TNF-alpha, interleukin-1alpha/beta, cytokine-induced neutrophil chemoattractant, interferon-inducible protein 10, macrophage inflammatory proteins 1alpha and 2, macrophage chemotactic protein 1, and P-selectin. However, there were no significant differences between PVG (C-) and PVG (C+) rats. Immunoperoxidase staining showed a similar increase of lung infiltration by CD11b/c(+) leukocytes in both rat strains. We therefore conclude that the described synergism between TNF-alpha and the MAC of the complement system on the induction of endothelial adhesion molecules is dispensable for inflammatory processes during endotoxin-mediated septic shock in vivo.


Subject(s)
Complement C6/physiology , Complement Membrane Attack Complex/physiology , Lipopolysaccharides/toxicity , Pneumonia/etiology , Animals , Chemokines/biosynthesis , Complement C6/deficiency , Cytokines/biosynthesis , Humans , Macrophage-1 Antigen/physiology , Male , RNA, Messenger/analysis , Rabbits , Rats , Tumor Necrosis Factor-alpha/physiology
15.
J Am Soc Nephrol ; 10(11): 2323-31, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10541291

ABSTRACT

Accumulating evidence suggests that the generation of complement activation products from filtered complement components in urine with nonselective proteinuria leads to tubulointerstitial disease, resulting in progressive loss of renal function. To elucidate the role of C5b-9 in complement-mediated effects on renal tubular cells exposed to proteinuric urine, equivalent levels of proteinuria were induced (using the aminonucleoside of puromycin) in normocomplementemic and genetically C6-deficient piebald viral glaxo (PVG) rats. Semiquantitative histologic analysis revealed that complement-sufficient animals developed more severe tubulointerstitial disease than did C6-deficient rats. Amelioration of tubulointerstitial damage in C6-deficient animals was confirmed by studies with three independent markers of tubular damage, i.e., vimentin, osteopontin, and proliferating cell nuclear antigen. More tubular epithelial cells expressed osteopontin (an early marker of tubular injury) in normocomplementemic rats, compared with C6-deficient rats, at both days 7 and 12. Staining of vimentin in the tubules, near areas of tubular damage, was increased in normocomplementemic rats at day 12, and more proliferating cell nuclear antigen-positive tubular cells were observed at day 12 in complement-sufficient animals. The tubulointerstitial damage in complement-sufficient rats was also associated with greater accumulation of extracellular matrix (fibronectin) at day 12. These studies document for the first time an important role for C6, and therefore C5b-9, in the pathogenesis of nonimmunologic tubulointerstitial injury induced by proteinuria. These findings suggest that C5b-9 formation resulting from proteinuria contributes to the loss of nephron function by damaging the tubulointerstitium and that prevention of C5b-9 formation in tubules could slow the deterioration of renal function.


Subject(s)
Complement C6/physiology , Complement Membrane Attack Complex/physiology , Kidney Tubules/pathology , Nephrotic Syndrome/pathology , Animals , Complement C6/deficiency , Fibronectins/biosynthesis , Male , Nephrotic Syndrome/metabolism , Proteinuria/metabolism , Proteinuria/pathology , Rats , Vimentin/biosynthesis
16.
J Biol Chem ; 274(45): 31811-8, 1999 Nov 05.
Article in English | MEDLINE | ID: mdl-10542204

ABSTRACT

In order to elucidate the function of complement component C6, truncated C6 molecules were expressed recombinantly. These were either deleted of the factor I modules (FIMs) (C6des-748-913) or both complement control protein (CCP) modules and FIMs (C6des-611-913). C6des-748-913 exhibited approximately 60-70% of the hemolytic activity of full-length C6 when assayed for Alternative Pathway activity, but when measured for the Classical Pathway, C6des-748-914 was only 4-6% as effective as C6. The activity difference between C6 and C6des-748-913 for the two complement pathways can be explained by a greater stability of newly formed metastable C5b* when produced by the Alternative Pathway compared with that made by the Classical Pathway. The half-lives of metastable C5b* and the decay of (125)I-C5b measured from cells used to activate the Alternative Pathway were found to be about 5-12-fold longer than those same parameters derived from cells that had activated the Classical Pathway. (125)I-C5 binds reversibly to C6 in an ionic strength-dependent fashion, but (125)I-C5 binds only weakly to C6des-FIMs and not at all to C6des-CCP/FIMs. Therefore, although the FIMs are not required absolutely for C6 activity, these modules promote interaction of C6 with C5 enabling a more efficient bimolecular coupling ultimately leading to the formation of the C5b-6 complex.


Subject(s)
Complement C6/physiology , Complement Factor I/physiology , Animals , CHO Cells , Complement C6/chemistry , Complement C9/chemistry , Complement C9/physiology , Complement Factor I/chemistry , Complement Pathway, Alternative , Complement Pathway, Classical , Cricetinae , Humans , Rabbits , Sheep , Structure-Activity Relationship
17.
Herz ; 23(3): 163-7, 1998 May.
Article in German | MEDLINE | ID: mdl-9646097

ABSTRACT

Why LDL entrapped in the subendothelium should trigger events leading to chronic inflammation and to arterial wall injury is a major enigma of modern medicine. Oxidation of LDL in vitro renders the molecule potentially atherogenic, and the concept that oxidation is the major single event underlying the transformation of LDL to a proinflammatory molecule dominates the world literature. Here, an alternative hypothesis on the pathogenesis of atherosclerosis will be presented. We have found that non-oxidative, enzymatic modification of LDL with ubiquitous enzymes (protease + cholesterol esterase + neuraminidase) also transforms the molecule to an atherogenic moiety. Enzymatically altered LDL (E-LDL) shares major properties in common with lipoproteins that have been isolated from atherosclerotic lesions. It activates complement via the alternative pathway and is recognized by a scavenger receptor on human macrophages, thus inducing foam cell formation. Uptake of E-LDL is accompanied by potent induction of MCP-1 synthesis and secretion. In contrast, E-LDL does not stimulate IL-1 or TNF-production and is only a weak inducer of IL-6. Monoclonal antibodies were produced that recognize neoepitopes on E-LDL, but that do not react with native or oxidized LDL. With the use of these antibodies, extensive deposition of E-LDL in very early atherosclerotic lesions was demonstrated. Activated complement components colocalized with E-LDL, corroborating the concept that subendothelially deposited LDL is enzymatically transformed to a complement activator at the earliest stages in lesion development. The pathogenetic relevance of unhalted complement activation in atherogenesis was demonstrated with the use of C6-deficient rabbits. It was found that C6-deficiency markedly protected against development of diet-induced atherosclerosis in the experimental animals. In sum, our hypothesis departs from the mainstream of atherosclerosis research and derives from the recognition that extracellular exposition of free cholesterol in LDL-particles by itself confers pro-inflammatory properties onto the lipoprotein molecule. We believe that the degrading enzymes are ubiquitously present in the extracellular matrix, so the only requirement for atherogenesis to occur is the deposition of large amounts of LDL. Oxidative processes or infections probably play only minor roles, and reduction of LDL plasma levels will predictably represent the single most important prophylactic measure against development and progression of atherosclerosis.


Subject(s)
Arteriosclerosis/immunology , Complement Activation/immunology , Coronary Artery Disease/immunology , Lipoproteins, LDL/immunology , Animals , Complement C6/deficiency , Complement C6/physiology , Complement Pathway, Alternative/immunology , Diet, Atherogenic , Endopeptidases/physiology , Endothelium, Vascular/immunology , Humans , Neuraminidase/physiology , Rabbits , Sterol Esterase/physiology
18.
J Heart Lung Transplant ; 17(5): 470-8, 1998 May.
Article in English | MEDLINE | ID: mdl-9628565

ABSTRACT

BACKGROUND: We have demonstrated previously that the terminal complement component C6 contributes to the acute rejection of ACI cardiac allografts by PVG recipients. ACI rats differ from PVG rats at major and minor histocompatibility antigens and ACI cardiac allografts stimulate vigorous alloantibody responses in PVG rats. We have now bred the C6 deficiency onto four PVG congenic rat strains to determine the effects of C6 on cardiac allograft survival across individual donor-recipient major histocompatibility complex (MHC) disparities. METHODS: Hearts from C6-deficient PVG.1A (RT1a) donors were transplanted heterotopically to fully MHC-incompatible C6-sufficient and C6-deficient PVG.1L (RT1(1)) recipients, as well as from C6-deficient PVG.R8 (RT1.AaBu) donors to MHC class I incompatible C6-sufficient and C6-deficient PVG. 1U (RT1.AuBu) recipients. RESULTS: Hearts from PVG.1A (C6-) female donors were rejected acutely (7 to 9 days; n = 5) by fully MHC disparate female PVG.1L (C6+) recipients, but they survived significantly longer in female PVG.1L (C6-) recipients (13 to >50 days; n = 6). Slightly better survival resulted in male PVG.1L (C6-) heart transplant recipients of male PVG.1A (C6-) hearts (19 to >50 days [n = 5] vs 6 to 9 days for C6+ male PVG.1L recipients [n = 10]). The C6 deficiency had an even greater effect in PVG.1U recipients of class I MHC disparate PVG.R8 hearts (>50 day survival in C6- PVG.1U recipients [n = 5] vs 6 to 7 days in C6+ recipients [n = 8]). The cardiac allografts elicited similarly vigorous immunoglobulin M and G alloantibody responses in the C6- and C6+ recipients as measured by flow cytometry. At the time of acute rejection, the hearts in the C6+ recipients demonstrated extensive vascular endothelial destruction. In contrast, rejection of hearts by C6- recipients was characterized by endothelialitis, but there was little destruction of the endothelium and limited proliferation of smooth muscle cells in the intima. CONCLUSIONS: These results demonstrate that the terminal complement component C6 can contribute to the rejection of class I or complete MHC-incompatible hearts in rats that have been characterized as "high" alloantibody responders.


Subject(s)
Complement C6/physiology , Graft Rejection/immunology , Heart Transplantation/immunology , Histocompatibility Antigens Class I/immunology , Animals , Complement C6/deficiency , Female , Isoantibodies/metabolism , Male , Phenotype , Rats , Rats, Inbred Strains , Sex Factors
19.
J Mol Cell Cardiol ; 30(1): 75-85, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9500866

ABSTRACT

Neutrophil accumulation and activation of the complement system with subsequent deposition of the cytolytic membrane attack complex (MAC) have been implicated in the pathogenesis of myocardial ischemia/reperfusion injury. The MAC, when present in high concentrations, promotes target cell lysis. However, relatively little is known about the potential modulatory role of sublytic concentrations of the MAC on nucleated cell function in vivo. In vitro studies demonstrated that the MAC regulates cell function by promoting the expression of pro-inflammatory mediators, including adhesion molecules and pro-inflammatory cytokines. We examined, using C6-deficient and C6-sufficient rabbits, the regulatory role of the MAC in mediating IL-8 expression and subsequent neutrophil recruitment in the setting of myocardial ischemia/reperfusion injury. C6-deficient and C6-sufficient rabbits were subjected to 30 min of regional myocardial ischemia followed by a period of reperfusion. In addition to a significant reduction in myocardial infarct size in C6-deficient animals, analysis of myocardial tissue demonstrated a decrease in neutrophil influx into the infarcted region. The reduction in neutrophil influx correlated with the decreased expression of the neutrophil chemotactic cytokine IL-8, as determined by ELISA and immunohistochemical analysis. The results derived from this study provide evidence that the MAC has an important function in mediating the recruitment of neutrophils to the reperfused myocardium through the local induction of IL-8.


Subject(s)
Complement C6/physiology , Complement Membrane Attack Complex/physiology , Interleukin-8/metabolism , Myocardial Ischemia/immunology , Myocardial Reperfusion Injury/immunology , Animals , Cell Movement , Leukocyte Count , Myocardial Infarction , Myocardial Ischemia/blood , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Neutrophils/cytology , Neutrophils/enzymology , Peroxidase/metabolism , Rabbits
20.
Transplantation ; 63(12): 1705-12, 1997 Jun 27.
Article in English | MEDLINE | ID: mdl-9210492

ABSTRACT

BACKGROUND: Factors that contribute to xenograft (Xg) rejection were investigated in complement C6-deficient (C-) PVG rats. METHODS: First and second hamster hearts were transplanted in C6-deficient and C6-sufficient PVG rats. Xenoantibody (XAb) formation, hemolytic C (CH50) activity and immunohistochemistry were studied. RESULTS: PVG C6-deficient rats rejected Xgs 3 days later than PVG C6-sufficient rats. Surprisingly, C activation participated in the rejection in PVG C- rats, as shown by partially recovered serum CH50 levels and deposition of C factors in the Xgs. As we found that cultured endothelial cells produced C6 in vitro, we hypothesized that Xg endothelial cells corrected the C6 defect in PVG C- rats. This was probably induced by IgM XAbs as: (1) it did not occur in immunosuppressed PVG C- rats in which XAb formation was prevented, and (2) transfer of IgM XAbs to naive, xenotransplanted PVG C- rats accelerated the recovery of CH50 and concomitantly Xg rejection. Thirty days after rejection of a first Xg, when no IgM XAbs or CH50 activity but high levels of IgG XAbs were detected in PVG C- rats, second Xgs underwent a hyperacute rejection. This time, complement was not involved, as no serum CH50 nor C deposition was found in the Xg. Instead, IgG antibody-dependent cellular cytotoxicity was involved as: (1) IgG XAbs were deposited in the Xg and (2) hyperacute rejection was induced in naive PVG C- rats by transfer of IgG XAbs, and (3) this rejection was delayed to 5+/-3 days if the adoptive hosts were first irradiated. CONCLUSIONS: In the face of a defect of host C factors, IgM XAb may induce cells of the Xg to secrete C factors which may correct the C defect of the host. Even if activation of lytic C can be prevented, IgG XAb may still provoke an acute Xg rejection by antibody-dependent cellular cytotoxicity.


Subject(s)
Complement C6/deficiency , Graft Rejection/immunology , Heart Transplantation/immunology , Animals , Cells, Cultured , Complement C6/physiology , Complement Membrane Attack Complex/physiology , Cricetinae , Fluorescent Antibody Technique, Indirect , Graft Survival , Humans , Rats , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...