Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Thromb Haemost ; 108(6): 1141-53, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23014597

ABSTRACT

Coagulation and complement regulators belong to two interactive systems constituting emerging mechanisms of diabetic nephropathy. Thrombomodulin (TM) regulates both coagulation and complement activation, in part through discrete domains. TM's lectin like domain dampens complement activation, while its EGF-like domains independently enhance activation of the anti-coagulant and cytoprotective serine protease protein C (PC). A protective effect of activated PC in diabetic nephropathy is established. We hypothesised that TM controls diabetic nephropathy independent of PC through its lectin-like domain by regulating complement. Diabetic nephropathy was analysed in mice lacking TM's lectin-like domain (TMLeD/LeD) and controls (TMwt/wt). Albuminuria (290 µg/mg vs. 166 µg/mg, p=0.03) and other indices of experimental diabetic nephropathy were aggravated in diabetic TMLeD/LeD mice. Complement deposition (C3 and C5b-9) was markedly increased in glomeruli of diabetic TMLeD/LeD mice. Complement inhibition with enoxaparin ameliorated diabetic nephropathy in TMLeD/LeD mice (e.g. albuminuria 85 µg/mg vs. 290 µg/mg, p<0.001). In vitro TM's lectin-like domain cell-autonomously prevented glucose-induced complement activation on endothelial cells and - notably - on podocytes. Podocyte injury, which was enhanced in diabetic TMLeD/LeD mice, was reduced following complement inhibition with enoxaparin. The current study identifies a novel mechanism regulating complement activation in diabetic nephropathy. TM's lectin-like domain constrains glucose-induced complement activation on endothelial cells and podocytes and ameliorates albuminuria and glomerular damage in mice.


Subject(s)
Diabetic Nephropathies/etiology , Thrombomodulin/chemistry , Thrombomodulin/physiology , Animals , Cell Line , Complement Activation/physiology , Complement Inactivator Proteins/chemistry , Complement Inactivator Proteins/deficiency , Complement Inactivator Proteins/genetics , Complement Inactivator Proteins/physiology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/physiopathology , Diabetic Nephropathies/physiopathology , Diabetic Nephropathies/prevention & control , Endothelial Cells/immunology , Endothelial Cells/pathology , Endothelial Cells/physiology , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Kidney Glomerulus/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/physiology , Podocytes/immunology , Podocytes/pathology , Podocytes/physiology , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thrombomodulin/deficiency , Thrombomodulin/genetics
3.
J Immunol ; 185(5): 3086-94, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20675597

ABSTRACT

Congenital and acquired deficiencies of complement regulatory proteins are associated with pathologic complement activation in several renal diseases. To elucidate the mechanisms by which renal tubular epithelial cells (TECs) control the complement system, we examined the expression of complement regulatory proteins by the cells. We found that Crry is the only membrane-bound complement regulator expressed by murine TECs, and its expression is concentrated on the basolateral surface. Consistent with the polarized localization of Crry, less complement activation was observed when the basolateral surface of TECs was exposed to serum than when the apical surface was exposed. Furthermore, greater complement activation occurred when the basolateral surface of TECs from Crry(-/-)fB(-/-) mice was exposed to normal serum compared with TECs from wild-type mice. Complement activation on the apical and basolateral surfaces was also greater when factor H, an alternative pathway regulatory protein found in serum, was blocked from interacting with the cells. Finally, we injected Crry(-/-)fB(-/-) and Crry(+/+)fB(-/-) mice with purified factor B (an essential protein of the alternative pathway). Spontaneous complement activation was seen on the tubules of Crry(-/-)fB(-/-) mice after injection with factor B, and the mice developed acute tubular injury. These studies indicate that factor H and Crry regulate complement activation on the basolateral surface of TECs and that factor H regulates complement activation on the apical surface. However, congenital deficiency of Crry or reduced expression of the protein on the basolateral surface of injured cells permits spontaneous complement activation and tubular injury.


Subject(s)
Complement Factor H/physiology , Complement Inactivator Proteins/physiology , Epithelial Cells/immunology , Kidney Tubules/immunology , Receptors, Complement/physiology , Animals , Cells, Cultured , Complement Factor H/biosynthesis , Complement Factor H/deficiency , Complement Inactivator Proteins/deficiency , Complement Pathway, Alternative/immunology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Kidney Tubules/cytology , Kidney Tubules/metabolism , Membrane Proteins/biosynthesis , Membrane Proteins/deficiency , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding/immunology , Receptors, Complement/biosynthesis , Receptors, Complement/deficiency , Receptors, Complement 3b
4.
J Immunol ; 179(6): 4187-92, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17785858

ABSTRACT

Complement activation is a central component of inflammation and sepsis and can lead to significant tissue injury. Complement factors are serum proteins that work through a cascade of proteolytic reactions to amplify proinflammatory signals. Inter-alpha-trypsin inhibitor (IaI) is an abundant serum protease inhibitor that contains potential complement-binding domains, and has been shown to improve survival in animal sepsis models. We hypothesized that IaI can bind complement and inhibit complement activation, thus ameliorating complement-dependent inflammation. We evaluated this hypothesis with in vitro complement activation assays and in vivo in a murine model of complement-dependent lung injury. We found that IaI inhibited complement activation through the classical and alternative pathways, inhibited complement-dependent phagocytosis in vitro, and reduced complement-dependent lung injury in vivo. This novel function of IaI provides a mechanistic explanation for its observed salutary effects in sepsis and opens new possibilities for its use as a treatment agent in inflammatory diseases.


Subject(s)
Alpha-Globulins/physiology , Complement Activation/immunology , Complement Inactivator Proteins/physiology , Complement System Proteins/toxicity , Lung/immunology , Lung/pathology , Alpha-Globulins/deficiency , Alpha-Globulins/genetics , Alpha-Globulins/metabolism , Animals , Complement Activation/genetics , Complement Inactivator Proteins/deficiency , Complement Inactivator Proteins/genetics , Complement Inactivator Proteins/metabolism , Complement System Proteins/metabolism , Female , Immune Complex Diseases/immunology , Immune Complex Diseases/metabolism , Immune Complex Diseases/pathology , Immune Complex Diseases/prevention & control , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis/immunology , Protein Binding/immunology , Protein Subunits/deficiency , Protein Subunits/genetics , Protein Subunits/metabolism , Protein Subunits/physiology
5.
J Immunol ; 175(3): 1813-9, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16034123

ABSTRACT

Streptococcus pneumoniae is a common cause of septicemia in the immunocompetent host. To establish infection, S. pneumoniae has to overcome host innate immune responses, one component of which is the complement system. Using isogenic bacterial mutant strains and complement-deficient immune naive mice, we show that the S. pneumoniae virulence factor pneumolysin prevents complement deposition on S. pneumoniae, mainly through effects on the classical pathway. In addition, using a double pspA-/ply- mutant strain we demonstrate that pneumolysin and the S. pneumoniae surface protein PspA act in concert to affect both classical and alternative complement pathway activity. As a result, the virulence of the pspA-/ply- strain in models of both systemic and pulmonary infection is greatly attenuated in wild-type mice but not complement deficient mice. The sensitivity of the pspA-/ply- strain to complement was exploited to demonstrate that although early innate immunity to S. pneumoniae during pulmonary infection is partially complement-dependent, the main effect of complement is to prevent spread of S. pneumoniae from the lungs to the blood. These data suggest that inhibition of complement deposition on S. pneumoniae by pneumolysin and PspA is essential for S. pneumoniae to successfully cause septicemia. Targeting mechanisms of complement inhibition could be an effective therapeutic strategy for patients with septicemia due to S. pneumoniae or other bacterial pathogens.


Subject(s)
Bacterial Proteins/physiology , Complement Inactivator Proteins/physiology , Complement System Proteins/metabolism , Heat-Shock Proteins/physiology , Pneumococcal Infections/immunology , Sepsis/immunology , Streptococcus pneumoniae/immunology , Streptolysins/physiology , Animals , Bacterial Proteins/pharmacology , Complement C1q/deficiency , Complement C1q/genetics , Complement C1q/physiology , Complement C3/deficiency , Complement C3/metabolism , Complement C3/physiology , Complement Factor B/deficiency , Complement Factor B/genetics , Complement Inactivator Proteins/deficiency , Complement Inactivator Proteins/pharmacology , Complement Pathway, Classical/genetics , Complement Pathway, Classical/immunology , Complement System Proteins/deficiency , Complement System Proteins/physiology , Drug Synergism , Heat-Shock Proteins/deficiency , Heat-Shock Proteins/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumococcal Infections/genetics , Pneumococcal Infections/microbiology , Sepsis/genetics , Sepsis/microbiology , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism , Streptolysins/deficiency , Streptolysins/pharmacology , Virulence Factors/deficiency , Virulence Factors/pharmacology , Virulence Factors/physiology
7.
Am J Trop Med Hyg ; 69(4): 360-5, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14640493

ABSTRACT

Red blood cells (RBCs) infected with Plasmodium falciparum are protected from complement-mediated lysis by surface membrane glycosyl-phosphatidylinositol (GPI)-anchored proteins, which include decay accelerating factor (DAF or CD55) and CD59. To determine if P. falciparum avoids or replicates less efficiently in GPI protein-deficient cells at a higher risk for complement-mediated lysis, we compared P. falciparum infectivity among control RBCs with those from subjects with paroxysmal nocturnal hemoglobinuria (PNH), a condition in which RBCs express variable levels of DAF (negative and positive) and CD59 (negative [-], intermediate [I], and high [H]). Co-cultures of 19 matched samples of control and PNH RBCs were infected with P. falciparum to directly compare parasitic invasion. Each PNH RBC sample was then assessed for P. falciparum infectivity across the spectrum of GPI protein deficiency. Identification methods included biotin-streptavidin for RBC populations, fluorescein isothiocyanate-labeled antibodies to DAF and CD59, hydroethidine for parasite DNA, and flow cytometry. The mean +/- SD parasitemias in co-cultured PNH and control RBCs were 24.7 +/- 6.9% versus 21.0 +/- 5.9% (P = 0.12). For individual PNH samples, parasitemias were significantly higher in DAF (-) cells versus DAF (+) cells (25.0 +/- 8.9% versus 19.1 +/- 8.7%; P < 0.001) and in CD59 (-) cells versus I/H cells (22.5 +/- 6.4% versus 17.6 +/- 4.2%; P < 0.0003). Across the CD59 spectrum, mean parasitemias were highest in CD59 (-) cells (24.5 +/- 6.4%), followed by CD59-H cells (19.5 +/- 5.4%), and CD59-I cells (16.4 +/- 4.8%). Expression of DAF in 12 (63%) of 19 infected PNH samples was reduced. Thus, P. falciparum does not selectively avoid RBCs with fewer GPI proteins and parasite replication in PNH cells is at least as robust as in normal RBCs.


Subject(s)
Complement Inactivator Proteins/deficiency , Erythrocyte Membrane/metabolism , Hemoglobinuria, Paroxysmal/blood , Membrane Glycoproteins/deficiency , Membrane Proteins/deficiency , Plasmodium falciparum/physiology , Animals , Cell Division/physiology , DNA, Protozoan/analysis , Flow Cytometry , Glycosylphosphatidylinositols/metabolism , Humans
8.
Am J Pathol ; 157(3): 905-18, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10980130

ABSTRACT

This study investigated the capacity of neurons and astrocytes to spontaneously activate the complement system and control activation by expressing complement regulators. Human fetal neurons spontaneously activated complement through the classical pathway in normal and immunoglobulin-deficient serum and C1q binding was noted on neurons but not on astrocytes. A strong staining for C4, C3b, iC3b neoepitope and C9 neoepitope was also found on neurons. More than 40% of human fetal neurons were lysed when exposed to normal human serum in the presence of a CD59-blocking antibody, whereas astrocytes were unaffected. Significant reduction in neuronal cell lysis was observed after the addition of soluble complement receptor 1 at 10 microg/ml. Fetal neurons were stained for CD59 and CD46 and were negative for CD55 and CD35. In contrast, fetal astrocytes were strongly stained for CD59, CD46, CD55, and were negative for CD35. This study demonstrates that human fetal neurons activate spontaneously the classical pathway of complement in an antibody-independent manner to assemble the cytolytic membrane attack complex on their membranes, whereas astrocytes are unaffected. One reason for the susceptibility of neurons to complement-mediated damage in vivo may reside in their poor capacity to control complement activation.


Subject(s)
Complement Activation/physiology , Complement Inactivator Proteins/deficiency , Complement Pathway, Classical/physiology , Membrane Glycoproteins/deficiency , Neurons/physiology , Antibodies, Blocking/pharmacology , Antigens, CD/genetics , Antigens, CD/metabolism , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/physiology , Brain/cytology , Brain/embryology , Cells, Cultured , Complement C3-C5 Convertases/physiology , Complement Inactivator Proteins/genetics , Complement Inactivator Proteins/metabolism , Fetus , Fluorescent Antibody Technique, Indirect , Humans , Image Processing, Computer-Assisted , In Situ Hybridization , Membrane Cofactor Protein , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Neurons/cytology , Neurons/drug effects , RNA/analysis , RNA, Messenger/metabolism , Receptors, Complement/deficiency , Receptors, Complement/genetics , Receptors, Complement/metabolism , Reverse Transcriptase Polymerase Chain Reaction
9.
Clin Immunol Immunopathol ; 84(1): 95-7, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9191888

ABSTRACT

A case of hereditary angioneurotic edema (HANE) associated with Sjögren's syndrome is presented. One of the members of a pedigree of HANE due to deficiency of C1 inhibitor (C1INH) had a positive titer for anti-SS-A and anti-SS-B antibodies in the serum, complaining of symptom of dry eyes and dry mouth. A lip biopsy revealed lymphocytic infiltration of minor salivary glands. The patient had renal tubular acidosis (RTA). Thus the patient was diagnosed as suffering from Sjögren's syndrome with RTA.


Subject(s)
Angioedema/genetics , Angioedema/immunology , Sjogren's Syndrome/immunology , Complement Inactivator Proteins/deficiency , Complement Inactivator Proteins/genetics , Female , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Middle Aged , Pedigree , Sjogren's Syndrome/genetics
10.
Chin Med J (Engl) ; 106(5): 323-30, 1993 May.
Article in English | MEDLINE | ID: mdl-7691484

ABSTRACT

The surface phosphatidylinositol (PI)-linked proteins on membrane of paroxysmal nocturnal hemoglobinuria (PNH) erythrocytes (PNHE) was analysed by a flowcytometer (FACS 420). It was found that the loss of acetylcholinesterase (AchE) and decay accelerating factor (DAF), two PI-linked proteins, from cell membrane of PNHE was not synchronous. The hemolysis rates of DAF (-) and AchE (-) PNHE were much higher than that of mixed population in cobra-venom factor (CoF) lysis test. Intact PNHE remaining after CoF lysis had relatively lower immunofluorescent labeling rate of AchE on membrane in comparison with normal erythrocytes. It implied that this subpopulation, in spite of being insensitive to complement lysis, was still abnormal in terms of the amount of PI-linked protein on cell membrane. When these intact PNHE remaining after CoF lysis were incubated with activated polymorphonuclear leukocytes (PMN) for three hours, immunofluorescent labeling of AchE on PNHE was prominently decreased. This indicated that reactive oxidants released from activated PMN might injure PI-linked proteins.


Subject(s)
Acetylcholinesterase/metabolism , Antigens, CD/metabolism , Complement Inactivator Proteins/deficiency , Hemoglobinuria, Paroxysmal/blood , Membrane Glycoproteins/metabolism , Acetylcholinesterase/deficiency , Blood Proteins/deficiency , Blood Proteins/metabolism , CD55 Antigens , Complement Inactivator Proteins/metabolism , Erythrocyte Membrane/metabolism , Flow Cytometry , Hemoglobinuria, Paroxysmal/classification , Humans , Neutrophils/physiology
12.
Mem Inst Oswaldo Cruz ; 87 Suppl 4: 111-6, 1992.
Article in English | MEDLINE | ID: mdl-1285336

ABSTRACT

Schistosomula of Schistosoma mansoni became resistant to antibody-dependent complement damage in vitro after pre-incubation with normal human erythrocytes (NHuE) whatever the ABO or Rh blood group. Resistant parasites were shown to acquire host decay accelerating factor (DAF), a 70 kDa glycoprotein attached to the membrane of NHuE by a GPI anchor. IgG2a mAb anti-human DAF (IA10) immunoprecipitated a 70 kDa molecule from 125I-labeled schistosomula pre-incubated with NHuE and inhibited their resistance to complement-dependent killing in vitro. Incubation of schistosomula with erythrocytes from patients with paroxysmal nocturnal hemoglobinuria (PNHE) or SRBC, which are DAF-deficient, did not protect the parasites from complement lesion. Supernatant of 100,000 x g collected from NHuE incubated for 24 h in defined medium was shown to contain a soluble form of DAF and to protect schistosomula from complement killing. Schistosomula treated with trypsin before incubation with NHuE ghosts did not become resistant to complement damage. On the other hand, pre-treatment with chymotrypsin did not interfere with the acquisition of resistance by the schistosomula. These results indicate that, in vitro, NHuE DAF can be transferred to schistosomula in a soluble form and that the binding of this molecule to the parasite surface is dependent upon trypsin-sensitive chymotrypsin-insensitive polypeptide(s) present on the surface of the worm.


Subject(s)
Complement System Proteins/immunology , Schistosoma mansoni/physiology , Animals , Antigens, CD/metabolism , CD55 Antigens , Chymotrypsin/pharmacology , Complement Inactivator Proteins/deficiency , Complement Inactivator Proteins/metabolism , Culture Media/pharmacology , Erythrocytes/parasitology , Guinea Pigs , Helminth Proteins/metabolism , Hemoglobinuria, Paroxysmal/immunology , Hemoglobinuria, Paroxysmal/parasitology , Larva , Membrane Glycoproteins/metabolism , Models, Biological , Peptides/metabolism , Protein Binding , Schistosoma mansoni/drug effects , Schistosoma mansoni/growth & development , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/parasitology , Sheep , Trypsin/pharmacology
13.
Blood ; 78(12): 3291-7, 1991 Dec 15.
Article in English | MEDLINE | ID: mdl-1720702

ABSTRACT

A 38-year-old Russian woman (KZ) has been identified as the fourth proposita with the Inab blood group phenotype. Like the first two propositi, she has a chronic intestinal disorder and, as shown for the third proposita, her Inab phenotype is demonstrably inherited. KZ's serum contained anti-IFC, which reacted with a red blood cell (RBC) membrane component with an Mr of 70,000, which is decay accelerating factor (DAF). Her RBCs lacked all Cromer-related blood group antigens and DAF. Her RBCs were no more susceptible than normal control RBCs to lysis in acid lysis or in rabbit or human antibody-initiated complement lysis tests. Northern blots of total RNA isolated from KZ's Epstein-Barr virus-transformed lymphoblasts showed a marked reduction of DAF mRNA when compared with normal. Polymerase chain reaction (PCR) amplification of cDNA confirmed this reduced level of DAF mRNA. Sequencing of the PCR product showed a 44-nucleotide deletion in the mRNA close to the short consensus repeats IIIa/IIIb intron/exon boundary. This deletion results in a change in the reading frame that places a termination codon six amino acids after the deletion. The putative translation product would lack a glycosyl phosphatidyl-inositol linkage site and, therefore, would not be membrane-bound in the RBC.


Subject(s)
Blood Group Antigens/genetics , Complement Inactivator Proteins/deficiency , Erythrocytes/immunology , Membrane Proteins/deficiency , Phenotype , Adult , Amino Acid Sequence , Antibodies/metabolism , Base Sequence , Blood Group Antigens/immunology , Blotting, Northern , CD55 Antigens , Complement C3/metabolism , Complement Inactivator Proteins/genetics , DNA/chemistry , Erythrocytes/chemistry , Erythrocytes/physiology , Female , Hemolysis , Humans , Immunoblotting , Membrane Proteins/genetics , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/analysis , RNA, Messenger/chemistry
15.
Immunol Lett ; 29(3): 205-9, 1991 Aug.
Article in English | MEDLINE | ID: mdl-1722775

ABSTRACT

We have assessed levels of surface-expressed complement regulatory proteins, decay-accelerating factor (DAF) and membrane cofactor protein (MCP) on cells from patients with hematological malignancies. Neither malignant cells nor unaffected nucleated blood cells from the patients lacked MCP. On the other hand, complete deficiency of DAF was found in 2/10 of non-Hodgkin's lymphoma (NHL), while none of the 38 patients with acute nonlymphocytic leukemia (ANLL) (14 cases), chronic myelogenous leukemia (CML) (6 cases), acute lymphocytic leukemia (ALL) (12 cases) and chronic lymphocytic leukemia (CLL) (6 cases) lacked DAF. The two patients with DAF-negative NHL had no history of paroxysmal nocturnal hemoglobinuria (PNH), and their peripheral blood cells were DAF-positive. One DAF-negative NHL exhibited T cell markers and the other those of B cell. In both cases, treatment of the DAF-negative lymphoma cells with antibody against MCP (M177) followed by Mg(2+)-EGTA-serum resulted in efficient deposition of homologous C3. These results infer that some NHL specifically lack DAF and, through treatment with M177, are targeted by homologous C3.


Subject(s)
Antigens, CD , Complement Inactivator Proteins/deficiency , Lymphoma, Non-Hodgkin/immunology , Membrane Proteins/deficiency , Antibodies, Monoclonal/immunology , Antigens, Surface/immunology , Blood Proteins/deficiency , CD55 Antigens , Complement C3/immunology , Flow Cytometry , Humans , Immunophenotyping , Leukemia, Myeloid/immunology , Membrane Cofactor Protein , Membrane Glycoproteins/analysis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology
16.
Rinsho Ketsueki ; 32(6): 612-7, 1991 Jun.
Article in Japanese | MEDLINE | ID: mdl-1716325

ABSTRACT

The significance of the deficiency of the major complement-regulatory membrane proteins, decay-accelerating factor (DAF) and CD59, to the lysis of paroxysmal nocturnal hemoglobinuria (PNH) red blood cells was investigated. DAF and CD59 were demonstrated to be deficient simultaneously on affected PNH red blood cells (PNH-III) by two-color FACS analysis. At least in some patients with PNH, PNH-I was also revealed to be deficient partially in DAF. Purified DAF and CD59 ameliorated the complement sensitivity of PNH red blood cells, partially and completely, respectively. Functional blocking of these molecules on nomrla human red cells by monoclonal antibodies to DAF and CD59 rendered A or AB type blood cells complement-sensitive but not O or B blood type blood cells. The differences of complement-sensitivity among blood types were revealed to reside on the step of binding of C9 to C5b-8, i. e. C9 can bind to C5b-8 more on A type blood cells than on O type blood cells. We conclude that the deficiency of DAF and CD59 play a major role for the complement sensitivity of PNH red blood cells and that other factors reported to be deficient in PNH do less than these two proteins.


Subject(s)
Complement Inactivator Proteins/deficiency , Erythrocyte Membrane/metabolism , Hemoglobinuria, Paroxysmal/metabolism , Membrane Proteins/deficiency , Antigens, CD/deficiency , CD55 Antigens , CD59 Antigens , Erythrocyte Membrane/immunology , Hemoglobinuria, Paroxysmal/immunology , Humans , Membrane Glycoproteins/deficiency
18.
Br J Haematol ; 74(4): 508-13, 1990 Apr.
Article in English | MEDLINE | ID: mdl-1693286

ABSTRACT

Glycosyl-phosphatidylinositol (GPI) anchored membrane proteins have been reported to be deficient on affected paroxysmal nocturnal haemoglobinuria (PNH) blood cells. In the present study we investigated the deficiency of several GPI anchored membrane proteins on PNH neutrophils (PMN) and monocytes from 10 patients with PNH. Decay-accelerating factor (DAF) and Fc gamma R-III (CD16) on PMN, DAF and CD14 on monocytes, were investigated by two-colour immunofluorocytometry. Neutrophil alkaline phosphatase activity was also assayed on PNH neutrophils. Normal human PMN were always shown phenotypically to be DAF+/CD16+. A DAF-/CD16- subpopulation of PMN was demonstrated in all the patients studied. In six out of the 10 patients, deficiencies of DAF and CD16 were found simultaneously on affected PNH PMN. The percentage of DAF- PMN showed a positive correlation with the neutrophil alkaline phosphatase (NAP) score. However, it should be noted that, in four out of the 10 patients with PNH, a DAF+/CD16- subpopulation of PMN was also clearly found. This may indicate that the deficiencies of DAF and CD16 on PNH PMN are heterogeneous. Normal human monocytes were demonstrated to be DAF+/CD14+, whereas PNH monocytes consisted of subpopulations of DAF+/CD14+ and DAF-/CD14-. In the same patients with PNH, the deficiencies of DAF on PMN and monocytes correlated well with each other. These results suggest that, at least in some patients with PNH, the mechanisms which induce the membrane defects of PNH blood cells are heterogeneous.


Subject(s)
Antigens, Differentiation/analysis , Complement Inactivator Proteins/deficiency , Glycolipids/blood , Hemoglobinuria, Paroxysmal/immunology , Membrane Proteins/deficiency , Neutrophils/immunology , Phosphatidylinositols/blood , Receptors, Fc/analysis , Adult , Antigens, Differentiation, Myelomonocytic/analysis , CD55 Antigens , Child , Erythrocytes/immunology , Female , Glycosylphosphatidylinositols , Humans , Lipopolysaccharide Receptors , Male , Middle Aged , Monocytes/immunology , Receptors, IgG
20.
Br J Haematol ; 73(2): 248-53, 1989 Oct.
Article in English | MEDLINE | ID: mdl-2479410

ABSTRACT

No episodes of clinically significant in vivo haemolysis have been reported in individuals with a novel form of decay accelerating factor (DAF) deficiency (Inab phenotype), nor do functional in vitro assays for complement-mediated haemolysis show the extreme sensitivity to lysis characteristic of paroxysmal nocturnal haemoglobinuria (PNH) erythrocytes. DAF appears to be totally deficient in the Inab erythrocytes as judged by immunochemical and functional assays. Unlike PNH, the only other described DAF deficiency (where several other phosphatidylinositol (PI)-linked membrane proteins are also absent), the only protein lacking from Inab erythrocytes appears to be DAF. The Inab phenotype seems to be an inherited specific defect in DAF whereas PNH is an acquired defect in the mechanism of insertion of PI-linked proteins into cell membranes. These findings support the view that susceptibility of PNH erythrocytes to in vivo and in vitro complement-mediated haemolysis is not due simply to DAF deficiency but to either the combined lack of several membrane proteins or to deficiency of other regulatory proteins such as the membrane attack complex inhibitor/homologous restriction factor (MIP/HRF). The findings also raise questions as to the role of erythrocyte DAF.


Subject(s)
Complement Inactivator Proteins/deficiency , Complement System Proteins/immunology , Hemolysis , CD55 Antigens , Complement C3/metabolism , Erythrocyte Membrane/analysis , Hemagglutination , Humans , Immunoblotting , Membrane Proteins , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...