Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Publication year range
1.
Gastroenterology ; 141(3): 918-28, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21699774

ABSTRACT

BACKGROUND & AIMS: CD46 is a C3b/C4b binding complement regulator and a receptor for several human pathogens. We examined the interaction between CD46 and Helicobacter pylori (a bacterium that colonizes the human gastric mucosa and causes gastritis), peptic ulcers, and cancer. METHODS: Using gastric epithelial cells, we analyzed a set of H pylori strains and mutants for their ability to interact with CD46 and/or influence CD46 expression. Bacterial interaction with full-length CD46 and small CD46 peptides was evaluated by flow cytometry, fluorescence microscopy, enzyme-linked immunosorbent assay, and bacterial survival analyses. RESULTS: H pylori infection caused shedding of CD46 into the extracellular environment. A soluble form of CD46 bound to H pylori and inhibited growth, in a dose- and time-dependent manner, by interacting with urease and alkyl hydroperoxide reductase, which are essential bacterial pathogenicity-associated factors. Binding of CD46 or CD46-derived synthetic peptides blocked the urease activity and ability of bacteria to survive in acidic environments. Oral administration of one CD46 peptide eradicated H pylori from infected mice. CONCLUSIONS: CD46 is an antimicrobial agent that can eradicate H pylori. CD46 peptides might be developed to treat H pylori infection.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gastric Mucosa/metabolism , Helicobacter pylori/drug effects , Membrane Cofactor Protein/pharmacology , Urease/drug effects , Urease/metabolism , Animals , Anti-Bacterial Agents/therapeutic use , Cell Line , Disease Models, Animal , Dose-Response Relationship, Drug , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Gastric Mucosa/cytology , Gastric Mucosa/microbiology , Helicobacter Infections/drug therapy , Helicobacter Infections/metabolism , Helicobacter pylori/growth & development , Helicobacter pylori/metabolism , Humans , Membrane Cofactor Protein/metabolism , Membrane Cofactor Protein/therapeutic use , Mice , Mice, Mutant Strains , Peroxiredoxins/drug effects , Peroxiredoxins/metabolism , Time Factors , Treatment Outcome
2.
Gene Ther ; 18(6): 613-21, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21307887

ABSTRACT

Activation of the alternative pathway of the complement system has been implicated in the pathogenesis of age-related macular degeneration. Membrane attack complex (MAC) has been identified mainly on the Bruch's membrane and drusen underlying the retinal pigment epithelium (RPE). Membrane cofactor protein (CD46) preferentially regulates the alternative pathway of complement. The aim of this study was to evaluate the potential of increasing CD46 expression on RPE cells using an adenovirus as a gene therapy approach to reduce alternative pathway-mediated damage to RPE cells. We generated a recombinant adenovirus vector expressing human CD46 (hCD46) and delivered the vector to murine hepatocytes and RPE cells in vitro. After incubation in human serum in conditions in which the classical pathway of complement was blocked, we measured alternative pathway-mediated damage of these cells by quantifying lysis and MAC formation. Adenovirus expressing hCD46 was delivered to the subretinal space of adult mice, and 1 week later, ocular flat mounts were challenged with human serum and the levels of complement-mediated damage was quantified. Adenovirus-mediated delivery of hCD46 localizes to the basal and lateral surfaces of RPE cells where it offers protection from alternative pathway-mediated damage, but not classical, allowing the classical pathway to function unhindered.


Subject(s)
Complement Pathway, Alternative/immunology , Genetic Therapy , Macular Degeneration/genetics , Macular Degeneration/immunology , Membrane Cofactor Protein/genetics , Retinal Pigment Epithelium/metabolism , Adenoviridae/genetics , Adult , Animals , Cell Line , Complement Membrane Attack Complex/metabolism , Complement Pathway, Alternative/genetics , Genetic Vectors , Hepatocytes/metabolism , Humans , Membrane Cofactor Protein/pharmacology , Mice , Retina/embryology , Retina/metabolism , Retinal Pigment Epithelium/immunology
3.
J Asthma ; 47(4): 367-73, 2010 May.
Article in English | MEDLINE | ID: mdl-20528588

ABSTRACT

BACKGROUND: The suppressive cytokine interleukin-10 (IL-10) plays a central role in disease control and clinical therapies of asthma. CD46 was recently identified as costimulatory molecule in inducing IL-10-producing regulatory T cells type 1 (Tr1) from CD4(+) T cells. Alterations in CD46 costimulation pathway were shown to be associated with multiple sclerosis and hemodialysis. In this study, the authors investigated alterations in CD46 costimulatory pathway in asthma. METHODS: CD4(+)CD25(+) regulatory T cells (Tregs) and CD4(+)CD25(-) T cells were isolated from peripheral blood mononuclear cells (PBMCs) of asthma patients (n = 13) and healthy subjects (n = 17). Both subsets of CD4(+) T cells were cultured alone or cocultured at a ratio of 1:10 under stimulation with CD3/CD28 or CD3/CD46, and the production of IL-10 in the supernatants was assessed by enzyme-linked immunosorbent assay (ELISA), the proliferation rates of the cells were determined with thymidine incorporation assay. RESULTS: Levels of IL-10 in the supernatants were higher in undivided CD4(+) T cells and 1:10 cocultured CD4(+)CD25(+) Tregs/CD4(+)CD25(-) T cells than in CD4(+)CD25(+) Tregs or CD4(+)CD25(-) T cells alone, either under CD46 or under CD28 costimulation, both in healthy controls (n = 9) and in asthma patients (n = 7). Under anti-CD3/CD46 stimulation, IL-10 production in undivided CD4(+) T cells and cocultured T cells from asthma patients was lower than that in healthy controls. When treated with glucocorticoids, IL-10 production in undivided CD4(+) T cells and 1:10 cocultured CD4(+) T cells was not different between asthma patients (n = 6) and healthy controls (n = 8). The proliferation rates and the surface expression of CD46 were not different in T cells from both groups. CONCLUSIONS: This study identified a new functional defect of CD4(+)CD25(+) Tregs in inducing IL-10 production from CD4(+)CD25(-) T cells under CD46 costimulation in asthma patients, which may be involved in the pathogenesis of allergic asthma.


Subject(s)
Asthma/immunology , CD4-Positive T-Lymphocytes/metabolism , Interleukin-10/biosynthesis , Interleukin-2 Receptor alpha Subunit/metabolism , Adult , Asthma/metabolism , CD28 Antigens/metabolism , Cell Proliferation , Cells, Cultured , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Female , Glucocorticoids/pharmacology , Humans , Male , Membrane Cofactor Protein/metabolism , Membrane Cofactor Protein/pharmacology , T-Lymphocytes, Regulatory/metabolism
4.
Sheng Wu Gong Cheng Xue Bao ; 24(2): 220-5, 2008 Feb.
Article in Chinese | MEDLINE | ID: mdl-18464604

ABSTRACT

Recombinant expression vector pcDNA3-DAFMCP-DP containing human membrane complement regulatory proteins (hCRPs) decay accelerating factor (DAF) and membrane cofactor protein (MCP) cDNA was constructed by using two independent promoters. After transfected into NIH3T3 cells by calcium phosphate-DNA precipitate method, NIH3T3 pcDNA3-DAFMCP-DP transfectants were obtained by G418 selection. Extraneous genes integration was identified by PCR. The co-expression of human DAF and MCP at both mRNA and protein levels was confirmed by using RT-PCR and Western blot analysis. Human DAF and MCP cDNA were integrated into NIH3T3 pcDNA3-DAFMCP-DP genomic DNA after continuous 30 times passages, indicating that NIH3T3 pcDNA3-DAFMCP-DP were stable cell lines. Human C-mediated cytolysis assays showed that NIH3T3 cells transfected stably with pcDNA3-DAF, pcDNA3-MCP, and pcDNA3-DAFMCP-DP were protected from C-mediated damage and co-expressed human DAF and MCP provided more excellent protection against C-mediated attack, which was compared with either DAF or MCP alone. These results suggest that the dicistronic vector could improve the efficiency of multi-gene delivery and benefit the synergic effect of human membrane complement regulatory proteins DAF and MCP.


Subject(s)
CD55 Antigens/biosynthesis , Graft Rejection/prevention & control , Membrane Cofactor Protein/biosynthesis , 3T3 Cells , Animals , CD55 Antigens/genetics , CD55 Antigens/pharmacology , DNA, Complementary/genetics , Drug Synergism , Humans , Membrane Cofactor Protein/genetics , Membrane Cofactor Protein/pharmacology , Mice , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Transfection
5.
J Reprod Immunol ; 73(2): 135-147, 2007 Apr.
Article in English | MEDLINE | ID: mdl-16950517

ABSTRACT

CD46, a membrane complement regulator, has been implicated as pathogen receptor, T cell activator and contributor to spermatozoa-egg interactions. In man, a role in the fertilization process was suggested by its localization on the acrosome. In rodents, CD46 is expressed only on the spermatozoal acrosome, suggesting an essential role at this site. This restricted expression led us to ask whether immunization with CD46 would generate anti-CD46 antibody responses that might target spermatozoa and influence fertility. We immunized male and female rats with rat CD46. Strong immune responses were generated in all rats and immune sera stained CD46 in testis extracts and in situ in testis and sperm. Incubation of spermatozoa with immune sera caused deposition of immunoglobulin and C3b in an acrosome pattern and reduced motility. We mated immune male rats with naïve females and female immune rats with naïve males. The incidence of pregnancy and number of fetuses were not different in matings involving immune male or female rats compared to controls. Testis sections from immune rats revealed no immunoglobulin deposition on CD46-positive sperm precursors, suggesting that acrosomal CD46 was inaccessible in this location. A minority of spermatozoa harvested from epididymis of immune rats had immunoglobulin and C3b bound to the acrosome, suggesting that anti-CD46, present in genital tract fluids, bound after acrosome reaction. These data demonstrate that the restricted expression of CD46 allows strong anti-CD46 responses in rats that target spermatozoa in vitro and in vivo. The anti-CD46 response did not influence fertility, perhaps reflecting the considerable redundancy for fertilization in rodents.


Subject(s)
Acrosome/immunology , Autoantibodies/immunology , Membrane Cofactor Protein/pharmacology , Sperm Motility/immunology , Sperm-Ovum Interactions/immunology , Animals , Complement C3b/immunology , Female , Immunization , In Vitro Techniques , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Male , Membrane Cofactor Protein/adverse effects , Membrane Cofactor Protein/immunology , Pregnancy , Rats , Rats, Wistar , Sperm Motility/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology
6.
Acta Pharmacol Sin ; 26(8): 987-91, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16038633

ABSTRACT

AIM: To evaluate the protective role of membrane cofactor protein (MCP, CD46) on complement-dependent injury. METHODS: MCP was separated by ion exchange chromatography on a DEAE sephadex A-50 column from pig erythrocyte ghosts. Its protective effect was tested in models such as cobra venom factor (CVF)-induced platelet metamorphosis and aggregation, human serum-induced injury in isolated working guinea pig heart and reverse passive Arthus reaction. RESULTS: MCP inhibited CVF-induced platelet metamorphosis with an IC50 of 56.7 mg/L+/-2.6 mg/L, and prevented injury induced by activated complement in isolated working guinea pig hearts. In the rat model of reverse Arthus reaction, MCP relieved the skin lesions induced by immune complexes. CONCLUSION: MCP has a protective effect against complement-dependent injury.


Subject(s)
Blood Platelets/drug effects , Complement System Proteins/physiology , Membrane Cofactor Protein/pharmacology , Platelet Aggregation/drug effects , Animals , Arthus Reaction/immunology , Arthus Reaction/prevention & control , Blood Platelets/physiology , Complement System Proteins/pharmacology , Dose-Response Relationship, Drug , Elapid Venoms/pharmacology , Erythrocytes/chemistry , Female , Guinea Pigs , Heart/drug effects , Heart/physiology , Humans , In Vitro Techniques , Membrane Cofactor Protein/isolation & purification , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/immunology , Ovalbumin/immunology , Rats , Rats, Sprague-Dawley , Sheep , Skin/drug effects , Skin/immunology , Skin/pathology , Swine , Zymosan/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL