Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
J Reprod Immunol ; 142: 103204, 2020 11.
Article in English | MEDLINE | ID: mdl-33130539

ABSTRACT

Detailed morphological characterization of testicular leukocytes in the adult CX3CR1 gfp/+ transgenic mouse identified two distinct CX3CR1 + mononuclear phagocyte (macrophage and dendritic cell) populations: stellate/dendriform cells opposed to the seminiferous tubules (peritubular), and polygonal cells associated with Leydig cells (interstitial). Using confocal microscopy combined with stereological enumeration of CX3CR1gfp/+ cells established that there were twice as many interstitial cells (68%) as peritubular cells (32%). Flow cytometric analyses of interstitial cells from mechanically-dissociated testes identified multiple mononuclear phagocyte subsets based on surface marker expression (CX3CR1, F4/80, CD11c). These cells comprised 80% of total intratesticular leukocytes, as identified by CD45 expression. The remaining leukocytes were CD3+ (T lymphocytes) and NK1.1+ (natural killer cells). Functional phenotype assessment using CD206 (an anti-inflammatory/M2 marker) and MHC class II (an activation marker) identified a potentially tolerogenic CD206+MHCII+ sub-population (12% of total CD45+ cells). Rare testicular subsets of CX3CR1 +CD11c+F4/80+ (4.3%) mononuclear phagocytes and CD3+NK1.1+ (3.1%) lymphocytes were also identified for the first time. In order to examine the potential for the immunoregulatory cytokine, activin A to modulate testicular immune cell populations, testes from adult mice with reduced activin A (Inhba+/-) or elevated activin A (Inha+/-) were assessed using flow cytometry. Although the proportion of F4/80+CD11b+ leukocytes (macrophages) was not affected, the frequency of CD206+MHCII+cells was significantly lower and CD206+MHCII- correspondingly higher in Inha+/- testes. This shift in expression of MHCII in CD206+ macrophages indicates that changes in circulating and/or local activin A influence resident macrophage activation and phenotype and, therefore, the immunological environment of the testis.


Subject(s)
Activins/metabolism , Inhibin-beta Subunits/metabolism , Leukocytes, Mononuclear/immunology , Macrophage Activation , Testis/immunology , Activins/analysis , Activins/genetics , Animals , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Cell Separation , Flow Cytometry , Inhibin-beta Subunits/analysis , Inhibin-beta Subunits/genetics , Leukocytes, Mononuclear/metabolism , Male , Mice , Mice, Transgenic , Testis/cytology
2.
Andrology ; 5(4): 763-770, 2017 07.
Article in English | MEDLINE | ID: mdl-28544640

ABSTRACT

Testicular germ cell tumours (TGCT) typically contain high numbers of infiltrating immune cells, yet the functional nature and consequences of interactions between GCNIS (germ cell neoplasia in situ) or seminoma cells and immune cells remain unknown. A co-culture model using the seminoma-derived TCam-2 cell line and peripheral blood mononuclear cells (PBMC, n = 7 healthy donors) was established to investigate how tumour and immune cells each contribute to the cytokine microenvironment associated with TGCT. Three different co-culture approaches were employed: direct contact during culture to simulate in situ cellular interactions occurring within seminomas (n = 9); indirect contact using well inserts to mimic GCNIS, in which a basement membrane separates the neoplastic germ cells and immune cells (n = 3); and PBMC stimulation prior to direct contact during culture to overcome the potential lack of immune cell activation (n = 3). Transcript levels for key cytokines in PBMC and TCam-2 cell fractions were determined using RT-qPCR. TCam-2 cell fractions showed an immediate increase (within 24 h) in several cytokine mRNAs after direct contact with PBMC, whereas immune cell fractions did not. The high levels of interleukin-6 (IL6) mRNA and protein associated with TCam-2 cells implicate this cytokine as important to seminoma physiology. Use of PBMCs from different donors revealed a robust, repeatable pattern of changes in TCam-2 and PBMC cytokine mRNAs, independent of potential inter-donor variation in immune cell responsiveness. This in vitro model recapitulated previous data from clinical TGCT biopsies, revealing similar cytokine expression profiles and indicating its suitability for exploring the in vivo circumstances of TGCT. Despite the limitations of using a cell line to mimic in vivo events, these results indicate how neoplastic germ cells can directly shape the surrounding tumour microenvironment, including by influencing local immune responses. IL6 production by seminoma cells may be a practical target for early diagnosis and/or treatment of TGCT.


Subject(s)
Cell Communication , Germ Cells/metabolism , Interleukin-6/metabolism , Leukocytes, Mononuclear/metabolism , Seminoma/metabolism , Seminoma/pathology , Testicular Neoplasms/metabolism , Tumor Microenvironment , Cell Line, Tumor , Cell Survival , Coculture Techniques , Culture Media, Conditioned/metabolism , Germ Cells/pathology , Humans , Interleukin-6/genetics , Leukocytes, Mononuclear/pathology , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Seminoma/genetics , Testicular Neoplasms/genetics , Testicular Neoplasms/pathology
3.
Am J Transplant ; 9(12): 2716-26, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19845582

ABSTRACT

Xenotransplantation of porcine islets into diabetic non-human primates is characterized by (i) an initial massive graft loss possibly due to the instant blood-mediated inflammatory reaction and (ii) the requirement of intensive, clinically unfriendly immunosuppressive therapy. We investigated whether the transgenic expression of a human complement-regulatory protein (hCD46) on porcine islets would improve the outcome of islet xenotransplantation in streptozotocin-induced diabetic Cynomolgus monkeys. Immunosuppression consisted of thymoglobulin, anti-CD154 mAb for costimulation blockade, and mycophenolate mofetil. Following the transplantation of islets from wild-type pigs (n = 2) or from 1,3-galactosyltransferase gene-knockout pigs (n = 2), islets survived for a maximum of only 46 days, as evidenced by return to hyperglycemia and the need for exogenous insulin therapy. The transplantation of islets from hCD46 pigs resulted in graft survival and insulin-independent normoglycemia in four of five monkeys for the 3 months follow-up of the experiment. One normalized recipient, selected at random, was followed for >12 months. Inhibition of complement activation by the expression of hCD46 on the pig islets did not substantially reduce the initial loss of islet mass, rather was effective in limiting antibody-mediated rejection. This resulted in a reduced need for immunosuppression to preserve a sufficient islet mass to maintain normoglycemia long-term.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Experimental/physiopathology , Islets of Langerhans Transplantation/methods , Membrane Cofactor Protein/genetics , Transplantation, Heterologous , Animals , Animals, Genetically Modified , Diabetes Mellitus, Experimental/surgery , Female , Macaca fascicularis , Male , Swine
4.
J Viral Hepat ; 15(10): 761-72, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18637077

ABSTRACT

SUMMARY: Serum-free culture conditions to generate immature human monocyte-derived DC (Mo-DC) were optimized, and the parameters that influence their maturation after exposure to lipopeptides containing CD4(+) and CD8(+) T-cell epitopes were examined. The lipopeptides contained a single CD4(+) helper T-cell epitopes, one of a number of human leucocyte antigen (HLA)-A2-restricted cytotoxic T-cell epitope and the lipid Pam2Cys. To ensure complete maturation of the Mo-DC, we examined (i) the optimal lipopeptide concentration, (ii) the optimal Mo-DC density and (iii) the appropriate period of exposure of the Mo-DC to the lipopeptides. The results showed that a high dose of lipopeptide (30 microm) was no more efficient at upregulating maturation markers on Mo-DC than a low dose (6 microm). There was an inverse relationship between Mo-DC concentration and the mean fluorescence intensity of maturation markers. In addition, at the higher cell concentrations, the chemotactic capacity of the Mo-DC towards a cognate ligand, CCL21, was reduced. Thus, high cell concentrations during lipopeptide exposure were detrimental to Mo-DC maturation and function. The duration of exposure of Mo-DC to the lipopeptides had little effect on phenotype, although Mo-DC exposed to lipopeptides for 48 rather than 4 h showed an increased ability to stimulate autologous peripheral blood mononuclear cells to release interferon-gamma in the absence of exogenous maturation factors. These findings reveal conditions for generating mature antigen-loaded DC suitable for targeted immunotherapy.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/immunology , Lipoproteins/immunology , Lymphocyte Activation , Peptides/immunology , T-Lymphocytes/immunology , Adult , Amino Acid Sequence , Cell Differentiation , Coculture Techniques , Culture Media, Serum-Free , Dendritic Cells/drug effects , Epitopes, T-Lymphocyte/chemistry , Humans , Immunologic Memory , Lipoproteins/chemical synthesis , Lipoproteins/chemistry , Male , Middle Aged , Monocytes/cytology , Peptides/chemical synthesis , Peptides/chemistry , T-Lymphocytes, Cytotoxic/immunology
5.
J Card Surg ; 16(6): 448-57, 2001.
Article in English | MEDLINE | ID: mdl-11925025

ABSTRACT

Xenotransplantation is being pursued vigorously to solve the shortage of allogeneic donor organs. Experimental studies of the major xenoantigen (Gal) and of complement regulation enable model xenografts to survive hyperacute rejection. When the Gal antigen is removed or reduced and complement activation is controlled, the major barriers to xenograft survival include unregulated coagulation within the graft and cellular reactions involving macrophages, neutrophils, natural killer (NK) cells, and T lymphocytes. Unlike allografts, where specific immune responses are the sole barrier to graft survival, molecular differences between xenograft and recipient that affect normal receptor-ligand interactions (largely active at the cell surface and which may not be immunogenic), are also involved in xenograft failure. Transgenic strategies provide the best options to control antigen expression, complement activation, and coagulation. Although the Gal antigen can be eliminated by gene knockout in mice, that outcome has only become a possibility in pigs due to the recent cloning of pigs after nuclear transfer. Instead, the use of transgenic glycosyl transferase enzymes and glycosidases, which generate alternative terminal carbohydrates on glycolipids and glycoproteins, has reduced antigen in experimental models. As a result, novel strategies are being tested to seek the most effective solution. Transgenic pigs expressing human complement-regulating proteins (DAF/CD55, MCP/CD46, or CD59) have revealed that disordered regulation of the coagulation system requires attention. There will undoubtedly be other molecular incompatibilities that need addressing. Xenotransplantation, however, offers hope as a therapeutic solution and provides much information about homeostatic mechanisms.


Subject(s)
Genetic Engineering , Transplantation, Heterologous/immunology , Animals , Blood Coagulation/physiology , Complement Activation/genetics , Complement Activation/immunology , Complement System Proteins/genetics , Complement System Proteins/immunology , Congresses as Topic/standards , Disaccharides/genetics , Disaccharides/immunology , Endothelium, Vascular/physiology , Gene Expression Regulation/physiology , Genetic Engineering/standards , Humans , Japan , Transplantation, Heterologous/standards
6.
Eur J Immunol ; 30(10): 2944-53, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11069077

ABSTRACT

The human anti-idiotypic antibody 105AD7 was isolated from a colorectal cancer patient receiving the anti-tumor antibody 791T/36 for radioimmuno-scintigraphy of liver metastases. We have mapped the binding site of 791T/36 to the first two small consensus repeat (SCR) domains of the complement regulatory protein (CD55) that is overexpressed by a wide range of solid tumors. Cloning of both antigen and anti-idiotype has identified the molecular basis of their mimicry. Amino acid homology has been identified between three complementarity-determining regions of 105AD7 and three regions of CD55 within the first two SCR domains. 791T/36 and anti-anti-idiotypic (Ab3) polyclonal antibodies raised against 105AD7 showed specific binding to these peptides. The antibodies were also found to bind synergistically to combinations of these peptides, indicating cooperativity between the peptides in stabilizing antibody binding. This also implies that the contact face on both CD55 antigen and 105AD7 is generated by the cooperation of several peptides positioned on two domains in each protein. Thus a human monoclonal anti-idiotypic antibody generated by a cancer patient is able to show both amino acid and structural homology with the complement regulatory protein CD55. These findings help identify the mechanism by which a human anti-idiotypic antibody is able to mimic a tumor-associated antigen and stimulate anti-tumor B and T cell responses.


Subject(s)
Adenocarcinoma/immunology , Antibodies, Anti-Idiotypic/therapeutic use , Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/immunology , Antigens, Neoplasm/chemistry , CD55 Antigens/chemistry , Colorectal Neoplasms/immunology , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/secondary , Adenocarcinoma/therapy , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/therapeutic use , Amino Acid Sequence , Animals , Antibodies, Anti-Idiotypic/chemistry , Antibodies, Anti-Idiotypic/genetics , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Neoplasm/biosynthesis , Antigen-Antibody Reactions , Antigens, CD/chemistry , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Binding Sites, Antibody , CD55 Antigens/genetics , CD55 Antigens/immunology , CHO Cells , Cloning, Molecular , Colorectal Neoplasms/therapy , Cricetinae , Genes, Immunoglobulin , Humans , Immune Sera/immunology , Immunity, Cellular , Immunoglobulin Variable Region/genetics , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/secondary , Membrane Cofactor Protein , Membrane Glycoproteins/chemistry , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Mimicry , Molecular Sequence Data , Peptide Fragments/chemistry , Protein Conformation , Protein Structure, Tertiary , Radioimmunodetection , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Sequence Alignment , Sequence Homology, Amino Acid , Transfection
8.
Transplantation ; 69(6): 1128-36, 2000 Mar 27.
Article in English | MEDLINE | ID: mdl-10762218

ABSTRACT

BACKGROUND: Recombinant soluble forms of complement regulatory molecules, including the human complement regulatory protein CD46 (rsCD46), have been shown to inhibit hyperacute transplant rejection (HAR) and protect against complement-mediated inflammatory tissue damage. Similarly, recombinant soluble forms of the immunoglobulin receptor FcgammaRII (rsFcgammaRII) can attenuate antibody-mediated inflammatory responses. We have produced and tested the function of novel recombinant chimeric proteins that incorporate the functional domains of both CD46 (membrane cofactor protein, MCP) and the low affinity human IgG receptor FcgammaRII (CD32). METHODS: Two recombinant soluble chimeric proteins (CD46:FcR and FcR:CD46) were designed and produced using a human cell expression system. Their ability to protect cells against complement-mediated lysis (through the CD46 domain) and bind human IgG (through the Fc receptor domain) was assessed in vitro. They were also tested in vivo in the rat reverse passive Arthus reaction and a murine model of hyperacute cardiac transplant rejection. RESULTS: In vitro, the functional domains of the chimeric proteins each retained their activity. In vivo, the serum half-life of the recombinant chimeric proteins in mice was more than either rsCD46 or rsFcgammaRII. In the rat reverse passive Arthus reaction, intradermal injection of each recombinant protein substantially reduced inflammatory skin edema (>50%) and polymorphonuclear neutrophil infiltration (>90%). In the hyperacute rejection model, i.v. treatment with FcR:CD46 prevented complement-mediated rejection, macroscopic bruising, edema, and thrombosis more effectively than rsCD46. CONCLUSIONS: CD46/FcgammaRII bifunctional proteins have an improved ability to control complement-mediated hyperacute graft rejection and have therapeutic potential in other conditions involving antibody-mediated inflammation.


Subject(s)
Antigens, CD/therapeutic use , Complement Inactivator Proteins/therapeutic use , Graft Rejection/prevention & control , Membrane Glycoproteins/therapeutic use , Receptors, IgG/therapeutic use , Animals , Antigens, CD/genetics , Complement Inactivator Proteins/genetics , Electrophoresis, Polyacrylamide Gel , Humans , Immunodominant Epitopes/genetics , Membrane Cofactor Protein , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Structure, Tertiary/physiology , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/immunology , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use , Sodium Dodecyl Sulfate , Solubility
9.
Immunology ; 97(2): 264-71, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10447741

ABSTRACT

Standard methods to generate autoimmune reactions in mice, by immunization with antigens emulsified with adjuvants, stimulate strong helper (CD4) T-cell and antibody responses but are not reported to induce cytolytic CD8 T cells. The aim of this study was to assess whether specific autoreactive CD8 T cells could be readily generated after immunization with a 'weak' autoantigen in adjuvant. Mice were immunized intraperitoneally three times with the E3 subunit of the mitochondrial 2-oxoacid dehydrogenase enzyme complexes (dihydrolipoamide dehydrogenase) emulsified with Freund's complete adjuvant. Splenic and lymph node lymphocytes were harvested after 14 days for in vitro functional studies. T lymphocytes were tested for proliferative responses and cytotoxicity against antigen-loaded isogeneic target cells. An autoreactive cytolytic T lymphocyte (CTL) response was detectable only after the in vitro restimulation of lymphocytes with E3 antigen-loaded syngeneic splenocytes. These CTL were identified as H-2-restricted CD8+ T cells. A proliferative response to E3 was demonstrable against antigen-pulsed syngeneic splenocytes. Immunized mice also generated strong antibody responses to E3. Liver histology showed portal infiltrates interpreted as a response of the liver to a non-specific immunological stimulus. It is concluded that autoreactive cytolytic T cells can be generated experimentally upon appropriate stimulation of the immune system, and can be identified in vitro upon release from the controlling mechanisms that are likely to regulate them in vivo.


Subject(s)
Autoimmunity , Dihydrolipoamide Dehydrogenase/immunology , Freund's Adjuvant , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Culture Techniques , Cell Division/immunology , Cytotoxicity, Immunologic , Female , Immunization , Liver/immunology , Liver/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H
10.
Reprod Fertil Dev ; 10(7-8): 683-96, 1998.
Article in English | MEDLINE | ID: mdl-10612477

ABSTRACT

The transplantation of organs and tissues between animal species, or xenotransplantation, is the focus of a growing field of research, owing primarily to the increasing shortage of allogeneic donor organs. The pig stands out as the most suitable donor animal for humans; however, xenografts (e.g. pig organs) used for human transplantation are normally destroyed by the host within minutes by hyperacute xenograft rejection. An improved understanding of the immune recognition and rejection of xenografts has resulted in new therapies that can partially overcome hyperacute rejection (HAR), delayed xenograft rejection (DXR) or acute vascular xenograft rejection. Strategies to diminish immunogenicity following xenotransplantation can be divided into two approaches: those directed at the recipient (e.g. antibodies or complement depletion or inhibition and tolerance induction) and those directed at the donor (e.g. transgenic modifications to express human complement-regulatory proteins or removal or displacement of alphaGal epitopes). DXR is likely to be controlled by transgenic inhibition of endothelial cell activation (e.g. inhibition of NF-kappaB). Transgenic pigs required for xenotransplantation will soon be generated at a greater efficiency and precision using nuclear transfer and cloning when compared to pronuclear injection. Of greater significance is that nuclear transfer offers the ability to target gene insertion selectively to specific gene loci and to delete specific genes in the pig. Experimental pig-to-primate organ xenotransplantation is currently under way, and results show increased transplant function from minutes to days and weeks. The final therapeutic regimen that allows survival of a discordant xenograft is likely to involve a combination of 'modified' functional genes in the donor organ, the development of immunological tolerance to pig antigens and administration of novel therapeutic agents, including immunosuppressants, that can control natural killer (NK) cell and monocyte mediated responses.


Subject(s)
Transplantation, Heterologous/methods , Transplantation, Heterologous/trends , Animals , Animals, Genetically Modified , Cloning, Organism , Complement Inactivator Proteins/therapeutic use , Embryo, Mammalian/cytology , Gene Expression , Graft Rejection/etiology , Graft Rejection/immunology , Graft Rejection/prevention & control , Humans , Immune Tolerance , Immunosuppression Therapy , Nuclear Transfer Techniques , Primates , Stem Cells/cytology , Swine
11.
Eur J Immunol ; 27(3): 726-34, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9079815

ABSTRACT

CD46 (membrane cofactor protein) is a human cell-surface regulator of activated complement and a receptor for the measles virus. A CD46 transgenic mouse line with an expression pattern similar to that of human tissues has been produced, to develop an animal model of (i) the control of complement activation by complement regulators in hyperacute rejection of xenografts, and (ii) measles virus infection. The mouse line was made using a CD46 minigene that includes promoter sequence and the first two introns of genomic CD46, which was coinjected into mouse ova with chicken lysozyme matrix attachment region DNA. A high level of CD46 expression in homozygotic transgenic mice was obtained with spleen cells having approximately 75% of the level found on human peripheral blood mononuclear cells. CD46 was detected in all tissues examined by immunohistochemistry, radioimmunoassay and Western blotting, showing that these mice were suitable for transplantation and measles virus infection studies. It also indicated that the transgene included the important regulatory elements of the CD46 promoter. Transgenic spleen cells were significantly protected in vitro from human complement activated by either the classical or alternative pathways and from alternative pathway rat complement. Furthermore, transgenic mouse hearts transplanted to rats regulated complement deposition in an in vivo model of antibody-dependent hyperacute xenograft rejection. Similar to human lymphocytes, transgenic lymphoblasts could be infected in vitro with measles virus; infected cells expressed viral proteins and produced infectious viral particles. The data demonstrate the suitability of this minigene for obtaining high-level CD46 expression sufficient for enhanced resistance of transgenic cells to complement attack and for obtaining wide tissue distribution of CD46, analogous to human tissues and, therefore, useful for comparative studies.


Subject(s)
Antigens, CD/physiology , Measles/immunology , Membrane Glycoproteins/physiology , Transplantation, Heterologous , Acute Disease , Animals , Complement Pathway, Alternative , Complement System Proteins/metabolism , Graft Rejection/immunology , Humans , Measles virus/growth & development , Measles virus/immunology , Membrane Cofactor Protein , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Rats , Tissue Distribution
12.
Eur J Biochem ; 238(1): 221-30, 1996 May 15.
Article in English | MEDLINE | ID: mdl-8665941

ABSTRACT

Specific sequences in the coding region of CD46 (membrane cofactor protein) transcripts have been shown to have a marked effect on translation. Two A+T-rich regions of CD46 cDNA were altered by mutation without changing the CD46 amino acid sequence (silent nucleotide substitution). In one region, the A+T content was reduced from 78% to 55% and in the other a putative polyadenylation addition sequence was disrupted. In each example, mutated sequences transfected into COS-7 cells produced significantly more soluble or cell surface protein (up to a 20-fold increase) than wild-type sequences. The amount of cellular plasmid DNA and CD46 mRNA was not increased, suggesting that the effect was not due to increased transfection efficiency, or transcript synthesis or stability. Biosynthetically labelled transfected cells showed an increase in translation rate but cell-free in vitro translation studies demonstrated that wild-type and mutated transcripts were translated with similar efficiency. The data show that translation of CD46 is affected by specific mRNA coding sequences, 400-540 bases from the initiation codon, and suggest that these sequences require the structural integrity of the cell to exert their effect.


Subject(s)
Antigens, CD/chemistry , Antigens, CD/genetics , Enhancer Elements, Genetic/genetics , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Protein Biosynthesis , Adenosine/chemistry , Adenosine/genetics , Animals , Antigens, CD/biosynthesis , Base Sequence , Cells, Cultured , DNA, Complementary/chemistry , DNA, Complementary/genetics , Membrane Cofactor Protein , Membrane Glycoproteins/biosynthesis , Models, Genetic , Molecular Sequence Data , Mutation , Plasmids/chemistry , Plasmids/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Thymidine/chemistry , Thymidine/genetics , Transfection
13.
Immunology ; 87(3): 348-54, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8778018

ABSTRACT

Human CD46 (membrane cofactor protein) is a type 1 glycoprotein that functions to protect autologous cells from complement-mediated damage by binding C3b and C4b for their factor I-mediated cleavage. We now describe the production and function of recombinant soluble CD46 (rsCD46), which was produced as a truncated form by mutagenesis using the splice overlap extension polymerase chain reaction, by inserting a translational stop codon into the CD46 cDNA at the junction of the transmembrane and extracellular domains. After transfection of an expression construct into 293-EBNA (Epstein-Barr nuclear antigen)-transformed cells, secretion of rsCD46 protein was detected by immunoradiometric assay using monoclonal antibodies. Following a single-step immunoaffinity purification, the protein resolved as a single band of approximately 56,000 MW on sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE). The purified rsCD46 (51 micrograms/ml) protected Chinese hamster ovary (CHO) cells from lysis initiated by a high titre rabbit anti-CHO antibody and complement from rabbit or human. The protection was specifically mediated by rsCD46 because the monoclonal antibody M177, which blocks interaction between CD46 and C3b/C4b, abrogated the protection. The results demonstrate that rsCD46 is effective as a fluid-phase regulator of complement activation on cell surfaces, even when initiated by the classical complement pathway. The in vivo efficacy of rsCD46 was investigated using a mouse heart to rat xenograft model. Administration of a bolus injection of rsCD46 was effective at delaying hyperacute graft rejection. These data suggest that rsCD46 may have a role as a therapeutic agent.


Subject(s)
Antigens, CD/immunology , Complement Activation/immunology , Complement System Proteins/immunology , Membrane Glycoproteins/immunology , Mutagenesis, Insertional , Animals , Antigens, CD/genetics , Antigens, CD/isolation & purification , Base Sequence , CHO Cells , Cricetinae , DNA Primers/genetics , Female , Genetic Vectors , Graft Rejection/immunology , Graft Rejection/therapy , Humans , Immunotherapy , Male , Membrane Cofactor Protein , Membrane Glycoproteins/genetics , Membrane Glycoproteins/isolation & purification , Mice , Models, Biological , Molecular Sequence Data , Polymerase Chain Reaction , Rats , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification
14.
Eur J Immunol ; 26(3): 578-85, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8605924

ABSTRACT

The human cell surface complement regulatory proteins CD46 (MCP), CD55 (DAF) and CD35 (CR1) protect autologous cells from complement-mediated damage by inhibiting C3 and C5 convertases. This regulatory potential has previously been exploited in the treatment of some models of inflammatory injury by the generation of recombinant soluble (rs) proteins, such as rsCD55 and rsCD35 . More recently, we have shown that rsCD46 inhibits complement activation in the fluid phase. In this report, the ability of rsCD46, rsD55 and rsCD35 to regulate human complement activation mediated by the classical pathway in vitro was clearly demonstrated by all three soluble proteins; however, rsCD35 was a more effective inhibitor than either rsCD46 or rsCD55. A combination of rsCD46+ rsCD55 was more potent than either of these proteins alone. Cell lysis via alternative pathway activation in vitro was efficiently regulated by rsCD46 and rsCD35 to a similar extent, whereas rsCD55 was not effective. Assays of rsCD46 in vivo have previously not been possible due to difficulties in expressing sufficient quantities of protein. This limitation has been overcome and now we report the ability of rsCD46 to inhibit immune complex-mediated inflammation in a rat using the reverse passive Arthus reaction model. Administration of rsCD46 significantly reduced the size of lesion, and histological examination showed a reduction in inflammatory infiltrate and edema. These data suggest that rsCD46, in addition to rsCd55 and rsCD35, may be useful a therapeutic agent.


Subject(s)
Antigens, CD/physiology , CD55 Antigens/physiology , Membrane Glycoproteins/physiology , Receptors, Complement 3b/physiology , Recombinant Proteins/pharmacology , Animals , Antigens, CD/administration & dosage , Arthus Reaction/immunology , Base Sequence , CD55 Antigens/pharmacology , Cell Line , Complement Inactivator Proteins/physiology , Complement Pathway, Alternative/drug effects , Complement Pathway, Classical/drug effects , Drug Combinations , Humans , Membrane Cofactor Protein , Membrane Glycoproteins/administration & dosage , Molecular Sequence Data , Rats , Recombinant Proteins/administration & dosage , Solubility
15.
J Immunol ; 155(11): 5089-94, 1995 Dec 01.
Article in English | MEDLINE | ID: mdl-7594517

ABSTRACT

CTLs that recognize tumor Ags have been described in mice and humans, particularly for melanoma. These CTLs are CD8+, which is MHC-restricted. In contrast, in human carcinomas of the breast, pancreas, or ovary, and in multiple myeloma, CD8+ CTLs have been described that lyse targets expressing human MUC1 in a non-MHC-restricted manner. On the basis of these observations, we immunized mice with conjugates of mannan-human fusion protein, human mucin 1 (MUC1), which produced CD8+ CTLs. In contrast to the human anti-MUC1 CTLs found in cancer patients, the murine anti-MUC1 CTLs were clearly MHC-restricted, e.g., in inbred mice of the H-2-b, d, k, s, or z haplotypes; the H-2 restriction was also confirmed in H-2 congenic strains. Tests of H-2 recombinant strains demonstrated that MUC1 peptides were able to associate with D or K class I molecules of the b, d, or k haplotypes. Mice lacking MHC-class I molecules made weak CTL responses that were H-2Db-restricted, and in the class I H-2Kbm1 mutant strain, CTL restriction was also shown. Finally, cold target inhibition studies demonstrated that Kb and Db are recognized similarly, but Kk is less well recognized. Thus, anti-MUC1 CTLs induced by immunization of mice are different from those obtained from patients. The immunization of cancer patients with MUC1 peptides is now undergoing clinical trials and it will be of interest to observe whether the CTLs induced are HLA-restricted, not restricted, or whether both types of CTLs are produced.


Subject(s)
Antigens, Neoplasm/immunology , Major Histocompatibility Complex , Mucin-1/genetics , Mucin-1/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, Neoplasm/genetics , Binding, Competitive/immunology , Cytotoxicity Tests, Immunologic , H-2 Antigens/genetics , H-2 Antigens/immunology , Humans , Mannans/metabolism , Mice , Mice, Inbred Strains , Mucin-1/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , beta 2-Microglobulin/immunology
16.
Proc Natl Acad Sci U S A ; 92(22): 10128-32, 1995 Oct 24.
Article in English | MEDLINE | ID: mdl-7479739

ABSTRACT

The induction of CD8+ cytotoxic T lymphocytes (CTLs) is desirable for immunization against many diseases, and recombinant-synthetic peptide antigens are now favored agents to use. However, a major problem is how to induce CTLs, which requires a T1-type response to such synthetic antigens. We report that T1-type (generating high CTL, low antibody) or T2-type (the reciprocal) responses can be induced by conjugation of the antigen to the carbohydrate polymer mannan: T1 responses are selected by using oxidizing conditions; T2 responses are selected by using reducing conditions for the conjugation. Using human MUC1 as a model antigen in mice, immunization with oxidized mannan-MUC1 fusion protein (ox-M-FP) led to complete tumor protection (challenge up to 5 x 10(7) MUC1+ tumor cells), CTLs, and a high CTL precursor (CTLp) frequency (1/6900), whereas immunization with reduced mannan-MUC1 FP (red-M-FP) led to poor protection after challenge with only 10(6) MUC1+ tumor cells, no CTLs, and a low CTLp frequency (1/87,800). Ox-M-FP selects for a T1 response (mediated here by CD8+ cells) with high interferon gamma (IFN-gamma) secretion, no interleukin 4 (IL-4), and a predominant IgG2a antibody response; red-M-FP selects for a T2-type response with IL-4 production and a high predominant IgG1 antibody response but no IFN-gamma.


Subject(s)
Mannans/immunology , Mucin-1/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/immunology , 3T3 Cells , Animals , Antibody Formation , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin G/classification , Interferon-gamma/biosynthesis , Interleukin-4/biosynthesis , Mice , Mice, Inbred BALB C , Oxidation-Reduction , Spleen/immunology , Tumor Cells, Cultured
17.
J Gastroenterol Hepatol ; 10(5): 491-7, 1995.
Article in English | MEDLINE | ID: mdl-8963023

ABSTRACT

The aim of this study was to investigate liver histology in mice after immunization with the conserved self molecule dihydrolipoamide dehydrogenase, E3, a subunit of the mitochondrial 2-OADC enzyme family identified as the M2 autoantigen in the liver disease, primary biliary cirrhosis. Mice were immunized by a novel procedure. The autoantigen E3 was introduced by pinocytosis into hypertonically treated syngeneic lymphoid cells to facilitate intracellular antigen processing and presentation and the generation of a cytolytic T cell response. Liver sections were examined and scored for evidence of an inflammatory response by two independent procedures: standard microscopy with visual scoring, and automated scanning with computerized scoring. There was a close correlation between read-outs of liver histology by standard microscopy and automated scanning, using the index of mononuclear cellular infiltrations in hepatic portal tracts. Such infiltrates were prominent in the immunized mice, but, unexpectedly, the degree of infiltration was similar in mice injected with autoantigen (E3)-loaded syngeneic cells, or syngeneic cells treated only with hypertonic medium. The equivalent changes in the liver with the experimental and control protocol is indicative of the reactivity of the liver to any provocative immune stimulus, and is cautionary for protocols designed for the induction of autoimmune liver disease in experimental animals.


Subject(s)
Immunization , Liver Circulation , Lymphocytes/immunology , Portal System , Animals , Autoantigens/immunology , Autoimmune Diseases/pathology , Culture Media/pharmacology , Cytotoxicity Tests, Immunologic , Female , Hypertonic Solutions/pharmacology , Liver/drug effects , Liver/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Time Factors
19.
Transplantation ; 58(8): 879-82, 1994 Oct 27.
Article in English | MEDLINE | ID: mdl-7524207

ABSTRACT

Hyperacute rejection, mediated by natural antibody, is the major barrier to xenotransplantation. The studies reported herein were aimed at evaluating antibody-mediated cytotoxicity and the role of the Gal alpha(1,3)Gal epitope, which we had previously demonstrated was the major epitope of pig cells detected by naturally occurring human antibodies. Also, we had shown that this epitope could be induced in non-expressing cells by the transfection of a cDNA clone encoding alpha(1,3)galactosyl transferase, the enzyme that produces this epitope. The importance of the Gal alpha(1,3)Gal epitope was supported by (1) sugar inhibition studies; (2) complete absorption of cytotoxic antibodies by melibiose-sepharose columns; and (3) the ability of normal human serum to lyse COS cells after transfection with a cDNA clone encoding alpha(1,3)galactosyl transferase. These findings strongly suggest that the majority of cytotoxic human antibodies that would recognize a xenogeneic graft are directed to the Gal alpha(1,3)Gal epitope.


Subject(s)
Epitopes/immunology , Galactosyltransferases/immunology , Transplantation, Heterologous/immunology , Animals , Antibodies, Heterophile/immunology , Antibody-Dependent Cell Cytotoxicity , Cell Line , Galactosyltransferases/genetics , Graft Rejection/immunology , Humans , Immunoglobulin M/immunology , Swine , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...