Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Dev Immunol ; 2013: 186420, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24066003

RESUMO

The measurement of vaccine-induced humoral and CD4(+) and CD8(+) cellular immune responses represents an important correlate of vaccine efficacy. Accurate and reliable assays evaluating such responses are therefore critical during the clinical development phase of vaccines. T cells play a pivotal role both in coordinating the adaptive and innate immune responses and as effectors. During the assessment of cell-mediated immunity (CMI) in subjects participating in a large-scale influenza vaccine trial, we identified the expansion of an IFN-γ-producing CD3(+)CD4(-)CD8(-) γδ (+) T cell population in the peripheral blood of 90/610 (15%) healthy subjects. The appearance of CD3(+)CD4(-)CD8(-) γδ (+) T cells in the blood of subjects was transient and found to be independent of the study cohort, vaccine group, subject gender and ethnicity, and ex vivo restimulation conditions. Although the function of this population and relevance to vaccination are unclear, their inclusion in the total vaccine-specific T-cell response has the potential to confound data interpretation. It is thus recommended that when evaluating the induction of IFN-γ-producing CD4(+) and CD8(+) immune responses following vaccination, the CD3(+)CD4(-)CD8(-) γδ (+) T cells are either excluded or separately enumerated from the overall frequency determination.


Assuntos
Leucócitos Mononucleares/imunologia , Subpopulações de Linfócitos T/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Complexo CD3/metabolismo , Antígenos CD4/metabolismo , Antígenos CD8/metabolismo , Estudos de Coortes , Humanos , Imunidade Celular/imunologia , Imunofenotipagem , Vacinas contra Influenza/imunologia , Influenza Humana/prevenção & controle , Leucócitos Mononucleares/metabolismo , Pessoa de Meia-Idade , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Subpopulações de Linfócitos T/metabolismo , Fatores de Tempo , Vacinação , Potência de Vacina , Adulto Jovem
2.
Vaccine ; 22(19): 2391-5, 2004 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-15193400

RESUMO

ISCOMATRIX adjuvant is capable of inducing broad and potent humoral and cellular immune responses. The components are well defined and the manufacturing process is simple and robust. Many vaccines containing the ISCOMATRIX adjuvant have been tested in a range of animal models, including human and non-human primates. Strong antibody and T cell responses have been induced in these studies. The antibody response is often achieved with lesser amounts of antigen than other adjuvant systems and the maximal responses have also been reached more quickly. Both CD4+ and CD8+ T cell responses are induced with the cytotoxic T lymphocyte responses being very long lived. Additionally, ISCOMATRIX adjuvant can be used in vaccines for induction of mucosal immune responses. This review provides an overview of the immune responses that can be elicited using ISCOMATRIX vaccines and the current state of knowledge regarding the mechanism of action of this adjuvant.


Assuntos
Adjuvantes Imunológicos/farmacologia , Formação de Anticorpos/efeitos dos fármacos , Imunidade Celular/efeitos dos fármacos , Vacinas/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Humanos
3.
Transgenic Res ; 10(6): 523-31, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11817540

RESUMO

A retrospective analysis of transgenesis rates obtained in seven pronuclear microinjection programs was undertaken to determine if a relationship existed between the amount of DNA injected and transgenesis rates in the pig. Logistic regression analysis showed that as the concentration of DNA injected increased from 1 to 10 ng/microl, the number of transgenics when expressed as a proportion of the number liveborn (integration rate) increased from 4% to an average of 26%. A similar relationship was found when the number of molecules of DNA injected per picolitre was analysed. No evidence was obtained to suggest either parameter influenced integration rate in mice when the same constructs were injected. The number of transgenics liveborn when expressed as a proportion of ova injected (efficiency rate), increased as DNA concentration increased up to 7.5 ng/microl and then decreased at 10 ng/microl for both species suggesting that at this concentration DNA (or possible contaminants) may have influenced embryo survival. The relationship between efficiency and the number of molecules injected per picolitre was complex suggesting that the concentration at which DNA was injected was a better determinant of integration and efficiency rates. In conclusion, the present study suggests that transgenes need to be injected at concentrations of between 5 and 10 ng/microl to maximise integration and efficiency rates in pigs.


Assuntos
Animais Geneticamente Modificados , Técnicas de Transferência de Genes , Camundongos Transgênicos , Animais , Southern Blotting , DNA/análise , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Estudos Retrospectivos , Suínos , Transgenes
4.
Xenotransplantation ; 7(4): 237-46, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11081758

RESUMO

Pig-to-primate vascularized xenografts undergo hyperacute rejection (HAR). This results from pre-formed xenoreactive antibodies directed against galactose-alpha1,3-galactose (alphaGal) in the donor organ and activation of the complement cascade. We describe an in vivo murine model of HAR using a BALB/c mice system devoid of histocompatibility or complement differences between donor and recipient to investigate in isolation, the effects of alphaGal epitope and anti-alphaGal antibody interactions in causing rejection of vascularized heart transplants. Gal KO mice were immunized with rabbit red blood cell membranes to induce high anti-alphaGal antibody titers that were predominantly IgM by ELISA (enzyme-linked immunosorbent assay). When alphaGal-expressing mice hearts were transplanted heterotopically into these recipients (n= 12), 67% of grafts rejected within 24 h, the majority within 16 h with histological features of HAR. In contrast, none of the grafts in the non-immunized Gal KO recipient control group (n=11) underwent HAR. Interestingly, approximately 50% of the remaining grafts in both the immunized and non-immunized Gal KO recipient group were rejected between 7 and 27 days by a rejection process characterized by a dense infiltrate of macrophage/monocytes, perivascular cuffing and tissue destruction similar to recent descriptions of delayed xenograft rejection (DXR). In addition, some grafts (21.5%) continued to survive in the immunized Gal KO recipients despite the presence of anti-alphaGal antibody and normal complement activity and these showed well-preserved myocardium when harvested whilst still functioning well at days 30 or 90. No rejection was seen when Gal KO donors were used in this system (n=4), nor when alphaGal-expressing BALB/c hearts were transplanted into alphaGal-expressing BALB/c recipients (n=5). This in vivo small animal model offers the opportunity to test a variety of strategies to overcome HAR prior to more resource intensive pig-to-primate studies, and may provide insights into the processes similar to DXR and accommodation.


Assuntos
Dissacarídeos/imunologia , Galactosiltransferases/metabolismo , Rejeição de Enxerto/imunologia , Transplante de Coração/imunologia , Doença Aguda , Animais , Anticorpos Heterófilos/sangue , Arteríolas/patologia , Vasos Coronários/patologia , Epitopos/imunologia , Transfusão de Eritrócitos , Galactosiltransferases/deficiência , Galactosiltransferases/genética , Rejeição de Enxerto/patologia , Transplante de Coração/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout , Coelhos
5.
Transplantation ; 70(6): 963-8, 2000 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-11014650

RESUMO

BACKGROUND: It has been proposed that hyperacute rejection (HAR) of pig-to-primate vascularized xenografts is due in large part to ineffective regulation of recipient complement by pig complement regulatory proteins (CRPs), and indeed transgenic expression of human CRPs in pigs can prevent hyperacute rejection. However, at least one pig CRP (CD59) efficiently regulates human complement in vitro, suggesting that it is the level of expression of a particular CRP(s) rather than cross-species incompatibility that explains the HAR of porcine xenografts. We investigated the relative effectiveness of transgenically expressed pig and human CD59 in providing protection of mouse hearts from human complement in an ex vivo setting. METHODS: Transgenic mice expressing pig CD59 or human CD59 under the control of the human ICAM-2 promoter, which restricts expression in tissues to vascular endothelium, were used. Hearts from mice expressing similar levels of pig CD59 or human CD59 were perfused ex vivo with 10% human plasma and heart function was monitored for 60 min. Sections of perfused hearts were examined for deposition of the membrane attack complex (MAC). RESULTS: Control nontransgenic hearts (n=5) were rapidly affected by the addition of human plasma, with mean function falling to less than 10% of the initial level within 15 min. In contrast, hearts expressing either pig CD59 (n=6) or human CD59 (n=8) were protected from plasma-induced injury, maintaining 31 and 35% function, respectively, after 60 min of perfusion. MAC deposition was markedly reduced in both pig CD59 and human CD59 transgenic hearts compared to nontransgenic control hearts. CONCLUSIONS: When highly expressed on endothelium in transgenic mice, pig CD59 provided equivalent protection to human CD59 in a model of human complement-mediated xenograft rejection. Thus supranormal expression of endogenous porcine CRPs may be a feasible alternative to the expression of human CRPs in preventing HAR of pig-to-primate xenografts.


Assuntos
Antígenos CD59/farmacologia , Proteínas Inativadoras do Complemento/farmacologia , Transplante de Coração/imunologia , Animais , Rejeição de Enxerto/prevenção & controle , Humanos , Imuno-Histoquímica , Antígeno de Macrófago 1/metabolismo , Camundongos , Camundongos Transgênicos , Miocárdio/imunologia , Miocárdio/metabolismo , Perfusão , Suínos , Transgenes/fisiologia
6.
Transplantation ; 69(12): 2504-15, 2000 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-10910270

RESUMO

BACKGROUND: The genetic modification of pigs is a powerful strategy that may ultimately enable successful xenotransplantation of porcine organs into humans. METHODS: Transgenic pigs were produced by microinjection of gene constructs for human complement regulatory proteins CD55 and CD59 and the enzyme alpha1,2-fucosyltransferase (H-transferase, HT), which reduces expression of the major xenoepitope galactose-alpha1,3-galactose (alphaGal). Kidneys from CD55/HT and CD55/CD59/HT transgenic pigs were transplanted into nephrectomised, nonimmunosuppressed adult baboons. RESULTS: In several lines of transgenic pigs, CD55 and CD59 were expressed strongly in all tissues examined, whereas HT expression was relatively weak and did not significantly reduce alphaGal. Control nontransgenic kidneys (n=4) grafted into baboons were hyperacutely rejected within 1 hr. In contrast, kidneys from CD55/HT pigs (n=2) were rejected after 30 hr, although kidneys from CD55/CD59/HT pigs (n=6) maintained function for up to 5 days. In the latter grafts, infiltration by macrophages, T cells, and B cells was observed at days 3 and 5 posttransplantation. The recipients developed thrombocytopenia and abnormalities in coagulation, manifested in increased clotting times and an elevation in the plasma level of the fibrin degradation product D-dimer, within 2 days of transplantation. Treatment with low molecular weight heparin prevented profound thrombocytopenia but not the other aspects of coagulopathy. CONCLUSIONS: Strong expression of CD55 and CD59 completely protected porcine kidneys from hyperacute rejection and allowed a detailed analysis of xenograft rejection in the absence of immunosuppression. Coagulopathy appears to be a common feature of pig-to-baboon renal transplantation and represents yet another major barrier to its clinical application.


Assuntos
Transtornos da Coagulação Sanguínea/etiologia , Antígenos CD59/fisiologia , Fucosiltransferases/fisiologia , Rejeição de Enxerto , Transplante de Rim/imunologia , Transplante Heterólogo/imunologia , Animais , Antígenos CD59/análise , Antígenos CD59/genética , Fucosiltransferases/genética , Imuno-Histoquímica , Terapia de Imunossupressão , Rim/patologia , Transplante de Rim/efeitos adversos , Camundongos , Papio , Suínos , Galactosídeo 2-alfa-L-Fucosiltransferase
7.
Subcell Biochem ; 32: 281-310, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10392000

RESUMO

Although originally generated to test the effect of eliminating the alpha-Gal epitope on HAR, it is becoming increasingly clear that GalT KO mice offer a convenient and inexpensive model to investigate many aspects of the anti-xenorgraft immune response. Clearly, not all aspects of anti-xenograft rejection responses are identical in mice and primates, which should be kept in mind when interpreting results of GalT KO mouse studies. However, with this and other mouse models it is possible to test a large number of variables, which is impractical for both logistical and financial reasons with primates. Furthermore the short gestation time and large litter size of mice means that genetic strategies targeting different aspects of the anti-xenograft immune response can be combined and subsequently tested to identify the optimal combination of genetic and therapeutic approaches to achieve long term xenograft survival. In this regard the GalT KO mouse has been and will continue to be a valuable small animal model for the study of all facets of xenograft rejection involving anti-Gal antibodies.


Assuntos
Galactosiltransferases/deficiência , Transplante Heterólogo/imunologia , Animais , Galactosiltransferases/genética , Camundongos , Camundongos Knockout
8.
Proc Natl Acad Sci U S A ; 96(13): 7312-7, 1999 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-10377411

RESUMO

Parasites have evolved a plethora of strategies to ensure their survival. The intracellular parasite Theileria parva secures its propagation and spreads through the infected animal by infecting and transforming T cells, inducing their continuous proliferation and rendering them metastatic. In previous work, we have shown that the parasite induces constitutive activation of the transcription factor NF-kappaB, by inducing the constitutive degradation of its cytoplasmic inhibitors. The biological significance of NF-kappaB activation in T. parva-infected cells, however, has not yet been defined. Cells that have been transformed by viruses or oncogenes can persist only if they manage to avoid destruction by the apoptotic mechanisms that are activated on transformation and that contribute to maintain cellular homeostasis. We now demonstrate that parasite-induced NF-kappaB activation plays a crucial role in the survival of T. parva-transformed T cells by conveying protection against an apoptotic signal that accompanies parasite-mediated transformation. Consequently, inhibition of NF-kappaB nuclear translocation and the expression of dominant negative mutant forms of components of the NF-kappaB activation pathway, such as IkappaBalpha or p65, prompt rapid apoptosis of T. parva-transformed T cells. Our findings offer important insights into parasite survival strategies and demonstrate that parasite-induced constitutive NF-kappaB activation is an essential step in maintaining the transformed phenotype of the infected cells.


Assuntos
Apoptose , Proteínas de Ligação ao Cálcio , Linfócitos T/patologia , Linfócitos T/parasitologia , Theileria parva/parasitologia , Theileriose/imunologia , Animais , Apoptose/imunologia , Bovinos , Linhagem Celular , Proteínas de Ligação a DNA/imunologia , Humanos , Proteínas I-kappa B , Glicoproteínas de Membrana/imunologia , NF-kappa B/imunologia , Proteínas do Tecido Nervoso/imunologia , Sinaptotagmina I , Sinaptotagminas , Linfócitos T/imunologia , Theileriose/patologia
9.
Xenotransplantation ; 5(3): 184-90, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9741456

RESUMO

High-level endothelial expression of the human complement regulatory factor CD59 has been shown to protect transgenic mouse hearts from human complement-mediated injury in an ex vivo perfusion model. In this study we examine whether co-expression of CD55 provides additional protection. CD55/CD59 double-transgenic mice were generated by co-injection of CD55 and CD59 expression constructs driven by the human intercellular adhesion molecule 2 (ICAM-2) promoter. A line was established from one mouse that exhibited strong expression of CD55 and CD59 on vascular endothelium in the heart and other transplantable organs. An ex vivo perfusion model was used to compare hearts from these CD55/CD59 mice with hearts from a previously established line, which expressed CD59 at a similar level to the double transgenic line. CD59 hearts displayed prolonged survival compared to wild-type hearts during perfusion with 40% human plasma and maintained approximately 20% maximum work after 60 min. CD55/CD59 hearts were further protected, with work maintained at 35% of the maximum level after 60 min. The data demonstrate that high-level endothelial co-expression of CD55 and CD59 provides greater protection from human complement-mediated injury in this model than expression of CD59 alone.


Assuntos
Antígenos CD55/imunologia , Antígenos CD59/imunologia , Ativação do Complemento/imunologia , Endotélio Vascular/imunologia , Traumatismo por Reperfusão Miocárdica/imunologia , Animais , Antígenos CD55/biossíntese , Antígenos CD55/genética , Antígenos CD59/biossíntese , Antígenos CD59/genética , Endotélio Vascular/patologia , Humanos , Camundongos , Camundongos Transgênicos , Miocárdio/imunologia , Miocárdio/patologia
10.
Transplantation ; 66(11): 1401-6, 1998 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-9869079

RESUMO

BACKGROUND: Complement activation plays a pivotal role in hyperacute xenograft rejection. In humans, activation of complement is regulated by a number of cell surface regulatory proteins. Membrane cofactor protein (CD46) is one such regulator that protects cells by acting as a cofactor for the factor I-mediated cleavage of C3b and C4b. Transgenic animals expressing human CD46 may provide organs that are resistant to complement attack. However, attempts to generate mice expressing human CD46 using cDNA-based constructs have been largely unsuccessful. METHODS: Transgenic mice expressing a glycosylphosphatidyl inositol (GPI)-linked form of CD46 were generated by microinjection of a hybrid CD46/CD55 cDNA under the control of the human intercellular adhesion molecule-2 promoter. Expression of CD46-GPI on the vascular endothelium was determined by immunohistochemistry. The ability of CD46-GPI to protect mouse tissues from human complement attack was determined using an ex vivo isolated perfused heart model. RESULTS: Three founder animals expressing CD46-GPI were identified. Histological analysis showed strong and uniform expression of CD46-GPI on the vascular endothelium of all organs examined. Ex vivo perfusion of transgenic mouse hearts with human plasma showed a reduction in C3c deposition and a slightly prolonged function compared with controls. CONCLUSIONS: High-level expression of CD46-GPI was achieved in transgenic mice by using a modified cDNA-based construct. The CD46-GPI was functional, providing some protection from complement-mediated damage in the ex vivo model, and may be useful in xenotransplantation if expressed in combination with CD55 and CD59.


Assuntos
Antígenos CD/genética , Antígenos CD/fisiologia , Glicosilfosfatidilinositóis/genética , Glicosilfosfatidilinositóis/fisiologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiologia , Animais , Moléculas de Adesão Celular/genética , Corantes , Proteínas Inativadoras do Complemento/genética , Proteínas Inativadoras do Complemento/fisiologia , Endotélio Vascular/citologia , Fluoresceína-5-Isotiocianato , Expressão Gênica , Glicosilfosfatidilinositóis/imunologia , Humanos , Proteína Cofatora de Membrana , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas
11.
Transplantation ; 66(6): 748-54, 1998 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-9771838

RESUMO

BACKGROUND: The key role of anti-galactose alpha1,3-galactose (anti-alphaGal) xenoantibodies in initiating hyperacute xenograft rejection has been clearly demonstrated using a variety of in vitro and in vivo approaches. However, the role of anti-alphaGal antibodies in mediating post-hyperacute rejection mechanisms, such as antibody-dependent cellular cytoxicity, remains to be determined, primarily because of the lack of a small animal model with which to study this phenomena. METHODS: Hearts from wild-type mice were transplanted heterotopically into alpha1,3-galactosyltransferase knockout (Gal KO) mice, which like humans develop antibodies to the disaccharide galactose alpha1,3-galactose (Gal). At the time of rejection, hearts were examined histologically to determine the mechanism of rejection. RESULTS: Hearts from wild-type mice transplanted into high-titer anti-alphaGal recipients were rejected in 8-13 days. Histological examination demonstrated a cellular infiltrate consisting of macrophages (80-90%), natural killer cells (5-10%), and T cells (1-5%). In contrast, wild-type hearts transplanted into low anti-Gal titer recipients demonstrated prolonged (>90 day) survival. However, a significant proportion (30-40%) of these underwent a minor rejection episode between 10 and 13 days, but then recovered ("accommodated"). CONCLUSIONS: The results of this study suggest that the Gal KO mouse is a useful small animal vascularized allograft model, in which the role of anti-alphaGal antibody in graft rejection can be studied in isolation from other rejection mechanisms. The titer of anti-alphaGal antibody was found to be the critical determinant of rejection. The histopathological features of rejection in this model are very similar to other models of delayed xenograft rejection, in both the timing and composition of the cellular infiltrate. The Gal KO mouse therefore provides a new rodent model, which will aid in the identification of the distinct components involved in the pathogenesis of delayed xenograft rejection.


Assuntos
Anticorpos/imunologia , Dissacarídeos/imunologia , Galactosiltransferases/genética , Rejeição de Enxerto/imunologia , Transplante Heterólogo/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Feminino , Galactosiltransferases/metabolismo , Rejeição de Enxerto/genética , Rejeição de Enxerto/patologia , Transplante de Coração/imunologia , Leishmania major/imunologia , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Miocárdio/patologia
12.
Transplantation ; 65(12): 1599-604, 1998 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-9665076

RESUMO

BACKGROUND: Organs from transgenic animals with high-level endothelial expression of the human complement regulatory factors CD55 and CD59 are significantly protected from human complement-mediated injury. Elimination or reduction of the major xenoepitope alphaGal, achieved by knocking out the alpha1,3-galactosyltransferase gene (Gal KO) or expressing human alpha1,2-fucosyltransferase (H transferase or HTF), also affords protection, although to a lesser degree. In this study, we examined whether the protection provided by strong CD55 and CD59 expression can be augmented by the Gal KO or HTF modifications. METHODS: Hearts from four groups of mice (wild type, CD55/CD59, CD55/CD59/HTF, and CD55/CD59/Gal KO) were perfused ex vivo with 40% human plasma. Mean heart work for each group was compared over a 60-min period. RESULTS: Wild-type hearts ceased to function effectively within 15 min of plasma addition. CD55/CD59 hearts displayed prolonged survival and maintained approximately 10% maximum work at the end of perfusion. Introduction of Gal KO or HTF onto the CD55/CD59 background resulted in a further prolongation, with work maintained at 20-30% of the maximum level. CONCLUSIONS: We used an ex vivo model to demonstrate that eliminating alphaGal expression further prolongs the function of mouse hearts expressing high levels of CD55 and CD59. In addition, we showed that reducing alphaGal by expressing HTF is equally as effective in prolonging CD55/CD59 heart function as knocking out Gal transferase, thus providing a feasible strategy for translating these advances to the pig.


Assuntos
Antígenos CD55/análise , Antígenos CD59/análise , Fucosiltransferases/fisiologia , Galactosiltransferases/fisiologia , Rejeição de Enxerto/prevenção & controle , Transplante de Coração , Animais , Galactosiltransferases/genética , Humanos , Camundongos , Camundongos Knockout , Miocárdio/imunologia , Miocárdio/patologia , Transplante Heterólogo , Galactosídeo 2-alfa-L-Fucosiltransferase
13.
J Biol Chem ; 273(19): 11737-44, 1998 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-9565596

RESUMO

The expression of intercellular adhesion molecule 2 (ICAM-2) in adult tissues is restricted to vascular endothelial cells and megakaryocytes. We have previously shown that the endothelial-specific in vivo activity of the human ICAM-2 promoter is contained within a small (0.33-kilobase (kbp)) 5'-flanking region of the gene. Here we describe the in vitro characterization of this region. The ICAM-2 promoter is TATA-less, and transcription in endothelial cells initiates at four sites. Reporter gene expression directed by the promoter was 125-fold greater than vector alone in bovine aortic endothelial cells but less than 2-fold vector alone in non-endothelial (COS) cells, confirming that specificity in vivo was paralleled in vitro. The addition of 2.7 kbp of 5'-flanking region to the 0.33-kbp fragment had no effect on promoter activity or specificity. The mutation of an Sp1 motif centered at base pair -194 or an eight-base pair palindrome at -268 each reduced promoter activity by 70%. Mutation of GATA motifs at -145 and -53 reduced promoter activity by 78 and 61%, respectively. Specific binding of bovine aortic endothelial cells nuclear proteins to the Sp1 and GATA sites was demonstrated by gel shift analysis. Promoter activity in COS cells was transactivated 3-4-fold by overexpression of GATA-2. The results presented here suggest that transcription from the ICAM-2 promoter in endothelial cells is regulated by the interplay of several positive-acting factors and provide the basis for further analysis of endothelial-specific gene expression.


Assuntos
Antígenos CD/genética , Moléculas de Adesão Celular/genética , Endotélio/fisiologia , Animais , Sequência de Bases , Sítios de Ligação , Análise Mutacional de DNA , Proteínas de Ligação a DNA/metabolismo , Fatores de Ligação de DNA Eritroide Específicos , Fator de Transcrição GATA2 , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Sequências Reguladoras de Ácido Nucleico , Mapeamento por Restrição , Homologia de Sequência do Ácido Nucleico , Fator de Transcrição Sp1/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional
14.
Transplantation ; 65(6): 826-31, 1998 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-9539095

RESUMO

BACKGROUND: Hyperacute rejection of discordant xenografts is dependent on activation of the complement system of the recipient. Transgenic expression of recipient complement regulatory factors in donor tissue has proved to be a promising approach to dealing with hyperacute rejection, although the relationship between the level of complement regulatory factor expression and the degree of protection is not well established. Here, we examine this relationship using CD59 transgenic mouse hearts in an ex vivo model of xenograft rejection. METHODS: The level of expression of CD59 in two lines of transgenic mice, in which CD59 is expressed under the control of either the murine H2Kb (MHC class I) promoter (line CA-17) or the endothelium-specific human intercellular adhesion molecule-2 promoter (line 237-7), was compared by immunohistochemistry and flow cytometry. Hearts from both groups and wild-type controls were perfused ex vivo with human plasma, and mean heart work for each group was compared over a 60-min period. RESULTS: CD59 expression on cardiac endothelial cells isolated from homozygous CA-17 mice was 25- to 30-fold lower than that on cardiac endothelial cells from heterozygous 237-7 mice. CA-17 hearts perfused with 6% human plasma exhibited a reduction in deposition of the membrane attack complex, but not a prolongation of function, compared with nontransgenic mouse hearts. In contrast, 237-7 hearts showed significantly prolonged function during perfusion with 20% plasma. CONCLUSIONS: High-level endothelial-specific expression of CD59 was effective in prolonging the function of mouse hearts perfused with 20% human plasma, whereas low-level, broader expression did not provide protection from 6% plasma.


Assuntos
Antígenos CD59/metabolismo , Endotélio Vascular/imunologia , Rejeição de Enxerto , Transplante de Coração/imunologia , Animais , Complemento C3c/metabolismo , Complemento C9/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Perfusão , Transplante Heterólogo
15.
Transplantation ; 65(6): 832-7, 1998 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-9539096

RESUMO

BACKGROUND: The expression of human alpha1,2-fucosyltransferase (H-transferase, HT) has been proposed as an alternative strategy to alpha1,3-galactosyltransferase (GT) gene knockout, which is not currently feasible in pigs, to reduce the galactose-alpha1,3-galactose (Gal) epitope expression. HT expression has recently been shown in transgenic mice and pigs to significantly reduce Gal expression on a variety of cells; however, its ability to do so on endothelial cells and its effectiveness at prolonging xenograft survival are yet to be determined. METHODS: HT-transgenic, Gal knockout (Gal KO) mice, and mice containing both genetic modifications (HT-transgenic/Gal KO) were tested for H-substance and Gal expression on splenocytes and endothelial cells by flow cytometric analysis. In addition, the hearts of these mice were perfused ex vivo with 6% human plasma, and the effect on cardiac function was determined. RESULTS AND CONCLUSION: H-substance expression was detected on both splenocytes and endothelial cells of HT-transgenic mice. The level of H-substance expression was not affected by the presence or absence of GT in the cells, consistent with HT being dominant over GT. The ability of HT expression to reduce Gal expression was highly variable depending on the cell type. Gal expression on splenocytes was almost completely eliminated, whereas on endothelial cells, substantial Gal remained despite a 70% reduction. When perfused ex vivo with human plasma, hearts from HT-transgenic, Gal KO, and HT-transgenic/Gal KO mice demonstrated a similar prolongation in survival, compared with wild-type controls. Therefore, as far as hyperacute rejection is concerned, HT expression may be as effective as Gal KO in protecting against xenoantibody and complement mediated injury. However, the effect of residual Gal on non-hyperacute rejection responses remains to be determined.


Assuntos
Fucosiltransferases/metabolismo , Rejeição de Enxerto , Transplante de Coração/imunologia , Lectinas de Plantas , Animais , Antígenos de Superfície/imunologia , Dissacarídeos/imunologia , Dissacarídeos/metabolismo , Endotélio Vascular/enzimologia , Endotélio Vascular/imunologia , Humanos , Lectinas , Camundongos , Camundongos Transgênicos , Miocárdio/citologia , Miocárdio/imunologia , Baço/enzimologia , Baço/imunologia , Transplante Heterólogo , Galactosídeo 2-alfa-L-Fucosiltransferase
16.
BioDrugs ; 9(3): 219-34, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18020562

RESUMO

Xenotransplantation offers an alternative source of organs to solve the current critical shortage of donor organs required for patients with end-stage kidney, heart and liver disease. For social, ethical and logistical purposes, pigs appear to be the most appropriate donor animal. The immunological barriers to xenotransplantation are greater than in allotransplantation because of the presence of preformed natural antibodies in the serum of the recipient. The rapid binding of antibody to donor endothelial cells is followed by complement activation, cell damage and vascular thrombosis. Antirejection therapies aimed at reducing the level of antibody, complement activity and cell-mediated immunity in the recipient may result in a significant increase in complications such as infections and malignancies compared with allotransplantation. Transgenic technology may permit modification of the donor organ, enabling it to evade the rapid antibody- and complement-mediated destruction. The main strategies to prevent xenotransplant rejection have been to reduce expression of 'Gal', the major target epitope for natural antibody, and to inhibit complement activation. Transgenic animals expressing membrane-bound inhibitors of the complement pathway and enzymes that compete for Gal synthesis have been generated. Both approaches provide limited protection, and preliminary experiments in vitro suggest that a combination approach may reduce antibody- and complement-mediated cellular damage.

17.
Trends Cardiovasc Med ; 8(7): 319-25, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14987557

RESUMO

Over the past 20 years, the mortality and morbidity associated with cardiac allotransplantation has fallen significantly, providing a viable treatment for patients with terminal cardiac failure. Unfortunately, the increase in the number of patients who could benefit from cardiac transplantation has not been matched with an increase in the number of organs available for transplantation. Thus, many patients with cardiac failure die waiting for a suitable organ, unlike patients with renal failure, who can be maintained on dialysis while waiting for a kidney. Although the development of artificial hearts may provide a life-sustaining bridging therapy until a donor organ becomes available, the quality of life associated with cardiac prostheses is currently less than that following successful cardiac allotransplantation.

18.
Transplantation ; 64(6): 882-8, 1997 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-9326415

RESUMO

BACKGROUND: Hyperacute rejection (HAR) currently prevents the use of pigs as organ donors for humans. It is now generally accepted that the key instigators of HAR are naturally occurring xenoantibodies against the terminal disaccharide galactose alpha1,3-galactose (Gal), and the species incompatibility between human complement and porcine complement regulatory molecules. Using two in vitro models and an ex vivo mouse heart perfusion model, we have shown previously that cells and tissues from Gal knockout (Gal KO) and transgenic mice expressing the human cell surface complement regulator decay-accelerating factor (DAF/CD55) are partially, but not completely, protected from human complement-mediated injury. METHODS: In the present study, Gal KO mice were crossed with DAF transgenic mice and bred to homozygosity (DAF/Gal KO). Isolated splenocytes were incubated with human serum, and the protective effect of DAF and Gal KO was assessed by measuring complement deposition and cell lysis. Hearts perfused ex vivo with human plasma were examined for human antibody and complement deposition, and assessed functionally by measuring work performed by the heart. RESULTS: Splenocytes from DAF/Gal KO mice were found to be more resistant to complement-mediated injury than cells from either DAF transgenic or Gal KO mice. In addition, hearts from DAF/Gal KO mice, when perfused with human plasma, displayed prolonged survival compared with hearts from Gal KO mice. This was associated with a reduction in the extent of endothelial deposition of IgG, IgM, and complement C3b. CONCLUSIONS: These findings demonstrate that expression of human DAF in association with elimination of the Gal epitope provides added protection from complement-mediated injury in these models of HAR.


Assuntos
Antígenos CD55/biossíntese , Proteínas do Sistema Complemento/toxicidade , Galactosiltransferases/deficiência , Transplante Heterólogo , Animais , Antígenos CD55/genética , Sobrevivência Celular , Células Cultivadas , Complemento C3/metabolismo , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Epitopos , Galactosiltransferases/genética , Homozigoto , Humanos , Linfócitos/citologia , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Miocárdio , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese , Baço/imunologia , Suínos
19.
Transplantation ; 64(2): 197-204, 1997 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-9256173

RESUMO

BACKGROUND: Inactivation of the alpha1,3-galactosyltransferase (GalT) gene by homologous recombination (knockout [KO] mice) and competition for the enzyme's N-acetyllactosamine substrate by transgenically expressed alpha1,2-fucosyltransferase (H-transferase) are two genetic approaches to elimination of the Gal alpha1,3Gal (alphaGal) epitope, which is the major xenoantigen in pigs against which humans have preformed antibodies. Such genetic manipulations often have unpredictable results. METHODS: A panel of 19 selected lectins was used to characterize the changes in cell surface glycosylation in GalT KO and H-transferase transgenic mice, compared with nontransgenic littermate controls. RESULTS: GalT KO mice showed complete elimination of the alphaGal epitope, as reported previously. Surprisingly, however, this was associated with only a modest increase in N-acetyllactosamine residues and had little other effect on the pattern of lectin binding. In contrast, the pattern of lectin binding to H-transferase transgenic mouse cells was more profoundly disturbed and indicated, in addition to the expected expression of H substance and suppression of the alphaGal epitope, that there was a marked reduction in alpha2,3-sialylation and exposure of the normally cryptic antigens, sialylated Tn and Forssman antigens. Similar changes in lectin reactivity with porcine aortic endothelial cells were induced by neuraminidase treatment. CONCLUSIONS: Lectins were able to bind underlying carbohydrate structures (sialylated Tn and Forssman antigens) that are normally cryptic antigens on H-transferase transgenic mouse spleen and cardiac endothelial cells, probably as a consequence of the reduction in the electronegativity of the cell surface due to reduced sialylation. As humans have preformed anti-Tn and anti-Forssman antibodies, it is possible that these structures may become targets of the xenograft rejection process, including hyperacute rejection.


Assuntos
Fucosiltransferases/genética , Galactosiltransferases/genética , Camundongos Knockout/genética , Camundongos Knockout/metabolismo , Camundongos Transgênicos/genética , Camundongos Transgênicos/metabolismo , Transplante Heterólogo/imunologia , Adsorção , Animais , Aorta/metabolismo , Sangue/metabolismo , Sequência de Carboidratos , Membrana Celular/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Epitopos/genética , Galactosiltransferases/imunologia , Glicosilação , Humanos , Imuno-Histoquímica , Lectinas/metabolismo , Camundongos , Miocárdio/química , Miocárdio/citologia , Baço/química , Baço/citologia , Suínos
20.
Transplantation ; 63(7): 1021-5, 1997 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-9112358

RESUMO

BACKGROUND: It is now generally accepted that complement activation is critical for the hyperacute rejection of xenografts. Activation of the classical pathway as the result of the interaction of xenoreactive IgM xenoantibodies with the vascular endothelium has been observed in all species combinations examined to date. A number of studies using a variety of species combinations have also implicated alternate pathway involvement; however, these studies do not enable a conclusion to be drawn as to whether the alternate pathway can be activated in the complete absence of classical pathway activation. METHODS: In this study, human plasma was depleted of both Clq and factor D and then reconstituted with purified Clq or factor D to restore the classical and alternate complement pathways, respectively. The ability of these modified plasmas to prosecute hyperacute rejection was then examined using an ex vivo isolated mouse heart perfusion model based on the Langendorff system. RESULTS AND CONCLUSIONS: In the mouse to human species combination, both the classical and alternate pathways of complement are independently capable of initiating complement activation and mediating xenograft rejection.


Assuntos
Via Alternativa do Complemento/fisiologia , Via Clássica do Complemento/fisiologia , Rejeição de Enxerto/imunologia , Imunologia de Transplantes , Animais , Débito Cardíaco , Frequência Cardíaca , Humanos , Camundongos , Miocárdio , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...