Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Trixie A. Shinkel is active.

Publication


Featured researches published by Trixie A. Shinkel.


Transplantation | 1996

The alpha-1,3-galactosyltransferase knockout mouse. Implications for xenotransplantation.

Rick G. Tearle; Margaret J. Tange; Zara L. Zannettino; Marina Katerelos; Trixie A. Shinkel; Bryce J. W. van Denderen; Andrew Lonie; Ian Lyons; Mark B. Nottle; Timothy Cox; Christiane Becker; Anita M. Peura; Peter L. Wigley; Robert J. Crawford; Allan J. Robins; Martin J. Pearse; Anthony J. F. d'Apice

Organ xenografts in discordant combinations such as pig-to-man undergo hyperacute rejection due to the presence of naturally occurring human anti-pig xenoantibodies. The galactose alpha(1,3)-galactose epitope on glycolipids and glycoproteins is the major porcine xenoantigen recognized by these xenoantibodies. This epitope is formed by alpha(1,3)-galactosyltransferase, which is present in all mammals except man, apes, and Old World monkeys. We have generated mice lacking this major xenoantigen by inactivating the alpha(1,3)-galactosyltransferase gene. These mice are viable and have normal organs but develop cataracts. Substantially less xenoantibody from human serum binds to cells and tissues of these mice compared with normal mice. Similarly, there is less activation of human complement on cells from mice lacking the galactose alpha(1,3)-galactose epitope. These mice confirm the importance of the galactose alpha(1,3)-galactose epitope in human xenoreactivity and the logic of continuing efforts to generate pigs that lack this epitope as a source of donor organs.


Transplantation | 2000

Renal xenografts from triple-transgenic pigs are not hyperacutely rejected but cause coagulopathy in non-immunosuppressed baboons.

Peter J. Cowan; Atousa Aminian; Helen Barlow; A. A. Brown; C.G Chen; Nella Fisicaro; David M. A. Francis; David J. Goodman; Wenruo Han; Margarita Kurek; Mark B. Nottle; Martin J. Pearse; Evelyn Salvaris; Trixie A. Shinkel; G. V. Stainsby; A. B. Stewart; Anthony J. F. d'Apice

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.


Transplantation | 1997

Changes In Cell Surface Glycosylation In α1,3-galactosyltransferase Knockout And α1,2-fucosyltransferase Transgenic Mice

Trixie A. Shinkel; Chao-Guang Chen; Evelyn Salvaris; Timothy R. Henion; Helen Barlow; Uri Galili; Martin Pearse; Anthony J. F. d'Apice

BACKGROUND Inactivation of the alpha1,3-galactosyltransferase (GalT) gene by homologous recombination (knockout [KO] mice) and competition for the enzymes 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.


American Journal of Transplantation | 2002

Protective Effects of Recombinant Human Antithrombin III in Pig-to-Primate Renal Xenotransplantation

Peter J. Cowan; Atousa Aminian; Helen Barlow; A. A. Brown; Karen M. Dwyer; Robin Filshie; Nella Fisicaro; David M. A. Francis; H. Gock; David J. Goodman; J. Katsoulis; Simon C. Robson; Evelyn Salvaris; Trixie A. Shinkel; A. B. Stewart; Anthony J. F. d'Apice

Delayed rejection of pig kidney xenografts by primates is associated with vascular injury that may be accompanied by a form of consumptive coagulopathy in recipients. Using a life‐supporting pig‐to‐baboon renal xenotransplantation model, we have tested the hypothesis that treatment with recombinant human antithrombin III would prevent or at least delay the onset of rejection and coagulopathy. Non‐immunosuppressed baboons were transplanted with transgenic pig kidneys expressing the human complement regulators CD55 and CD59. Recipients were treated with an intravenous infusion of antithrombin III eight hourly (250 units per kg body weight), with or without low molecular weight heparin. Antithrombin‐treated recipients had preservation of normal renal function for 4–5 days, which was twice as long as untreated animals, and developed neither thrombocytopenia nor significant coagulopathy during this period. Thus, recombinant antithrombin III may be a useful therapeutic agent to ameliorate both early graft damage and the development of systemic coagulation disorders in pig‐to‐human xenotransplantation.


Transplantation | 1998

Anti-gal Antibody-mediated Allograft Rejection In α1,3-galactosyltransferase Gene Knockout Mice: A Model of Delayed Xenograft Rejection

Martin J. Pearse; Ewa Witort; Patricia L. Mottram; Wenruo Han; Lisa Murray-Segal; Margarita Romanella; Evelyn Salvaris; Trixie A. Shinkel; David J. Goodman; Anthony J. F. d'Apice

Background. The key role of anti-galactosea1,3-galactose (anti-aGal) 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-aGal 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 a1,3-galactosyltransferase knockout (Gal KO) mice, which like humans develop antibodies to the disaccharide galactosea1,3-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-aGal 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-aGal antibody in graft rejection can be studied in isolation from other rejection mechanisms. The titer of antiaGal 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.


Transplantation | 1998

Knock out of alpha1,3-galactosyltransferase or expression of alpha1,2-fucosyltransferase further protects CD55- and CD59-expressing mouse hearts in an ex vivo model of xenograft rejection.

Peter J. Cowan; Chao-Guang Chen; Trixie A. Shinkel; Nella Fisicaro; Evelyn Salvaris; Atousa Aminian; Margarita Romanella; Martin Pearse; Anthony J. F. d'Apice

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.


Xenotransplantation | 1996

Reduction in Gal-α1,3-Gal epitope expression in transgenic mice expressing human H-transferase

Chao-Guang Chen; Nella Fisicaro; Trixie A. Shinkel; Victoria Aitken; Marina Katerelos; Bryce J. W. van Denderen; Margaret J. Tange; Robert J. Crawford; Allan J. Robins; Martin J. Pearse; Anthony J. F. d'Apice

Abstract: There is now considerable evidence implicating anti‐Gal xenoantibodies as a key instigator in the hyperacute rejection of discordant xenografts. As a consequence it is generally held that elimination or reduction of the Gal/anti‐Gal component is critical to overcoming hyperacute rejection. We have recently shown that in mice inactivation of the GalT gene by homologous recombination completely eliminates the expression of the Gal‐epitope and that hearts from these mice demonstrate prolonged survival when perfused ex vivo with human plasma. Unfortunately this strategy is currently not feasible in pigs because the technology to isolate porcine embryonic stem cells, which are critical for homologous recombination, is not yet available. This study investigates an alternative competition‐based transgenic strategy to suppress the level of the Gal epitope by expression of H‐transferase (α1,2‐fucosyltransferase) an enzyme which has the same substrate specificity (lactobiose) as α1,3‐galactosyltransferase. In vitro transfection of murine cells with H‐transferase reduced Gal‐epitope expression by 80–90%. A similar reduction in Gal expression was observed on PBL and thymocytes from H‐transferase transgenic mice. This reduction in Gal epitope expression resulted in a marked reduction in the reactivity of these cells with human serum. In tissues from these mice the reduction in Gal expression was inversely proportional to the endogenous level of Gal. The results of this study support pursuing this strategy as a means to reduce the xenoantigenicity of porcine tissues.


Xenotransplantation | 2004

Enhanced expression of glutathione peroxidase protects islet beta cells from hypoxia-reoxygenation.

Diana A. Lepore; Trixie A. Shinkel; Nella Fisicaro; Tharun B. Mysore; Lucinda E. A. Johnson; Anthony J. F. d'Apice; Peter J. Cowan

Abstract:  The survival of pancreatic islet β–cell xenografts and allografts may be affected by damaging reactive oxygen and nitrogen species generated during hypoxia‐reoxygenation. Peroxynitrite, which is formed from superoxide and nitric oxide, appears to be an important mediator of β‐cell destruction. The intracellular antioxidant enzymes glutathione peroxidase‐1 (Gpx‐1) and copper–zinc superoxide dismutase (CuZn SOD) detoxify peroxynitrite and superoxide, respectively. The aim of this study was to examine whether enhanced expression of Gpx‐1 and/or CuZn SOD protected NIT‐1 mouse insulinoma cells from hypoxia–reoxygenation injury. Stable transfectants expressing human Gpx‐1 or CuZn SOD were isolated and tested for their resistance to hydrogen peroxide (H2O2) and menadione, which generates superoxide intracellularly. Clones expressing one or both enzymes were subjected to hypoxia in glucose‐free medium for 18 h, followed by reoxygenation in complete medium for 1.5 h. Cell viability was measured using the 3‐(4,5‐dimethylthiazol‐2‐yl)‐2,5‐diphenyltetrazoliumbromide (MTT) reduction assay. Increases of up to two fold in Gpx or total SOD activity protected NIT‐1 cells from H2O2 and menadione. Expression of Gpx‐1 significantly increased NIT‐1 survival following hypoxia‐reoxygenation (viability 65 ± 9% vs. control 15 ± 3%, P < 0.001) but CuZn SOD expression had no effect (15 ± 1%). Expression of both enzymes was no more protective (60 ± 6%) than expression of Gpx‐1 alone. Genetic manipulation of islet β cells to increase expression of Gpx‐1 may protect them from oxidative injury associated with the transplantation procedure.


Xenotransplantation | 2003

Targeting gene expression to endothelium in transgenic animals: a comparison of the human ICAM-2, PECAM-1 and endoglin promoters.

Peter J. Cowan; Trixie A. Shinkel; Nella Fisicaro; James W. Godwin; Carmelo Bernabeu; Nuria Almendro; Carlos Rius; Andrew Lonie; Mark B. Nottle; Peter L. Wigley; Kathy Paizis; Martin J. Pearse; Anthony J. F. d'Apice

Abstract: It is highly likely that successful pig‐to‐human xenotransplantation of vascularized organs will require genetic modification of the donor pig, and in particular of donor vascular endothelium. Promoters are generally tested in transgenic mice before generating transgenic pigs. Several promoters have been used to drive endothelial cell‐specific expression in mice but none have yet been tested in pigs. We compared the promoters of three human genes that are predominantly expressed in vascular endothelium: intercellular adhesion molecule 2 (ICAM‐2), platelet endothelial cell adhesion molecule 1 (PECAM‐1) and endoglin. Expression of human complement regulatory proteins (hCRPs), directed by each of the promoters in mice, was largely restricted to vascular endothelium and leukocyte subpopulations. However, expression from the PECAM‐1 promoter was weak in liver and non‐uniform in the small vessels of heart, kidney, and lung. Conversely, expression from the endoglin promoter was consistently strong in the small vessels of these organs but was absent in larger vessels. The ICAM‐2 promoter, which produced strong and uniform endothelial expression in all organs examined, was therefore used to generate hCRP transgenic pigs. Leukocytes from 57 pigs containing at least one intact transgene were tested for transgene expression by flow cytometry. Forty‐seven of these transgenic pigs were further analyzed by immunohistochemical staining of liver biopsies, and 18 by staining of heart and kidney sections. Only two of the pigs showed expression, which appeared to be restricted to vascular endothelium in heart and kidney but was markedly weaker than in transgenic mice produced with the same batch of DNA. Thus, in this case, promoter performance in mice and pigs was not equivalent. The weak expression driven by the human ICAM‐2 promoter in pigs relative to mice suggests the need for additional regulatory elements to achieve species‐specific gene expression in pigs.


Transplantation | 1996

Expression of functional decay-accelerating factor (CD55) in transgenic mice protects against human complement-mediated attack.

B. J. W. Van Denderen; Martin J. Pearse; Marina Katerelos; Mark B. Nottle; Z.-T. Du; Atousa Aminian; W. R. Adam; A. Shenoy-Scaria; D. M. Lublin; Trixie A. Shinkel; Anthony J. F. d'Apice

Transgenic mice expressing human CD55 were generated by microinjection of a CD55-minigene under the control of the mouse H2K(b) (MHC class I) promoter. Offspring were tested for transgene integration by PCR analysis, and for CD55 expression on peripheral blood leukocytes (PBLs) by flow cytometry. Expression levels of 15 founders ranged from 30 to 80% of that on human neutrophils. Immunohistochemical analysis of kidney, heart, liver, and lung tissue demonstrated staining for CD55 on endothelial surfaces as well as general diffuse staining throughout the tissues. The capacity of the transgenically expressed CD55 to prevent human C3 deposition on the surface of mouse splenocytes was assessed by flow cytometry. Cells from hemizygous mice incubated with 10% fresh human serum as a source of natural antibody and complement bound approximately 65% less C3 than control littermates. No further protection was seen using cells from homozygous littermates, and the protective effect was abrogated by prior incubation with an OFFi-CD55 monoclonal antibody. Similarly, transgenic mice were afforded significant protection from human serum-mediated lysis, determined using an LDH release assay. Hearts perfused with human plasma showed no increase in survival time in a modified Langendorff perfusion system, however deposition of human C3c was greatly reduced in transgenic hearts.

Collaboration


Dive into the Trixie A. Shinkel's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter J. Cowan

St. Vincent's Health System

View shared research outputs
Top Co-Authors

Avatar

Martin J. Pearse

St. Vincent's Health System

View shared research outputs
Top Co-Authors

Avatar

Atousa Aminian

St. Vincent's Health System

View shared research outputs
Top Co-Authors

Avatar

Nella Fisicaro

St. Vincent's Health System

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Helen Barlow

St. Vincent's Health System

View shared research outputs
Top Co-Authors

Avatar

David J. Goodman

St. Vincent's Health System

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge