Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Ann Surg ; 274(3): 473-480, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34238812

RESUMEN

OBJECTIVE: Pig-to-primate renal xenotransplantation is plagued by early antibody-mediated graft loss which precludes clinical application of renal xenotransplantation. We evaluated whether temporary complement inhibition with anti-C5 antibody Tesidolumab could minimize the impact of early antibody-mediated rejection in rhesus monkeys receiving pig kidneys receiving costimulatory blockade-based immunosuppression. METHODS: Double (Gal and Sda) and triple xenoantigen (Gal, Sda, and SLA I) pigs were created using CRISPR/Cas. Kidneys from DKO and TKO pigs were transplanted into rhesus monkeys that had the least reactive crossmatches. Recipients received anti-C5 antibody weekly for 70 days, and T cell depletion, anti-CD154, mycophenolic acid, and steroids as baseline immunosuppression (n = 7). Control recipients did not receive anti-C5 therapy (n = 10). RESULTS: Temporary anti-C5 therapy reduced early graft loss secondary to antibody-mediated rejection and improved graft survival (P < 0.01). Deleting class I MHC (SLA I) in donor pigs did not ameliorate early antibody-mediated rejection (table). Anti-C5 therapy did not allow for the use of tacrolimus instead of anti-CD154 (table), prolonging survival to a maximum of 62 days. CONCLUSION: Inhibition of the C5 complement subunit prolongs renal xenotransplant survival in a pig to non-human primate model.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales/farmacología , Rechazo de Injerto/inmunología , Rechazo de Injerto/prevención & control , Inmunosupresores/farmacología , Trasplante de Riñón , Trasplante Heterólogo , Animales , Animales Modificados Genéticamente , Profilaxis Antibiótica , Tolerancia Inmunológica , Macaca mulatta , Modelos Animales , Rituximab/farmacología , Porcinos , Tacrolimus/farmacología
2.
Ann Surg ; 268(4): 564-573, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30048323

RESUMEN

OBJECTIVE: Xenotransplantation using pig organs could end the donor organ shortage for transplantation, but humans have xenoreactive antibodies that cause early graft rejection. Genome editing can eliminate xenoantigens in donor pigs to minimize the impact of these xenoantibodies. Here we determine whether an improved cross-match and chemical immunosuppression could result in prolonged kidney xenograft survival in a pig-to-rhesus preclinical model. METHODS: Double xenoantigen (Gal and Sda) knockout (DKO) pigs were created using CRISPR/Cas. Serum from rhesus monkeys (n = 43) was cross-matched with cells from the DKO pigs. Kidneys from the DKO pigs were transplanted into rhesus monkeys (n = 6) that had the least reactive cross-matches. The rhesus recipients were immunosuppressed with anti-CD4 and anti-CD8 T-cell depletion, anti-CD154, mycophenolic acid, and steroids. RESULTS: Rhesus antibody binding to DKO cells is reduced, but all still have positive CDC and flow cross-match. Three grafts were rejected early at 5, 6, and 6 days. Longer survival was achieved in recipients with survival to 35, 100, and 435 days. Each of the 3 early graft losses was secondary to IgM antibody-mediated rejection. The 435-day graft loss occurred secondary to IgG antibody-mediated rejection. CONCLUSIONS: Reducing xenoantigens in donor pigs and chemical immunosuppression can be used to achieve prolonged renal xenograft survival in a preclinical model, suggesting that if a negative cross-match can be obtained for humans then prolonged survival could be achieved.


Asunto(s)
Antígenos Heterófilos/inmunología , Supervivencia de Injerto/inmunología , Terapia de Inmunosupresión/métodos , Inmunosupresores/farmacología , Trasplante de Riñón , Animales , Animales Modificados Genéticamente , Antígenos Heterófilos/efectos de los fármacos , Modelos Animales de Enfermedad , Quimioterapia Combinada , Supervivencia de Injerto/efectos de los fármacos , Inmunoglobulina M/inmunología , Macaca mulatta , Porcinos , Trasplante Heterólogo
3.
J Surg Res ; 229: 28-40, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29937002

RESUMEN

BACKGROUND: Tools for genome editing in pigs are improving rapidly so that making precise cuts in DNA for the purposes of deleting genes is straightforward. Development of means to replace pig genes with human genes with precision is very desirable for the future development of donor pigs for xenotransplantation. MATERIALS AND METHODS: We used Cas9 to cut pig thrombomodulin (pTHBD) and replace it with a plasmid containing a promoterless antibiotic selection marker and the exon for human thrombomodulin. PhiC31 recombinase was used to remove the antibiotic selection marker to create porcine aortic endothelial cells expressing human instead of pTHBD, driven by the endogenous pig promoter. RESULTS: The promoterless selection cassette permitted efficient enrichment of cells containing correctly inserted transgene. Recombinase treatment of selected cells excised the resistance marker permitting expression of the human transgene by the endogenous pTHBD promoter. Gene regulation was maintained after gene replacement because pig endogenous promoter was kept intact in the correct position. CONCLUSIONS: Cas9 and recombinase technology make orthotopic human for pig gene exchange feasible and pave the way for creation of pigs with human genes that can be expressed in the appropriate tissues preserving gene regulation.


Asunto(s)
Edición Génica/métodos , Porcinos/genética , Trombomodulina/genética , Recolección de Tejidos y Órganos/métodos , Trasplante Heterólogo , Animales , Animales Modificados Genéticamente/genética , Bacteriófagos/genética , Sistemas CRISPR-Cas/genética , Células Cultivadas , Células Endoteliales , Cultivo Primario de Células , Recombinasas/genética , Transfección/métodos , Proteínas Virales/genética
4.
Transgenic Res ; 25(5): 751-9, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27100221

RESUMEN

UNLABELLED: The future of solid organ transplantation is challenged by an increasing shortage of available allografts. Xenotransplantation of genetically modified porcine organs offers an answer to this problem. Strategies of genetic modification have 'humanized' the porcine model towards clinical relevance. Most notably, these approaches have aimed at either antigen reduction or human transgene expression. The object of this study was to evaluate the relative effects of both antigen reduction and direct complement regulation on the human-anti-porcine complement dependent cytotoxicity response. Genetically modified animals were created through CRISPR/Cas9-directed mutation and human transgene delivery. Pigs doubly deficient in GGTA1 and CMAH genes were compared to pigs of the same background that expressed a human complement regulatory protein (hCRP). A third animal was made deficient in GGTA1, CMAH and B4GalNT2 gene expression. Cells from these animals were subjected to measures of human antibody binding and antibody-mediated complement-dependent cytotoxicity by flow cytometry. Human IgG and IgM antibody binding was unchanged between the double knockout and the transgenic hCRP double knockout pig. IgG and IgM binding was reduced by 49.1 and 43.2 % respectively by silencing the B4GalNT2 gene. Compared to the double knockout, human anti-porcine cytotoxicity was reduced by 8 % with the addition of a hCRP (p = .032); It was reduced by 21 % with silencing the B4GalNT2 gene (p = .012). CONCLUSIONS: Silencing the GGTA1, CMAH and B4GalNT2 genes in pigs achieved a significant antigen reduction. Changing the porcine carbohydrate profile effectively mediates human antibody-mediated complement dependent cytoxicity.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Citotoxicidad Inmunológica , Galactosiltransferasas/genética , Oxigenasas de Función Mixta/genética , N-Acetilgalactosaminiltransferasas/genética , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/inmunología , Sistemas CRISPR-Cas/genética , Proteínas del Sistema Complemento/biosíntesis , Proteínas del Sistema Complemento/genética , Regulación de la Expresión Génica , Humanos , Trasplante de Órganos , Porcinos/inmunología , Trasplante Heterólogo
5.
Xenotransplantation ; 23(2): 106-16, 2016 03.
Artículo en Inglés | MEDLINE | ID: mdl-27106872

RESUMEN

BACKGROUND: The Galα(1,3)Gal epitope (α-GAL), created by α-1,3-glycosyltransferase-1 (GGTA1), is a major xenoantigen causing hyperacute rejection in pig-to-primate and pig-to-human xenotransplantation. In response, GGTA1 gene-deleted pigs have been generated. However, it is unclear whether there is a residual small amount of α-Gal epitope expressed in GGTA1(-/-) pigs. Isoglobotrihexosylceramide synthase (iGb3s), another member of the glycosyltransferase family, catalyzes the synthesis of isoglobo-series glycosphingolipids with an α-GAL-terminal disaccharide (iGb3), creating the possibility that iGb3s may be a source of α-GAL epitopes in GGTA1(-/-) animals. The objective of this study was to examine the impact of silencing the iGb3s gene (A3GalT2) on pig-to-primate and pig-to-human immune cross-reactivity by creating and comparing GGTA1(-/-) pigs to GGTA1(-/-) - and A3GalT2(-/-) -double-knockout pigs. METHODS: We used the CRISPR/Cas 9 system to target the GGTA1 and A3GalT2 genes in pigs. Both GGTA1 and A3GalT2 genes are functionally inactive in humans and baboons. CRISPR-treated cells used directly for somatic cell nuclear transfer produced single- and double-gene-knockout piglets in a single pregnancy. Once grown to maturity, the glycosphingolipid profile (including iGb3) was assayed in renal tissue by normal-phase liquid chromatography. In addition, peripheral blood mononuclear cells (PBMCs) were subjected to (i) comparative cross-match cytotoxicity analysis against human and baboon serum and (ii) IB4 staining for α-GAL/iGb3. RESULTS: Silencing of the iGb3s gene significantly modulated the renal glycosphingolipid profile and iGb3 was not detected. Moreover, the human and baboon serum PBMC cytotoxicity and α-GAL/iGb3 staining were unchanged by iGb3s silencing. CONCLUSIONS: Our data suggest that iGb3s is not a contributor to antibody-mediated rejection in pig-to-primate or pig-to-human xenotransplantation. Although iGb3s gene silencing significantly changed the renal glycosphingolipid profile, the effect on Galα3Gal levels, antibody binding, and cytotoxic profiles of baboon and human sera on porcine PBMCs was neutral.


Asunto(s)
Galactosa/metabolismo , Galactosiltransferasas/genética , Rechazo de Injerto/genética , Xenoinjertos/inmunología , Trasplante Heterólogo , Enfermedad Aguda , Animales , Animales Modificados Genéticamente , Sistemas CRISPR-Cas/genética , Galactosiltransferasas/metabolismo , Técnicas de Inactivación de Genes/métodos , Humanos , Leucocitos Mononucleares/inmunología , Papio , Porcinos , Trasplante Heterólogo/métodos
6.
J Surg Res ; 200(2): 698-706, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26375504

RESUMEN

BACKGROUND: Thrombocytopenia may represent a significant challenge to the clinical application of solid-organ xenotransplantation. When studied in a pig-to-primate model, consumptive coagulopathy has challenged renal xenografts. New strategies of genetic manipulation have altered porcine carbohydrate profiles to significantly reduce human antibody binding to pig cells. As this process continues to eliminate immunologic barriers to clinical xenotransplantation, the relationship between human platelets and pig organs must be considered. METHODS: Genetically modified pigs that were created by the CRISPR/Cas9 system with α-1,3-galactosyltransferase (GGTA1)(-/-) or GGTA1(-/-) cytidine monophosphate-N-acetylneuraminic acid hydroxylase(-/-) phenotype, as well as domestic pigs, were used in this study. Autologous porcine platelets were isolated from donor animal blood collection, and human platelets were obtained from a blood bank. Platelets were fluorescently labeled and in a single-pass model, human, or autologous platelets were perfused through porcine organs at a constant concentration and controlled temperature. Platelet uptake was measured by sampling venous output and measuring sample florescence against input florescence. In vitro study of the interaction between human platelets and porcine endothelial cells was accomplished by immunohistochemical stain and confocal microscopy. RESULTS: Differences between human and autologous platelet loss through the porcine kidney were not significant in any genetic background tested (WT P = 0.15, GGTA1(-/-)P = 0.12, GGTA1(-/-) cytidine monophosphate-N-acetylneuraminic acid hydroxylase(-/-)P = 0.25). The unmodified porcine liver consumed human platelets in a single-pass model of platelet perfusion in fewer than 10 min. WT suprahepatic inferior vena cava fluoresce reached a maximum of 76% of input fluoresce within the human platelet cohort and was significantly lower than the autologous platelet control cohort (P = 0.001). Confocal microscopic analysis did not demonstrate a significant association between human platelets and porcine renal endothelial cells compared with porcine liver endothelial positive controls. CONCLUSIONS: Our results suggest that in the absence of immunologic injury, human platelets respond in a variable fashion to organ-specific porcine endothelial surfaces. Human platelets are not removed from circulation by exposure to porcine renal endothelium but are removed by unmodified porcine hepatic endothelium. Kidneys possessing genetic modifications currently relevant to clinical xenotransplantation failed to consume human platelets in an isolated single-pass model. Human platelets did not exhibit significant binding to renal endothelial cells by in vitro assay.


Asunto(s)
Animales Modificados Genéticamente , Plaquetas/inmunología , Trasplante de Riñón/métodos , Complicaciones Posoperatorias/prevención & control , Sus scrofa/genética , Trombocitopenia/prevención & control , Trasplante Heterólogo/métodos , Animales , Plaquetas/metabolismo , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Endotelio/inmunología , Endotelio/metabolismo , Galactosiltransferasas/genética , Galactosiltransferasas/inmunología , Técnicas de Inactivación de Genes , Humanos , Riñón/inmunología , Riñón/metabolismo , Hígado/inmunología , Hígado/metabolismo , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/inmunología , Distribución Aleatoria , Sus scrofa/inmunología , Porcinos , Trombocitopenia/etiología
7.
J Immunol ; 193(11): 5751-7, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25339675

RESUMEN

Pigs are emerging as important large animal models for biomedical research, and they may represent a source of organs for xenotransplantation. The MHC is pivotal to the function of the immune system in health and disease, and it is particularly important in infection and transplant rejection. Pigs deficient in class I MHC could serve as important reagents to study viral immunity as well as allograft and xenograft rejection. In this study, we report the creation and characterization of class I MHC knockout pigs using the Cas9 nuclease and guide RNAs. Pig fetal fibroblasts were genetically engineered using Cas9 and guide RNAs, and class I MHC(-) cells were then used as nuclear donors for somatic cell nuclear transfer. We produced three piglets devoid of all cell surface class I proteins. Although these animals have reduced levels of CD4(-)CD8(+) T cells in peripheral blood, the pigs appear healthy and are developing normally. These pigs are a promising reagent for immunological research.


Asunto(s)
Fibroblastos/fisiología , Técnicas de Silenciamiento del Gen/métodos , Antígenos de Histocompatibilidad Clase I/genética , Porcinos/inmunología , Linfocitos T/inmunología , Trasplante Heterólogo , Virosis/inmunología , Animales , Células Cultivadas , Endonucleasas/metabolismo , Ingeniería Genética , Rechazo de Injerto , Modelos Animales , Técnicas de Transferencia Nuclear , ARN Guía de Kinetoplastida/genética , Porcinos/genética
8.
Xenotransplantation ; 22(3): 194-202, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25728481

RESUMEN

BACKGROUND: Simultaneous inactivation of pig GGTA1 and CMAH genes eliminates carbohydrate xenoantigens recognized by human antibodies. The ß4GalNT2 glycosyltransferase may also synthesize xenoantigens. To further characterize glycan-based species incompatibilities, we examined human and non-human primate antibody binding to cells derived from genetically modified pigs lacking these carbohydrate-modifying genes. METHODS: The Cas9 endonuclease and gRNA were used to create pigs lacking GGTA1, GGTA1/CMAH, or GGTA1/CMAH/ß4GalNT2 genes. Peripheral blood mononuclear cells were isolated from these animals and examined for binding to IgM and IgG from humans, rhesus macaques, and baboons. RESULTS: Cells from GGTA1/CMAH/ß4GalNT2 deficient pigs exhibited reduced human IgM and IgG binding compared to cells lacking both GGTA1 and CMAH. Non-human primate antibody reactivity with cells from the various pigs exhibited a slightly different pattern of reactivity than that seen in humans. Simultaneous inactivation of the GGTA1 and CMAH genes increased non-human primate antibody binding compared to cells lacking either GGTA1 only or to those deficient in GGTA1/CMAH/ß4GalNT2. CONCLUSIONS: Inactivation of the ß4GalNT2 gene reduces human and non-human primate antibody binding resulting in diminished porcine xenoantigenicity. The increased humoral immunity of non-human primates toward GGTA1-/CMAH-deficient cells compared to pigs lacking either GGTA1 or GGTA1/CMAH/ß4GalNT2 highlights the complexities of carbohydrate xenoantigens and suggests potential limitations of the non-human primate model for examining some genetic modifications. The progressive reduction of swine xenoantigens recognized by human immunoglobulin through inactivation of pig GGTA1/CMAH/ß4GalNT2 genes demonstrates that the antibody barrier to xenotransplantation can be minimized by genetic engineering.


Asunto(s)
Antígenos Heterófilos/inmunología , Galactosiltransferasas/genética , Leucocitos Mononucleares/inmunología , Trasplante Heterólogo , Animales , Animales Modificados Genéticamente , Antígenos Heterófilos/genética , Rechazo de Injerto/genética , Rechazo de Injerto/inmunología , Humanos , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Unión Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Porcinos
9.
Xenotransplantation ; 22(1): 20-31, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25178170

RESUMEN

BACKGROUND: Manipulating the pig genome to increase compatibility with human biology may facilitate the clinical application of xenotransplantation. Genetic modifications to pig cells have been made by sequential recombination in fetal fibroblasts and liver-derived cells followed by cross-breeding or somatic cell nuclear transfer. The generation of pigs for research or organ donation by these methods is slow, expensive and requires technical expertise. A novel system incorporating the bacterial nuclease Cas9 and single-guide RNA targeting a 20 nucleotide site within a gene can be expressed from a single plasmid leading to a double-strand break and gene disruption. Coexpression of multiple unique single-guide RNA can modify several genetic loci in a single step. We describe a process for increasing the efficiency of selecting cells with multiple genetic modifications. METHODS: We used the CRISPR/Cas system to target the GGTA1, CMAH and putative iGb3S genes in pigs that have been naturally deleted in humans. Cells lacking galactose α-1,3 galactose (α-Gal) were negatively selected by an IB4 lectin/magnetic bead. α-Gal negative multiplexed single-guide RNA-treated cells were used for somatic cell nuclear transfer (SCNT) and transferred to fertile sows. We examined the levels of α-Gal and Neu5Gc expression of 32 day fetuses and piglets and analyzed the targeted genes by DNA sequencing. RESULTS: Liver-derived cells treated with multiple single-guide RNA and selected for an α-Gal null phenotype were significantly more likely to also carry mutations in simultaneously targeted genes. Multiplex single-guide RNA-treated cells used directly for SCNT without further genetic selection produced piglets with deletions in the targeted genes but also created double- and triple-gene KO variations. CRISPR/Cas-treated cells grew normally and yielded normal liters of healthy piglets via somatic cell nuclear transfer. CONCLUSIONS: The CRISPR/Cas system allows targeting of multiple genes in a single reaction with the potential to create pigs of one genetic strain or multiple genetic modifications in a single pregnancy. The application of this phenotypic selection strategy with multiplexed sgRNA and the Cas9 nuclease has accelerated our ability to produce and evaluate pigs important to xenotransplantation.


Asunto(s)
Sistemas CRISPR-Cas , Galactosiltransferasas/genética , Técnicas de Inactivación de Genes , Oxigenasas de Función Mixta/genética , Técnicas de Transferencia Nuclear , ARN Guía de Kinetoplastida/genética , Sus scrofa/genética , Animales , Antígenos Heterófilos/genética , Biotinilación , Femenino , Eliminación de Gen , Vectores Genéticos , Hepatocitos/citología , Separación Inmunomagnética , Fenotipo , Lectinas de Plantas/metabolismo , Embarazo , Estreptavidina , Porcinos
10.
Xenotransplantation ; 22(3): 203-10, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25728617

RESUMEN

BACKGROUND: The lethal thrombocytopenia that accompanies liver xenotransplantation is a barrier to clinical application. Human platelets are bound by the asialoglycoprotein receptor (ASGR) on pig sinusoidal endothelial cells and phagocytosed. Inactivation of the ASGR1 gene in donor pigs may prevent xenotransplantation-induced thrombocytopenia. METHODS: Transcription activator-like effector nucleases (TALENs) were targeted to the ASGR1 gene in pig liver-derived cells. ASGR1 deficient pig cells were used for somatic cell nuclear transfer (SCNT). ASGR1 knock out (ASGR1-/-) fetal fibroblasts were used to produce healthy ASGR1 knock out piglets. Human platelet uptake was measured in ASGR1+/+ and ASGR1-/- livers. RESULTS: Targeted disruption of the ASGR1 gene with TALENs eliminated expression of the receptor. ASGR1-/- livers phagocytosed fewer human platelets than domestic porcine livers during perfusion. CONCLUSIONS: The use of TALENs in liver-derived cells followed by SCNT enabled the production of healthy homozygous ASGR1 knock out pigs. Livers from ASGR1-/- pigs exhibit decreased human platelet uptake. Deletion of the ASGR1 gene is a viable strategy to diminish platelet destruction in pig-to-human xenotransplantation.


Asunto(s)
Receptor de Asialoglicoproteína/metabolismo , Plaquetas/metabolismo , Hígado/citología , Trasplante Heterólogo , Animales , Receptor de Asialoglicoproteína/genética , Células Endoteliales/metabolismo , Técnicas de Inactivación de Genes/métodos , Hepatocitos/metabolismo , Humanos , Técnicas de Transferencia Nuclear , Porcinos , Trombocitopenia/inmunología
11.
Xenotransplantation ; 21(4): 376-84, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24986655

RESUMEN

BACKGROUND: Pig erythrocytes are potentially useful to solve the worldwide shortage of human blood for transfusion. Domestic pig erythrocytes, however, express antigens that are bound by human preformed antibodies. Advances in genetic engineering have made it possible to rapidly knock out the genes of multiple xenoantigens, namely galactose α1,3 galactose (aGal) and N-glycolylneuraminic acid (Neu5Gc). We have recently targeted the GGTA1 and CMAH genes with zinc finger endonucleases resulting in double knockout pigs that no longer express aGal or Neu5Gc and attract significantly fewer human antibodies. In this study, we characterized erythrocytes from domestic and genetically modified pigs, baboons, chimpanzees, and humans for binding of human and baboon natural antibody, and complement-mediated lysis. METHODS: Distribution of anti-Neu5Gc IgG and IgM in pooled human AB serum was analyzed by ELISA. Erythrocytes from domestic pigs (Dom), aGal knockout pigs (GGTA1 KO), aGal and Neu5Gc double knockout pigs (GGTA1/CMAH KO), baboons, chimpanzees, and humans were analyzed by flow cytometry for aGal and Neu5Gc expression. In vitro comparative analysis of erythrocytes was conducted with pooled human AB serum and baboon serum. Total antibody binding was accessed by hemagglutination; complement-dependent lysis was measured by hemolytic assay; IgG or IgM binding to erythrocytes was characterized by flow cytometry. RESULTS: The pooled human AB serum contained 0.38 µg/ml anti-Neu5Gc IgG and 0.085 µg/ml anti-Neu5Gc IgM. Both Gal and Neu5Gc were not detectable on GGTA1/CMAH KO erythrocytes. Hemagglutination of GGTA1/CMAH KO erythrocytes with human serum was 3.5-fold lower compared with GGTA1 KO erythrocytes, but 1.6-fold greater when agglutinated with baboon serum. Hemolysis of GGTA1/CMAH KO erythrocytes by human serum (25%) was reduced 9-fold compared with GGTA1 KO erythrocytes, but increased 1.64-fold by baboon serum. Human IgG binding was reduced 27-fold on GGTA1/CMAH KO erythrocytes compared with GGTA1 KO erythrocytes, but markedly increased 3-fold by baboon serum IgG. Human IgM binding was decreased 227-fold on GGTA1/CMAH KO erythrocytes compared with GGTA1 KO erythrocytes, but enhanced 5-fold by baboon serum IgM. CONCLUSIONS: Removal of aGal and Neu5Gc antigens from pig erythrocytes significantly reduced human preformed antibody-mediated cytotoxicity but may have complicated future in vivo analysis by enhancing reactivity from baboons. The creation of the GGTA1/CMAH KO pig has provided the xenotransplantation researcher with organs and cells that attract fewer human antibodies than baboon and our closest primate relative, chimpanzee. These finding suggest that while GGTA1/CMAH KO erythrocytes may be useful for human transfusions, in vivo testing in the baboon may not provide a direct transplantation to the clinic.


Asunto(s)
Transfusión de Eritrocitos/métodos , Galactosiltransferasas/deficiencia , Técnicas de Inactivación de Genes/veterinaria , Oxigenasas de Función Mixta/deficiencia , Sus scrofa/sangre , Sus scrofa/genética , Trasplante Heterólogo/métodos , Animales , Animales Modificados Genéticamente , Anticuerpos Heterófilos/sangre , Transfusión de Eritrocitos/efectos adversos , Eritrocitos/inmunología , Galactosiltransferasas/sangre , Galactosiltransferasas/genética , Humanos , Inmunidad Innata , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Oxigenasas de Función Mixta/sangre , Oxigenasas de Función Mixta/genética , Modelos Animales , Papio , Primates , Trasplante Heterólogo/efectos adversos
12.
Medicina (B Aires) ; 74(2): 99-103, 2014.
Artículo en Español | MEDLINE | ID: mdl-24736251

RESUMEN

We compared the use of evidence-based secondary prevention drugs for coronary artery disease at hospital discharge and 3 years of follow-up in a group of patients associated to an integrated network of health services. We conducted a retrospective group study that included 125 patients under 80 years of age who were hospitalized for acute coronary syndrome. McNemar's test was used to compare values at baseline and 3 years. The mean age of of participants was 63.7 years (SD ± 10.08) and 65.6% (95% CI 56.6-73.9) of male sex. The average follow-up time was 2.94 years (SD ± 0.25). The use of secondary prevention drugs for coronary heart disease decreased at 3 years of follow-up: anti-platelet 97.6 to 88.0% (p = 0.012), beta-blockers 94.4 to 84.8% (p = 0.021) and statins 83.7 to 91.2% (p = 0.035). Patients medicated with a combination of anti-platelet, beta blockers and statins showed a decrease from 86.4 to 66.3% (p < 0.0001). It is necessary to study the causes for the decreased adherence to long-term cardio-protective drugs.


Asunto(s)
Síndrome Coronario Agudo/prevención & control , Cumplimiento de la Medicación , Prevención Secundaria , Síndrome Coronario Agudo/tratamiento farmacológico , Antagonistas Adrenérgicos beta/administración & dosificación , Anciano , Inhibidores de la Enzima Convertidora de Angiotensina/administración & dosificación , Estudios de Cohortes , Quimioterapia Combinada , Femenino , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Masculino , Persona de Mediana Edad , Inhibidores de Agregación Plaquetaria/administración & dosificación , Estudios Retrospectivos
13.
Xenotransplantation ; 20(5): 277-91, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24033743

RESUMEN

BACKGROUND: The temporary or long-term xenotransplantation of pig organs into people would save thousands of lives each year if not for the robust human antibody response to pig carbohydrates. Genetically engineered pigs deficient in galactose α1,3 galactose (gene modified: GGTA1) and N-glycolylneuraminic acid (gene modified: CMAH) have significantly improved cell survival when challenged by human antibody and complement in vitro. There remains, however, a significant portion of human antibody binding. METHODS: To uncover additional xenoantigens, we compared the asparagine-linked (N-linked) glycome from serum proteins of humans, domestic pigs, GGTA1 knockout pigs, and GGTA1/CMAH knockout pigs using mass spectrometry. Carbohydrate structures were determined with assistance from GlycoWorkbench, Cartoonist, and SimGlycan software by comparison to existing database entries and collision-induced dissociation fragmentation data. RESULTS: Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS) analysis of reduced and solid-phase permethylated glycans resulted in the detection of high-mannose, hybrid, and complex type N-linked glycans in the 1000-4500 m/z ion range. GGTA1/CMAH knockout pig samples had increased relative amounts of high-mannose, incomplete, and xylosylated N-linked glycans. All pig samples had significantly higher amounts of core and possibly antennae fucosylation. CONCLUSIONS: We provide for the first time a comparison of the serum protein glycomes of the human, domestic pig, and genetically modified pigs important to xenotransplantation.


Asunto(s)
Antígenos Heterófilos/inmunología , Secuencia de Carbohidratos/genética , Ácido N-Acetilneuramínico Citidina Monofosfato/inmunología , Galactosiltransferasas/genética , Polisacáridos/química , Animales , Antígenos Heterófilos/genética , Secuencia de Carbohidratos/fisiología , Carbohidratos/química , Carbohidratos/inmunología , Galactosa/inmunología , Galactosiltransferasas/inmunología , Técnicas de Inactivación de Genes , Humanos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Sus scrofa/inmunología , Porcinos , Trasplante Heterólogo/métodos
14.
Xenotransplantation ; 20(1): 27-35, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23384142

RESUMEN

BACKGROUND: Clinical xenotransplantation is not possible because humans possess antibodies that recognize antigens on the surface of pig cells. Galα-1,3-Gal (Gal) and N-glycolylneuraminic acid (Neu5Gc) are two known xenoantigens. METHODS: We report the homozygous disruption of the α1, 3-galactosyltransferase (GGTA1) and the cytidine monophosphate-N-acetylneuraminic acid hydroxylase (CMAH) genes in liver-derived female pig cells using zinc-finger nucleases (ZFNs). Somatic cell nuclear transfer (SCNT) was used to produce healthy cloned piglets from the genetically modified liver cells. Antibody-binding and antibody-mediated complement-dependent cytotoxicity assays were used to examine the immunoreactivity of pig cells deficient in Neu5Gc and Gal. RESULTS: This approach enabled rapid production of a pig strain deficient in multiple genes without extensive breeding protocols. Immune recognition studies showed that pigs lacking both CMAH and GGTA1 gene activities reduce the humoral barrier to xenotransplantation, further than pigs lacking only GGTA1. CONCLUSIONS: This technology will accelerate the development of pigs for xenotransplantation research.


Asunto(s)
Disacáridos/inmunología , Ácidos Neuramínicos/inmunología , Sus scrofa/genética , Sus scrofa/inmunología , Trasplante Heterólogo/inmunología , Animales , Anticuerpos Heterófilos/metabolismo , Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos Heterófilos/inmunología , Antígenos Heterófilos/metabolismo , Secuencia de Bases , Células Cultivadas , ADN/genética , Disacáridos/deficiencia , Femenino , Galactosiltransferasas/deficiencia , Galactosiltransferasas/genética , Técnicas de Inactivación de Genes/métodos , Humanos , Leucocitos Mononucleares/inmunología , Oxigenasas de Función Mixta/deficiencia , Oxigenasas de Función Mixta/genética , Ácidos Neuramínicos/metabolismo , Sus scrofa/metabolismo
15.
J Surg Res ; 181(1): e39-45, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22795272

RESUMEN

BACKGROUND: Genetic modification of the pig has been hampered by inefficiency of homologous recombination and unavailability of pig embryonic stem cells. Engineered zinc finger nuclease (ZFN)-mediated genetic modification in somatic cells combined with somatic cell nuclear transfer (SCNT) technology provides a new approach for targeted modification in pig genome. In this study, we used a ZFN pair to disrupt porcine α-1,3, galactosyltransferase (GGTA1) gene in liver-derived cells (LDC). ZFN-treated LDC were used as nuclear donors to produce fetuses and piglets via SCNT. All cloned fetuses and piglets showed biallelic knockout of GGTA1 gene. MATERIALS AND METHODS: A ZFN pair was designed to target exon 8 of pig GGTA1 gene. LDC were transfected with GGTA1 ZFN plasmids. SURVEYOR assay was used to evaluate the ZFN activity in LDC. GGTA1 gene knockout cells (GTKO) were obtained by counter-selection and used as nuclear donors for SCNT. The cloned fetuses and piglets were characterized by DNA sequencing. Expression of α-Gal epitope was further examined by flow cytometry and confocal microscopy. RESULTS: SURVEYOR assay revealed 6.48% ZFN activity in LDC. GTKO cells were obtained by counter-selection 10 d after ZFN transfection. A total of six fetuses and 13 piglets were produced by SCNT. All fetuses and piglets had biallelic mutations in the ZFN targeted region and were negative for α-Gal epitope. CONCLUSIONS: Biallelic GGTA1 gene disruption in LDC was generated efficiently by ZFN. GTKO fetuses were produced from ZFN-treated LDC by SCNT. GTKO piglets were obtained by SCNT of ZFN-treated LDC or recloning of fetal fibroblasts from GTKO fetuses. With longer lifespan and robust growth rate, LDC has the potential to endure multiple genetic modifications in vitro without going to SCNT, which could accelerate the production of genetically modified pig organs for xenotransplantation.


Asunto(s)
Endonucleasas/genética , Galactosiltransferasas/genética , Técnicas de Inactivación de Genes/métodos , Hígado/citología , Técnicas de Transferencia Nuclear , Dedos de Zinc , Animales , Porcinos , Trasplante Heterólogo
16.
J Surg Res ; 184(2): e37-42, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23890396

RESUMEN

BACKGROUND: The process of selecting transgenic cells has been one of the bottlenecks in the generation of transgenic animals by somatic cell nuclear transfer (SCNT). In particular, selection for the Gal double-knockout (Gal-DKO) genotype has been time consuming and inefficient. The objective of this work was to generate a highly efficient system to select Gal-DKO cells to be used in SCNT without affecting the efficiency in production of Gal-null pigs. MATERIALS AND METHODS: Fetal liver-derived cells deficient in Gal-expression were initially selected by fluorescence-activated cell sorting (FACS) using IB4 conjugated to a fluorescent dye. Cells recovered by FACS were cultured and expanded, followed by a second round of selection using streptavidin magnetic beads and IB4 lectin biotin. RESULTS: Recovery efficiency of target cells was 0.04% for the first selection using FACS and 0.3% for the second round by magnetic beads. Full reprogramming was obtained on selected Gal-DKO cells after FACS and magnetic beads selection, when used for SCNT to produce the Gal-null piglets. Cells obtained from magnetic beads developed 48 colonies; the Gal-null genotype was found in 44 of them (91.7%). Three of these colonies were used to generate piglets by SCNT. From three recipients receiving embryos, two became pregnant and produced 17 piglets, all of them DKO. CONCLUSIONS: Sequential selection of Gal-DKO cells by FACS/magnetic beads is a highly efficient system to generate null cells. Selected cells were successfully used to generate healthy double-knockout piglets by SCNT.


Asunto(s)
Galactosiltransferasas/deficiencia , Hígado/citología , Técnicas de Transferencia Nuclear , Animales , Animales Modificados Genéticamente , Células Cultivadas , Citometría de Flujo , Galactosiltransferasas/genética , Hígado/embriología , Magnetismo , Modelos Animales , Porcinos
17.
Xenotransplantation ; 19(2): 92-101, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22497511

RESUMEN

BACKGROUND: Human preformed antibodies continue to recognize porcine xenografts, despite the advent of α-galactosyltransferase knockout (GTKO) pigs. This study examined the potential reactivity of human preformed IgG and IgM antibodies toward antigens in the GTKO pig liver. METHODS: Human serum was analyzed for the concentration of IgG, IgM, anti-αgal antibody, anti-non-αgal antibody and cytotoxicity toward domestic and GTKO fibroblasts and liver sinusoidal endothelial cells (LSEC). We detected preformed antibodies in human serum directed toward GTKO pig liver cells and tissue samples using advanced proteomic techniques. The targets of preformed antibodies were identified by MALDI TOF TOF mass spectrometry and validated by confocal microscopy, immunoblot, and immunoprecipitation. RESULTS: Human serum used in this study contained 2.06 µg/ml IgG and 0.013 µg/ml IgM directed toward GTKO fibroblasts. Human IgG and IgM bound to GTKO LSEC in a dose-dependent manner and were cytotoxic. We detected 357 protein spots recognized by human IgG and 233 by human IgM. Two hundred and nineteen proteins were common to both human IgG and IgM. Mass spectrometry identified numerous immunoreactive proteins, of which 19 were membrane proteins on liver cells. The most significant to this study were α-enolase, CFTR, and E-cadherin, which were abundant in GTKO pig tissues and expressed on the surface of GTKO LSEC. Human IgG captured α-enolase, CFTR, and E-cadherin by immunoprecipitation validating the proteomic identification. CONCLUSION: These experiments indicate that several membrane antigens in GTKO pigs could be recognized directly by human IgG or IgM. Further studies on the contribution of these antigens to antibody-mediated xenograft rejection are necessary.


Asunto(s)
Anticuerpos Heterófilos/sangre , Galactosiltransferasas/deficiencia , Galactosiltransferasas/genética , Sus scrofa/genética , Sus scrofa/inmunología , Animales , Animales Modificados Genéticamente , Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos Heterófilos , Proteínas del Sistema Complemento/metabolismo , Células Endoteliales/inmunología , Galactosiltransferasas/inmunología , Técnicas de Inactivación de Genes , Rechazo de Injerto/etiología , Rechazo de Injerto/inmunología , Hepatocitos/inmunología , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Proteómica , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Trasplante Heterólogo/efectos adversos , Trasplante Heterólogo/inmunología , Trisacáridos/inmunología
18.
Medicina (B Aires) ; 82(2): 192-199, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35417382

RESUMEN

The COVID-19 pandemic elicited important changes in community habits and behaviors, including a distancing of people from the healthcare system. The objective of this work was to understand the causes that gave rise to changes in behavior from an individual perspective in the Argentine Republic. We performed a cross-sectional, web-based survey using an online questionnaire. The survey was distributed via the WhatsApp application for smartphones among subjects across the Argentine territory using a combination of convenience and snowball sampling. We received 6176 responses. Almost 70% of respondents manifested fear of visiting a physician. One third of respondents manifested having a desire or need in at least one occasion of visiting a physician but, of these, 48% avoided it. The main reasons for avoiding visits were: 1) a recommendation of staying home (40%); 2) lack of access to the physician (35%); and 3) fear of contagion (26%). The most common form of consultation was through unconventional means (e-mail, telephone, or WhatsApp). One of 5 respondents had difficulties to obtain a medication prescription and 5% stopped the use of at least one medication. Regarding healthy habits, almost 2/3 of those surveyed stated that they became more sedentary; 11% of hypertensive patients increased their consumption of salt and 15% saw their blood pressure values increase, while 16% of dyslipidemic patients showed increased consumption of fats.


La pandemia de COVID-19 determinó un importante cambio de los hábitos y comportamientos comunitarios, entre ellos se observó un distanciamiento de la gente del sistema de salud y el abandono de hábitos saludables. El objetivo de este trabajo fue comprender las causas que dieron lugar a dichos cambios de comportamiento desde una perspectiva individual y evaluar el impacto en el control de los factores de riesgo cardiovasculares. Realizamos una encuesta utilizando un cuestionario en línea y distribuida mediante la aplicación WhatsApp entre personas de la Argentina utilizando una combinación de muestreo por conveniencia y en "bola de nieve". Recibimos 6176 respuestas, casi el 70% de los encuestados manifestaron temor de realizar una consulta médica. Un tercio de los encuestados manifestó haber tenido necesidad de ir al médico en al menos una oportunidad desde el inicio de la pandemia, pero de éstos, el 48% evitó la visita. Las principales razones para evitarla: 1) la recomendación de quedarse en casa (40%); 2) difi cultad para acceder al sistema de salud (35%); y 3) miedo al contagio (26%). La forma de consulta más común fue a través de medios no convencionales (correo electrónico, teléfono o WhatsApp). Uno de cada 5 encuestados tuvo dificultades para obtener una receta y el 5% dejó de tomar al menos un medicamento. En cuanto a los hábitos saludables, casi 2/3 de los encuestados afirmaron volverse más sedentarios, el 11% de los hipertensos aumentó su consumo de sal y el 15% aumentó sus valores de presión arterial, mientras que el 16% de los dislipidémicos mostró un mayor consumo de grasas.


Asunto(s)
COVID-19 , COVID-19/epidemiología , Estudios Transversales , Atención a la Salud , Humanos , Pandemias , SARS-CoV-2
19.
Xenotransplantation ; 18(4): 245-51, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21848542

RESUMEN

BACKGROUND: Porcine liver xenografts represent a potential solution to the organ shortage, but thrombocytopenia occurs within minutes to hours after xenotransplantation, preventing clinical application. Recently, it was discovered that porcine liver sinusoidal endothelial cells (LSEC) bind and phagocytose human platelets. We examined the role of ASGR1 in binding and removing human platelets by the pig liver endothelium. METHODS: Primary porcine enriched LSEC (eLSEC) were characterized by flow cytometry, immunoblot, quantitative PCR, and immunohistochemistry using confocal microscopy. Phagocytosis inhibition assays using anti-ASGR1 and an ASGR1 substrate were performed. ASGR1 was targeted for siRNA knockdown, and ASGR1-reduced cells were tested for human platelet binding and phagocytosis. RESULTS: ASGR1 is expressed by eLSEC. Human platelet binding and phagocytosis by porcine eLSEC was inhibited by asialofetuin, but not fetuin, suggesting an interaction with galactose ß1-4 N-acetyl glucosamine. Anti-ASGR1 antibodies inhibited human platelet binding in a dose-dependent manner. Knockdown experiments using siRNA reduced ASGR1 expression in asynchronous primary eLSEC by 40%-80%. There was a 20% reduction in translated protein significantly correlated with a 21% decrease in human platelet binding. CONCLUSIONS: ASGR1 on porcine eLSEC mediates phagocytosis of xenogeneic platelets.


Asunto(s)
Receptor de Asialoglicoproteína/metabolismo , Plaquetas/metabolismo , Células Endoteliales/metabolismo , Hígado/citología , Fagocitosis/fisiología , Trasplante Heterólogo , Animales , Receptor de Asialoglicoproteína/genética , Células Cultivadas , Humanos , Transfusión de Plaquetas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Porcinos
20.
Transgenic Res ; 19(5): 869-87, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20099029

RESUMEN

This study was undertaken to characterize the effects of constitutive expression of the hedgehog transcriptional activator, Gli2, in porcine skin. The keratinocyte-specific human transgene, K5-hGli2 Delta N, was used to produce transgenic porcine lines via somatic cell nuclear transfer techniques. In mice, K5-hGli2 Delta N induces epithelial downgrowths resembling basal cell carcinomas. Our porcine model also developed these basal cell carcinoma-like lesions, however gross tumor development was not appreciated. In contrast to the murine model, diffuse epidermal changes as well as susceptibility to cutaneous infections were seen in the swine model. Histologic analysis of transgenic piglets revealed generalized epidermal changes including: epidermal hyperplasia (acanthosis), elongated rete ridges, parakeratotic hyperkeratosis, epidermal neutrophilic infiltration, capillary loop dilation and hypogranulosis. By 2 weeks of age, the transgenic piglets developed erythematic and edematous lesions at high contact epidermal areas and extensor surfaces of distal limb joints. Despite antibiotic treatment, these lesions progressed to a deep bacterial pyoderma and pigs died or were euthanized within weeks of birth. Non-transgenic littermates were phenotypically normal by gross and histological analysis. In summary, constitutive expression of the human hGli2 Delta N in keratinocytes, results in cutaneous changes that have not been reported in the K5-hGli2 Delta N murine model. These findings indicate a need for a multiple species animal model approach in order to better understand the role of Gli2 in mammalian skin.


Asunto(s)
Animales Modificados Genéticamente , Epidermis/patología , Factores de Transcripción de Tipo Kruppel/fisiología , Proteínas Nucleares/fisiología , Enfermedades Cutáneas Infecciosas/etiología , Porcinos/metabolismo , Animales , Animales Modificados Genéticamente/genética , Animales Lactantes , Epidermis/metabolismo , Femenino , Fibroblastos/metabolismo , Predisposición Genética a la Enfermedad , Folículo Piloso/metabolismo , Humanos , Queratinas/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Ratones , Proteínas Nucleares/genética , Fenotipo , Piodermia/etiología , Proteínas Recombinantes de Fusión/fisiología , Enfermedades Cutáneas Infecciosas/patología , Especificidad de la Especie , Sus scrofa , Transgenes , Proteína Gli2 con Dedos de Zinc
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA