Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Xenotransplantation ; 22(3): 203-10, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25728617

RESUMO

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.


Assuntos
Receptor de Asialoglicoproteína/metabolismo , Plaquetas/metabolismo , Fígado/citologia , Transplante Heterólogo , Animais , Receptor de Asialoglicoproteína/genética , Células Endoteliais/metabolismo , Técnicas de Inativação de Genes/métodos , Hepatócitos/metabolismo , Humanos , Técnicas de Transferência Nuclear , Suínos , Trombocitopenia/imunologia
2.
Xenotransplantation ; 22(1): 20-31, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25178170

RESUMO

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.


Assuntos
Sistemas CRISPR-Cas , Galactosiltransferases/genética , Técnicas de Inativação de Genes , Oxigenases de Função Mista/genética , Técnicas de Transferência Nuclear , RNA Guia de Cinetoplastídeos/genética , Sus scrofa/genética , Animais , Antígenos Heterófilos/genética , Biotinilação , Feminino , Deleção de Genes , Vetores Genéticos , Hepatócitos/citologia , Separação Imunomagnética , Fenótipo , Lectinas de Plantas/metabolismo , Gravidez , Estreptavidina , Suínos
3.
Xenotransplantation ; 20(1): 27-35, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23384142

RESUMO

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.


Assuntos
Dissacarídeos/imunologia , Ácidos Neuramínicos/imunologia , Sus scrofa/genética , Sus scrofa/imunologia , Transplante Heterólogo/imunologia , Animais , Anticorpos Heterófilos/metabolismo , Citotoxicidade Celular Dependente de Anticorpos , Antígenos Heterófilos/imunologia , Antígenos Heterófilos/metabolismo , Sequência de Bases , Células Cultivadas , DNA/genética , Dissacarídeos/deficiência , Feminino , Galactosiltransferases/deficiência , Galactosiltransferases/genética , Técnicas de Inativação de Genes/métodos , Humanos , Leucócitos Mononucleares/imunologia , Oxigenases de Função Mista/deficiência , Oxigenases de Função Mista/genética , Ácidos Neuramínicos/metabolismo , Sus scrofa/metabolismo
4.
J Surg Res ; 184(2): 1123-33, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23673165

RESUMO

BACKGROUND: Antibody-mediated rejection continues to be an obstacle for xenotransplantation despite development of α1,3-galactosyltransferase knockout (GTKO) pigs. Fibronectin (Fn) from GTKO pigs was identified as a xenoantigen in baboons. N-glycolylneuraminic acid (Neu5Gc), similar to galactose α1,3-galactose, is an antigenic carbohydrate found in pigs. We evaluated human antibody reactivity and performed initial antigenic epitope characterization of Fn from GTKO pigs. MATERIALS AND METHODS: GTKO pig aortic endothelial cells (AEC) were isolated and assessed for antibody-mediated complement-dependent cytotoxicity (CDC). Human and GTKO pig Fn were purified and analyzed using immunoblots. GTKO pig and human AEC absorbed human sera were assessed for CDC and anti-GTKO pig Fn antibodies. GTKO pig proteins were assessed for Neu5Gc. Immunoaffinity-purified human IgG anti-GTKO pig (hIgG-GTKOp) Fn using a GTKO pig Fn column were evaluated for cross-reactivity with other proteins. RESULTS: GTKO pig AEC had greater human antibody binding, complement deposition and CDC compared with allogeneic human AEC. Human sera absorbed with GTKO pig AEC resulted in diminished anti-GTKO pig Fn antibody. Neu5Gc was identified on GTKO pig Fn and other proteins. The hIgG-GTKOp Fn cross-reacted with multiple GTKO pig proteins and was enriched with anti-Neu5Gc antibody. CONCLUSIONS: Removal of antigenic epitopes from GTKO pig AEC would improve xenograft compatibility. GTKO pig Fn has antigenic epitopes, one identified as Neu5Gc, which may be responsible for pathology and cross-reactivity of hIgG-GTKOp Fn. Genetic knockout of Neu5Gc appears necessary to address significance and identification of non-Neu5Gc GTKO pig Fn antigenic epitopes.


Assuntos
Antígenos Heterófilos/imunologia , Fibronectinas/imunologia , Galactosiltransferases/deficiência , Galactosiltransferases/genética , Suínos/imunologia , Transplante Heterólogo , Animais , Animais Geneticamente Modificados , Anticorpos/imunologia , Aorta/citologia , Aorta/imunologia , Células Cultivadas , Reações Cruzadas/imunologia , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Epitopos/imunologia , Técnicas de Inativação de Genes , Humanos , Modelos Animais , Suínos/genética
5.
Xenotransplantation ; 18(4): 245-51, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21848542

RESUMO

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.


Assuntos
Receptor de Asialoglicoproteína/metabolismo , Plaquetas/metabolismo , Células Endoteliais/metabolismo , Fígado/citologia , Fagocitose/fisiologia , Transplante Heterólogo , Animais , Receptor de Asialoglicoproteína/genética , Células Cultivadas , Humanos , Transfusão de Plaquetas , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Suínos
6.
Xenotransplantation ; 17(5): 350-61, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20955292

RESUMO

BACKGROUND: Pig liver xenotransplantation could offset the shortage of livers available for orthotopic liver transplantation. Studies in pig and baboon liver xenografts revealed the main obstacle to be a lethal thrombocytopenia that occurred within minutes to hours of transplantation. METHODS: We have created a model of xenotransplantation-induced thrombocytopenia using ex vivo pig liver perfusion with human platelets. Thrombocytopenia was examined using fluorescently labeled platelets during the ex vivo perfusion and coculture with primary liver sinusoidal endothelial cells (LSEC). RESULTS: Ex vivo liver perfusion revealed that 93% of human platelets were removed from circulation after 15 min. Endothelial cells and platelets were not activated based on tissue factor release into the perfusate. Biopsies from the ex vivo perfusion at 15 and 30 min and in vitro analysis indicated that human platelets are phagocytosed by pig LSEC and degraded in phagosomes. Sixty to 120 min after the addition of platelets to the ex vivo perfusion system, we observed platelet fragments and degraded platelets in hepatocytes. Platelet phagocytosis was not mediated by opsonization as Fc blocking had no effect on platelet phagocytosis. In vitro uptake of human platelets by primary LSEC cultures peaked at 15 min followed by a greater than 55% decrease in platelet fluorescence after 3 h. Primary pig LSEC phagosomes containing human platelets were colocalized with lysosomes positive for lysosome-associated membrane protein-1 (LAMP1), indicating the formation of mature phagosomes within pig LSEC. CONCLUSIONS: Our observation of pig LSEC phagocytosis of human platelets describes a novel mechanism of large-particle uptake in the liver. The creation of a model system to study xenotransplantation-induced thrombocytopenia makes possible the investigation into mechanisms that mediate platelet loss.


Assuntos
Plaquetas/metabolismo , Fígado/citologia , Fígado/metabolismo , Trombocitopenia/sangue , Transplante Heterólogo/efeitos adversos , Animais , Plaquetas/citologia , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Humanos , Transplante de Fígado , Perfusão , Fagocitose , Trombocitopenia/etiologia
7.
Am J Med Sci ; 353(2): 132-136, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28183413

RESUMO

Publications in peer-reviewed biomedical journals are essential for sharing knowledge and advancing healthcare. This article will articulate a 5-step approach for developing and publishing a manuscript, and provide academic clinicians with an instructional tool they can provide to their protégés and junior faculty. The authors attempt to distill existing advice for preparing manuscripts, which is found in myriad formats, combine these tutorials with their collective experience and present this approach for developing and publishing successfully a manuscript in a peer-reviewed journal. The 5 steps identified instruct would-be authors to (1) know their material and determine their audience; (2) outline their manuscript; (3) be ethically vigilant; (4) develop individual sections and submit their manuscript and (5) respond to reviewers׳ comments. This article describes each of these steps in detail. Rewards of publishing articles include recognition by peers and supervisors, contribution to academic promotion and dissemination of information to the medical community.


Assuntos
Manuscritos como Assunto , Editoração , Pesquisa Biomédica , Humanos , Revisão por Pares , Pesquisadores
8.
Transplantation ; 100(3): 533-7, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26906938

RESUMO

BACKGROUND: Disrupting the porcine GGTA1 and CMAH genes [double knockout (DKO)] that produce the gal-α(1,3)-gal and N-glycolylneuraminic acid xenoantigens reduces human antibody binding to porcine peripheral blood mononuclear cells. It is important to examine rejection pathways at an organ-specific level. The object of this study is to evaluate the human preformed antibody reactivity against DKO renal microvascular endothelial cells (RMEC) in vitro. METHODS: Characteristics of DKO RMEC were analyzed using flow cytometry. Human IgG/M binding to primary RMEC, immortalized RMEC (iRMEC), and iRMEC-deficient in B4GALNT2 genes were examined using flow cytometric crossmatch assay. RESULTS: Porcine RMEC expressed gal-α(1,3)-gal, N-glycolylneuraminic acid, and Dolichos biflorus agglutinin glycans recognized by human preexisting antibodies in humans. Antigenicity of DKO RMEC was lower than GGTA1 KO RMEC. The disruption of B4GALNT2 gene in DKO iRMEC further reduced human IgG/IgM binding. CONCLUSIONS: Silencing the porcine GGTA1, CMAH, and B4GALNT2 genes is an effective strategy to reduce human preformed antibody binding to RMEC. Porcine RMEC will be a useful reagent for the further study of xenoimmunology.


Assuntos
Antígenos Heterófilos/imunologia , Células Endoteliais/imunologia , Rim/irrigação sanguínea , Microvasos/imunologia , Animais , Animais Geneticamente Modificados , Antígenos Heterófilos/metabolismo , Células Cultivadas , Células Endoteliais/metabolismo , Galactosiltransferases/deficiência , Galactosiltransferases/genética , Galactosiltransferases/imunologia , Técnicas de Inativação de Genes , Sobrevivência de Enxerto , Humanos , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Imunoglobulina M/imunologia , Imunoglobulina M/metabolismo , Microvasos/citologia , Microvasos/metabolismo , Oxigenases de Função Mista/deficiência , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/imunologia , N-Acetilgalactosaminiltransferases/deficiência , N-Acetilgalactosaminiltransferases/genética , N-Acetilgalactosaminiltransferases/imunologia , Fenótipo , Suínos , Transfecção
9.
Transplantation ; 92(7): 739-44, 2011 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-21836538

RESUMO

BACKGROUND: Hepatic failure has been treated successfully with clinical extracorporeal perfusions of porcine livers. However, dog-to-pig and pig-to-baboon liver xenotransplant models have resulted in severe bleeding secondary to liver xenograft-induced thrombocytopenia. Kupffer cells (KC) are abundant phagocytic cells in the liver. KC express the CD11b/CD18 receptor, which has been implicated in chilled platelet binding and phagocytosis through interaction with platelet surface proteins and carbohydrates. We sought to identify the role of KC CD18 in liver xenograft-induced thrombocytopenia. METHODS: Primary pig KC were characterized by flow cytometry, immunoblots, and quantitative polymerase chain reaction. Pig KC were used in inhibition assays with fluorescently labeled human platelets. The CD18 receptor was targeted for siRNA knockdown. RESULTS: Domestic and α1,3-galactosyltransferase double knockout porcine KC cultures were approximately 92% positive for CD18 as detected by quantitative polymerase chain reaction and flow cytometry. Use of CD18 blocking antibodies resulted in reduction of human platelet binding and phagocytosis. Additionally, asialofetuin, not fetuin, inhibited platelet phagocytosis suggesting the involvement of an oligosaccharide-binding site. Furthermore, reduced CD18 expression by siRNA resulted in decreased human platelet binding. CONCLUSIONS: Our data suggest that primary pig KC bind and phagocytose human platelets with involvement of CD18. Further understanding and modification of CD18 expression in pigs may result in a liver xenograft with reduced thrombocytopenic effects, which could be used as a bridge to allogeneic liver transplantation.


Assuntos
Plaquetas/fisiologia , Antígenos CD18/fisiologia , Citofagocitose/fisiologia , Células de Kupffer/fisiologia , Animais , Animais Geneticamente Modificados , Assialoglicoproteínas/farmacologia , Antígenos CD18/efeitos dos fármacos , Antígenos CD18/genética , Células Cultivadas , Citofagocitose/efeitos dos fármacos , Fetuínas/farmacologia , Humanos , Transplante de Fígado/efeitos adversos , Modelos Animais , RNA Interferente Pequeno/farmacologia , Suínos , Porco Miniatura , Trombocitopenia/etiologia , Transplante Heterólogo/efeitos adversos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA