Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 205
Filtrar
1.
Sci Rep ; 11(1): 13131, 2021 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-34162938

RESUMEN

Xenotransplantation (cross-species transplantation) using genetically-engineered pig organs offers a potential solution to address persistent organ shortage. Current evaluation of porcine genetic modifications is to monitor the nonhuman primate immune response and survival after pig organ xenotransplantation. This measure is an essential step before clinical xenotransplantation trials, but it is time-consuming, costly, and inefficient with many variables. We developed an efficient approach to quickly examine human-to-pig xeno-immune responses in vitro. A porcine endothelial cell was characterized and immortalized for genetic modification. Five genes including GGTA1, CMAH, ß4galNT2, SLA-I α chain, and ß2-microglobulin that are responsible for the production of major xenoantigens (αGal, Neu5Gc, Sda, and SLA-I) were sequentially disrupted in immortalized porcine endothelial cells using CRISPR/Cas9 technology. The elimination of αGal, Neu5Gc, Sda, and SLA-I dramatically reduced the antigenicity of the porcine cells, though the cells still retained their ability to provoke human natural killer cell activation. In summary, evaluation of human immune responses to genetically modified porcine cells in vitro provides an efficient method to identify ideal combinations of genetic modifications for improving pig-to-human compatibility, which should accelerate the application of xenotransplantation to humans.


Asunto(s)
Animales Modificados Genéticamente/inmunología , Antígenos Heterófilos/inmunología , Células Endoteliales/inmunología , Porcinos/inmunología , Trasplante Heterólogo/métodos , Animales , Anticuerpos Heterófilos/inmunología , Reacciones Antígeno-Anticuerpo , Antígenos Heterófilos/genética , Sistemas CRISPR-Cas , Degranulación de la Célula , Línea Celular Transformada , Citocinas/farmacología , Células Endoteliales/efectos de los fármacos , Galactosiltransferasas/genética , Galactosiltransferasas/inmunología , Técnicas de Inactivación de Genes , Rechazo de Injerto/inmunología , Rechazo de Injerto/prevención & control , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Células Asesinas Naturales/inmunología , Hígado/citología , Activación de Linfocitos , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/inmunología , N-Acetilgalactosaminiltransferasas/genética , N-Acetilgalactosaminiltransferasas/inmunología , Microglobulina beta-2/genética , Microglobulina beta-2/inmunología
2.
Parasit Vectors ; 14(1): 303, 2021 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-34090497

RESUMEN

BACKGROUND: An epitope, Galα1-3Galß1-4GlcNAc-R, termed α-gal, is present in glycoconjugates of New World monkeys (platyrrhines) and other mammals but not in hominoids and Old World monkeys (catarrhines). The difference is due to the inactivation of α1-3 galactosyl transferase (α1-3 GT) genes in catarrhines. Natural antibodies to α-gal are therefore developed in catarrhines but not platyrrhines and other mammals. Hypersensitivity reactions are commonly elicited by mosquito and tick vector bites. IgE antibodies against α-gal cause food allergy to red meat in persons who have been exposed to tick bites. Three enzymes synthesising the terminal α1-3-linked galactose in α-gal, that are homologous to mammalian α and ß1-4 GTs but not mammalian α1-3 GTs, were recently identified in the tick vector Ixodes scapularis. IgG and IgM antibodies to α-gal are reported to protect against malaria because mosquito-derived sporozoites of malaria parasites express α-gal on their surface. This article explores the possibility that the α-gal in sporozoites are acquired from glycoconjugates synthesised by mosquitoes rather than through de novo synthesis by sporozoites. METHODS: The presence of proteins homologous to the three identified tick α1-3 GTs and mammalian α1-3 GTs in two important mosquito vectors, Aedes aegypti and Anopheles gambiae, as well as Plasmodium malaria parasites, was investigated by BLASTp analysis to help clarify the source of the α-gal on sporozoite surfaces. RESULTS: Anopheles gambiae and Ae. aegypti possessed several different proteins homologous to the three I. scapularis proteins with α1-3 GT activity, but not mammalian α1-3 GTs. The putative mosquito α1-3 GTs possessed conserved protein domains characteristic of glycosyl transferases. However, the genus Plasmodium lacked proteins homologous to the three I. scapularis proteins with α1-3 GT activity and mammalian α1-3 GTs. CONCLUSIONS: The putative α1-3 GTs identified in the two mosquito vectors may synthesise glycoconjugates containing α-gal that can be transferred to sporozoite surfaces before they are inoculated into skin during blood feeding. The findings merit further investigation because of their implications for immunity against malaria, hypersensitivity to mosquito bites, primate evolution, and proposals for immunisation against α-gal.


Asunto(s)
Galactosiltransferasas/genética , Hipersensibilidad , Proteínas de Insectos/inmunología , Ixodes/química , Malaria/inmunología , Mosquitos Vectores/química , Mordeduras de Garrapatas/inmunología , Alérgenos/inmunología , Animales , Vectores de Enfermedades , Galactosiltransferasas/inmunología , Humanos , Hipersensibilidad/prevención & control , Inmunoglobulina E/inmunología , Proteínas de Insectos/genética , Ixodes/enzimología , Ixodes/genética , Ixodes/inmunología , Malaria/prevención & control , Mosquitos Vectores/enzimología , Mosquitos Vectores/genética
3.
Eur J Immunol ; 51(6): 1412-1422, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33576494

RESUMEN

Heterologous polyclonal antibodies might represent an alternative to the use of convalescent plasma or monoclonal antibodies (mAbs) in coronavirus disease (COVID-19) by targeting multiple antigen epitopes. However, heterologous antibodies trigger human natural xenogeneic antibody responses particularly directed against animal-type carbohydrates, mainly the N-glycolyl form of the neuraminic acid (Neu5Gc) and the α1,3-galactose, potentially leading to serum sickness or allergy. Here, we immunized cytidine monophosphate-N-acetylneuraminic acid hydroxylase and α1,3-galactosyl-transferase (GGTA1) double KO pigs with the Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike receptor binding domain to produce glyco-humanized polyclonal neutralizing antibodies lacking Neu5Gc and α1,3-galactose epitopes. Animals rapidly developed a hyperimmune response with anti-SARS-CoV-2 end-titers binding dilutions over one to a million and end-titers neutralizing dilutions of 1:10 000. The IgG fraction purified and formulated following clinical Good Manufacturing Practices, named XAV-19, neutralized spike/angiotensin converting enzyme-2 interaction at a concentration <1 µg/mL, and inhibited infection of human cells by SARS-CoV-2 in cytopathic assays. We also found that pig GH-pAb Fc domains fail to interact with human Fc receptors, thereby avoiding macrophage-dependent exacerbated inflammatory responses and a possible antibody-dependent enhancement. These data and the accumulating safety advantages of using GH-pAbs in humans warrant clinical assessment of XAV-19 against COVID-19.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , COVID-19/inmunología , COVID-19/terapia , SARS-CoV-2/inmunología , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/inmunología , Anticuerpos Neutralizantes/genética , Anticuerpos Neutralizantes/farmacología , Anticuerpos Antivirales/genética , Anticuerpos Antivirales/farmacología , COVID-19/genética , Galactosiltransferasas/deficiencia , Galactosiltransferasas/inmunología , Células HEK293 , Humanos , Inmunización Pasiva , SARS-CoV-2/genética , Ácidos Siálicos/genética , Ácidos Siálicos/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología , Porcinos , Sueroterapia para COVID-19
4.
Laryngoscope ; 131(1): E196-E202, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32427353

RESUMEN

OBJECTIVES: The objective of this study is to determine the value of the anti- glycoprotein-N-acetylgalactosamine 3-beta-galactosyltransferase 1 (C1GALT1) autoantibody as a biomarker for distant metastasis and good response to immune checkpoint inhibitors in patients with head and neck squamous cell carcinoma (HNSCC). METHODS: In this retrospective study with a median follow-up of 55.7 months, 186 HNSCC patients were enrolled between July 2013 and August 2014. Data were analyzed between April 2018 and November 2019. Titers of autoantibody against the C1GALT1 peptide were measured by ELISA. Student t test, Kaplan-Meier analysis, and univariate and multivariate Cox proportional hazard models were used to evaluate the association of anti-C1GALT1 autoantibody titer with clinicopathologic factors, survival, and response to immunotherapy. RESULTS: Our results showed that high levels of the anti-C1GALT1 autoantibody is an independent marker for distant metastasis and poor disease-specific survivals in HNSCC patients. In 19 recurrent or metastatic (R/M) HNSCC patients who have received nivolumab or pembrolizumab, higher autoantibody titers are associated with a better treatment response. CONCLUSION: We propose that the anti-C1GALT1 autoantibody can serve as a novel biomarker for distant metastasis in HNSCC patients. It is also useful in individualized medicine for R/M HNSCC patients who are considering immunotherapy. LEVEL OF EVIDENCE: IV Laryngoscope, 131:E196-E202, 2021.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Autoanticuerpos/sangre , Galactosiltransferasas/inmunología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Nivolumab/uso terapéutico , Carcinoma de Células Escamosas de Cabeza y Cuello/sangre , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Biomarcadores/sangre , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Carcinoma de Células Escamosas de Cabeza y Cuello/mortalidad , Carcinoma de Células Escamosas de Cabeza y Cuello/secundario , Tasa de Supervivencia , Resultado del Tratamiento
5.
Transplantation ; 105(2): 318-327, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32796494

RESUMEN

BACKGROUND: Natural preformed anti-pig IgM/IgG antibodies in primates play an important role in xenograft rejection. As it is not clear how IgE and IgA engage in the immune system in xenotransplantation, we investigated natural preformed and elicited anti-pig IgE/IgA in naive primates and after xenotransplantation in nonhuman primates. METHODS: The binding of IgM/IgG/IgE/IgA antibodies to red blood cells (RBCs) from wild-type (WT), α1,3-galactosyltransferase gene-knockout (GTKO), and GTKO/cytidine monophospho-N-acetylneuraminic acid hydroxylase gene-knockout/ß-1,4 N-acetylgalactosaminyltransferase 2 gene-knockout (ie, triple-knockout pigs) pigs were measured by flow cytometry in naive human (n = 50) and baboon (n = 14) sera. Antibody binding to WT and GTKO pig RBCs (pRBCs) was also measured in the sera of baboons (nonsensitized n = 7, sensitized n = 2) and rhesus monkeys (nonsensitized n = 2, sensitized n = 11) following WT or GTKO pig organ/tissue xenotransplantation. Deposition of IgM/IgG/IgE/IgA in the grafts was detected by immunohistochemistry. RESULTS: The majority of humans had natural preformed IgM/IgG/IgE/IgA to WT and GTKO pRBCs. In contrast, IgM/IgG/IgE/IgA to triple-knockout pRBCs were present at lower levels and frequency (P < 0.01). Baboons also had IgM/IgG/IgE/IgA antibodies against WT pRBCs, but fewer to GTKO and triple-knockout (P < 0.01). After xenotransplantation into nonhuman primates, when IgM/IgG increased, IgE/IgA also increased, but to a lesser extent. In addition to IgM/IgG, IgE or IgA deposition was observed in rejected pig xenografts. CONCLUSIONS: Primates develop serum anti-pig IgE/IgA antibodies both naturally and during xenograft rejection. The pathophysiological role, if any, of anti-pig IgE/IgA antibodies remains unknown.


Asunto(s)
Anticuerpos Heterófilos/sangre , Eritrocitos/inmunología , Rechazo de Injerto/inmunología , Inmunoglobulina A/sangre , Inmunoglobulina E/sangre , Trasplante Heterólogo/efectos adversos , Animales , Animales Modificados Genéticamente , Galactosiltransferasas/genética , Galactosiltransferasas/inmunología , Rechazo de Injerto/sangre , Rechazo de Injerto/patología , Humanos , Macaca mulatta , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/inmunología , N-Acetilgalactosaminiltransferasas/genética , N-Acetilgalactosaminiltransferasas/inmunología , Papio , Especificidad de la Especie , Sus scrofa/genética , Sus scrofa/inmunología
6.
Transplantation ; 104(8): 1566-1573, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32732833

RESUMEN

BACKGROUND: Xenogeneic organ transplantation has been proposed as a potential approach to fundamentally solve organ shortage problem. Xenogeneic immune responses across species is one of the major obstacles for clinic application of xeno-organ transplantation. The generation of glycoprotein galactosyltransferase α 1, 3 (GGTA1) knockout pigs has greatly contributed to the reduction of hyperacute xenograft rejection. However, severe xenograft rejection can still be induced by xenoimmune responses to the porcine major histocompatibility complex antigens swine leukocyte antigen class I and class II. METHODS: We simultaneously depleted GGTA1, ß2-microglobulin (ß2M), and major histocompatibility complex class II transactivator (CIITA) genes using clustered regularly interspaced short palindromic repeats and CRISPR-associated proteins technology in Bamma pig fibroblast cells, which were further used to generate GGTA1ß2MCIITA triple knockout (GBC-3KO) pigs by nuclear transfer. RESULTS: The genotype of GBC-3KO pigs was confirmed by polymerase chain reaction and Sanger sequencing, and the loss of expression of α-1,3-galactose, SLA-I, and SLA-II was demonstrated by flow cytometric analysis using fluorescent-conjugated lectin from bandeiraea simplicifolia, anti-ß2-microglobulin, and swine leukocyte antigen class II DR antibodies. Furthermore, mixed lymphocyte reaction assay revealed that peripheral blood mononuclear cells from GBC-3KO pigs were significantly less effective than (WT) pig peripheral blood mononuclear cells in inducing human CD3CD4 and CD3CD8 T-cell activation and proliferation. In addition, GBC-3KO pig skin grafts showed a significantly prolonged survival in immunocompetent C57BL/6 mice, when compared with wild-type pig skin grafts. CONCLUSIONS: Taken together, these results demonstrate that elimination of GGTA1, ß2M, and CIITA genes in pigs can effectively alleviate xenogeneic immune responses and prolong pig organ survival in xenogenesis. We believe that this work will facilitate future research in xenotransplantation.


Asunto(s)
Rechazo de Injerto/prevención & control , Xenoinjertos/inmunología , Trasplante de Órganos/métodos , Trasplante Heterólogo/métodos , Aloinjertos/provisión & distribución , Animales , Animales Modificados Genéticamente/inmunología , Sistemas CRISPR-Cas/genética , Modelos Animales de Enfermedad , Femenino , Galactosiltransferasas/genética , Galactosiltransferasas/inmunología , Técnicas de Inactivación de Genes/métodos , Genes MHC Clase II/genética , Genes MHC Clase II/inmunología , Rechazo de Injerto/inmunología , Supervivencia de Injerto/genética , Supervivencia de Injerto/inmunología , Xenoinjertos/trasplante , Humanos , Masculino , Ratones , Trasplante de Órganos/efectos adversos , Porcinos/genética , Porcinos/inmunología , Trasplante Heterólogo/efectos adversos , Microglobulina beta-2/genética , Microglobulina beta-2/inmunología
7.
Int J Biol Macromol ; 161: 875-890, 2020 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-32535205

RESUMEN

This study reports a ≅12.5 kDa protein tetrachloro-1,4-benzoquinone reductase (CpsD) from Bacillus cereus strain AOA-CPS1 (BcAOA). CpsD is purified to homogeneity with a total yield of 35% and specific activity of 160 U·mg-1 of protein. CpsD showed optimal activity at pH 7.5 and 40 °C. The enzyme was found to be functionally stable between pH 7.0-7.5 and temperature between 30 °C and 35 °C. CpsD activity was enhanced by Fe2+ and inhibited by sodium azide and SDS. CpsD followed Michaelis-Menten kinetic exhibiting an apparent vmax, Km, kcat and kcat/Km values of 0.071 µmol·s-1, 94 µmol, 0.029 s-1 and 3.13 × 10-4 s-1·µmol-1, respectively, for substrate tetrachloro-1,4-benzoquinone. The bioinformatics analysis indicated that CpsD belongs to the PCD/DCoH superfamily, with specific conserved protein domains of pterin-4α-carbinolamine  dehydratase (PCD). This study proposed that CpsD catalysed the reduction of tetrachloro-1,4-benzoquinone to tetrachloro-p-hydroquinone and released the products found in phenylalanine hydroxylation system (PheOHS) via a Ping-Pong or atypical ternary mechanism; and regulate expression of phenylalanine 4-monooxygenase by blocking reverse flux in BcAOA PheOHS using a probable Yin-Yang mechanism. The study also concluded that CpsD may play a catalytic and regulatory role in BcAOA PheOHS and pentachlorophenol degradation pathway.


Asunto(s)
Bacillus cereus/metabolismo , Proteínas Bacterianas/inmunología , Cloranilo/metabolismo , Galactosiltransferasas/inmunología , Hidroxilación/fisiología , Pentaclorofenol/metabolismo , Fenilalanina/metabolismo , Cinética , Oxidorreductasas/metabolismo
8.
J Immunol ; 204(7): 1998-2005, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32144163

RESUMEN

Mice have been used as accepted tools for investigating complex human diseases and new drug therapies because of their shared genetics and anatomical characteristics with humans. However, the tissues in mice are different from humans in that human cells have a natural mutation in the α1,3 galactosyltransferase (α1,3GT) gene and lack α-Gal epitopes on glycosylated proteins, whereas mice and other nonprimate mammals express this epitope. The lack of α-Gal epitopes in humans results in the loss of immune tolerance to this epitope and production of abundant natural anti-Gal Abs. These natural anti-Gal Abs can be used as an adjuvant to enhance processing of vaccine epitopes to APCs. However, wild-type mice and all existing humanized mouse models cannot be used to test the efficacy of vaccines expressing α-Gal epitopes because they express α-Gal epitopes and lack anti-Gal Abs. Therefore, in an effort to bridge the gap between the mouse models and humans, we developed a new humanized mouse model that mimics humans in that it lacks α-Gal epitopes and secretes human anti-Gal Abs. The new humanized mouse model (Hu-NSG/α-Galnull) is designed to be used for preclinical evaluations of viral and tumor vaccines based on α-Gal epitopes, human-specific immune responses, xenotransplantation studies, and in vivo biomaterials evaluation. To our knowledge, our new Hu-NSG/α-Galnull is the first available humanized mouse model with such features.


Asunto(s)
Anticuerpos/inmunología , Epítopos/inmunología , Galactosiltransferasas/inmunología , alfa-Galactosidasa/inmunología , Animales , Vacunas contra el Cáncer/inmunología , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Trasplante Heterólogo/métodos
9.
Xenotransplantation ; 27(2): e12567, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31762117

RESUMEN

BACKGROUND: Engineering of α-Galactosyltransferase gene-knockout pigs circumvented hyperacute rejection of pig organs after xenotransplantation in non-human primates. Overcoming this hurdle revealed the importance of non-α-Gal carbohydrate antigens in the immunobiology of acute humoral xenograft rejection. METHODS: This study analyzed serum from seven naïve cynomolgus monkeys (blood type O/B/AB = 3/2/2) for the intensity of natural IgM and IgG signals using carbohydrate antigen microarray, which included historically reported α-Gal and non-α-Gal carbohydrate antigens with various modifications. RESULTS: The median (range) of IgM and IgG signals were 12.71 (7.23-16.38) and 9.05 (7.23-15.90), respectively. The highest IgM and IgG signals with narrowest distribution were from mono- and disaccharides, followed by modified structures. Natural anti-α-Gal antibody signals were medium to high in IgM (11.2-15.9) and medium in IgG (8.5-11.6) spectra, and was highest with Lac core structure (Galα1-3Galß1-4Glc, iGb3) and lowest with LacNAc core structure (Galα1-3Galß1-4GlcNAc). Similar signal intensities (up to 15.8 in IgM and up to 11.8 in IgG) were observed for historically detected natural non-α-Gal antigens, which included Tn antigen, T antigen, GM2 glycolipid, and Sda antigen. The hierarchical clustering analysis revealed the presence of clusters of anti-A antibodies and was capable of distinguishing between the blood group B and AB non-human primates. CONCLUSIONS: The results presented here provide the most comprehensive evaluation of natural antibodies present in cynomolgus monkeys.


Asunto(s)
Anticuerpos/sangre , Antígenos Heterófilos/inmunología , Rechazo de Injerto/inmunología , Xenoinjertos/inmunología , Animales , Anticuerpos/inmunología , Disacáridos/inmunología , Galactosiltransferasas/inmunología , Macaca fascicularis , Primates , Trasplante Heterólogo/métodos
10.
Xenotransplantation ; 26(4): e12516, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30989742

RESUMEN

Xenotransplantation research has made considerable progress in recent years, largely through the increasing availability of pigs with multiple genetic modifications. We suggest that a pig with nine genetic modifications (ie, currently available) will provide organs (initially kidneys and hearts) that would function for a clinically valuable period of time, for example, >12 months, after transplantation into patients with end-stage organ failure. The national regulatory authorities, however, will likely require evidence, based on in vitro and/or in vivo experimental data, to justify the inclusion of each individual genetic modification in the pig. We provide data both from our own experience and that of others on the advantages of pigs in which (a) all three known carbohydrate xenoantigens have been deleted (triple-knockout pigs), (b) two human complement-regulatory proteins (CD46, CD55) and two human coagulation-regulatory proteins (thrombomodulin, endothelial cell protein C receptor) are expressed, (c) the anti-apoptotic and "anti-inflammatory" molecule, human hemeoxygenase-1 is expressed, and (d) human CD47 is expressed to suppress elements of the macrophage and T-cell responses. Although many alternative genetic modifications could be made to an organ-source pig, we suggest that the genetic manipulations we identify above will all contribute to the success of the initial clinical pig kidney or heart transplants, and that the beneficial contribution of each individual manipulation is supported by considerable experimental evidence.


Asunto(s)
Animales Modificados Genéticamente/genética , Rechazo de Injerto/prevención & control , Porcinos/genética , Trasplante Heterólogo , Animales , Animales Modificados Genéticamente/inmunología , Antígeno CD47/genética , Antígeno CD47/inmunología , Antígenos CD55/genética , Antígenos CD55/inmunología , Receptor de Proteína C Endotelial/genética , Receptor de Proteína C Endotelial/inmunología , Galactosiltransferasas/deficiencia , Galactosiltransferasas/genética , Galactosiltransferasas/inmunología , Técnicas de Sustitución del Gen , Técnicas de Inactivación de Genes , Rechazo de Injerto/inmunología , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/inmunología , Humanos , Proteína Cofactora de Membrana/genética , Proteína Cofactora de Membrana/inmunología , Oxigenasas de Función Mixta/deficiencia , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/inmunología , N-Acetilgalactosaminiltransferasas/deficiencia , N-Acetilgalactosaminiltransferasas/genética , N-Acetilgalactosaminiltransferasas/inmunología , Porcinos/inmunología , Trombomodulina/genética , Trombomodulina/inmunología
11.
Xenotransplantation ; 26(4): e12504, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30825348

RESUMEN

The humoral barrier has been the limiting factor in moving xenotransplantation towards the clinic. Improvements in somatic cell nuclear transfer and genome editing, particularly CRISPR-Cas9, have made it possible to create pigs with multiple glycan xenoantigen deletions for the purposes of reducing xenoreactive antibody binding to the xenografted organ. Recent studies have also considered the aetiology and existence of antibodies directed at the swine leucocyte antigen (SLA) complex, and potential genetic engineering strategies to avoid these antibodies. Evaluation of xenoreactive antibody binding is very important for the advancement of xenotransplantation, because if patients do not have any detectable xenoreactive antibody, then it is reasonable to expect that cellular rejection and not antibody-mediated rejection (AMR) will be the next hurdle to clinical application.


Asunto(s)
Antígenos Heterófilos/inmunología , Galactosiltransferasas/inmunología , Técnicas de Inactivación de Genes , Rechazo de Injerto/prevención & control , Oxigenasas de Función Mixta/inmunología , N-Acetilgalactosaminiltransferasas/inmunología , Porcinos/inmunología , Trasplante Heterólogo , Animales , Animales Modificados Genéticamente/inmunología , Anticuerpos Heterófilos/biosíntesis , Anticuerpos Heterófilos/inmunología , Reacciones Antígeno-Anticuerpo , Antígenos Heterófilos/genética , Epítopos/inmunología , Galactosiltransferasas/deficiencia , Galactosiltransferasas/genética , Ingeniería Genética , Rechazo de Injerto/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Oxigenasas de Función Mixta/deficiencia , Oxigenasas de Función Mixta/genética , N-Acetilgalactosaminiltransferasas/deficiencia , N-Acetilgalactosaminiltransferasas/genética , Inmunología del Trasplante
12.
Xenotransplantation ; 26(3): e12498, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30770572

RESUMEN

BACKGROUND: Old World non-human primates (OWNHPs) are used for preclinical pig-to-NHP studies. However, like pigs, OWNHPs express Neu5Gc, and therefore do not develop natural anti-Neu5Gc antibodies. New World NHPs (NWNHPs) have been reported not to express Neu5Gc. We investigated the potential of NWNHPs in xenotransplantation research. METHODS: We investigated expression of Gal, Neu5Gc, and Sda antigens on RBCs and PBMCs from humans, selected OWNHPs, and capuchin monkeys (a NWNHP). Serum anti-Gal and anti-Neu5Gc IgM and IgG levels were measured by ELISA. Binding of primate serum IgM and IgG to pig RBCs was measured by flow cytometry. RESULTS: (a) Neither humans, OWNHPs, or capuchin monkeys expressed Gal on their RBCs, but capuchins expressed Gal on PBMCs. Humans and capuchins did not express Neu5Gc on either RBCs or PBMCs, but OWNHPs expressed Neu5Gc on both cells. Sda was not expressed on any RBCs or PBMCs. (b) By ELISA, human and OWNHP, but not capuchin, sera showed IgM and IgG binding to Gal. Human and capuchin, but not OWNHP, sera demonstrated some binding to Neu5Gc. (c) Anti-Sda IgM/IgG antibodies were detected in OWNHP sera. Knockout of Sda on pig RBCs did not significantly reduce human and capuchin antibody binding. CONCLUSION: Capuchin monkeys could be surrogates for humans in experiments using RBCs, islets, neuronal cells, etc, from triple-knockout pigs (but may be too small to be used as recipients of pig organ grafts).


Asunto(s)
Carbohidratos/inmunología , Galactosiltransferasas/inmunología , Xenoinjertos/inmunología , Trasplante Heterólogo , Animales , Animales Modificados Genéticamente , Carbohidratos/genética , Cebus , Galactosiltransferasas/genética , Técnicas de Inactivación de Genes , Rechazo de Injerto/inmunología , Humanos , Platirrinos , Porcinos
13.
Sci Rep ; 8(1): 15424, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30337555

RESUMEN

Alpha-Gal (Gal) epitopes present in animal tissues are known to be the key xenoantigens that elicit xenorejection. However, a standardized method to determine Gal epitope in animal tissue-derived biomaterials does not exist. Herein, a standardized method for quantitative detection of Gal antigen was established based on an ELISA inhibition assay with Gal antibody. In this method, the key optimized experimental conditions were: (1) Gal-antigen positive and negative reference materials were developed, and used as positive and negative control in the test system, respectively; (2) A mixture of artificial Gal-BSA antigen plus Gal-negative matrix was used as the calibration standard sample, making it has similar composition with test sample; and (3) The lysis buffer was combined with the homogenate to expose the Gal antigen as much as possible. The results from validation and application experiments showed that the standardized method had good reproducibility (RSD = 12.48%), and the lower detection limit (LDL) is ~7.1 × 1011 Gal epitopes/reaction. This method has been further developed into a detection Kit (Meitan 70101, China), and it has been developed as a standard method for detecting remnant immunogen of animal tissue derived medical devices, and as the industry standard has been released in China. (YY/T 1561-2017).


Asunto(s)
Alérgenos/análisis , Materiales Biocompatibles/metabolismo , Disacáridos/inmunología , Ensayo de Inmunoadsorción Enzimática/veterinaria , Epítopos/análisis , Alérgenos/inmunología , Alérgenos/metabolismo , Animales , Disacáridos/metabolismo , Ensayo de Inmunoadsorción Enzimática/normas , Epítopos/inmunología , Galactosiltransferasas/inmunología , Galactosiltransferasas/metabolismo , Porcinos
14.
Biomaterials ; 187: 93-104, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30312852

RESUMEN

Whole organ tissue engineering is a promising approach to address organ shortages in many applications, including lung transplantation for patients with chronic pulmonary disease. Engineered lungs may be derived from animal sources after removing cellular content, exposing the extracellular matrix to serve as a scaffold for recellularization with human cells. However, the use of xenogeneic tissue sources in human transplantation raises concerns due to the presence of the antigenic Gal epitope. In the present study, lungs from wild type or α-Gal knockout pigs were harvested, decellularized, and implanted subcutaneously in a non-human primate model to evaluate the host immune response. The decellularized porcine implants were compared to a sham surgery control, as well as native porcine and decellularized macaque lung implants. The results demonstrated differential profiles of circulating and infiltrating immune cell subsets and histological outcomes depending on the implanted tissue source. Upon implantation, the decellularized α-Gal knockout lung constructs performed similarly to the decellularized wild type lung constructs. However, upon re-implantation into a chronic exposure model, the decellularized wild type lung constructs resulted in a greater proportion of infiltrating CD45+ cells, including CD3+ and CD8+ cytotoxic T-cells, likely mediated by an increase in production of Gal-specific antibodies. The results suggest that removal of the Gal epitope can potentially reduce adverse inflammatory reactions associated with chronic exposure to engineered organs containing xenogeneic components.


Asunto(s)
Galactosiltransferasas/genética , Enfermedades Pulmonares/terapia , Pulmón/citología , Andamios del Tejido , Inmunidad Adaptativa , Animales , Materiales Biocompatibles , Galactosiltransferasas/inmunología , Técnicas de Inactivación de Genes , Humanos , Inmunidad Humoral , Enfermedades Pulmonares/inmunología , Macaca mulatta , Porcinos , Ingeniería de Tejidos , Trasplante , Trasplante Heterólogo
15.
Artif Cells Nanomed Biotechnol ; 46(sup3): S359-S369, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30207744

RESUMEN

Homeotransplantation of bones for replacement therapy have been demonstrated reliably in clinical data. However, human donor bones applicable for homeotransplantation are in short supply, which facilitates the search for suitable alternatives, such as xenografts grafts. The α-Gal antigen-related immune risk of xenografts directly affects the safety and effectiveness of the biomaterials and limits their applications in the clinic. The immune risk can be prevented by depletion or breaking anti-Gal antibody prior to transplant. Therefore, how to assess the immune risk of the bone substitutes and select the reliable animal research model become extremely important. In this study, we prepared lyophilized bone substitutes (T1) and Guanghao Biotech bone substitutes (T2, animal-derived biomaterials with α-Gal antigen decreased), aimed to assess the immune risk of xenografts bone substitutes on GGTA1 knockout mice. The α-Gal antigen contents of T1 and T2 were firstly detected by ELISA method in vitro. The bone substitutes were then implanted subcutaneously into GGTA1 knockout mice for 2, 4 and 12 weeks, respectively. The total serum antibody levels, anti-α-Gal antibody levels, inflammatory cytokine and splenic lymphocyte surface molecules were detected and histology analysis of skin and thymus were performed to systematically evaluate the immune response caused by the T1 and T2 bone substitutes in mice. In vitro results showed that the amount of α-Gal epitopes in T1 bone substitutes was significantly higher than T2 bone substitutes, and the clearance rate of α-Gal antigen in T2 bone substitutes achieved about 55.6%. Results of antibody level in vivo showed that the T1 bone substitutes group possessed significantly higher total IgG, IgM, IgA and anti-α-Gal IgG levels than T2 and control group, while T2 group showed no significant changes of these indexes compared with control. In terms of inflammatory cytokines, T1 bone substitutes showed evidently higher levels of IL-4, IL-12P70 and IL-10 than T2 and control, while T2 group was comparable to control. No changes in the levels of splenic lymphocyte surface molecules were found in the three groups (T1, T2 and control group) during the experimental periods. The pathological results demonstrated that the inflammatory response in T2 group was lighter than the T1 group, which was in accordance with the inflammatory cytokines levels. The above results indicated that the process of antigen removal effectively reduced the α-Gal antigens content in T2 bone substitutes, which caused little immune response in vivo and could be used as bone healing materials. This study also demonstrated that GGTA1 knockout mice can be used as a routine tool to assess the immune risk of animal-derived biomaterials.


Asunto(s)
Matriz Ósea , Trasplante Óseo , Galactosiltransferasas/deficiencia , Animales , Anticuerpos/inmunología , Matriz Ósea/inmunología , Matriz Ósea/patología , Matriz Ósea/trasplante , Sustitutos de Huesos/farmacología , Galactosiltransferasas/inmunología , Xenoinjertos , Ratones , Ratones Noqueados , Trasplante Heterólogo
16.
Acta Biomater ; 72: 196-205, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29631050

RESUMEN

Bioprosthetic heart valves (BHVs) originating from pigs are extensively used for heart valve replacement in clinics. However, recipient immune responses associated with chronic calcification lead to structural valve deterioration (SVD) of BHVs. Two well-characterized epitopes on porcine BHVs have been implicated in SVD, including galactose-α1,3-galactose (αGal) and N-glycolylneuraminic acid (Neu5Gc) whose synthesis are catalyzed by α(1,3) galactosyltransferase (encoded by the GGTA1 gene) and CMP-Neu5Ac hydroxylase (encoded by the CMAH gene), respectively. It has been reported that BHV from αGal-knockout pigs are associated with a significantly reduced immune response by human serum. Moreover, valves from αGal/Neu5Gc-deficient pigs could further reduce human IgM/IgG binding when compared to BHV from αGal-knockout pigs. Recently, another swine xenoantigen, Sd(a), produced by ß-1,4-N-acetyl-galactosaminyl transferase 2 (ß4GalNT2), has been identified. To explore whether tissue from GGTA1, CMAH, and ß4GalNT2 triple gene-knockout (TKO) pigs would further minimize human antibody binding to porcine pericardium, TKO pigs were successfully produced by CRISPR/Cas9 mediated gene targeting. Our results showed that the expression of αGal, Neu5G and Sd(a) on TKO pigs was negative, and that human IgG/IgM binding to pericardium was minimal. Moreover, the analysis of collagen composition and physical characteristics of porcine pericardium from the TKO pigs indicated that elimination of the three xenoantigens had no significant impact on the physical proprieties of porcine pericardium. Our results demonstrated that TKO pigs would be an ideal source of BHVs. STATEMENT OF SIGNIFICANCE: Surgical heart valve replacement is an established lifesaving treatment for diseased heart valve. Bioprosthetic heart valves (BHVs) made from glutaraldehyde-fixed porcine or bovine tissues are widely used in clinics but exhibit age-dependent structural valve degeneration (SVD) which is associated with the immune response against BHVs. Three major xenoantigens present on commercial BHVs, Galactosea α1,3 galactose (αGal), N-glycolylneuraminic acid (Neu5Gc) and glycan products of ß-1,4-N-acetyl-galactosaminyl transferase 2 (ß4GalNT2) are eliminated through CRISPR/Cas9 mediated gene targeting in the present study. The genetically modified porcine pericardium showed reduced immunogenicity but comparable collagen composition and physical characteristics of the pericardium from wild-type pigs. Our data suggested that BHVs from TKO pigs is a promising alternative for currently available BHVs from wild-type pigs.


Asunto(s)
Animales Modificados Genéticamente , Antígenos Heterófilos/genética , Bioprótesis , Galactosiltransferasas/genética , Eliminación de Gen , Prótesis Valvulares Cardíacas , Oxigenasas de Función Mixta/genética , N-Acetilgalactosaminiltransferasas/genética , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/inmunología , Antígenos Heterófilos/inmunología , Galactosiltransferasas/inmunología , Humanos , Oxigenasas de Función Mixta/inmunología , N-Acetilgalactosaminiltransferasas/inmunología , Porcinos
17.
Transplantation ; 102(5): e195-e204, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29266033

RESUMEN

BACKGROUND: We investigated in vitro whether HLA highly sensitized patients with end-stage renal disease will be disadvantaged immunologically after a genetically engineered pig kidney transplant. METHODS: Blood was drawn from patients with a calculated panel-reactive antibody (cPRA) 99% to 100% (Gp1, n = 10) or cPRA 0% (Gp2, n = 12), and from healthy volunteers (Gp3, n = 10). Serum IgM and IgG binding was measured (i) to galactose-α1-3 galactose and N-glycolylneuraminic acid glycans by enzyme-linked immunosorbent assay, and (ii) to pig red blood cell, pig aortic endothelial cells, and pig peripheral blood mononuclear cell from α1,3-galactosyltransferase gene-knockout (GTKO)/CD46 and GTKO/CD46/cytidine monophosphate-N-acetylneuraminic acid hydroxylase-knockout (CMAHKO) pigs by flow cytometry. (iii) T-cell and B-cell phenotypes were determined by flow cytometry, and (iv) proliferation of T-cell and B-cell carboxyfluorescein diacetate succinimidyl ester-mixed lymphocyte reaction. RESULTS: (i) By enzyme-linked immunosorbent assay, there was no difference in IgM or IgG binding to galactose-α1-3 galactose or N-glycolylneuraminic acid between Gps1 and 2, but binding was significantly reduced in both groups compared to Gp3. (ii) IgM and IgG binding in Gps1 and 2 was also significantly lower to GTKO/CD46 pig cells than in healthy controls, but there were no differences between the 3 groups in binding to GTKO/CD46/CMAHKO cells. (iii and iv) Gp1 patients had more memory T cells than Gp2, but there was no difference in T or B cell proliferation when stimulated by any pig cells. The proliferative responses in all 3 groups were weakest when stimulated by GTKO/CD46/CMAHKO pig peripheral blood mononuclear cell. CONCLUSIONS: (i) End-stage renal disease was associated with low antipig antibody levels. (ii) Xenoreactivity decreased with increased genetic engineering of pig cells. (iii) High cPRA status had no significant effect on antibody binding or T-cell and B-cell response.


Asunto(s)
Galactosiltransferasas/genética , Antígenos HLA/inmunología , Fallo Renal Crónico/cirugía , Trasplante de Riñón/métodos , Proteína Cofactora de Membrana/genética , Oxigenasas de Función Mixta/genética , Animales , Animales Modificados Genéticamente , Linfocitos B/inmunología , Estudios de Casos y Controles , Células Cultivadas , Galactosiltransferasas/deficiencia , Galactosiltransferasas/inmunología , Rechazo de Injerto/genética , Rechazo de Injerto/inmunología , Rechazo de Injerto/prevención & control , Antígenos HLA/sangre , Xenoinjertos , Humanos , Memoria Inmunológica , Isoanticuerpos/sangre , Isoanticuerpos/inmunología , Fallo Renal Crónico/sangre , Fallo Renal Crónico/diagnóstico , Fallo Renal Crónico/inmunología , Trasplante de Riñón/efectos adversos , Activación de Linfocitos , Proteína Cofactora de Membrana/deficiencia , Proteína Cofactora de Membrana/inmunología , Oxigenasas de Función Mixta/deficiencia , Oxigenasas de Función Mixta/inmunología , Sus scrofa , Linfocitos T/inmunología
18.
PLoS One ; 12(10): e0184901, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29077749

RESUMEN

OBJECTIVES: Single-agent immunotherapy is ineffective against poorly immunogenic cancers, including pancreatic ductal adenocarcinoma (PDAC). The aims of this study were to demonstrate the feasibility of production of novel autologous tumor lysate vaccines from resected PDAC tumors, and verify vaccine safety and efficacy. METHODS: Fresh surgically resected tumors obtained from human patients were processed to enzymatically synthesize α-gal epitopes on the carbohydrate chains of membrane glycoproteins. Processed membranes were analyzed for the expression of α-gal epitopes and the binding of anti-Gal, and vaccine efficacy was assessed in vitro and in vivo. RESULTS: Effective synthesis of α-gal epitopes was demonstrated after processing of PDAC tumor lysates from 10 different patients, and tumor lysates readily bound an anti-Gal monoclonal antibody. α-gal(+) PDAC tumor lysate vaccines elicited strong antibody production against multiple tumor-associated antigens and activated multiple tumor-specific T cells. The lysate vaccines stimulated a robust immune response in animal models, resulting in tumor suppression and a significant improvement in survival without any adverse events. CONCLUSIONS: Our data suggest that α-gal(+) PDAC tumor lysate vaccination may be a practical and effective new immunotherapeutic approach for treating pancreatic cancer.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Carcinoma Ductal Pancreático/terapia , Epítopos/inmunología , Galactosiltransferasas/inmunología , Inmunoterapia/métodos , Neoplasias Pancreáticas/terapia , Anciano , Anciano de 80 o más Años , Animales , Anticuerpos Monoclonales/inmunología , Vacunas contra el Cáncer/inmunología , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Mucina-1/inmunología
19.
Sci Rep ; 7(1): 10024, 2017 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-28855711

RESUMEN

When hyperacute rejection is avoided by deletion of Gal expression in the pig, delayed xenograft rejection (DXR) becomes a major immunologic barrier to successful xenotransplantation. This study was to investigate the potential antigens involved in DXR. We isolated primary renal microvascular endothelial cells (RMEC) and aortic endothelial cells (AEC) from a GGTA1/CMAH double-knockout (DKO) pig (and a GGTA1-KO pig) and immunized cynomolgus monkeys with both of these cells. After sensitization, monkey serum antibody binding and cytotoxicity to RMEC was significantly higher than to AEC(p < 0.05), suggesting that RMEC are more immunogenic than AEC. Transcriptome sequencing of GGTA1/CMAH DKO pigs indicated that the expression of 1,500 genes was higher in RMEC than in AEC, while expression of 896 genes was lower. Next, we selected 101 candidate genes expressed only in pig RMEC, but not in pig AEC or in monkey or human RMEC. When these genes were knocked out individually in GGTA1/CMAH DKO RMEC, 32 genes were associated with reduced antibody binding, indicating that these genes might be primary immunologic targets involved in DXR. These genes may be important candidates for deletion in producing pigs against which there is a reduced primate immune response in pig kidney xenograft.


Asunto(s)
Antígenos/genética , Galactosiltransferasas/inmunología , Rechazo de Injerto/genética , Xenoinjertos/inmunología , Transcriptoma , Animales , Antígenos/inmunología , Células Cultivadas , Células Endoteliales/inmunología , Endotelio Vascular/inmunología , Femenino , Galactosiltransferasas/genética , Rechazo de Injerto/inmunología , Humanos , Riñón/irrigación sanguínea , Macaca fascicularis , Porcinos
20.
Monoclon Antib Immunodiagn Immunother ; 36(3): 95-103, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28636500

RESUMEN

IgA nephropathy (IgAN) is a globally common primary glomerulonephritis characterized by an elevated level of serum IgA and immune complex deposition in the mesangial area. In the serum of patients with IgAN, the hinge region of IgA1 immunoglobulin contains aberrantly glycosylated O-glycans deficient in galactose, which is normally added to the core 1 O-glycan structure by core 1 synthase, glycoprotein-N-acetylgalactosamine 3-beta-galactosyltransferase 1 (C1GALT1), the key enzyme in the process of glycosylation. It is unknown if single-nucleotide polymorphisms rs1047763 and rs1008898 of C1GALT1 increase the risk of IgAN. We enrolled 5 subjects in this meta-analysis, including a total of 1693 IgAN patients and 1864 control subjects. We performed meta-analysis on associations between rs1047763, rs1008898, and IgAN using the allele model, dominant model, recessive model, and additive model. We found that there was no relationship between rs1047763 and rs1008898 in C1GALT1 and susceptibility to IgAN.


Asunto(s)
Galactosiltransferasas/genética , Glomerulonefritis por IGA/genética , Inmunoglobulina A/genética , Polimorfismo de Nucleótido Simple , Alelos , Estudios de Casos y Controles , Galactosiltransferasas/inmunología , Expresión Génica , Frecuencia de los Genes , Glomerulonefritis por IGA/inmunología , Glomerulonefritis por IGA/patología , Glicosilación , Humanos , Inmunoglobulina A/química , Células Mesangiales/inmunología , Células Mesangiales/patología , Modelos Genéticos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...