Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 624
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 97(2): e0168422, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36651747

RESUMEN

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant is becoming a dominant circulator and has several mutations in the spike glycoprotein, which may cause shifts of immunogenicity, so as to result in immune escape and breakthrough infection among the already infected or vaccinated populations. It is unclear whether infection with Omicron could generate adequate cross-variant protection. To investigate this possibility, we used Syrian hamsters as an animal model for infection of SARS-CoV-2. The serum from Omicron BA.1 variant-infected hamsters showed a significantly lower neutralization effect against infection of the same or different SARS-CoV-2 variants than the serum from Beta variant-infected hamsters. Furthermore, the serum from Omicron BA.1 variant-infected hamsters were insufficient to protect against rechallenge of SARS-CoV-2 Prototype, Beta and Delta variants and itself. Importantly, we found that rechallenge with different SARS-CoV-2 lineages elevated cross-variant serum neutralization titers. Overall, our findings indicate a weakened immunogenicity feature of Omicron BA.1 variant that can be overcome by rechallenge of a different SARS-CoV-2 lineages. Our results may lead to a new guideline in generation and use of the vaccinations to combat the pandemic of SARS-CoV-2 Omicron variant and possible new variants. IMPORTANCE The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant causes breakthrough infections among convalescent patients and vaccinated populations. However, Omicron does not generate robust cross-protective responses. Here, we investigate whether heterologous SARS-CoV-2 challenge is able to enhance antibody response in a sensitive animal model, namely, Syrian hamster. Of note, a heterologous challenge of Beta and Omicron BA.1 variant significantly broadens the breadth of SARS-CoV-2 neutralizing responses against the prototype, Beta, Delta, and Omicron BA.1 variants. Our findings confirm that vaccination strategy with heterologous antigens might be a good option to protect against the evolving SARS-CoV-2.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Cricetinae , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Antígenos Heterófilos/inmunología , Infección Irruptiva , COVID-19/prevención & control , Mesocricetus , SARS-CoV-2/genética , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Inmunogenicidad Vacunal
2.
Xenotransplantation ; 31(3): e12872, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38924560

RESUMEN

Attack of donor tissues by pre-formed anti-pig antibodies is well known to cause graft failure in xenotransplantation. Genetic engineering of porcine donors to eliminate targets of these pre-formed antibodies coupled with advances in immunosuppressive medicines have now made it possible to achieve extended survival in the pre-clinical pig-to-non-human primate model. Despite these improvements, antibodies remain a risk over the lifetime of the transplant, and many patients continue to have pre-formed donor-specific antibodies even to highly engineered pigs. While therapeutics exist that can help mitigate the detrimental effects of antibodies, they act broadly potentially dampening beneficial immunity. Identifying additional xenoantigens may enable more targeted approaches, such as gene editing, to overcome these challenges by further eliminating antibody targets on donor tissue. Because we have found that classical class I swine leukocyte antigens are targets of human antibodies, we now examine whether related pig proteins may also be targeted by human antibodies. We show here that non-classical class I swine leukocyte proteins (SLA-6, -7, -8) can be expressed at the surface of mammalian cells and act as antibody targets.


Asunto(s)
Antígenos Heterófilos , Antígenos de Histocompatibilidad Clase I , Trasplante Heterólogo , Animales , Porcinos , Trasplante Heterólogo/métodos , Antígenos Heterófilos/inmunología , Humanos , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Rechazo de Injerto/inmunología , Animales Modificados Genéticamente
3.
Acta Biochim Biophys Sin (Shanghai) ; 53(6): 707-718, 2021 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-33963824

RESUMEN

The major etiologic agent that causes acute gastroenteritis worldwide in young animals and children is Group A rotavirus. Currently, commercially available vaccines do not often prevent porcine rotavirus (PRV) infection. In this study, we evaluated the efficacy of oral recombinant Lactobacillus vaccine against PRV in a mouse model. Lactobacillus plantarum NC8 was used as the host strain, and bacterial vectors were constructed, because the NC8 isolated has shown the capability to survive gastric transit and to colonize the intestinal tract of humans and other mammals. To explore the immunological mechanisms, lactic acid bacterial vectors were used to express VP7 antigen from PRV. We constructed an L. plantarum strain with surface-displayed VP7, named NC8-pSIP409-pgsA-VP7-DCpep. The expressed recombinant protein had a molecular weight of ∼37 kDa. The strain was used to immunize BALB/c mice to evaluate their immunomodulatory characteristics. Mice were orally immunized with recombinant L. plantarum NC8-pSIP409-pgsA-VP7-DCpep at a dose of 2 × 109 colony forming units/200 µl. The results showed that NC8-pSIP409-pgsA-VP7-DCpep significantly stimulated the differentiation of dendritic cells (DCs) in Peyer's patches (PPs) and increased the serum levels of IL-4 and IFN-γ, as measured by enzyme-linked immunosorbent assay in mice treated with NC8-pSIP409-pgsA-VP7-DCpep. Compared to the empty vector group, NC8-pSIP409-pgsA-VP7-DCpep significantly increased the production of B220+ B cells in mesenteric lymph nodes (MLNs) and PPs and also increased the titer levels of the VP7-specific antibodies, including IgG and sIgA. The administration of NC8-pSIP409-pgsA-VP7-DCpep mediated relatively broad cellular responses. This study reveals that clear alternatives exist for PRV control strategies and provides information on PRV infection.


Asunto(s)
Anticuerpos Antivirales/inmunología , Antígenos Virales/genética , Antígenos Virales/inmunología , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Ingeniería Genética/métodos , Inmunización/métodos , Inmunogenicidad Vacunal , Lactobacillus plantarum/genética , Lactobacillus plantarum/metabolismo , Vacunas Sintéticas/administración & dosificación , Animales , Antígenos Heterófilos/genética , Antígenos Heterófilos/inmunología , Antígenos Heterófilos/metabolismo , Antígenos Virales/metabolismo , Linfocitos B/inmunología , Proteínas de la Cápside/metabolismo , Citocinas/sangre , Femenino , Genes Virales , Inmunoglobulina A Secretora/inmunología , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Rotavirus/inmunología , Rotavirus/metabolismo , Porcinos , Vacunas Sintéticas/inmunología
4.
J Virol ; 93(18)2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31243134

RESUMEN

Neutralization by antibodies and complement limits the effective dose and thus the therapeutic efficacy of oncolytic viruses after systemic application. We and others previously showed that pseudotyping of oncolytic rhabdoviruses such as maraba virus and vesicular stomatitis virus (VSV) with the lymphocytic choriomeningitis virus glycoprotein (LCMV-GP) results in only a weak induction of neutralizing antibodies. Moreover, LCMV-GP-pseudotyped VSV (VSV-GP) was significantly more stable in normal human serum (NHS) than VSV. Here, we demonstrate that depending on the cell line used for virus production, VSV-GP showed different complement sensitivities in nonimmune NHS. The NHS-mediated titer reduction of VSV-GP was dependent on activation of the classical complement pathway, mainly by natural IgM antibodies against xenoantigens such as galactose-α-(1,3)-galactose (α-Gal) or N-glycolylneuraminic acid (Neu5Gc) expressed on nonhuman production cell lines. VSV-GP produced on human cell lines was stable in NHS. However, VSV-GP generated in transduced human cells expressing α-Gal became sensitive to NHS. Furthermore, GP-specific antibodies induced complement-mediated neutralization of VSV-GP independently of the producer cell line, suggesting that complement regulatory proteins potentially acquired by the virus during the budding process are not sufficient to rescue the virus from antibody-dependent complement-mediated lysis. Thus, our study points to the importance of a careful selection of cell lines for viral vector production for clinical use.IMPORTANCE Systemic application aims to deliver oncolytic viruses to tumors as well as to metastatic lesions. However, we found that xenoantigens incorporated onto the viral surface from nonhuman production cell lines are recognized by natural antibodies in human serum and that the virus is thereby inactivated by complement lysis. Hence, to maximize the effective dose, careful selection of cell lines for virus production is crucial.


Asunto(s)
Virus de la Coriomeningitis Linfocítica/inmunología , Estomatitis Vesicular/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología , Células A549 , Animales , Anticuerpos Neutralizantes/inmunología , Antígenos Heterófilos/inmunología , Línea Celular , Chlorocebus aethiops , Proteínas del Sistema Complemento/inmunología , Cricetinae , Vectores Genéticos , Glicoproteínas/genética , Humanos , Ratones , Viroterapia Oncolítica/métodos , Virus Oncolíticos/metabolismo , Virus de la Estomatitis Vesicular Indiana/genética , Virus de la Estomatitis Vesicular Indiana/fisiología , Vesiculovirus/genética
5.
Glycoconj J ; 37(4): 485-498, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32542517

RESUMEN

Animal bioprosthetic heart valves (BHV) are used to replace defective valves in patients with valvular heart disease. Especially young BHV recipients may experience a structural valve deterioration caused by an immune reaction in which α-Gal and Neu5Gc are potential target antigens. The expression of these and other carbohydrate antigens in animal tissues used for production of BHV was explored. Protein lysates of porcine aortic and pulmonary valves, and porcine, bovine and equine pericardia were analyzed by Western blotting using anti-carbohydrate antibodies and lectins. N-glycans were released by PNGase F digestion and O-glycans by ß-elimination. Released oligosaccharides were analyzed by liquid chromatography - tandem mass spectrometry. In total, 102 N-glycans and 40 O-glycans were identified in animal heart tissue lysates. The N- and O-glycan patterns were different between species. α-Gal and Neu5Gc were identified on both N- and O-linked glycans, N,N´-diacetyllactosamine (LacdiNAc) on N-glycans only and sulfated O-glycans. The relative amounts of α-Gal-containing N-glycans were higher in bovine compared to equine and porcine pericardia. In contrast to the restricted number of proteins carrying α-Gal and LacdiNAc, the distribution of proteins carrying Neu5Gc-determinants varied between species and between different tissues of the same species. Porcine pericardium carried the highest level of Neu5Gc-sialylated O-glycans, and bovine pericardium the highest level of Neu5Gc-sialylated N-glycans. The identified N- and O-linked glycans, some of which may be immunogenic and remain in BHVs manufactured for clinical use, could direct future genetic engineering to prevent glycan expression rendering the donor tissues less immunogenic in humans.


Asunto(s)
Antígenos Heterófilos/análisis , Antígenos Heterófilos/inmunología , Miocardio/metabolismo , Animales , Antígenos Heterófilos/metabolismo , Válvula Aórtica/metabolismo , Bovinos , Caballos , Immunoblotting , Antígenos del Grupo Sanguíneo de Lewis/metabolismo , Pericardio/metabolismo , Polisacáridos/química , Polisacáridos/metabolismo , Válvula Pulmonar/metabolismo , Porcinos , Espectrometría de Masas en Tándem
6.
Xenotransplantation ; 27(6): e12629, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32697003

RESUMEN

Progress has been made in overcoming antibody-mediated rejection of porcine xenografts by deleting pig genes that produce unique carbohydrate epitopes. Pigs deficient in galactose α-1,3 galactose (gene modified: GGTA1) and neu5Gc (gene modified: CMAH) have reduced levels of human antibody binding. Previously we identified α-fucose as a glycan that was expressed in high levels on cells of GGTA1/CMAH KO pigs. To validate the α-fucose phenotype observed previously we compared lectin affinity toward human and pig serum glycoproteins by dot blot analysis and confocal microscopy. Human anti-fucose antibody isolated by affinity chromatography was tested for specificity to L-fucose by custom macroarray. The affinity and cytotoxicity of the isolated human anti-fucose antibody toward human and GGTA1/CMAH KO pig PBMCs was determined by flow cytometry. Dot blot and confocal analysis support out previous findings that α-fucose is more highly expressed in pigs than humans. Pig kidney glomeruli and tubules contain abundant α-fucose and may represent focal sites for anti-α-fucose antibody binding. The Isolated human anti-fucose IgA, IgG and IgM bound to GGTA1/CMAH KO pig PBMC and were cytotoxic. Interestingly, the isolated human IgG cross reacted with the methyl pentose, L-rhamnose. Human anti-fucose antibody bound and was cytotoxic to GGTA1/CMAH KO pig peripheral blood monocytes. We have shown that α-fucose is an abundant target for cytotoxic human antibody in the organs of genetically modified pigs important to xenotransplantation.


Asunto(s)
Animales Modificados Genéticamente , Antígenos Heterófilos/inmunología , Fucosa , Trasplante Heterólogo , Animales , Fucosa/inmunología , Galactosiltransferasas , Técnicas de Inactivación de Genes , Humanos , Leucocitos Mononucleares , Oxigenasas de Función Mixta , Porcinos
7.
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
8.
Xenotransplantation ; 26(4): e12523, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31074044

RESUMEN

BACKGROUND: Differences in quality and strength of immune responses between individuals are mainly due to polymorphisms in major histocompatibility complex (MHC) molecules. Focusing on MHC class-II, we asked whether the intensity of human anti-pig T-cell responses is influenced by genetic variability in the human HLA-DRB1 and/or the porcine SLA-DRB1 locus. METHODS: ELISpot assays were performed using peripheral blood mononuclear cells (PBMCs) from 62 HLA-DRB1-typed blood donors as responder and the porcine B cell line L23 as stimulator cells. Based on the frequency of IFN-γ-secreting cells, groups of weak, medium, and strong responder individuals were defined. Mixed lymphocyte reaction (MLR) assays were performed to study the stimulatory capacity of porcine PBMCs expressing different SLA-DRB1 alleles. RESULTS: Concerning the MHC class-II configuration of human cells, we found a significant overrepresentation of HLA-DRB1*01 alleles in the medium/strong responder group as compared to individuals showing weak responses to stimulation with L23 cells. Evaluation of the role of MHC class-II variability in porcine stimulators revealed that cells expressing SLA-DRB1*06 alleles triggered strong proliferation in approximately 70% of humans. Comparison of amino acid sequences indicated that strong human anti-pig reactivity may be associated with a high rate of similarity between human and pig HLA/SLA-DRB1 alleles. CONCLUSION: Variability in human and porcine MHC determines the intensity of individual human anti-pig T-cell responses. MHC typing and cross-matching of prospective recipients of xenografts and donor pigs could be relevant to select for donor-recipient combinations with minimal anti-porcine immunity.


Asunto(s)
Antígenos Heterófilos/inmunología , Variación Biológica Individual , Cadenas HLA-DRB1/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Porcinos/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Linfocitos T/inmunología , Alelos , Secuencia de Aminoácidos , Animales , Genotipo , Cadenas HLA-DRB1/genética , Haplotipos , Humanos , Interferón gamma/biosíntesis , Células Asesinas Naturales/metabolismo , Leucocitos Mononucleares/inmunología , Activación de Linfocitos , Ratas , Alineación de Secuencia , Homología de Secuencia de Aminoácido
9.
Xenotransplantation ; 26(5): e12524, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31115108

RESUMEN

Two well-characterized carbohydrate epitopes are absent in humans but present in other mammals. These are galactose-α1,3-galactose (αGal) and N-glycolylneuraminic acid (Neu5Gc) which are introduced by the activities of two enzymes including α(1,3) galactosyltransferase (encoded by the GGTA1 gene) and CMP-Neu5Gc hydroxylase (encoded by the CMAH gene) that are inactive in humans but present in cattle. Hence, bovine-derived products are antigenic in humans who receive bioprosthetic heart valves (BHVs) or those that suffer from red meat syndrome. Using programmable nucleases, we disrupted (knockout, KO) GGTA1 and CMAH genes encoding for the enzymes that catalyse the synthesis of αGal and Neu5Gc, respectively, in both male and female bovine fibroblasts. The KO in clonally selected fibroblasts was detected by polymerase chain reaction (PCR) and confirmed by Sanger sequencing. Selected fibroblasts colonies were used for somatic cell nuclear transfer (SCNT) to produce cloned embryos that were implanted in surrogate recipient heifers. Fifty-three embryos were implanted in 33 recipients heifers; 3 pregnancies were carried to term and delivered 3 live calves. Primary cell cultures were established from the 3 calves and following molecular analyses confirmed the genetic deletions. FACS analysis showed the double-KO phenotype for both antigens confirming the mutated genotypes. Availability of such cattle double-KO model lacking both αGal and Neu5Gc offers a unique opportunity to study the functionality of BHV manufactured with tissues of potentially lower immunogenicity, as well as a possible new clinical approaches to help patients with red meat allergy syndrome due to the presence of these xenoantigens in the diet.


Asunto(s)
Animales Modificados Genéticamente , Antígenos Heterófilos/metabolismo , Citidina Monofosfato/análogos & derivados , Galactosa/metabolismo , Galactosiltransferasas/genética , Técnicas de Inactivación de Genes , Oxigenasas de Función Mixta/genética , Ácidos Neuramínicos/metabolismo , Animales , Antígenos Heterófilos/inmunología , Bioprótesis , Bovinos , Citidina Monofosfato/inmunología , Citidina Monofosfato/metabolismo , Femenino , Fibroblastos/inmunología , Hipersensibilidad a los Alimentos/inmunología , Galactosa/inmunología , Galactosiltransferasas/deficiencia , Prótesis Valvulares Cardíacas , Humanos , Masculino , Oxigenasas de Función Mixta/deficiencia , Ácidos Neuramínicos/inmunología , Trasplante Heterólogo
10.
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
11.
Curr Opin Organ Transplant ; 24(5): 535-542, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31348015

RESUMEN

PURPOSE OF REVIEW: The use of genetically modified pigs has resulted in prolonged xenograft organ survival, overcoming the initial barriers that lead to hyperacute rejection and immediate loss of the graft. The purpose of the present review is to revisit the xenogeneic response and the pathologic changes in the xenograft organ in the context of recent publications of large animal studies that highlight existing challenges. RECENT FINDINGS: Transgenic modifications that have included complement regulatory proteins and coagulation regulatory proteins have prolonged xenograft survival in pig to nonhuman primate kidneys, livers, and hearts. Modifications of immunosuppressive regimens such as the addition of mTOR inhibition and costimulatory blockade have also led to better outcomes. Antibody-mediated rejection and thrombotic microangiopathy persist as primary challenges to the field and require further systematic exploration. SUMMARY: The efforts to overcome the natural antibody response to xenoantigens are largely sufficient. There is great opportunity for designing immunosuppression protocols and for detecting early coagulopathies, complement activation, and donor-specific antibody response. With graft survival prolongation, there is also a greater need to understand mechanisms and to enhance diagnostic tools for pathologic evaluation.


Asunto(s)
Animales Modificados Genéticamente , Rechazo de Injerto/patología , Trasplante de Órganos/métodos , Trasplante Heterólogo/métodos , Animales , Antígenos Heterófilos/inmunología , Modelos Animales de Enfermedad , Rechazo de Injerto/inmunología , Supervivencia de Injerto/inmunología , Humanos , Inmunosupresores , Porcinos
12.
J Infect Dis ; 217(11): 1798-1808, 2018 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-29415180

RESUMEN

Background: BCG vaccination is associated with a reduction in all-cause infant mortality in high-mortality settings. The underlying mechanisms remain uncertain, but long-term modulation of the innate immune response (trained immunity) may be involved. Methods: Whole-blood specimens, collected 7 days after randomization from 212 neonates enrolled in a randomized trial of neonatal BCG vaccination, were stimulated with killed pathogens and Toll-like receptor (TLR) ligands to interrogate cytokine responses. Results: BCG-vaccinated infants had increased production of interleukin 6 (IL-6) in unstimulated samples and decreased production of interleukin 1 receptor antagonist, IL-6, and IL-10 and the chemokines macrophage inflammatory protein 1α (MIP-1α), MIP-1ß, and monocyte chemoattractant protein 1 (MCP-1) following stimulation with peptidoglycan (TLR2) and R848 (TLR7/8). BCG-vaccinated infants also had decreased MCP-1 responses following stimulation with heterologous pathogens. Sex and maternal BCG vaccination status interacted with neonatal BCG vaccination. Conclusions: Neonatal BCG vaccination influences cytokine responses to TLR ligands and heterologous pathogens. This effect is characterized by decreased antiinflammatory cytokine and chemokine responses in the context of higher levels of IL-6 in unstimulated samples. This supports the hypothesis that BCG vaccination modulates the innate immune system. Further research is warranted to determine whether there is an association between these findings and the beneficial nonspecific (heterologous) effects of BCG vaccine on all-cause mortality.


Asunto(s)
Antígenos Heterófilos/inmunología , Vacuna BCG/inmunología , Citocinas/inmunología , Receptores Toll-Like/inmunología , Adulto , Quimiocina CCL2/inmunología , Quimiocina CCL3/inmunología , Quimiocina CCL4/inmunología , Femenino , Humanos , Interleucina-10/inmunología , Interleucina-6/inmunología , Ligandos , Masculino , Receptor Toll-Like 2/inmunología , Vacunación/métodos
13.
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
14.
Biochem Biophys Res Commun ; 480(3): 474-478, 2016 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-27773813

RESUMEN

The number of patients in need of organ transplantation is continuously on the rise. However, because of organ donor shortage, xenotransplantation has been highlighted as an alternative. Among the various porcine organs and tissues, porcine islets are considered to be the best-matching implantable candidates for clinical application based on recent progress in nonhuman primate pre-clinical studies. Nevertheless, before initiation of clinical trials, it should be confirmed whether the requisite xeno-antigen sensitization would have a deleterious effect on subsequent allo-transplantation or vice versa. Therefore, in the present study, the survival rate of islets grafted in naïve recipients was compared with that in cross-sensitized recipients. Enzyme-linked immunosorbent spot, fluorescence-activated cell sorting, and immunohistochemistry were conducted to assess the cellular and humoral immune responses. The survival days of Balb/c mouse islets transplanted into B6 mice that had been previously sensitized with porcine cells (i.e., xeno-sensitized) showed no significant difference from that of naïve B6 mice. Moreover, the survival days of porcine islets transplanted into allo-antigen (Balb/c)-sensitized B6 recipients was not significantly different from that in naïve B6 mice. Furthermore, our data provide the first demonstration that the cellular xenogeneic immune response (against porcine antigen) measured by an enzyme-linked immunosorbent spot assay is not cross-reactive to the allogeneic immune responses in a murine islet transplantation model. These results suggest that clinical application of islet xenotransplantation is not likely to have a deleterious effect on subsequent allogeneic islet transplantation.


Asunto(s)
Antígenos Heterófilos/inmunología , Reacciones Cruzadas/inmunología , Supervivencia de Injerto/inmunología , Trasplante de Islotes Pancreáticos/métodos , Islotes Pancreáticos/inmunología , Isoantígenos/inmunología , Animales , Anticuerpos Heterófilos/inmunología , Inmunización/métodos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Porcinos
15.
Basic Res Cardiol ; 111(4): 39, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27154491

RESUMEN

Pre-clinical and clinical data have unequivocally demonstrated the usefulness of decellularized heart valve (HV) matrices implanted for HV replacement therapy. However, human donor valves applicable for decellularization are in short supply, which prompts the search for suitable alternatives, such as porcine grafts. Since decellularization might be insufficient to remove all xenoantigens, we analysed the interaction of human preformed antibodies with decellularized porcine HV in vitro to assess potential immune reactions upon implantation. Detergent-decellularized pulmonary HV from German Landrace wild-type (wt) or α1,3-galactosyltransferase knockout (GGTA1-KO) pigs were investigated by inhibition ELISA and GSL I-B4 staining to localize and quantify matrix-bound αGal epitopes, which represent the most prominent xenoantigen. Additionally, preformed human xenoantibodies were affinity purified by perfusing porcine kidneys. Binding of purified human antibodies to decellularized HV was investigated by inhibition ELISA. Furthermore, binding of human plasma proteins to decellularized matrices was determined by western blot. Decellularized human pulmonary artery served as controls. Decellularization of wt HV led to a reduction of αGal epitopes by 70 %. Residual epitopes were associated with the subendothelial extracellular matrix. As expected, no αGal epitopes were found on decellularized GGTA1-KO matrix. The strongest binding of preformed human anti-pig antibodies was found on wt matrices, whereas GGTA1-KO matrices bound similar or even fewer xenoantibodies than human controls. These results demonstrate the suitability of GGTA1-KO pigs as donors for decellularized heart valves for human patients. Besides the presence of αGal antibodies on decellularized heart valves, no further preformed xenoantibodies against porcine matrix were detected in tested human sera.


Asunto(s)
Anticuerpos Heterófilos/inmunología , Galactosiltransferasas/deficiencia , Prótesis Valvulares Cardíacas , Válvulas Cardíacas/inmunología , Xenoinjertos/inmunología , Animales , Antígenos Heterófilos/inmunología , Bioprótesis , Western Blotting , Técnica del Anticuerpo Fluorescente , Técnicas de Inactivación de Genes , Humanos , Porcinos
16.
Transfusion ; 56(10): 2495-2501, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27383738

RESUMEN

BACKGROUND: Complement has significant status in the field of transfusion medicine. The accepted stability profile of complement is based on historical studies of diluted human serum hemolyzing rabbit heterophile antibody-sensitized sheep red blood cells (RBCs). Contemporary tools are available to reevaluate these historical observations using human heterophile antibodies, undiluted serum, and antigen-modified human RBCs. STUDY DESIGN AND METHODS: Human RBCs were made into "animal-like" kodecytes with heterophile Galα3Galß4GlcNAcß function-spacer-lipid constructs. These α-Gal-kodecytes were prepared with an antigen dilution capable of consistently producing 50% antibody-mediated hemolysis against human α1-3galactose heterophile antibodies and undiluted standardized serum. Standardized human serum aliquots from a two-donor pool stored at -85, -20, 4, 22, and 37°C for durations of up to 150 days were evaluated for loss of hemolytic activity. Where practical methodologic procedures were aligned with historical studies. RESULTS: Comparison of the historical assay with the α-Gal-kodecyte assay against complement activity standards showed concordance. However, in most scenarios complement was found to be more than twice as stable as generally accepted. At least 60% of complement hemolytic activity was observed in serum stored at 22°C for 1 week or 2 months at 4°C. No loss of hemolytic activity was observed after 5 months' storage at temperatures below -20°C. CONCLUSIONS: An alternative method using undiluted serum and modified human RBCs observed that classical-pathway complement hemolytic activity in stored human serum is at least twice as stable as previously accepted.


Asunto(s)
Anticuerpos Heterófilos/inmunología , Vía Clásica del Complemento , Proteínas del Sistema Complemento/química , Hemólisis/inmunología , Animales , Antígenos Heterófilos/inmunología , Activación de Complemento , Proteínas del Sistema Complemento/inmunología , Galactosa/inmunología , Humanos , Estabilidad Proteica
17.
Xenotransplantation ; 23(5): 370-80, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27511593

RESUMEN

BACKGROUND: Glutaraldehyde-fixed bioprosthetic heart valves (GBHVs) derived from wild-type (WT, genetically unmodified) pigs are widely used clinically for heart valve replacement. There is evidence that their failure is related to an immune response. The use of valves from genetically engineered pigs that do not express specific pig antigens may prolong GBHV survival. Our aims were to determine (i) expression of Gal and NeuGc on heart (aortic and pulmonary) valves and pericardium of WT, α1,3-galactosyltransferase gene knockout (GTKO) and GTKO/N-glycolylneuraminic acid gene-knockout (GTKO/NeuGcKO) pigs in comparison with three different commercially available GBHVs and (ii) to determine human antibody binding to these tissues. METHODS: Wild-type, GTKO/CD46, and GTKO/CD46/NeuGcKO pig valves and pericardium were tested (i) fresh and (ii) after fixation with glutaraldehyde (0.02%, 0.2%, 2%). Sections of GBHVs, fresh and fixed valves, and pericardium were stained for Gal and NeuGc expression, and for human IgM and IgG antibody binding. RESULTS: Gal and NeuGc expression was high on all GBHVs and WT pig valves/pericardium, but was absent after antigen-specific-knockout. There was no difference in antigen expression or antibody binding among WT aortic, pulmonary valves, and pericardium as well as GBHVs. Glutaraldehyde fixation did not alter expression of Gal or NeuGc. After incubation with human serum, human IgM and IgG bound to all GBHVs and WT pig valves/pericardium. Valves from GTKO/CD46 pigs and, particularly, GTKO/CD46/NeuGcKO pigs (with/without glutaraldehyde fixation) showed less IgM and IgG binding. CONCLUSION: Compared to WT pigs, GTKO/CD46/NeuGcKO pigs would be preferable sources of GBHVs, because the absence of Gal/NeuGc expression reduces human antibody binding.


Asunto(s)
Antígenos Heterófilos/inmunología , Válvulas Cardíacas/inmunología , Trasplante Heterólogo , Animales , Animales Modificados Genéticamente , Bioprótesis , Técnicas de Inactivación de Genes/métodos , Válvulas Cardíacas/patología , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Porcinos , Trasplante Heterólogo/métodos
18.
J Virol ; 88(11): 6243-54, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24648461

RESUMEN

UNLABELLED: Prime-boost immunization regimens have proven efficacious at generating robust immune responses. However, whether the level of replication of the boosting antigen impacts the magnitude and protective efficacy of vaccine-elicited immune responses remains unclear. To evaluate this, we primed mice with replication-defective adenovirus vectors expressing the lymphocytic choriomeningitis virus (LCMV) glycoprotein (GP), followed by boosting with either LCMV Armstrong, which is rapidly controlled, or LCMV CL-13, which leads to a more prolonged exposure to the boosting antigen. Although priming of naive mice with LCMV CL-13 normally results in T cell exhaustion and establishment of chronic infection, boosting with CL-13 resulted in potent recall CD8 T cell responses that were greater than those following boosting with LCMV Armstrong. Furthermore, following the CL-13 boost, a greater number of anamnestic CD8 T cells localized to the lymph nodes, exhibited granzyme B expression, and conferred improved protection against Listeria and vaccinia virus challenges compared with the Armstrong boost. Overall, our findings suggest that the replicative capacity of the boosting antigen influences the protective efficacy afforded by prime-boost vaccine regimens. These findings are relevant for optimizing vaccine candidates and suggest a benefit of robustly replicating vaccine vectors. IMPORTANCE: The development of optimal prime-boost vaccine regimens is a high priority for the vaccine development field. In this study, we compared two boosting antigens with different replicative capacities. Boosting with a more highly replicative vector resulted in augmented immune responses and improved protective efficacy.


Asunto(s)
Inmunidad Heteróloga/inmunología , Inmunización Secundaria/métodos , Vacunas Virales/inmunología , Replicación Viral/fisiología , Adenoviridae , Animales , Antígenos Heterófilos/inmunología , Linfocitos T CD8-positivos/inmunología , Femenino , Citometría de Flujo , Vectores Genéticos , Glicoproteínas/metabolismo , Estimación de Kaplan-Meier , Listeria/inmunología , Virus de la Coriomeningitis Linfocítica/metabolismo , Ratones , Ratones Endogámicos C57BL , Estadísticas no Paramétricas , Virus Vaccinia/inmunología
19.
Xenotransplantation ; 22(5): 368-78, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26314946

RESUMEN

BACKGROUND: The objective of this study was to investigate the humoral immune response to xenogeneic antigens administered during the fetal state utilizing a baboon-to-pig model. METHODS: Nine fetuses from an alpha-1,3-galactosyltransferase gene knockout (GalT-KO) MGH-miniature swine sow underwent transuterine ultrasound-guided intraportal injection of T-cell depleted baboon bone marrow (B-BM) at mid-gestation. Two juvenile GalT-KO swine undergoing direct B-BM intraportal injection were used as controls. RESULTS: Postnatal humoral tolerance was induced in the long-term surviving piglets as demonstrated by the absence of any antibody response to baboon donor cells. In addition, a second intraportal B-BM administration at 2.5 months post-birth led to no antibody formation despite re-exposure to xenogeneic antigens. This B-cell unresponsiveness was abrogated only when the animal was exposed subcutaneously to third-party xenogeneic and allogeneic antigens, suggesting that the previously achieved humoral non-responsiveness was donor specific. In comparison, the two juvenile GalT-KO control swine demonstrated increasing anti-baboon IgM and IgG levels following intraportal injection. CONCLUSIONS: In summary, xenogeneic B-cell tolerance was induced through in utero intraportal exposure to donor cells and this tolerance persisted following postnatal rechallenge with donor B-BM, but was lost on exposure to third-party antigen, possibly as a result of cross-reactive antibody formation.


Asunto(s)
Antígenos Heterófilos/inmunología , Linfocitos B/inmunología , Trasplante de Médula Ósea/métodos , Inmunidad Humoral , Papio/inmunología , Porcinos/inmunología , Trasplante Heterólogo/métodos , Animales , Anticuerpos Heterófilos/inmunología , Femenino , Tolerancia Inmunológica , Embarazo , Efectos Tardíos de la Exposición Prenatal/inmunología
20.
Xenotransplantation ; 22(2): 85-94, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25308416

RESUMEN

Human beings do not synthesize the glycolyl form of the sialic acid (Neu5Gc) and only express the acetylated form of the sugar, whereas a diet-based intake of Neu5Gc provokes a natural immunization and production of anti-Neu5Gc antibodies in human serum. However, Neu5Gc is expressed on mammal glycoproteins and glycolipids in most organs and cells. We review here the relevance of Neu5Gc and anti-Neu5Gc antibodies in the context of xenotransplantation and the use of animal-derived molecules and products, as well as the possible consequences of a long-term exposure to anti-Neu5Gc antibodies in recipients of xenografts. In addition, the importance of an accurate estimation of the anti-Neu5Gc response following xenotransplantation and the future contribution of knockout animals mimicking the human situation are also assessed.


Asunto(s)
Anticuerpos Heterófilos/sangre , Ácidos Neuramínicos/inmunología , Trasplante Heterólogo/efectos adversos , Animales , Animales de Laboratorio , Antígenos Heterófilos/genética , Antígenos Heterófilos/inmunología , Humanos , Inmunidad Innata , Modelos Animales , Trasplante de Órganos/efectos adversos , Primates , Sus scrofa/genética , Sus scrofa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA