Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
Sci Rep ; 11(1): 13131, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34162938

ABSTRACT

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.


Subject(s)
Animals, Genetically Modified/immunology , Antigens, Heterophile/immunology , Endothelial Cells/immunology , Swine/immunology , Transplantation, Heterologous/methods , Animals , Antibodies, Heterophile/immunology , Antigen-Antibody Reactions , Antigens, Heterophile/genetics , CRISPR-Cas Systems , Cell Degranulation , Cell Line, Transformed , Cytokines/pharmacology , Endothelial Cells/drug effects , Galactosyltransferases/genetics , Galactosyltransferases/immunology , Gene Knockout Techniques , Graft Rejection/immunology , Graft Rejection/prevention & control , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Killer Cells, Natural/immunology , Liver/cytology , Lymphocyte Activation , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/immunology , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/immunology , beta 2-Microglobulin/genetics , beta 2-Microglobulin/immunology
2.
Acta Biochim Biophys Sin (Shanghai) ; 53(6): 707-718, 2021 May 21.
Article in English | MEDLINE | ID: mdl-33963824

ABSTRACT

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.


Subject(s)
Antibodies, Viral/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , Capsid Proteins/genetics , Capsid Proteins/immunology , Genetic Engineering/methods , Immunization/methods , Immunogenicity, Vaccine , Lactobacillus plantarum/genetics , Lactobacillus plantarum/metabolism , Vaccines, Synthetic/administration & dosage , Animals , Antigens, Heterophile/genetics , Antigens, Heterophile/immunology , Antigens, Heterophile/metabolism , Antigens, Viral/metabolism , B-Lymphocytes/immunology , Capsid Proteins/metabolism , Cytokines/blood , Female , Genes, Viral , Immunoglobulin A, Secretory/immunology , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Rotavirus/immunology , Rotavirus/metabolism , Swine , Vaccines, Synthetic/immunology
3.
Transpl Immunol ; 67: 101409, 2021 08.
Article in English | MEDLINE | ID: mdl-34015463

ABSTRACT

Xenotransplantation, using genetically-modified pigs for clinical organ transplantation, is a solution to the organ shortage. The biggest barrier to clinical implementation is the antigenicity of pig cells. Humans possess preformed antibody to pig cells that initiate antibody-mediated rejection of pig organs in primates. Advances in genetic engineering have led to the development of a pig lacking the three known glycan xenoantigens (triple-knockout [TKO] pigs). A significant number of human sera demonstrate no antibody binding to TKO pig cells. As a result of the TKO pig's low antigen expression, survival of life-supporting pig organs in immunosuppressed nonhuman primates has significantly increased, and hope has been renewed for clinical trials of xenotransplantation. It is important to understand the context in which xenotransplantation's predecessor, allotransplantation, has been successful, and the steps needed for the success of xenotransplantation. Successful allotransplantation has been based on two main immunological approaches - (i) adequate immunosuppressive therapy, and (ii) careful histocompatibility matching. In vivo studies suggest that the available immunosuppressive regimens are adequate to suppress the human anti-pig cellular response. Methods to evaluate and screen patients for the first clinical xenotransplantation trial are the next challenge. The goal of this review is to summarize the history of histocompatibility testing, and the available tools that can be utilized to determine xenograft histocompatibility.


Subject(s)
Antigens, Heterophile/genetics , HLA Antigens/genetics , Histocompatibility Antigens Class I/genetics , Histocompatibility Testing/methods , Polysaccharides/genetics , Transplantation, Heterologous , Animals , Animals, Genetically Modified , Antibodies, Heterophile/blood , Antigens, Heterophile/immunology , Cells, Cultured , Gene Knockout Techniques , Histocompatibility , Humans , Polysaccharides/immunology , Swine , Tissue and Organ Procurement
4.
Transplantation ; 105(2): 300-307, 2021 02 01.
Article in English | MEDLINE | ID: mdl-32433239

ABSTRACT

Advances in genetic engineering, particularly CRISPR/Cas9, have resulted in the development of a triple glycan-knockout (TKO) pig. There is minimal human antipig antibody binding to TKO pig cells. The TKO background has decreased antibody binding to a sufficiently low level that any additional xenoantigens expressed on the cells can now be more easily detected. One of these xenoantigens is the swine major histocompatibility complex, termed swine leukocyte antigens (SLA). SLA are the homolog to HLAs, a protein complex expressed on human tissue capable of stimulating the development of new antibodies in allotransplantation. These antibodies can result in graft failure through hyperacute, acute, or chronic rejection. Our knowledge of SLA, particularly in the last 5 years, has grown considerably. The presence, cause, and methods to detect anti-SLA antibodies will need to be carefully considered for the first clinical trial of xenotransplantation. The focus of this review is to summarize the role of SLA in xenotransplantation and consider whether it will prove to be a major barrier. Techniques are now available to mutate target SLA amino acids to ensure that cross-reactive anti-HLA antibodies no longer bind to SLA on the cells of the organ-source pigs. While deletion of SLA expression is possible, it would render the pig at risk for infectious complications. The ideal organ-source pig for HLA highly sensitized recipients may therefore be 1 with site-specific mutations to eliminate cross-reactive binding.


Subject(s)
Antibodies, Heterophile/blood , Antigens, Heterophile/immunology , Graft Rejection/immunology , Histocompatibility Antigens Class I/immunology , Sus scrofa/immunology , Transplantation, Heterologous , Animals , Animals, Genetically Modified , Antibody Specificity , Antigens, Heterophile/genetics , Graft Rejection/blood , Graft Rejection/prevention & control , Graft Survival , Histocompatibility Antigens Class I/genetics , Humans , Species Specificity , Sus scrofa/genetics , Transplantation Tolerance , Transplantation, Heterologous/adverse effects
5.
Viruses ; 12(1)2020 01 03.
Article in English | MEDLINE | ID: mdl-31947825

ABSTRACT

Zika virus (ZIKV) emerged in the Americas in 2015, presenting unique challenges to public health. Unlike other arboviruses of the Flaviviridae family, it is transmissible by sexual contact, which facilitates the spread of the virus into new geographic areas. Additionally, ZIKV can be transmitted from mother to fetus, causing microcephaly and other severe developmental abnormalities. Reliable and easy-to-work-with clones of ZIKV expressing heterologous genes will significantly facilitate studies aimed at understanding the virus pathogenesis and tissue tropism. Here, we developed and characterized two novel approaches for expression of heterologous genes of interest in the context of full-length ZIKV genome and compared them to two previously published strategies for ZIKV-mediated gene expression. We demonstrated that among the four tested viruses expressing nLuc gene, the virus constructed using a newly developed approach of partial capsid gene duplication (PCGD) attained the highest titer in Vero cells and resulted in the highest level of nLuc expression. Suitability of the PCGD approach for expression of different genes of interest was validated by replacing nLuc sequence with that of eGFP gene. The generated constructs were further characterized in cell culture. Potential applications of ZIKV clones stably expressing heterologous genes include development of detection assays, antivirals, therapeutics, live imaging systems, and vaccines.


Subject(s)
Reverse Genetics/methods , Zika Virus/genetics , Animals , Antigens, Heterophile/genetics , Capsid Proteins/genetics , Chlorocebus aethiops , Cloning, Molecular , DNA Transposable Elements , Gene Expression , Genes, Viral , Genome, Viral , Green Fluorescent Proteins/genetics , Humans , Vero Cells , Virus Replication
6.
Immunogenetics ; 71(7): 479-487, 2019 07.
Article in English | MEDLINE | ID: mdl-31270568

ABSTRACT

Xenotransplantation of pig organs into people may help alleviate the critical shortage of donors which faces organ transplantation. Unfortunately, human antibodies vigorously attack pig tissues preventing the clinical application of xenotransplantation. The swine leukocyte antigens (SLA), homologs of human HLA molecules, can be xenoantigens. SLA molecules, encoded by genes in the pig major histocompatibility complex, contribute to protective immune responses in pig. Therefore, simply inactivating them through genome engineering could reduce the ability of the human immune system to surveil transplanted pig organs for infectious disease or the development of neoplasms. A potential solution to this problem is to identify and modify epitopes in SLA proteins to eliminate their contribution to humoral xenoantigenicity while retaining their biosynthetic competence and ability to contribute to protective immunity. We previously showed that class II SLA proteins were recognized as xenoantigens and mutating arginine at position 55 to proline, in an SLA-DQ beta chain, could reduce human antibody binding. Here, we extend these observations by creating several additional point mutants at position 55. Using a panel of monoclonal antibodies specific for class II SLA proteins, we show that these mutants remain biosynthetically competent. Examining antibody binding to these variants shows that point mutagenesis can reduce, eliminate, or increase antibody binding to class II SLA proteins. Individual mutations can have opposite effects on antibody binding when comparing samples from different people. We also performed a preliminary analysis of creating point mutants near to position 55 to demonstrate that manipulating additional residues also affects antibody reactivity.


Subject(s)
Antibodies, Monoclonal/metabolism , Epitopes/genetics , Histocompatibility Antigens Class I/metabolism , Animals , Antigens, Heterophile/genetics , Arginine/genetics , HEK293 Cells , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Mutagenesis, Site-Directed , Point Mutation , Swine
7.
Xenotransplantation ; 26(4): e12504, 2019 07.
Article in English | MEDLINE | ID: mdl-30825348

ABSTRACT

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.


Subject(s)
Antigens, Heterophile/immunology , Galactosyltransferases/immunology , Gene Knockout Techniques , Graft Rejection/prevention & control , Mixed Function Oxygenases/immunology , N-Acetylgalactosaminyltransferases/immunology , Swine/immunology , Transplantation, Heterologous , Animals , Animals, Genetically Modified/immunology , Antibodies, Heterophile/biosynthesis , Antibodies, Heterophile/immunology , Antigen-Antibody Reactions , Antigens, Heterophile/genetics , Epitopes/immunology , Galactosyltransferases/deficiency , Galactosyltransferases/genetics , Genetic Engineering , Graft Rejection/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Mixed Function Oxygenases/deficiency , Mixed Function Oxygenases/genetics , N-Acetylgalactosaminyltransferases/deficiency , N-Acetylgalactosaminyltransferases/genetics , Transplantation Immunology
8.
Transplantation ; 102(12): 1974-1982, 2018 12.
Article in English | MEDLINE | ID: mdl-30247446

ABSTRACT

For a clinical trial today, what might realistically be the optimal pig among those currently available? Deletion of expression of the 3 pig carbohydrate antigens, against which humans have natural (preformed) antibodies (triple-knockout pigs), should form the basis of any clinical trial. However, because both complement and coagulation can be activated in the absence of antibody, the expression of human complement- and coagulation-regulatory proteins is likely to be important in protecting the graft further. Any genetic manipulation that might reduce inflammation of the graft, for example, expression of hemeoxygenase-1 or A20, may also be beneficial to the long-term survival of the graft. The transgene for human CD47 is likely to have a suppressive effect on monocyte/macrophage and T-cell activity. Furthermore, deletion of xenoantigen expression and expression of a human complement-regulatory protein are both associated with a reduced T-cell response. Although there are several other genetic manipulations that may reduce the T-cell response further, it seems likely that exogenous immunosuppressive therapy, particularly if it includes costimulation blockade, will be sufficient. We would therefore suggest that, with our present knowledge and capabilities, the optimal pig might be a triple-knockout pig that expressed 1 or more human complement-regulatory proteins, 1 or more human coagulation-regulatory proteins, a human anti-inflammatory transgene, and CD47. Absent or minimal antibody binding is important, but we suggest that the additional insertion of protective human transgenes will be beneficial, and may be essential.


Subject(s)
Clinical Trials as Topic , Graft Rejection/prevention & control , Graft Survival , Heart Transplantation/methods , Kidney Transplantation/methods , Sus scrofa/genetics , Animals , Animals, Genetically Modified , Antigens, Heterophile/genetics , Antigens, Heterophile/immunology , Blood Coagulation Factors/genetics , Blood Coagulation Factors/immunology , CD47 Antigen/genetics , CD47 Antigen/immunology , Complement System Proteins/genetics , Complement System Proteins/immunology , Genotype , Graft Rejection/genetics , Graft Rejection/immunology , Heart Transplantation/adverse effects , Humans , Kidney Transplantation/adverse effects , Phenotype , Risk Factors , Sus scrofa/immunology , Transplantation Tolerance , Transplantation, Heterologous
9.
Acta Biomater ; 72: 196-205, 2018 05.
Article in English | MEDLINE | ID: mdl-29631050

ABSTRACT

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.


Subject(s)
Animals, Genetically Modified , Antigens, Heterophile/genetics , Bioprosthesis , Galactosyltransferases/genetics , Gene Deletion , Heart Valve Prosthesis , Mixed Function Oxygenases/genetics , N-Acetylgalactosaminyltransferases/genetics , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/immunology , Antigens, Heterophile/immunology , Galactosyltransferases/immunology , Humans , Mixed Function Oxygenases/immunology , N-Acetylgalactosaminyltransferases/immunology , Swine
10.
EBioMedicine ; 18: 204-215, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28302457

ABSTRACT

Conventional HIV T cell vaccine strategies have not been successful in containing acute peak viremia, nor in providing long-term control. We immunized rhesus macaques intramuscularly and rectally using a heterologous adenovirus vectored SIV vaccine regimen encoding normally weakly immunogenic tat, vif, rev and vpr antigens fused to the MHC class II associated invariant chain. Immunizations induced broad T cell responses in all vaccinees. Following up to 10 repeated low-dose intrarectal challenges, vaccinees suppressed early viral replication (P=0.01) and prevented the peak viremia in 5/6 animals. Despite consistently undetectable viremia in 2 out of 6 vaccinees, all animals showed evidence of infection induced immune responses indicating that infection had taken place. Vaccinees, with and without detectable viremia better preserved their rectal CD4+ T cell population and had reduced immune hyperactivation as measured by naïve T cell depletion, Ki-67 and PD-1 expression on T cells. These results indicate that vaccination towards SIV accessory antigens vaccine can provide a level of acute control of SIV replication with a suggestion of beneficial immunological consequences in infected animals of unknown long-term significance. In conclusion, our studies demonstrate that a vaccine encoding subdominant antigens not normally associated with virus control can exert a significant impact on acute peak viremia.


Subject(s)
Antigens, Heterophile/immunology , Genetic Vectors/immunology , Retroviruses, Simian/physiology , SAIDS Vaccines/immunology , Adenoviridae/genetics , Animals , Antigens, Heterophile/genetics , Antigens, Heterophile/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Genetic Vectors/genetics , Genetic Vectors/metabolism , HEK293 Cells , Humans , Macaca mulatta , Mice , Viremia/immunology , Viremia/prevention & control , Virus Replication/physiology
11.
Transplantation ; 101(4): e86-e92, 2017 04.
Article in English | MEDLINE | ID: mdl-28114170

ABSTRACT

BACKGROUND: Antipig antibodies are a barrier to clinical xenotransplantation. We evaluated antibody binding of waitlisted renal transplant patients to 3 glycan knockout (KO) pig cells and class I swine leukocyte antigens (SLA). METHODS: Peripheral blood mononuclear cells from SLA identical wild type (WT), α1, 3-galactosyltransferase (GGTA1) KO, GGTA1/ cytidine monophosphate-N-acetylneuraminic acid hydroxylase (CMAH) KO, and GGTA1/ CMAH /b1,4 N-acetylgalactosaminyl transferase (B4GalNT2) KO pigs were screened for human antibody binding using flow cytometric crossmatch (FCXM). Sera from 820 patients were screened on GGTA1/CMAH/B4GalNT2 KO cells and a subset with elevated binding was evaluated further. FCXM was performed on SLA intact cells and GGTA1/SLA class I KO cells after depletion with WT pig RBCs to remove cell surface reactive antibodies, but leave SLA antibodies. Lastly, human and pig reactive antibodies were eluted and tested for cross-species binding and reactivity to single-antigen HLA beads. RESULTS: Sequential glycan KO modifications significantly reduce antibody binding of waitlisted patients. Sera exhibiting elevated binding without reduction after depletion with WT RBCs demonstrate reduced binding to SLA class I KO cells. Human IgG, eluted from human and pig peripheral blood mononuclear cells, interacted across species and bound single-antigen HLA beads in common epitope-restricted patterns. CONCLUSIONS: Many waitlisted patients have minimal xenoreactive antibody binding to the triple KO pig, but some HLA antibodies in sensitized patients cross-react with class I SLA. SLA class I is a target for genome editing in xenotransplantation.


Subject(s)
Antibodies, Heterophile/blood , Antigens, Heterophile/immunology , Galactosyltransferases/immunology , Gene Knockout Techniques , Histocompatibility Antigens Class II/immunology , Immunity, Humoral , Immunoglobulin G/blood , Kidney Transplantation , Mixed Function Oxygenases/immunology , N-Acetylgalactosaminyltransferases/immunology , Waiting Lists , Animals , Animals, Genetically Modified , Antigens, Heterophile/genetics , Cross Reactions , Flow Cytometry , Galactosyltransferases/deficiency , Galactosyltransferases/genetics , Genotype , HLA Antigens/immunology , Histocompatibility , Histocompatibility Antigens Class I , Histocompatibility Antigens Class II/genetics , Histocompatibility Testing/methods , Humans , Mixed Function Oxygenases/deficiency , Mixed Function Oxygenases/genetics , N-Acetylgalactosaminyltransferases/deficiency , N-Acetylgalactosaminyltransferases/genetics , Phenotype , Protein Binding , Swine , Transplantation, Heterologous
12.
Transplantation ; 101(3): 517-523, 2017 03.
Article in English | MEDLINE | ID: mdl-27379553

ABSTRACT

BACKGROUND: The rapidly improving tools of genetic engineering may make it possible to overcome the humoral immune barrier that prevents xenotransplantation. We hypothesize that levels of human antibody binding to donor tissues from swine must approximate the antibody binding occurring in allotransplantation. It is uncertain if this is an attainable goal. Here we perform an initial analysis of this issue by comparing human antibody binding to red blood cells (RBC) isolated from knockout swine and to allogeneic or autologous human RBC. METHODS: Human sera were incubated with RBC isolated from various genetically engineered swine or from humans. The level of IgG and IgM binding to these cells were compared using either flow cytometry or a novel mass spectrometric assay. RESULTS: Mass spectroscopic quantitation of human antibody binding demonstrated that as few as 3 gene inactivations can reduce the levels human antibody binding to swine RBC that is as low as autologous human RBC. Flow cytometry showed that RBC from 2-gene knockout swine exhibited less human antibody binding than human blood group O allogeneic RBC in 22% of tested sera. Deletion of a third gene from pigs resulted in 30% of human samples having less IgG and IgM RBC xenoreactivity than alloreactivity. CONCLUSIONS: Xenoantigenicity of swine RBC can be eliminated via gene disruption. These results suggest that the gene knockout approach may be able reduce antigenicity in other pig tissues to levels that enable the xenotransplantation humoral barrier to be overcome.


Subject(s)
Antigens, Heterophile/genetics , Antigens, Heterophile/immunology , Erythrocytes/immunology , Gene Knockout Techniques , Histocompatibility , Swine/immunology , Animals , Animals, Genetically Modified , Antigens, Heterophile/blood , Binding Sites, Antibody , Flow Cytometry , Graft Survival , Humans , Immunity, Humoral , Isoantibodies/blood , Isoantibodies/immunology , Protein Binding , Swine/blood , Swine/genetics , Tandem Mass Spectrometry , Transplantation Tolerance , Transplantation, Heterologous
13.
Methods Mol Biol ; 1349: 225-37, 2016.
Article in English | MEDLINE | ID: mdl-26458839

ABSTRACT

Paramyxoviruses are able to stably express a wide-variety of heterologous antigens at relatively high levels in various species and are consequently considered as potent gene delivery vehicles. A single vaccination is frequently sufficient for the induction of robust humoral and cellular immune responses. Here we provide detailed methods for the construction and application of Newcastle disease virus (NDV)-based vector vaccines. The in silico design and in vitro rescue as well as the in vivo evaluation of NDV based vectors are described in this chapter.


Subject(s)
Antigens, Heterophile/genetics , Immunity, Cellular/genetics , Newcastle Disease/prevention & control , Newcastle disease virus/genetics , Vaccination/methods , Animals , Antigens, Heterophile/administration & dosage , Antigens, Heterophile/immunology , Computer Simulation , Gene Transfer Techniques , Genetic Vectors , Immunity, Cellular/immunology , Newcastle Disease/virology , Newcastle disease virus/immunology , Poultry/virology , Reverse Genetics , Vaccination/veterinary
14.
Xenotransplantation ; 22(4): 302-9, 2015.
Article in English | MEDLINE | ID: mdl-26130164

ABSTRACT

The longest survival of a non-human primate with a life-supporting kidney graft to date has been 90 days, although graft survival > 30 days has been unusual. A baboon received a kidney graft from an α-1,3-galactosyltransferase gene-knockout pig transgenic for two human complement-regulatory proteins and three human coagulation-regulatory proteins (although only one was expressed in the kidney). Immunosuppressive therapy was with ATG+anti-CD20mAb (induction) and anti-CD40mAb+rapamycin+corticosteroids (maintenance). Anti-TNF-α and anti-IL-6R were administered. The baboon survived 136 days with a generally stable serum creatinine (0.6 to 1.6 mg/dl) until termination. No features of a consumptive coagulopathy (e.g., thrombocytopenia, decreased fibrinogen) or of a protein-losing nephropathy were observed. There was no evidence of an elicited anti-pig antibody response. Death was from septic shock (Myroides spp). Histology of a biopsy on day 103 was normal, but by day 136, the kidney showed features of glomerular enlargement, thrombi, and mesangial expansion. The combination of (i) a graft from a specific genetically engineered pig, (ii) an effective immunosuppressive regimen, and (iii) anti-inflammatory agents prevented immune injury and a protein-losing nephropathy, and delayed coagulation dysfunction. This outcome encourages us that clinical renal xenotransplantation may become a reality.


Subject(s)
Kidney Transplantation/methods , Transplantation, Heterologous/methods , Animals , Animals, Genetically Modified , Antigens, Heterophile/genetics , Complement System Proteins/genetics , Galactosyltransferases/deficiency , Galactosyltransferases/genetics , Galactosyltransferases/immunology , Gene Knockout Techniques , Genetic Engineering , Graft Survival/immunology , Humans , Immunosuppressive Agents/administration & dosage , Kidney/immunology , Kidney/pathology , Kidney Transplantation/adverse effects , Papio , Swine , Time Factors , Transplantation, Heterologous/adverse effects
15.
Xenotransplantation ; 22(3): 194-202, 2015.
Article in English | MEDLINE | ID: mdl-25728481

ABSTRACT

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


Subject(s)
Antigens, Heterophile/immunology , Galactosyltransferases/genetics , Leukocytes, Mononuclear/immunology , Transplantation, Heterologous , Animals , Animals, Genetically Modified , Antigens, Heterophile/genetics , Graft Rejection/genetics , Graft Rejection/immunology , Humans , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Swine
16.
Vaccine ; 33(15): 1880-9, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25701672

ABSTRACT

Live attenuated vaccines are cost effective approach for preventing a broad range of infectious diseases, and thus are of great interest. However, immune-defects can predispose the patient to infections by the vaccine candidate itself. So far, few live vaccine candidates have been designed specifically for immune compromised individuals. Recently, we reported a new Salmonella Typhimurium Z234-vaccine strain (Periaswamy et al., PLoS ONE 2012;7:e45433), which was specifically attenuated in the NADPH-oxidase deficient host. In the present study, the Z234-vaccine strain was further engineered to express heterologous antigen (Vibrio cholerae toxin antigen subunit-B, i.e. CtxB) with the intention of creating a vector for simultaneous protection against Cholera and Salmonellosis. The primary aim of this study was to ensure the expression of CtxB antigen by the recombinant vaccine strain Z234-pMS101. The antigen CtxB was expressed through Z234 as a fusion protein with N-terminal signal sequence of Salmonella outer protein (SopE), an effector protein from Salmonella under the control of SopE promoter. The CtxB-expressing plasmid construct pMS101 (pM968-pSopE-ctxB) was found to be stable both in vitro and in vivo. In an oral mouse infection model, the vaccine strain Z234-pMS101 efficiently colonized the host gut. The extent of protection was confirmed after challenging the immunized hosts with live V. cholerae. Vaccinated mice showed reduced gut colonization by V. cholerae. Further assessment of immunological parameters supported the possibility of conferring effective immune response by Z234-pMS101 vaccine strain. Overall, the Z234-pMS101 vaccine strain showed potential as a promising polyvalent vaccine candidate to protect against S. Typhimurium and V. cholerae infection simultaneously.


Subject(s)
Cholera Toxin/immunology , Cholera Vaccines/immunology , Cholera/prevention & control , Salmonella Infections, Animal/prevention & control , Salmonella Vaccines/immunology , Salmonella typhimurium/immunology , Animals , Antibodies, Bacterial/immunology , Antigens, Heterophile/genetics , Antigens, Heterophile/immunology , Cecum/microbiology , Cecum/pathology , Cholera Toxin/genetics , Disease Models, Animal , Feces/microbiology , Genetic Vectors , Immunity, Cellular , Immunocompromised Host , Mice , Mice, Inbred C57BL , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Salmonella typhimurium/growth & development , Vaccination , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Vibrio cholerae/growth & development
17.
Xenotransplantation ; 22(2): 102-11, 2015.
Article in English | MEDLINE | ID: mdl-25470239

ABSTRACT

BACKGROUND: Genetically modified pigs are a promising potential source of lung xenografts. Ex vivo xenoperfusion is an effective platform for testing the effect of new modifications, but typical experiments are limited by testing of a single genetic intervention and small sample sizes. The purpose of this study was to analyze the individual and aggregate effects of donor genetic modifications on porcine lung xenograft survival and injury in an extensive pig lung xenoperfusion series. METHODS: Data from 157 porcine lung xenoperfusion experiments using otherwise unmodified heparinized human blood were aggregated as either continuous or dichotomous variables. Lungs were wild type in 17 perfusions (11% of the study group), while 31 lungs (20% of the study group) had one genetic modification, 40 lungs (39%) had 2, and 47 lungs (30%) had 3 or more modifications. The primary endpoint was functional lung survival to 4 h of perfusion. Secondary analyses evaluated previously identified markers associated with known lung xenograft injury mechanisms. In addition to comparison among all xenografts grouped by survival status, a subgroup analysis was performed of lungs incorporating the GalTKO.hCD46 genotype. RESULTS: Each increase in the number of genetic modifications was associated with additional prolongation of lung xenograft survival. Lungs that exhibited survival to 4 h generally had reduced platelet activation and thrombin generation. GalTKO and the expression of hCD46, HO-1, hCD55, or hEPCR were associated with improved survival. hTBM, HLA-E, and hCD39 were associated with no significant effect on the primary outcome. CONCLUSION: This meta-analysis of an extensive lung xenotransplantation series demonstrates that increasing the number of genetic modifications targeting known xenogeneic lung injury mechanisms is associated with incremental improvements in lung survival. While more detailed mechanistic studies are needed to explore the relationship between gene expression and pathway-specific injury and explore why some genes apparently exhibit neutral (hTBM, HLA-E) or inconclusive (CD39) effects, GalTKO, hCD46, HO-1, hCD55, and hEPCR modifications were associated with significant lung xenograft protection. This analysis supports the hypothesis that multiple genetic modifications targeting different known mechanisms of xenograft injury will be required to optimize lung xenograft survival.


Subject(s)
Animals, Genetically Modified/immunology , Heterografts/immunology , Lung Transplantation/methods , Sus scrofa/genetics , Sus scrofa/immunology , Transplantation, Heterologous/methods , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Heterophile/genetics , Blood/immunology , Endothelial Protein C Receptor , Galactosyltransferases/deficiency , Galactosyltransferases/genetics , Galactosyltransferases/immunology , Gene Knockout Techniques , Graft Survival/genetics , Graft Survival/immunology , Heme Oxygenase-1/genetics , Heme Oxygenase-1/immunology , Humans , In Vitro Techniques , Lung Transplantation/adverse effects , Membrane Cofactor Protein/genetics , Membrane Cofactor Protein/immunology , Perfusion , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Transplantation, Heterologous/adverse effects
18.
Xenotransplantation ; 22(1): 20-31, 2015.
Article in English | MEDLINE | ID: mdl-25178170

ABSTRACT

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.


Subject(s)
CRISPR-Cas Systems , Galactosyltransferases/genetics , Gene Knockout Techniques , Mixed Function Oxygenases/genetics , Nuclear Transfer Techniques , RNA, Guide, Kinetoplastida/genetics , Sus scrofa/genetics , Animals , Antigens, Heterophile/genetics , Biotinylation , Female , Gene Deletion , Genetic Vectors , Hepatocytes/cytology , Immunomagnetic Separation , Phenotype , Plant Lectins/metabolism , Pregnancy , Streptavidin , Swine
19.
Xenotransplantation ; 22(2): 85-94, 2015.
Article in English | MEDLINE | ID: mdl-25308416

ABSTRACT

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.


Subject(s)
Antibodies, Heterophile/blood , Neuraminic Acids/immunology , Transplantation, Heterologous/adverse effects , Animals , Animals, Laboratory , Antigens, Heterophile/genetics , Antigens, Heterophile/immunology , Humans , Immunity, Innate , Models, Animal , Organ Transplantation/adverse effects , Primates , Sus scrofa/genetics , Sus scrofa/immunology
20.
Benef Microbes ; 6(3): 313-24, 2015.
Article in English | MEDLINE | ID: mdl-25245573

ABSTRACT

UNLABELLED: Lactic acid bacteria (LABs) are good candidates for the development of new oral vaccines and are attractive alternatives to attenuated pathogens. This review focuses on the use of wild-type and recombinant lactococci and lactobacilli with emphasis on their molecular design, immunomodulation and treatment of bacterial infections. The majority of studies related to recombinant LABs have focused on Lactococcus lactis, however, molecular tools have been successfully used for Lactobacillus spp. RESEARCH: Recombinant lactobacilli and lactococci have several health benefits, such as immunomodulation, restoration of the microbiota, synthesis of antimicrobial substances and inhibition of virulence factors. In addition, protective immune responses that are well tolerated are induced by the expression of heterologous antigens from recombinant probiotics.


Subject(s)
Antigens, Heterophile/genetics , Drug Delivery Systems/methods , Genetic Vectors/genetics , Lactobacillales/genetics , Vaccination/methods , Animals , Antigens, Heterophile/administration & dosage , Antigens, Heterophile/immunology , Drug Delivery Systems/trends , Genetic Vectors/immunology , Humans , Lactobacillales/metabolism , Vaccination/trends
SELECTION OF CITATIONS
SEARCH DETAIL
...