Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Xenotransplantation ; 31(3): e12851, 2024.
Article in English | MEDLINE | ID: mdl-38747130

ABSTRACT

BACKGROUND: The German Xenotransplantation Consortium is in the process to prepare a clinical trial application (CTA) on xenotransplantation of genetically modified pig hearts. In the CTA documents to the central and national regulatory authorities, that is, the European Medicines Agency (EMA) and the Paul Ehrlich Institute (PEI), respectively, it is required to list the potential zoonotic or xenozoonotic porcine microorganisms including porcine viruses as well as to describe methods of detection in order to prevent their transmission. The donor animals should be tested using highly sensitive detection systems. I would like to define a detection system as the complex including the actual detection methods, either PCR-based, cell-based, or immunological methods and their sensitivity, as well as sample generation, sample preparation, sample origin, time of sampling, and the necessary negative and positive controls. Lessons learned from the identification of porcine cytomegalovirus/porcine roseolovirus (PCMV/PRV) in the xenotransplanted heart in the recipient in the Baltimore study underline how important such systems are. The question is whether veterinary laboratories can supply such assays. METHODS: A total of 35 veterinary laboratories in Germany were surveyed for their ability to test for selected xenotransplantation-relevant viruses, including PCMV/PRV, hepatitis E virus, and porcine endogenous retrovirus-C (PERV-C). As comparison, data from Swiss laboratories and a laboratory in the USA were analyzed. Furthermore, we assessed which viruses were screened for in clinical and preclinical trials performed until now and during screening of pig populations. RESULTS: Of the nine laboratories that provided viral diagnostics, none of these included all potential viruses of concern, indeed, the most important assays confirmed in recent human trials, antibody detection of PCMV/PRV and screening for PERV-C were not available at all. The situation was similar in Swiss and US laboratories. Different viruses have been tested for in first clinical and preclinical trials performed in various countries. CONCLUSION: Based on these results it is necessary to establish special virological laboratories able to test for all xenotransplantation-relevant viruses using validated assays, optimally in the xenotransplantation centers.


Subject(s)
Transplantation, Heterologous , Animals , Transplantation, Heterologous/methods , Swine , Humans , Viruses/isolation & purification , Laboratories , Germany , Virus Diseases/diagnosis , Heart Transplantation , Heterografts/virology
2.
J Exp Med ; 218(7)2021 07 05.
Article in English | MEDLINE | ID: mdl-33988715

ABSTRACT

HIV-specific CD8+ T cells partially control viral replication and delay disease progression, but they rarely provide lasting protection, largely due to immune escape. Here, we show that engrafting mice with memory CD4+ T cells from HIV+ donors uniquely allows for the in vivo evaluation of autologous T cell responses while avoiding graft-versus-host disease and the need for human fetal tissues that limit other models. Treating HIV-infected mice with clinically relevant HIV-specific T cell products resulted in substantial reductions in viremia. In vivo activity was significantly enhanced when T cells were engineered with surface-conjugated nanogels carrying an IL-15 superagonist, but it was ultimately limited by the pervasive selection of a diverse array of escape mutations, recapitulating patterns seen in humans. By applying mathematical modeling, we show that the kinetics of the CD8+ T cell response have a profound impact on the emergence and persistence of escape mutations. This "participant-derived xenograft" model of HIV provides a powerful tool for studying HIV-specific immunological responses and facilitating the development of effective cell-based therapies.


Subject(s)
HIV Infections/immunology , HIV-1/immunology , Heterografts/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line , HEK293 Cells , HIV Infections/virology , Heterografts/virology , Humans , Immunotherapy/methods , Interleukin-15/immunology , Mice , Mutation/immunology , Viremia/immunology , Viremia/virology , Virus Replication/immunology
3.
Nat Commun ; 12(1): 2031, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33795676

ABSTRACT

Patient-derived xenografts are crucial for drug development but their use is challenged by issues such as murine viral infection. We evaluate the scope of viral infection and its impact on patient-derived xenografts by taking an unbiased data-driven approach to analyze unmapped RNA-Seq reads from 184 experiments. We find and experimentally validate the extensive presence of murine viral sequence reads covering entire viral genomes in patient-derived xenografts. The existence of viral sequences inside tumor cells is further confirmed by single cell sequencing data. Extensive chimeric reads containing both viral and human sequences are also observed. Furthermore, we find significantly changed expression levels of many cancer-, immune-, and drug metabolism-related genes in samples with high virus load. Our analyses indicate a need to carefully evaluate the impact of viral infection on patient-derived xenografts for drug development. They also point to a need for attention to quality control of patient-derived xenograft experiments.


Subject(s)
Genome, Viral/genetics , High-Throughput Nucleotide Sequencing/methods , Neoplasms/genetics , Sequence Analysis, DNA/methods , Xenograft Model Antitumor Assays/methods , Animals , Cell Line, Tumor , Gene Products, env/classification , Gene Products, env/genetics , Gene Products, gag/classification , Gene Products, gag/genetics , Heterografts/metabolism , Heterografts/virology , Humans , Mice , Neoplasms/classification , Neoplasms/virology , Phylogeny , Virus Diseases/genetics , Virus Diseases/virology
4.
Xenotransplantation ; 27(5): e12623, 2020 09.
Article in English | MEDLINE | ID: mdl-32596829

ABSTRACT

Many patients who would undergo organ transplantation cannot proceed due to the inability of human organ donation to satisfy medical needs. Xenotransplantation has the potential to offer unlimited availability of pig organs for transplantation, and pig-to-non-human primate models have demonstrated outcomes that may soon justify clinical trials. However, one of the unique ethical challenges faced by xenotransplantation is that the risk of introducing potential zoonotic disease into the community must be weighed along with the benefit to the patient. While most experts believe that zoonosis is manageable, apprehension over disease transmission from animal donors to human recipients remains a frequent concern of many who are undecided or opposed to clinical xenotransplantation. The COVID-19 pandemic represents a scenario (rapid worldwide spread of a highly contagious novel zoonotic disease with no natural defense in humans) that would seem to justify apprehension, especially in the United States, which has largely avoided previous pandemic outbreaks. However, there are many differences between zoonosis found in the wild or after xenotransplantation that favor the safety of the latter. Still, these differences, as well as the benefits of xenotransplantation, are not widely understood outside of the field. We must therefore ask what impact the COVID-19 pandemic will have on attitudes toward xenotransplantation.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/complications , Heterografts , Pneumonia, Viral/complications , Transplantation, Heterologous , COVID-19 , Heterografts/virology , Humans , Pandemics , SARS-CoV-2 , Tissue Donors/statistics & numerical data , Tissue and Organ Procurement/methods , Transplantation, Heterologous/ethics , United States
5.
Viruses ; 11(9)2019 08 29.
Article in English | MEDLINE | ID: mdl-31470671

ABSTRACT

Pigs are used as potential donor animals for xenotransplantation. However, porcine endogenous retrovirus (PERV), shown to infect both human and non-human primate (NHP) cells in vitro, presents a risk of transmission to humans in xenotransplantation. In this study, we analyzed PERV transmission in various organs after pig-to-NHP xenotransplantation. We utilized pig-to-NHP xenotransplant tissue samples obtained using two types of transgenic pigs from the National Institute of Animal Science (NIAS, Republic of Korea), and examined them for the existence of PERV genes in different organs via PCR and RT-PCR with specific primers. To determine PERV insertion into chromosomes, inverse PCR using PERV long terminal repeat (LTR) region-specific primers was conducted. The PERV gene was not detected in NHP organs in cardiac xenotransplantation but detected in NHP bladders in renal xenotransplantation. The insertion experiment confirmed that PERVs originate from porcine donor cells rather than integrated provirus in the NHP chromosome. We also demonstrate the presence of pig cells in the NHP bladder after renal xenotransplantation using specific-porcine mitochondrial DNA gene PCR. The PERV sequence was detected in the bladder of NHPs after renal xenotransplantation by porcine cell-microchimerism but did not integrate into the NHP chromosome.


Subject(s)
Endogenous Retroviruses/isolation & purification , Heterografts/virology , Kidney Transplantation/adverse effects , Transplantation, Heterologous/adverse effects , Urinary Bladder/virology , Animals , Animals, Genetically Modified , Chimerism , Cytochromes b/genetics , Endogenous Retroviruses/genetics , Genes, Viral/genetics , Heterografts/cytology , Humans , Macaca mulatta , Swine , Urinary Bladder/cytology
7.
J Gen Virol ; 100(4): 656-661, 2019 04.
Article in English | MEDLINE | ID: mdl-30767852

ABSTRACT

The release of porcine endogenous retrovirus (PERV) particles from pig cells is a potential risk factor during xenotransplantation by way of productively infecting the human transplant recipient. Potential countermeasures against PERV replication are restriction factors that block retroviral replication. SAMHD1 is a triphosphohydrolase that depletes the cellular pool of dNTPs in non-cycling cells starving retroviral reverse transcription. We investigated the antiviral activity of human SAMHD1 against PERV and found that SAMHD1 potently restricts its reverse transcription in human monocytes, monocyte-derived dendritic cells (MDDC), or macrophages (MDM) and in monocytic THP-1 cells. Degradation of SAMHD1 by SIVmac Vpx or CRISPR/Cas9 knock-out of SAMHD1 allowed for PERV reverse transcription. Addition of deoxynucleosides alleviated the SAMHD1-mediated restriction suggesting that SAMHD1-mediated degradation of dNTPs restricts PERV replication in these human immune cells. In conclusion, our findings highlight SAMHD1 as a potential barrier to PERV transmission from pig transplants to human recipients during xenotransplantation.


Subject(s)
Endogenous Retroviruses/physiology , Heterografts/metabolism , Heterografts/virology , SAM Domain and HD Domain-Containing Protein 1/metabolism , Animals , CRISPR-Cas Systems/physiology , Cell Line , HEK293 Cells , Humans , Macrophages/metabolism , Macrophages/virology , Monocytes/metabolism , Monocytes/virology , Reverse Transcription/physiology , Swine , THP-1 Cells , Transplantation, Heterologous/methods , Virus Replication/physiology
8.
Viruses ; 12(1)2019 12 29.
Article in English | MEDLINE | ID: mdl-31905731

ABSTRACT

Xenotransplantation using pig tissues and organs is under development in order to alleviate the increasing shortage of human transplants. Since xenotransplantation may be associated with the transmission of porcine microorganisms to the human recipient, the donor pigs should be carefully analyzed, especially for the presence of potentially zoonotic viruses. Göttingen Minipigs (GöMP) are potential donors of islet cells for the treatment of diabetes. Despite the fact that all animals produced at Ellegaard Göttingen Minipigs A/S carry porcine endogenous retroviruses (PERVs) in their genome and that very few animals were infected with porcine cytomegalovirus (PCMV), hepatitis E virus (HEV) and porcine lymphotropic herpesvirus (PLHV), no transmission of these viruses was observed in a preclinical trial transplanting GöMP islet cells into cynomolgus monkeys. Using a new comprehensive strategy, we then analyzed an isolated subpopulation of Göttingen Minipigs which remained at the University of Göttingen. We concentrated on 11 xenotransplantation-relevant viruses and combined co-incubation assays with susceptible human target cells and molecular biological methods to evaluate the risk posed by PERV. All animals in Göttingen carry PERV-A, PERV-B, and PERV-C in their genome but they are not infected with PCMV, PLHV and HEV. The difference may be explained by selection of negative animals and/or de novo infection. The PERV copy number was established using ddPCR (93 copies) and a human-tropic PERV-A/C was found released from PBMCs of one animal with a high expression of PERV-C.


Subject(s)
Endogenous Retroviruses/isolation & purification , Genome, Viral , Heterografts/virology , Swine Diseases/virology , Swine, Miniature/virology , Animals , Endogenous Retroviruses/classification , Female , Gene Dosage , HEK293 Cells , Humans , Male , Swine , Swine Diseases/transmission , Transplantation, Heterologous
9.
Sci Rep ; 8(1): 13930, 2018 09 17.
Article in English | MEDLINE | ID: mdl-30224769

ABSTRACT

Pediatric high-grade brain tumors and adult glioblastoma are associated with significant morbidity and mortality. Oncolytic herpes simplex virus-1 (oHSV) is a promising approach to target brain tumors; oHSV G207 and M032 (encodes human interleukin-12) are currently in phase I clinical trials in children with malignant supratentorial brain tumors and adults with glioblastoma, respectively. We sought to compare the sensitivity of patient-derived pediatric malignant brain tumor and adult glioblastoma xenografts to these clinically-relevant oHSV. In so doing we found that pediatric brain tumors were more sensitive to the viruses and expressed significantly more nectin-1 (CD111) than adult glioblastoma. Pediatric embryonal and glial tumors were 74-fold and 14-fold more sensitive to M002 and 16-fold and 6-fold more sensitive to G207 than adult glioblastoma, respectively. Of note, pediatric embryonal tumors were more sensitive than glial tumors. Differences in sensitivity may be due in part to nectin-1 expression, which predicted responses to the viruses. Treatment with oHSV resulted in prolonged survival in both pediatric and adult intracranial patient-dervied tumor xenograft models. Our results suggest that pediatric brain tumors are ideal targets for oHSV and that brain tumor expression of nectin-1 may be a useful biomarker to predict patient response to oHSV.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/virology , Herpesvirus 1, Human/genetics , Nectins/genetics , Oncolytic Viruses/genetics , Adolescent , Adult , Animals , Biomarkers, Tumor/genetics , Cell Line, Tumor , Child , Disease Models, Animal , Female , Glioblastoma/genetics , Glioblastoma/virology , Heterografts/virology , Humans , Male , Mice, Nude , Oncolytic Virotherapy/methods , Xenograft Model Antitumor Assays/methods
11.
Xenotransplantation ; 25(4): e12453, 2018 07.
Article in English | MEDLINE | ID: mdl-30264878

ABSTRACT

Routine large-scale xenotransplantation from pigs to humans is getting closer to clinical reality owing to several state-of-the-art technologies, especially the ability to rapidly engineer genetically defined pigs. However, using pig organs in humans poses risks including unwanted cross-species transfer of viruses and adaption of these pig viruses to the human organ recipient. Recent developments in the field of virology, including the advent of metagenomic techniques to characterize entire viromes, have led to the identification of a plethora of viruses in many niches. Single-stranded DNA (ssDNA) viruses are the largest group prevalent in virome studies in mammals. Specifically, the ssDNA viral genomes are characterized by a high rate of nucleotide substitution, which confers a proclivity to adapt to new hosts and cross-species barriers. Pig-associated ssDNA viruses include torque teno sus viruses (TTSuV) in the Anelloviridae family, porcine parvoviruses (PPV), and porcine bocaviruses (PBoV) both in the family of Parvoviridae, and porcine circoviruses (PCV) in the Circoviridae family, some of which have been confirmed to be pathogenic to pigs. The risks of these viruses for the human recipient during xenotransplantation procedures are relatively unknown. Based on the scant knowledge available on the prevalence, predilection, and pathogenicity of pig-associated ssDNA viruses, careful screening and monitoring are required. In the case of positive identification, risk assessments and strategies to eliminate these viruses in xenotransplantation pig stock may be needed.


Subject(s)
Circovirus/pathogenicity , DNA Viruses/pathogenicity , Heterografts/virology , Swine Diseases/prevention & control , Transplantation, Heterologous , Animals , Humans , Swine , Swine Diseases/virology
12.
Xenotransplantation ; 25(4): e12428, 2018 07.
Article in English | MEDLINE | ID: mdl-30264879

ABSTRACT

BACKGROUND: We established a Source Animal (barrier) Facility (SAF) for generating designated pathogen-free (DPF) pigs to serve as donors of viable organs, tissues, or cells for xenotransplantation into clinical patients. This facility was populated with caesarian derived, colostrum deprived (CDCD) piglets, from sows of conventional-specific (or specified) pathogen-free (SPF) health status in six cohorts over a 10-month period. In all cases, CDCD piglets fulfilled DPF status including negativity for porcine circovirus (PCV), a particularly environmentally robust and difficult to inactivate virus which at the time of SAF population was epidemic in the US commercial swine production industry. Two outbreaks of PCV infection were subsequently detected during sentinel testing. The first occurred several weeks after PCV-negative animals were moved under quarantine from the nursery into an animal holding room. The apparent origin of PCV was newly installed stainless steel penning, which was not sufficiently degreased thereby protecting viral particles from disinfection. The second outbreak was apparently transmitted via employee activities in the Caesarian-section suite adjacent to the barrier facility. In both cases, PCV was contained in the animal holding room where it was diagnosed making a complete facility depopulation-repopulation unnecessary. METHOD: Infectious PCV was eliminated during both outbreaks by the following: euthanizing infected animals, disposing of all removable items from the affected animal holding room, extensive cleaning with detergents and degreasing agents, sterilization of equipment and rooms with chlorine dioxide, vaporized hydrogen peroxide, and potassium peroxymonosulfate, and for the second outbreak also glutaraldehyde/quaternary ammonium. Impact on other barrier animals throughout the process was monitored by frequent PCV diagnostic testing. RESULT: After close monitoring for 6 months indicating PCV absence from all rooms and animals, herd animals were removed from quarantine status. CONCLUSION: Ten years after PCV clearance following the second outbreak, due to strict adherence to biosecurity protocols and based on ongoing sentinel diagnostic monitoring (currently monthly), the herd remains DPF including PCV negative.


Subject(s)
Circoviridae Infections/prevention & control , Circovirus/pathogenicity , Specific Pathogen-Free Organisms , Swine Diseases/prevention & control , Transplantation, Heterologous , Animals , Heterografts/virology , Swine , Swine Diseases/virology , Transplantation, Heterologous/instrumentation , Transplantation, Heterologous/methods
13.
Xenotransplantation ; 25(4): e12423, 2018 07.
Article in English | MEDLINE | ID: mdl-30264880

ABSTRACT

Post-transplantation infections are common in allograft recipients and should be expected in all immunocompromised hosts. Based on the need for immunosuppression in xenotransplantation, procedures developed to enhance safety in allotransplantation can be applied in future xenotransplantation clinical trials. Standardized approaches can be developed to guide the evaluation of common infectious syndromes in xenograft recipients. The opportunity created by screening of swine intended as xenograft donors has equal applicability to allotransplantation-notably broader screening strategies for allograft donors such as use of advanced sequencing modalities including broad-range molecular probes, microarrays, and high-throughput pyrosequencing. Considerations in management of allotransplant- and xenotransplant-associated infections are largely the same. Experience in xenotransplantation will continue to inform thinking regarding donor-derived infections in allotransplantation. We expect that experience in managing complex allotransplant recipients will similarly inform clinical trials in xenotransplantation.


Subject(s)
Heterografts/virology , Immunosuppression Therapy , Infections/virology , Tissue Donors , Transplantation, Heterologous , Animals , Graft Rejection/prevention & control , Graft Rejection/virology , Humans , Immunosuppression Therapy/methods
14.
Xenotransplantation ; 25(4): e12427, 2018 07.
Article in English | MEDLINE | ID: mdl-30264882

ABSTRACT

BACKGROUND: Recent advances in xenotransplantation have produced organs from pigs that are well tolerated in primate models because of genetic changes engineered to delete major antigens from donor animals. To ensure the safety of human transplant recipients, it will be essential to understand both the spectrum of infectious agents in donor pigs and their potential to be transmitted to immunocompromised transplant recipients. Equally important will be the development of new highly sensitive diagnostic methods for use in the detection of these agents in donor animals and for the monitoring of transplant recipients. METHODS: Herein, we report the development of a panel of 30 quantitative polymerase chain reaction (qPCR) assays for infectious agents with the potential to be transmitted to the human host. The reproducibility, sensitivity and specificity of each assay were evaluated and were found to exhibit analytic sensitivity that was similar to that of quantitative assays used to perform viral load testing of human viruses in clinical laboratories. RESULTS: This analytical approach was used to detect nucleic acids of infectious agents present in specimens from 9 sows and 22 piglets derived by caesarean section. The most commonly detected targets in adult animals were Mycoplasma species and two distinct herpesviruses, porcine lymphotrophic herpesvirus 2 and 3. A total of 14 piglets were derived from three sows infected with either or both herpesviruses, yet none tested positive for the viruses indicating that vertical transmission of these viruses is inefficient. CONCLUSIONS: The data presented demonstrate that procedures in place are highly sensitive and can specifically detect nucleic acids from target organisms in the panel, thus ensuring the safety of organs for transplantation as well as the monitoring of patients potentially receiving them.


Subject(s)
Herpesviridae/pathogenicity , Heterografts/virology , Swine Diseases/virology , Transplantation, Heterologous/adverse effects , Animals , Cytomegalovirus/genetics , Humans , Reproducibility of Results , Swine , Swine Diseases/diagnosis
15.
Xenotransplantation ; 25(4): e12449, 2018 07.
Article in English | MEDLINE | ID: mdl-30264883

ABSTRACT

For clinical xenotransplantation, transplants must be free of porcine cytomegalovirus (PCMV). Piglets become infected primarily in the perinatal period by the mother sow. While individual donor animals can be protected from infection by isolation husbandry, success is not guaranteed and this strategy poses the risk of undetected infections and raises animal welfare questions. Here, we present the establishment of a completely PCMV-negative pig herd for breeding donor animals for xenotransplantation. Eleven pregnant DanAvl Basic hybrid sows were purchased from a designated pathogen-free (DPF), PCMV-positive colony and transferred to a new pig facility at the Centre for Innovative Medical Models (CiMM) 4 weeks prior to farrowing. At the age of 24 hours, piglets were early-weaned and transferred to a commercially available Rescue Deck system dedicated to motherless rearing of piglets. Sows were removed from the facility. The PCMV status of F1-generation animals was determined at regular intervals over a period of 14 months by a sensitive real-time PCR-based detection method testing blood, nasal swabs and cultured peripheral blood mononuclear cells (PBMCs). F1 sows were used as recipients of genetically modified embryos to generate a xenotransplant donor herd. Offspring were tested for PCMV accordingly. All offspring have remained PCMV negative over the whole observation period of 14 months. A completely PCMV-negative pig herd for xenotransplantation has thus been successfully established.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus/genetics , Leukocytes, Mononuclear/virology , Transplantation, Heterologous , Animals , Animals, Genetically Modified , Cytomegalovirus/isolation & purification , Heterografts/virology , Swine , Tissue Donors , Weaning
18.
Transplant Proc ; 49(9): 2209-2214, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29149984

ABSTRACT

BACKGROUND: Xenotransplantations of pig corneas have become an attractive alternative to human corneas. Such xenotransplantations carry the danger, however, of transmission of porcine endogenous retroviruses (PERVs). Here, we investigated whether porcine corneas harbor viral DNA and RNA and whether transplantation to a nonhuman primate would lead to host PERV infection. METHODS: Monkey vein endothelial cells (MVECs) were inoculated with porcine aortic endothelial cell (PAEC) supernatants, and DNA and total RNA of MVECs were tested for PERV by polymerase chain reaction (PCR) and reverse transcription PCR (RT-PCR) assays. Corneas were harvested from Wuzhishan miniature pigs, and the presence of PERV proviral DNA and RNA was analyzed by PCR and RT-PCR, respectively. Fresh or dehydrated corneas were then transplanted to rhesus monkeys, and PERV proviral DNA and RNA were analyzed in host peripheral blood lymphocytes at 6 and 24 months. Furthermore, the presence of PERV sequences was analyzed in the transplant at 24 months. RESULTS: PCR analysis showed PERV transfection from PAECs to MVECs in vitro. PCR and RT-PCR gave positive signals for PERV subtypes A and B, but not PERV-C, regardless of how the corneas were prepared. No evidence was found for PERV transmission to the host, and the transplant had lost its viral signal at the end of the 24-month period. CONCLUSIONS: Regardless of cornea preparation and storage, PERV transmission from pig to host could not be detected, despite that the transplant was initially PERV-positive. The use of the Wuzhishan miniature pig as the donor may be advantageous because it lacks PERV-C and hence potentially infectious A/C recombinants.


Subject(s)
Cornea/virology , Corneal Transplantation/methods , Endogenous Retroviruses/genetics , Graft vs Host Disease/virology , Heterografts/virology , Transplantation, Heterologous/methods , Animals , Corneal Transplantation/adverse effects , DNA, Viral/analysis , Macaca mulatta , Polymerase Chain Reaction , RNA, Viral/analysis , Swine/virology , Swine, Miniature , Transplantation, Heterologous/adverse effects
19.
Virol J ; 14(1): 171, 2017 09 06.
Article in English | MEDLINE | ID: mdl-28874166

ABSTRACT

The composition of the porcine virome includes viruses that infect pig cells, ancient virus-derived elements including endogenous retroviruses inserted in the pig chromosomes, and bacteriophages that infect a broad array of bacteria that inhabit pigs. Viruses infecting pigs, among them viruses also infecting human cells, as well as porcine endogenous retroviruses (PERVs) are of importance when evaluating the virus safety of xenotransplantation. Bacteriophages associated with bacteria mainly in the gut are not relevant in this context. Xenotransplantation using pig cells, tissues or organs is under development in order to alleviate the shortage of human transplants. Here for the first time published data describing the viromes in different pigs and their relevance for the virus safety of xenotransplantation is analysed. In conclusion, the analysis of the porcine virome has resulted in numerous new viruses being described, although their impact on xenotransplantation is unclear. Most importantly, viruses with known or suspected zoonotic potential were often not detected by next generation sequencing, but were revealed by more sensitive methods.


Subject(s)
Heterografts/virology , Swine/virology , Transplantation, Heterologous , Virus Physiological Phenomena , Animals , Heterografts/standards , Humans , Public Health , Transplantation, Heterologous/adverse effects , Transplantation, Heterologous/standards , Viruses/classification
20.
Xenotransplantation ; 24(4)2017 07.
Article in English | MEDLINE | ID: mdl-28503733

ABSTRACT

BACKGROUND: The risk of xenozoonosis mainly by porcine endogenous retrovirus (PERV) has been considered as one of the main hurdles in xenotransplantation and therefore should be elucidated prior to the clinical use of porcine corneal grafts. Accordingly, an investigation was performed to analyze the infectivity of PERVs from porcine keratocytes to human cells, and the long-term risk of transmission of PERVs was determined using pig-to-non-human primate (NHP) corneal transplantation models. METHODS: The infectivity of PERVs from the SNU miniature pig keratocytes was investigated by coculture with a human embryonic kidney cell line. Twenty-two rhesus macaques underwent xenocorneal transplantation as follows: (i) group 1 (n=4): anterior lamellar keratoplasty (LKP) with freshly preserved porcine corneas, (ii) group 2 (n=5): anterior LKP with decellularized porcine corneas followed by penetrating keratoplasty (PKP) with allografts, (iii) group 3 (n=3): PKP under steroid-based immunosuppression, (iv) group 4 (n=4): PKP under anti-CD154 antibody-based immunosuppression, (v) group 5 (n=4): deep anterior LKP with freshly preserved porcine corneas under anti-CD40 antibody-based immunosuppression, and (vi) group 6 (n=2): PKP under anti-CD40 antibody-based immunosuppression. Postoperative blood samples were serially collected, and tissue samples were obtained from thirteen different organs at the end of each experiment. The existence of PERV DNA and RNA was investigated using PCR and RT-PCR. RESULTS: Using two independent in vitro infectivity tests, neither PERV pol nor pig mitochondrial cytochrome oxidase II was detected after 41 and 92 days of coculture, respectively. After xenocorneal transplantation, a total of 257 serial peripheral blood mononuclear cell samples, 34 serial plasma samples, and 282 tissue samples were obtained from the NHP recipients up to 1176 days post-transplantation. No PERV transmission was evident in any samples. CONCLUSIONS: Within the limits of this study, there is no evidence to support any risk of PERV transmission from porcine corneal tissues to NHP recipients, despite the existence of PERV-expressing cells in porcine corneas.


Subject(s)
Corneal Transplantation/adverse effects , Endogenous Retroviruses , Heterografts/virology , Leukocytes, Mononuclear/virology , Retroviridae Infections/transmission , Animals , CD40 Ligand/metabolism , Cell Line , Corneal Transplantation/methods , Macaca mulatta , Swine , Time , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...