Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.337
Filter
1.
Zebrafish ; 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39316469

ABSTRACT

Xenotransplantation of neuroblastoma cells into larval zebrafish allows the characterization of their in vivo tumorigenic abilities and high-throughput treatment screening. This established preclinical model traditionally relies on microinjection into the yolk or perivitelline space, leaving the engraftment ability of cells at the hindbrain ventricle (HBV) and pericardial space (PCS), sites valuable for evaluating metastasis, angiogenesis, and the brain microenvironment, unknown. To address this gap in knowledge, Casper zebrafish at 48 h postfertilization were microinjected with approximately 200 Kelly, Be(2)-C, SK-N-AS, or SY5Y cells into either the HBV or PCS. Fish were imaged at 1, 3, and 6 days postinjection and tumor growth was monitored at each timepoint. We hypothesized that engraftment ability and location preference would be cell line dependent. Kelly and SK-N-AS cells were able to engraft at both the HBV and PCS, with a near doubling in size of tumor volume during the 6 days observation period, with cells appearing to grow better in the HBV. Be(2)-C tumors remained static while SY5Y tumors decreased in size, with almost complete loss of volume at both sites. Therefore, the capability of neuroblastoma cell engraftment in zebrafish larvae is cell line dependent with a location preference.

2.
Cell Rep Med ; : 101744, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39317190

ABSTRACT

This study develops an observational model to assess kidney function recovery and xenogeneic immune responses in kidney xenotransplants, focusing on gene editing and immunosuppression. Two brain-dead patients undergo single kidney xenotransplantation, with kidneys donated by minipigs genetically modified to include triple-gene knockouts (GGTA1, ß4GalNT2, CMAH) and human gene transfers (hCD55 or hCD55/hTBM). Renal xenograft functions are fully restored; however, immunosuppression without CD40-CD154 pathway blockade is ineffective in preventing acute rejection by day 12. This rejection manifests as both T cell-mediated rejection and antibody-mediated rejection (AMR), confirmed by natural killer (NK) cell and macrophage infiltration in sequential xenograft biopsies. Despite donor pigs being pathogen free before transplantation, xenografts and recipient organs test positive for porcine cytomegalovirus/porcine roseolovirus (PCMV/PRV) by the end of the observation period, indicating reactivation and contributing to significant immunopathological changes. This study underscores the critical need for extended clinical observation and comprehensive evaluation using deceased human models to advance xenograft success.

3.
Stem Cell Res Ther ; 15(1): 325, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39334434

ABSTRACT

BACKGROUND: The uncultured adipose-derived stromal vascular fraction (SVF), consisting of adipose-derived stromal cells (ADSCs), M2 macrophages (M2Φ) and others, has shown therapeutic potential against osteoarthritis (OA), however, the mechanisms underlying its therapeutic effects remain unclear. Therefore, this study investigated the effects of the SVF on OA in a human-immunodeficient rat xenotransplantation model. METHODS: OA model was induced in the knees of female immunodeficient rats by destabilization of the medial meniscus. Immediately after the surgery, human SVF (1 × 105), ADSCs (1 × 104), or phosphate buffered saline as a control group were transplanted into the knees. At 4 and 8 weeks postoperatively, OA progression and synovitis were analyzed by macroscopic and histological analyses, and the expression of collagen II, SOX9, MMP-13, ADAMTS-5, F4/80, CD86 (M1), CD163 (M2), and human nuclear antigen (hNA) were evaluated immunohistochemically. In vitro, flow cytometry was performed to collect CD163-positive cells as M2Φ from the SVF. Chondrocyte pellets (1 × 105) were co-cultured with SVF (1 × 105), M2Φ (1 × 104), and ADSCs (1 × 104) or alone as a control group, and the pellet size was compared. TGF-ß, IL-10 and MMP-13 concentrations in the medium were evaluated using enzyme-linked immunosorbent assay. RESULTS: In comparison with the control and ADSC groups, the SVF group showed significantly slower OA progression and less synovitis with higher expression of collagen II and SOX9, lower expression of MMP-13 and ADAMTS-5, and lower F4/80 and M1/M2 ratio in the synovium. Only the SVF group showed partial expression of hNA-, CD163-, and F4/80-positive cells in the rat synovium. In vitro, the SVF, M2Φ, ADSC and control groups, in that order, showed larger pellet sizes, higher TGF-ß and IL-10, and lower MMP-13 concentrations. CONCLUSIONS: The M2Φ in the transplanted SVF directly affected recipient tissue, enhancing the secretion of growth factors and chondrocyte-protecting cytokines, and partially improving chondrocytes and joint homeostasis. These findings indicate that the SVF is as an effective option for regenerative therapy for OA, with mechanisms different from those of ADSCs.


Subject(s)
Adipose Tissue , Macrophages , Osteoarthritis , Animals , Humans , Rats , Female , Osteoarthritis/therapy , Osteoarthritis/pathology , Macrophages/metabolism , Adipose Tissue/cytology , Chondrocytes/metabolism , Chondrocytes/cytology , Stromal Vascular Fraction/metabolism , Disease Models, Animal
4.
Life (Basel) ; 14(9)2024 Sep 08.
Article in English | MEDLINE | ID: mdl-39337915

ABSTRACT

Blood analysis plays a pivotal role in assessing the health of laboratory animals, including pigs. This study investigated the hematological profiles of transgenic pigs of the MGH breed for xenotransplantation, focusing on the effect of housing conditions on blood parameters. A cohort of pigs was longitudinally monitored from 6 to 18 months of age in both conventional and specific pathogen-free (SPF) environments. Red blood cells (RBCs), hemoglobin (HGB), and white blood cells (WBCs) were analyzed using standardized hematology analyzers. The results revealed that RBC and HGB levels were consistently higher in SPF-housed pigs. Notably, WBC counts were significantly lower in SPF-housed pigs, suggesting that reduced pathogen exposure under SPF conditions effectively diminished immune system activation. These findings raise a novel question as to whether distinct hematological parameters of specific and/or designated PF pigs would be advantages for the success of clinical xenotransplantation trials.

6.
Clin Transplant Res ; 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39233453

ABSTRACT

The progress of transplantation has been propelled forward by animal experiments. Animal models have not only provided opportunities to understand complex immune mechanisms in transplantation but also served as a platform to assess therapeutic interventions. While small animals have been instrumental in uncovering new therapeutic concepts related to immunosuppression and immune tolerance, the progression to human trials has largely been driven by studies in large animals. Recent research has begun to explore the potential of porcine organs to address the shortage of available organs. The consistent progress in transplant immunology research can be attributed to a thorough understanding of animal models. This review provides a comprehensive overview of the available animal models, detailing their modifications, strengths, and weaknesses, as well as their historical applications, to aid researchers in selecting the most suitable model for their specific research needs.

8.
Xenotransplantation ; 31(5): e12883, 2024.
Article in English | MEDLINE | ID: mdl-39276074

ABSTRACT

This updated report highlights significant developments in the field of xenotransplantation since December 2023. Over the past 6 months, there has been a notable increase in discussions regarding the feasibility of clinical trials, with particular emphasis on their progression and associated ethical considerations. This review presents the most pertinent findings from December 2023 to June 2024.


Subject(s)
Heterografts , Transplantation, Heterologous , Transplantation, Heterologous/methods , Animals , Humans
9.
Front Immunol ; 15: 1455691, 2024.
Article in English | MEDLINE | ID: mdl-39346923

ABSTRACT

As an effective treatment for diabetes, islet transplantation has garnered significant attention and research in recent years. However, immune rejection and the toxicity of immunosuppressive drugs remain critical factors influencing the success of islet transplantation. While immunosuppressants are essential in reducing immune rejection reactions and can significantly improve the survival rate of islet transplants, improper use of these drugs can markedly increase mortality rates following transplantation. Additionally, the current availability of islet organ donations fails to meet the demand for organ transplants, making xenotransplantation a crucial method for addressing organ shortages. This review will cover the following three aspects: 1) the immune responses occurring during allogeneic islet transplantation, including three stages: inflammation and IBMIR, allogeneic immune response, and autoimmune recurrence; 2) commonly used immunosuppressants in allogeneic islet transplantation, including calcineurin inhibitors (Cyclosporine A, Tacrolimus), mycophenolate mofetil, glucocorticoids, and Bortezomib; and 3) early and late immune responses in xenogeneic islet transplantation and the immune effects of triple therapy (ECDI-fixed donor spleen cells (ECDI-SP) + anti-CD20 + Sirolimus) on xenotransplantation.


Subject(s)
Graft Rejection , Immunosuppressive Agents , Islets of Langerhans Transplantation , Transplantation, Heterologous , Islets of Langerhans Transplantation/methods , Islets of Langerhans Transplantation/adverse effects , Islets of Langerhans Transplantation/immunology , Humans , Immunosuppressive Agents/therapeutic use , Animals , Graft Rejection/immunology , Graft Rejection/prevention & control , Transplantation, Homologous , Graft Survival/drug effects , Graft Survival/immunology
10.
Xenotransplantation ; 31(4): e12879, 2024.
Article in English | MEDLINE | ID: mdl-39166818

ABSTRACT

Transplantation remains the preferred treatment for end-stage kidney disease but is critically limited by the number of available organs. Xenografts from genetically modified pigs have become a promising solution to the loss of life while waiting for transplantation. However, the current clinical model for xenotransplantation will require off-site procurement, leading to a period of ischemia during transportation. As of today, there is limited understanding regarding the preservation of these organs, including the duration of viability, and the associated molecular changes. Thus, our aim was to evaluate the effects of static cold storage (SCS) on α1,3-galactosyltransferase knockout (GGTA1 KO) kidney. After SCS, viability was further assessed using acellular sub-normothermic ex vivo perfusion and simulated transplantation with human blood. Compared to baseline, tubular and glomerular interstitium was preserved after 2 days of SCS in both WT and GGTA1 KO kidneys. Bulk RNA-sequencing demonstrated that only eight genes were differentially expressed after SCS in GGTA1 KO kidneys. During sub-normothermic perfusion, kidney function, reflected by oxygen consumption, urine output, and lactate production was adequate in GGTA1 KO grafts. During a simulated transplant with human blood, macroscopic and histological assessment revealed minimal kidney injury. However, GGTA1 KO kidneys exhibited higher arterial resistance, increased lactate production, and reduced oxygen consumption during the simulated transplant. In summary, our study suggests that SCS is feasible for the preservation of porcine GGTA1 KO kidneys. However, alternative preservation methods should be evaluated for extended preservation of porcine grafts.


Subject(s)
Galactosyltransferases , Kidney Transplantation , Kidney , Organ Preservation , Transplantation, Heterologous , Animals , Transplantation, Heterologous/methods , Kidney Transplantation/methods , Galactosyltransferases/genetics , Galactosyltransferases/deficiency , Swine , Organ Preservation/methods , Humans , Animals, Genetically Modified , Perfusion/methods , Heterografts , Cryopreservation/methods , Gene Knockout Techniques/methods , Mice
11.
Regen Med ; 19(6): 303-315, 2024 06 02.
Article in English | MEDLINE | ID: mdl-39177571

ABSTRACT

Aim: The escalating demand for corneal transplants significantly surpasses the available supply. To bridge this gap, we concentrated on ethical and sustainable corneal grafting sources. Our objective was to create viable corneal scaffolds from preserved slaughterhouse waste.Materials & methods: Corneas were extracted and decellularized from eyeballs that had been refrigerated for several days. These scaffolds underwent evaluation through DNA quantification, histological analysis, surface tension measurement, light propagation testing, and tensile strength assessment.Results: Both the native and acellular corneas (with ~90% DNA removed using a cost-effective and environmentally friendly surfactant) maintained essential optical and biomechanical properties for potential clinical use.Conclusion: Our method of repurposing slaughterhouse waste, stored at 4°C for several days, to develop corneal scaffolds offers a sustainable and economical alternative xenograft model.


[Box: see text].


Subject(s)
Abattoirs , Cornea , Tissue Scaffolds , Animals , Tissue Scaffolds/chemistry , Cornea/cytology , Sheep , Corneal Transplantation/methods , Tissue Engineering/methods
12.
ChemMedChem ; : e202400495, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39136593

ABSTRACT

A novel library of naphthoquinone derivatives (3-5aa) was synthesized and evaluated for their anticancer properties. Specifically, compounds 5i, 5l, 5o, 5q, 5r, 5s, 5t, and 5v demonstrated superior cytotoxic activity against the cancer cell lines that were studied. Notably, compound 5v displayed a greater cytotoxic effect on MCF-7 cells compared to the standard drug doxorubicin. To further investigate the mechanism of cytotoxic effect, additional anticancer studies were conducted with 5v in MCF-7 cells. The findings showed that 5v triggered cytotoxic effects in MCF-7 cells through the initiation of cell cycle arrest at the G1/S phase and necrosis. In vivo ecotoxicity studies indicated that 5v had lower toxicity towards zebrafish larvae (LC50 = 50.15 µM) and had an insignificant impact on cardiac functions. In vivo xenotransplantation of MCF-7 cells in zebrafish larvae demonstrated a significant reduction in tumour volume in the xenograft. Approximately 95% of the zebrafish larvae with 5v xenografts survived after 10 days of the treatment. Finally, a computational modelling study was conducted on four protein receptors, namely ER, EFGR, BRCA1, and VEFGR2. The findings highlight the importance of the aminonaphthoquinone moiety, amide linkage, and propyl thio moiety in enhancing the anticancer properties.

13.
Rev Cardiovasc Med ; 25(7): 271, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39139422

ABSTRACT

Cardiac xenotransplantation (cXT) has emerged as a solution to heart donor scarcity, prompting an exploration of its scientific, ethical, and regulatory facets. The review begins with genetic modifications enhancing pig hearts for human transplantation, navigating through immunological challenges, rejection mechanisms, and immune responses. Key areas include preclinical milestones, complement cascade roles, and genetic engineering to address hyperacute rejection. Physiological counterbalance systems, like human thrombomodulin and endothelial protein C receptor upregulation in porcine xenografts, highlight efforts for graft survival enhancement. Evaluating pig and baboon donors and challenges with non-human primates illuminates complexities in donor species selection. Ethical considerations, encompassing animal rights, welfare, and zoonotic disease risks, are critically examined in the cXT context. The review delves into immune control mechanisms with aggressive immunosuppression and clustered regularly interspaced palindromic repeats associated protein 9 (CRISPR/Cas9) technology, elucidating hyperacute rejection, complement activation, and antibody-mediated rejection intricacies. CRISPR/Cas9's role in creating pig endothelial cells expressing human inhibitor molecules is explored for rejection mitigation. Ethical and regulatory aspects emphasize the role of committees and international guidelines. A forward-looking perspective envisions precision medical genetics, artificial intelligence, and individualized heart cultivation within pigs as transformative elements in cXT's future is also explored. This comprehensive analysis offers insights for researchers, clinicians, and policymakers, addressing the current state, and future prospects of cXT.

14.
Biomedicines ; 12(8)2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39200391

ABSTRACT

The blockade of the CD40/CD40L immune checkpoint is considered essential for cardiac xenotransplantation. However, it is still unclear which single antibody directed against CD40 or CD40L (CD154), or which combination of antibodies, is better at preventing organ rejection. For example, the high doses of antibody administered in previous experiments might not be feasible for the treatment of humans, while thrombotic side effects were described for first-generation anti-CD40L antibodies. To address these issues, we conducted six orthotopic pig-to-baboon cardiac xenotransplantation experiments, combining a chimeric anti-CD40 antibody with an investigational long-acting PASylated anti-CD40L Fab fragment. The combination therapy effectively resulted in animal survival with a rate comparable to a previous study that utilized anti-CD40 monotherapy. Importantly, no incidence of thromboembolic events associated with the administration of the anti-CD40L PAS-Fab was observed. Two experiments failed early because of technical reasons, two were terminated deliberately after 90 days with the baboons in excellent condition and two were extended to 120 and 170 days, respectively. Unexpectedly, and despite the absence of any clinical signs, histopathology revealed fungal infections in all four recipients. This study provides, for the first time, insights into a combination therapy with anti-CD40/anti-CD40L antibodies to block this immune checkpoint.

15.
Xenotransplantation ; 31(4): e12880, 2024.
Article in English | MEDLINE | ID: mdl-39185772

ABSTRACT

OBJECTIVE: To evaluate the clinically relevant anti-CD40 antibody iscalimab for baseline immunosuppression in a preclinical pig-to-rhesus renal xenograft model. SUMMARY BACKGROUND DATA: CD40/CD40L co-stimulation blockade-based immunosuppression has been more successful than calcineurin-based protocols in prolonging xenograft survival in preclinical models. METHODS: GGTA1 knockout/CD55 transgenic pig kidneys were transplanted into rhesus monkeys (n = 6) receiving an iscalimab-based immunosuppressive regimen. RESULTS: Two grafts were lost early (22 and 26 days) because of ectatic donor ureters with otherwise normal histology. The other recipients survived 171, 315, 422, and 439 days with good renal function throughout the posttransplant course. None of the recipients experienced serious infectious morbidity. CONCLUSIONS: It may be reasonable to evaluate an iscalimab-based immunosuppressive regimen in clinical renal xenotransplantation.


Subject(s)
Graft Survival , Heterografts , Immunosuppressive Agents , Kidney Transplantation , Macaca mulatta , Transplantation, Heterologous , Animals , Transplantation, Heterologous/methods , Swine , Graft Survival/immunology , Graft Survival/drug effects , Kidney Transplantation/methods , Immunosuppressive Agents/pharmacology , Heterografts/immunology , Graft Rejection/immunology , Graft Rejection/prevention & control , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Animals, Genetically Modified , Antibodies, Monoclonal/pharmacology , Humans , Galactosyltransferases/genetics
16.
Xenotransplantation ; 31(4): e12881, 2024.
Article in English | MEDLINE | ID: mdl-39185796

ABSTRACT

BACKGROUND: The number of multigene-modified donor pigs for xenotransplantation is increasing with the advent of gene-editing technologies. However, it remains unclear which gene combination is suitable for specific organ transplantation. METHODS: In this study, we utilized CRISPR/Cas9 gene editing technology, piggyBac transposon system, and somatic cell cloning to construct GTKO/hCD55/hTBM/hCD39 four-gene-edited cloned (GEC) pigs and performed kidney transplantation from pig to rhesus monkey to evaluate the effectiveness of these GEC pigs. RESULTS: First, 107 cell colonies were obtained through drug selection, of which seven were 4-GE colonies. Two colonies were selected for somatic cell nuclear transfer (SCNT), resulting in seven fetuses, of which four were GGTA1 biallelic knockout. Out of these four, two fetuses had higher expression of hCD55, hTBM, and hCD39. Therefore, these two fetuses were selected for two consecutive rounds of cloning, resulting in 97 live piglets. After phenotype identification, the GGTA1 gene of these pigs was inactivated, and hCD55, hTBM, and hCD39 were expressed in cells and multiple tissues. Furthermore, the numbers of monkey IgM and IgG binding to the peripheral blood mononuclear cells (PBMCs) of the 4-GEC pigs were markedly reduced. Moreover, 4-GEC porcine PBMCs had greater survival rates than those from wild-type pigs through complement-mediated cytolysis assays. In pig-to-monkey kidney xenotransplantation, the kidney xenograft successfully survived for 11 days. All physiological and biochemical indicators were normal, and no hyperacute rejection or coagulation abnormalities were found after transplantation. CONCLUSION: These results indicate that the GTKO/hCD55/hTBM/hCD39 four-gene modification effectively alleviates immune rejection, and the pig kidney can functionally support the recipient monkey's life.


Subject(s)
Animals, Genetically Modified , Galactosyltransferases , Gene Editing , Kidney Transplantation , Transplantation, Heterologous , Animals , Transplantation, Heterologous/methods , Kidney Transplantation/methods , Swine , Gene Editing/methods , Galactosyltransferases/genetics , CRISPR-Cas Systems , Macaca mulatta , Nuclear Transfer Techniques , Heterografts , Humans , Graft Survival/immunology , Graft Rejection/immunology , Apyrase , Antigens, CD
17.
Sci Rep ; 14(1): 20109, 2024 08 29.
Article in English | MEDLINE | ID: mdl-39209914

ABSTRACT

The idea of utilizing unused oocytes present in the ovaries has been tested in various ways to produce offspring. However, only a limited number of studies succeeded in offspring generation. They include transplantation of ovaries into autologous or allogeneic animals, and acquisition of pups from oocytes obtained by transplanting mouse ovaries into immunodeficient rats. Here we report successful production of rat oocytes by transplanting rat ovaries under the kidney capsule of immunodeficient mice with addition of hormone administration to the mice. In addition, these oocytes were developed by in vitro fertilization, and transplanted into the oviducts of pseudopregnant rats, resulting in successful delivery of pups. The modified gene of the donor rat was confirmed to be correctly inherited to the pups. These results show that xenotransplantation of ovarian tissue makes it possible to leave offspring, beginning a new phase in developmental engineering.


Subject(s)
Oocytes , Ovary , Transplantation, Heterologous , Animals , Female , Transplantation, Heterologous/methods , Rats , Mice , Ovary/transplantation , Fertilization in Vitro/methods
18.
Article in English | MEDLINE | ID: mdl-39097214

ABSTRACT

BACKGROUND: Xenotransplantation has made significant advances recently using pigs genetically engineered to remove carbohydrate antigens, either alone or with addition of various human complement, coagulation, and anti-inflammatory ''transgenes''. Here we evaluated results associated with gene-edited (GE) pig hearts transplanted in baboons using an established costimulation-based immunosuppressive regimen and a cold-perfused graft preservation technique. METHODS: Eight baboons received heterotopic abdominal heart transplants from 3-GE (GalKO.ß4GalNT2KO.hCD55, n = 3), 9-GE (GalKO.ß4GalNT2KO.GHRKO.hCD46.hCD55. TBM.EPCR.hCD47. HO-1, n = 3) or 10-G (9-GE+CMAHKO, n = 2) pigs using Steen's cold continuous perfusion for ischemia minimization. Immunosuppression (IS) included induction with anti-thymocyte globulin and αCD20, ongoing αCD154, MMF, and tapered corticosteroid. RESULTS: All three 3-GE grafts functioned well initially, but failed within 5 days. One 9-GE graft was lost intraoperatively due to a technical issue and another was lost at POD 13 due to antibody mediated rejection (AMR) in a baboon with a strongly positive pre-operative cross-match. One 10-GE heart failed at POD113 with combined cellular and antibody mediated rejection. One 9-GE and one 10-GE hearts had preserved graft function with normal myocardium on protocol biopsies, but exhibited slowly progressive graft hypertrophy until elective necropsy at POD393 and 243 respectively. Elevated levels of IL-6, MCP-1, C-reactive protein, and human thrombomodulin were variably associated with conditioning, the transplant procedure, and clinically significant postoperative events. CONCLUSION: Relative to reference genetics without thrombo-regulatory and anti-inflammatory gene expression, 9- or 10-GE pig hearts exhibit promising performance in the context of a clinically applicable regimen including ischemia minimization and αCD154-based IS, justifying further evaluation in an orthotopic model.

19.
Animal Model Exp Med ; 7(4): 584-590, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38962826

ABSTRACT

BACKGROUND: Pig organ xenotransplantation is a potential solution for the severe organ shortage in clinic, while immunogenic genes need to be eliminated to improve the immune compatibility between humans and pigs. Current knockout strategies are mainly aimed at the genes causing hyperacute immune rejection (HAR) that occurs in the first few hours while adaptive immune reactions orchestrated by CD4 T cell thereafter also cause graft failure, in which process the MHC II molecule plays critical roles. METHODS: Thus, we generate a 4-gene (GGTA1, CMAH, ß4GalNT2, and CIITA) knockout pig by CRISPR/Cas9 and somatic cell nuclear transfer to compromise HAR and CD4 T cell reactions simultaneously. RESULTS: We successfully obtained 4KO piglets with deficiency in all alleles of genes, and at cellular and tissue levels. Additionally, the safety of our animals after gene editing was verified by using whole-genome sequencing and karyotyping. Piglets have survived for more than one year in the barrier, and also survived for more than 3 months in the conventional environment, suggesting that the piglets without MHC II can be raised in the barrier and then gradually mated in the conventional environment. CONCLUSIONS: 4KO piglets have lower immunogenicity, are safe in genomic level, and are easier to breed than the model with both MHC I and II deletion.


Subject(s)
Graft Rejection , N-Acetylgalactosaminyltransferases , Transplantation, Heterologous , Animals , Humans , Swine , N-Acetylgalactosaminyltransferases/genetics , Graft Rejection/genetics , Graft Rejection/immunology , Galactosyltransferases/genetics , Nuclear Proteins/genetics , Mixed Function Oxygenases/genetics , Animals, Genetically Modified , Gene Knockout Techniques , CRISPR-Cas Systems , Gene Editing , Trans-Activators
20.
Biomedicines ; 12(7)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-39062043

ABSTRACT

BACKGROUND: Patterns of humoral immune responses represent a major hurdle in terms of pig-to-human xenotransplantation approaches. The best-known xenogeneic glycan antigens present in pigs are the αGal (Galili antigen) and the non-human sialic acid Neu5Gc. As there are further differences between porcine and human cellular surface glycosylation, a much broader range of glycan epitopes with xeno-reactive relevance can be anticipated. Therefore, we set out to chemically modify porcine cellular surface glycans in a global approach by applying sodium periodate (NaIO4) oxidation. METHODS: Porcine endothelial cells were exposed to oxidation with 1 to 5 mM NaIO4 for different time periods at 37 °C or 4 °C and under static or dynamic conditions. The impact on cellular survival was determined by applying live/dead assays. Oxidation of αGal-epitopes was assessed by fluorescence microscopy-based quantification of isolectin-B4 (IL-B4) staining. Overall immunogenicity of porcine cells was determined by human serum antibody binding. RESULTS: Treatment of porcine endothelial cells and tissues with NaIO4 led to reduced binding of the αGal-specific IL-B4 and/or human serum antibodies. NaIO4 was revealed to be cytotoxic when performed at elevated temperatures and for a prolonged time. However, by applying 2 mM NaIO4 for 60 min at 4 °C, a high extent of cellular viability and a relevant reduction in detectable αGal epitope were observed. No differences were detected irrespectively on whether the cells were oxidized under static or flow conditions. CONCLUSIONS: Glycan epitopes on living cells can be oxidized with NaIO4 while maintaining their viability. Accordingly, this strategy holds promise to prevent immune reactions mediated by preformed anti-glycan antibodies.

SELECTION OF CITATIONS
SEARCH DETAIL