Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Xenotransplantation ; 28(2): e12663, 2021 03.
Article in English | MEDLINE | ID: mdl-33230864

ABSTRACT

BACKGROUND: Genetically modified pigs (GMP) have been developed to alleviate the shortage of donors in human islet transplantation and rejection. In this study, we characterized and compared the islets from GalTKO, GalTKO/hCD46, GalTKO/hCD46/hCD39, and wild-type (WT) neonatal pigs. METHODS: Islets were isolated from GMP and WT pig pancreases that have been packaged with ice pack for at least 24 hours. The difference in gene expression and function of islets were evaluated by microarray analysis and transplantation of islets under the kidney capsule of streptozotocin-induced diabetic immune-deficient mice, respectively. Blood glucose levels of these mice were monitored weekly post-transplantation for >100 days, and islet grafts were collected and evaluated for the presence of endocrine cells. RESULTS: The genes involved in extracellular components, cell adhesion, glucose metabolism, and inflammatory response are differentially expressed between GMP and WT pig islets. Variation in the ability of pig islets in correcting the diabetic state of the mouse recipients appears to be dependent on the pig donor. In addition, prolonged cold ischemia time had a negative effect on the transplant outcome. All normoglycemic mice were able to respond well to glucose challenge despite the initial differences in the ability of islet transplants to reverse their diabetic state. Islet xenografts of normoglycemic mice contained abundant insulin- and glucagon-positive cells. CONCLUSION: The effect of GMP and WT neonatal pig islet transplants on hyperglycemia in mice appears to be dependent on the pig donor, and prolonged cold ischemia time negatively affects the neonatal pig islet transplant outcome.


Subject(s)
Diabetes Mellitus, Experimental , Islets of Langerhans Transplantation , Islets of Langerhans , Animals , Cold Ischemia , Mice , Pancreas , Transplantation, Heterologous
2.
Xenotransplantation ; 19(5): 305-10, 2012.
Article in English | MEDLINE | ID: mdl-22970769

ABSTRACT

Serum anti-galactose-α1,3-galactose (Gal) IgM and IgG antibody levels were measured by ELISA in α1,3-galactosyltransferase gene-knockout (GTKO) pigs (78 estimations in 47 pigs). A low level of anti-Gal IgM was present soon after birth, and rose to a peak at 4-6 m, which was maintained thereafter even in the oldest pigs tested (at >2 yr). Anti-Gal IgG was also present at birth, peaked at 3 m, and after 6 m steadily decreased until almost undetectable at 20 m. No differences in this pattern were seen between pigs of different gender. Total IgM followed a similar pattern as anti-Gal IgM, but total IgG did not decrease after 6m. The data provide useful baseline data for future experimental studies in GTKO pigs, e.g., relating to the antibody response to WT pig allografts.


Subject(s)
Galactosyltransferases/deficiency , Galactosyltransferases/genetics , Sus scrofa/genetics , Sus scrofa/immunology , Trisaccharides/immunology , Age Factors , Animals , Female , Galactosyltransferases/immunology , Gene Knockout Techniques , Immunoglobulin G/blood , Immunoglobulin M/blood , Isoantibodies/blood , Male , Transplantation, Homologous
3.
Xenotransplantation ; 19(6): 342-54, 2012.
Article in English | MEDLINE | ID: mdl-23145497

ABSTRACT

BACKGROUND: The increasing availability of genetically engineered pigs is steadily improving the results of pig organ and cell transplantation in non-human primates (NHPs). Current techniques offer knockout of pig genes and/or knockin of human genes. Knowledge of normal values of hematologic, biochemical, coagulation, and other parameters in healthy genetically engineered pigs and NHPs is important, particularly following pig organ transplantation in NHPs. Furthermore, information on parameters in various NHP species may prove important in selecting the optimal NHP model for specific studies. METHODS: We have collected hematologic, biochemical, and coagulation data on 71 α1,3-galactosyltransferase gene-knockout (GTKO) pigs, 18 GTKO pigs additionally transgenic for human CD46 (GTKO.hCD46), four GTKO.hCD46 pigs additionally transgenic for human CD55 (GTKO.hCD46.hCD55), and two GTKO.hCD46 pigs additionally transgenic for human thrombomodulin (GTKO.hCD46.hTBM). RESULTS: We report these data and compare them with similar data from wild-type pigs and the three major NHP species commonly used in biomedical research (baboons, cynomolgus, and rhesus monkeys) and humans, largely from previously published reports. CONCLUSIONS: Genetic modification of the pig (e.g., deletion of the Gal antigen and/or the addition of a human transgene) (i) does not result in abnormalities in hematologic, biochemical, or coagulation parameters that might impact animal welfare, (ii) seems not to alter metabolic function of vital organs, although this needs to be confirmed after their xenotransplantation, and (iii) possibly (though, by no means certainly) modifies the hematologic, biochemical, and coagulation parameters closer to human values. This study may provide a good reference for those working with genetically engineered pigs in xenotransplantation research and eventually in clinical xenotransplantation.


Subject(s)
Blood Coagulation/physiology , Galactosyltransferases/metabolism , Transplantation, Heterologous/immunology , Animals , Animals, Genetically Modified/genetics , Galactosyltransferases/genetics , Gene Knockout Techniques/methods , Genetic Engineering/methods , Graft Rejection/genetics , Graft Rejection/immunology , Humans , Primates , Swine
4.
J Thorac Cardiovasc Surg ; 164(6): e411-e424, 2022 12.
Article in English | MEDLINE | ID: mdl-33933257

ABSTRACT

BACKGROUND: Galactose-α-1,3-galactose (alpha-gal) is a carbohydrate that is ubiquitously expressed in all mammals except for primates and humans. Patients can become sensitized to this antigen and develop alpha-gal syndrome (AGS), or a red meat allergy. Symptoms range from generalized gastroenteritis and malaise to anaphylaxis, and in endemic areas, the prevalence can be as high as 20%. Although AGS patients commonly avoid alpha-gal by avoiding meat, patients have also developed symptoms due to animal-derived medical products and devices. With the rise in transcatheter aortic valve replacement, we investigate the immunogenicity of common cardiac materials and valves. OBJECTIVE: To assess the in vitro immunoglobulin E response toward common medical products, including cardiac patch materials and bioprosthetic valves in patients with AGS. METHODS: Immunoblot and immunohistochemistry techniques were applied to assess immunoglobulin E reactivity to various mammalian derived tissues and medical products for patients with AGS. RESULTS: AGS serum showed strong reactivity to all of the commercially available, nonhuman products tested, including various decellularized cardiac patch materials and bioprosthetic aortic valves. AGS serum did not react to tissues prepared using alpha-gal knockout pigs. CONCLUSIONS: Despite commercial decellularization processes, alpha-gal continues to be present in animal-derived medical products, including bioprosthetic valves. Serum from patients with AGS demonstrates a strong affinity for these products in vitro. This may have serious potential implications for sensitized patients undergoing cardiac surgery, including early valve failure and accelerated coronary artery disease.


Subject(s)
Anaphylaxis , Food Hypersensitivity , Humans , Swine , Animals , Galactose , Immunoglobulin E , Anaphylaxis/diagnosis , Syndrome , Mammals
5.
Tissue Eng Part A ; 27(19-20): 1305-1320, 2021 10.
Article in English | MEDLINE | ID: mdl-33514288

ABSTRACT

Existing strategies for repair of major peripheral nerve injury (PNI) are inefficient at promoting axon regeneration and functional recovery and are generally ineffective for nerve lesions >5 cm. To address this need, we have previously developed tissue engineered nerve grafts (TENGs) through the process of axon stretch growth. TENGs consist of living, centimeter-scale, aligned axon tracts that accelerate axon regeneration at rates equivalent to the gold standard autograft in small and large animal models of PNI, by providing a newfound mechanism-of-action referred to as axon-facilitated axon regeneration (AFAR). To enable clinical-grade biomanufacturing of TENGs, a suitable cell source that is hypoimmunogenic, exhibits low batch-to-batch variability, and able to tolerate axon stretch growth must be utilized. To fulfill these requirements, a genetically engineered, FDA-approved, xenogeneic cell source, GalSafe® neurons, produced by Revivicor, Inc., have been selected to advance TENG biofabrication for eventual clinical use. To this end, sensory and motor neurons were harvested from genetically engineered GalSafe day 40 swine embryos, cultured in custom mechanobioreactors, and axon tracts were successfully stretch-grown to 5 cm within 25 days. Importantly, both sensory and motor GalSafe neurons were observed to tolerate established axon stretch growth regimes of ≥1 mm/day to produce continuous, healthy axon tracts spanning 1, 3, or 5 cm. Once stretch-grown, 1 cm GalSafe TENGs were transplanted into a 1 cm lesion in the sciatic nerve of athymic rats. Regeneration was assessed through histological measures at the terminal time point of 2 and 8 weeks. Neurons from GalSafe TENGs survived and elicited AFAR as observed when using wild-type TENGs. At 8 weeks postrepair, myelinated regenerated axons were observed in the nerve section distal to the injury site, confirming axon regeneration across the lesion. These experiments are the first to demonstrate successful harvest and axon stretch growth of GalSafe neurons for use as starting biomass for bioengineered nerve grafts as well as initial safety and efficacy in an established preclinical model-important steps for the advancement of clinical-grade TENGs for future regulatory testing and eventual clinical trials. Impact statement Biofabrication of tissue engineered medical products requires several steps, one of which is choosing a suitable starting biomass. To this end, we have shown that the clinical-grade, genetically engineered biomass-GalSafe® neurons-is a viable option for biomanufacturing of our tissue engineered nerve grafts (TENGs) to promote regeneration following major peripheral nerve injury. Importantly, this is a first step in clinical-grade TENG biofabrication, proving that GalSafe TENGs recapitulate the mechanism of axon-facilitated axon regeneration seen previously with research-grade TENGs.


Subject(s)
Axons , Nerve Regeneration , Animals , Motor Neurons , Myelin Sheath , Rats , Sciatic Nerve , Swine
6.
Biomaterials ; 276: 121029, 2021 09.
Article in English | MEDLINE | ID: mdl-34311317

ABSTRACT

Decellularized pig lungs recellularized with human lung cells offer a novel approach for organ transplantation. However, the potential immunogenicity of decellularized pig lungs following exposure to human tissues has not been assessed. We found that exposure of native lungs from wildtype and transgenic pigs lacking alpha (1,3)-galactosyltransferase (α-gal KO) to sera from normal healthy human volunteers demonstrated similar robust IgM and IgG immunoreactivity, comparably decreased in decellularized lungs. Similar results were observed with sera from patients who had previously undergone transcutaneous porcine aortic valve replacement (TAVR) or from patients with increased circulating anti-α-gal IgE antibodies (α-gal syndrome). Depleting anti-α-gal antibodies from the sera demonstrated both specificity of α-gal immunoreactivity and also residual immunoreactivity similar between wildtype and α-gal KO pig lungs. Exposure of human monocytes and macrophages to native wildtype lungs demonstrated greater induction of M2 phenotype than native α-gal KO pig lungs, which was less marked with decellularized lungs of either type. Overall, these results demonstrate that native wildtype and α-gal KO pig lungs provoke similar immune responses that are comparably decreased following decellularization. This provides a further platform for potential use of decellularized pig lungs in tissue engineering approaches and subsequent transplantation schemes but no obvious overall immunologic advantage of utilizing lungs obtained from α-gal KO pigs.


Subject(s)
Bioprosthesis , Heart Valve Prosthesis , Animals , Food Hypersensitivity , Galactosyltransferases/genetics , Humans , Lung , Swine , Transplantation, Heterologous
7.
Biomaterials ; 187: 93-104, 2018 12.
Article in English | MEDLINE | ID: mdl-30312852

ABSTRACT

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


Subject(s)
Galactosyltransferases/genetics , Lung Diseases/therapy , Lung/cytology , Tissue Scaffolds , Adaptive Immunity , Animals , Biocompatible Materials , Galactosyltransferases/immunology , Gene Knockout Techniques , Humans , Immunity, Humoral , Lung Diseases/immunology , Macaca mulatta , Swine , Tissue Engineering , Transplantation , Transplantation, Heterologous
8.
Spine J ; 6(1): 34-9, 2006.
Article in English | MEDLINE | ID: mdl-16413445

ABSTRACT

BACKGROUND CONTEXT: Several methods to sterilize allograft bone exist, including gamma irradiation and freeze-drying, which can alter the mechanical properties of the graft. Efforts are under way to develop a method for processing osseous allograft that maintains structural integrity. Herein is presented one such method. PURPOSE: To analyze the mechanical properties, compared with nontreated controls, of a novel sterilization process for allograft cortical bone. STUDY DESIGN/SETTING: A controlled biomechanical evaluation of allograft bone under various types of loading after a novel sterilization treatment. PATIENT SAMPLE: Not applicable; basic science. OUTCOME MEASURES: The load to failure was recorded for both the study and control groups, and statistical analysis of these results was performed. Significance level (alpha) and power (beta) were set to 0.05 and 0.90, respectively. Single-factor analysis of variance (ANOVA) was used to detect significant differences between the treated and untreated groups. A post-experimental power analysis was performed for each of the response variables. METHODS: Cortical tibia and femur samples from seven cadaveric donors (mean age 68.7 years) were treated with Biocleanse and compared with untreated samples with regard to density and strength. All samples were loaded to failure under diametral and biaxial compression, shear, and three-point bending. RESULTS: Statistical analysis was done on the density and failure stress for all modes of loading. ANOVA did not indicate a significant (p>.05) effect of treatment on the density except for the axial and biaxial specimens (p<.05). ANOVA analysis of failure stress demonstrated no significant differences (p>.05) between cortical bone treated with Biocleanse and untreated specimens under all four types of mechanical loading. Post-experimental power analysis revealed power to be greater than 0.9 for each test. CONCLUSIONS: Sterilization of allograft bone with Biocleanse does not significantly alter the mechanical properties when compared with untreated samples. The effect of this sterilization process on the osteoconductive and osteoinductive properties of allograft bone must be determined.


Subject(s)
Biomechanical Phenomena , Gamma Rays/therapeutic use , Graft Rejection/prevention & control , Sterilization/methods , Aged , Analysis of Variance , Bone Transplantation , Cadaver , Femur , Freeze Drying , Humans , Middle Aged , Probability , Reference Values , Risk Factors , Sensitivity and Specificity , Specimen Handling/methods , Stress, Mechanical , Tibia , Transplantation, Homologous
9.
Tissue Eng Part C Methods ; 22(8): 725-39, 2016 08.
Article in English | MEDLINE | ID: mdl-27310581

ABSTRACT

BACKGROUND: A novel potential approach for lung transplantation could be to utilize xenogeneic decellularized pig lung scaffolds that are recellularized with human lung cells. However, pig tissues express several immunogenic proteins, notably galactosylated cell surface glycoproteins resulting from alpha 1,3 galactosyltransferase (α-gal) activity, that could conceivably prevent effective use. Use of lungs from α-gal knock out (α-gal KO) pigs presents a potential alternative and thus comparative de- and recellularization of wild-type and α-gal KO pig lungs was assessed. METHODS: Decellularized lungs were compared by histologic, immunohistochemical, and mass spectrometric techniques. Recellularization was assessed following compartmental inoculation of human lung bronchial epithelial cells, human lung fibroblasts, human bone marrow-derived mesenchymal stromal cells (all via airway inoculation), and human pulmonary vascular endothelial cells (CBF) (vascular inoculation). RESULTS: No obvious differences in histologic structure was observed but an approximate 25% difference in retention of residual proteins was determined between decellularized wild-type and α-gal KO pig lungs, including retention of α-galactosylated epitopes in acellular wild-type pig lungs. However, robust initial recellularization and subsequent growth and proliferation was observed for all cell types with no obvious differences between cells seeded into wild-type versus α-gal KO lungs. CONCLUSION: These proof of concept studies demonstrate that decellularized wild-type and α-gal KO pig lungs can be comparably decellularized and comparably support initial growth of human lung cells, despite some differences in retained proteins. α-Gal KO pig lungs are a suitable platform for further studies of xenogeneic lung regeneration.


Subject(s)
Epithelial Cells/cytology , Fibroblasts/cytology , Galactosyltransferases/physiology , Lung/cytology , Mesenchymal Stem Cells/cytology , Regeneration/physiology , Animals , Animals, Genetically Modified , Cell Proliferation , Epithelial Cells/enzymology , Extracellular Matrix/enzymology , Fibroblasts/enzymology , Humans , Lung/enzymology , Mesenchymal Stem Cells/enzymology , Swine
10.
J Orthop Res ; 29(8): 1193-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21374710

ABSTRACT

Soft tissue allografts, such as the bone-patellar tendon-bone (BPTB) graft, have been frequently used for anterior cruciate ligament (ACL) reconstruction. As allografts are subjected to freezing and thawing for multiple cycles, the objective of this study was to measure the changes of the biomechanical properties of the human BPTB allograft after 4 and 8 freeze-thaw cycles in comparison to a single freeze-thaw cycle. Three BPTB specimens were procured from 21 human donors and divided into three groups: 1, 4, or 8 freeze-thaw cycles. Each freeze-thaw cycle consisted of freezing at -20 ± 10°C for more than 6 h and thawing at 22 ± 3°C for at least 6 h. Tensile testing of the BPTB specimens consisted of loading between 50 N and 250 N for 100 cycles and then loading to failure. Cyclic loading revealed a similar amount of creep (∼0.5 mm) among the three freeze-thaw cycles groups (p = 0.38). The stiffness of the BPTB graft for the 1, 4, and 8 freeze-thaw cycle groups were 244 ± 42 N/mm, 235 ± 39 N/mm, and 231 ± 40 N/mm, respectively (p = 0.43). Similar findings were obtained for the ultimate load of the BPTB graft (p = 0.14) and the tangent modulus of the PT substance (p = 0.41). The results of this study suggest that there would be little measurable effect on the structural properties of the BPTB graft or mechanical properties of the PT tissue substance following 8 freeze-thaw cycles. These BPTB allografts could potentially be re-frozen without a loss in their biomechanical properties, given appropriate storage and care.


Subject(s)
Bone-Patellar Tendon-Bone Grafting , Cryopreservation , Patellar Ligament , Adult , Aged , Biomechanical Phenomena , Female , Freezing , Humans , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL