Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 115
Filter
Add more filters

Publication year range
1.
Kidney Int ; 105(1): 84-98, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37839695

ABSTRACT

Clinical verification of adoptively transferred regulatory T cell (Treg) efficacy in transplantation remains challenging. Here, we examined the influence of autologous ex vivo-expanded polyclonal Tregs on kidney graft survival in a clinically relevant non-human primate model. Peripheral blood Tregs were isolated and expanded using artificial antigen presenting cells. Immunosuppression was comprised of tapered tacrolimus and CTLA4 immunoglobulin, in five animals each without or with Treg infusions. Escalating Treg doses were administered 6, 10, 13, 16, 20, 23, 27 and 30 days after transplant. Infused Tregs were monitored for Treg signature, anti-apoptotic (Bcl-2) and proliferation (Ki67) marker expression. Treg infusions prolonged median graft survival time significantly from 35 to 70 days. Treg marker (Ki67 and Bcl-2) expression by infused Tregs diminished after their infusion but remained comparable to that of circulating native Tregs. No major changes in circulating donor-reactive T cell responses or total Treg percentages, or in graft-infiltrating T cell subsets were observed with Treg infusion. However, Treg infusion was associated with significant increases in CD163 expression by circulating HLA-DR+ myeloid cells and elevated levels of circulating soluble CD163. Further, graft-infiltrating CD163+ cells were increased with Treg infusion. Thus, multiple Treg infusions were associated with M2-like myeloid cell enhancement that may mediate immunomodulatory, anti-inflammatory and graft reparative effects.


Subject(s)
Primates , T-Lymphocytes, Regulatory , Animals , Ki-67 Antigen/metabolism , Kidney , Allografts , Myeloid Cells , Proto-Oncogene Proteins c-bcl-2/metabolism
2.
Xenotransplantation ; 28(4): e12700, 2021 07.
Article in English | MEDLINE | ID: mdl-34036638

ABSTRACT

Pigs deficient in three glycosyltransferase enzymes (triple-knockout [TKO] pigs) and expressing "protective" human transgenes are likely sources of organs for transplantation into human recipients. Testing of human sera against red blood cells (RBCs) and peripheral blood mononuclear cells (PBMCs) from TKO pigs has revealed minimal evidence of natural antibody binding. However, unlike humans, baboons exhibit natural antibody binding to TKO pig cells. The xenoantigen specificities of these natural antibodies are postulated to be one or more carbohydrate moieties exposed when N-glycolylneuraminic acid (Neu5Gc) is deleted. The aim of this study was to compare the survival of renal grafts in baboons from pigs that either expressed Neu5Gc (GTKO pigs; Group1, n = 5) or did not express Neu5Gc (GTKO/CMAHKO [DKO] or TKO pigs; Group2, n = 5). An anti-CD40mAb-based immunosuppressive regimen was administered in both groups. Group1 kidneys functioned for 90-260 days (median 237, mean 196 days), with histopathological features of antibody-mediated rejection in two kidneys. Group2 kidneys functioned for 0-183 days (median 35, mean 57), with all of the grafts exhibiting histologic features of antibody-mediated rejection. These findings suggest that the absence of expression of Neu5Gc on pig kidneys impacts graft survival in baboon recipients.


Subject(s)
Kidney Transplantation , Animals , Animals, Genetically Modified , Graft Rejection , Leukocytes, Mononuclear , Neuraminic Acids , Papio , Swine , Transplantation, Heterologous
3.
Xenotransplantation ; 28(6): e12715, 2021 11.
Article in English | MEDLINE | ID: mdl-34644438

ABSTRACT

INTRODUCTION: Pigs deficient in three glycosyltransferase enzymes (triple-knockout [TKO] pigs, that is, not expressing the three known carbohydrate xenoantigens) and expressing 'protective' human transgenes are considered a likely source of organs for transplantation into human recipients. Some human sera have no or minimal natural antibody binding to red blood cells (RBCs) and peripheral blood mononuclear cells (PBMCs) from TKO pigs. However, all Old World monkeys exhibit natural antibody binding to TKO pig cells. The xenoantigen targets of Old World monkey natural antibodies are postulated to be carbohydrate moieties exposed when the expression of the carbohydrate N-glycolylneuraminic acid (Neu5Gc) is deleted. The aim of this study was to compare the survival in baboons and histopathology of renal grafts from pigs that either (a) expressed Neu5Gc (GTKO pigs; Group 1) or (b) did not express Neu5Gc (GTKO/CMAHKO [DKO] or TKO pigs; Group 2). METHODS: Life-supporting renal transplants were carried out using GTKO (n = 5) or DKO/TKO (n = 5) pig kidneys under an anti-CD40mAb-based immunosuppressive regimen. RESULTS: Group 1 baboons survived longer than Group 2 baboons (median 237 vs. 35 days; mean 196 vs. 57 days; p < 0.07) and exhibited histopathological features of antibody-mediated rejection in only two kidneys. Group 2 exhibited histopathological features of antibody-mediated rejection in all five grafts, with IgM and IgG binding to renal interstitial arteries and peritubular capillaries. Rejection-free survival was significantly longer in Group 1 (p < 0.05). CONCLUSIONS: The absence of expression of Neu5Gc on pig kidney grafts is associated with increased binding of baboon antibodies to pig endothelium and reduced graft survival.


Subject(s)
Kidney , Leukocytes, Mononuclear , Animals , Animals, Genetically Modified , Carbohydrates , Graft Rejection , Papio , Swine , Transplantation, Heterologous
4.
Am J Transplant ; 20(6): 1691-1702, 2020 06.
Article in English | MEDLINE | ID: mdl-31883190

ABSTRACT

Early phase clinical trials are evaluating the feasibility, safety, and therapeutic potential of ex vivo expanded regulatory T cells (Treg) in transplantation. A limitation is the paucity of naturally occurring Treg numbers in peripheral blood. Hence, protracted ex vivo expansion is required to obtain sufficient Treg in order to meet target cell doses. Because cytokine administration has been used successfully to mobilize immune cells to the peripheral blood in experimental and clinical studies, we hypothesized that granulocyte macrophage-colony-stimulating factor (GM-CSF) and granulocyte-CSF (G-CSF) administration would enhance Treg percentages in leukapheresis products of rhesus monkeys. Following combined GM-CSF and G-CSF administration, the incidence of Treg in peripheral blood and leukapheresis products was elevated significantly, where approximately 3.7 × 106 /kg CD4+ CD25hi Foxp3hi or 6.8 × 106 /kg CD4+ CD25hi CD127lo Treg can be collected from individual products. Mobilized Treg expressed a comparable repertoire of surface markers, chemokine receptors, and transcription factors to naïve monkey peripheral blood Treg. Furthermore, when expanded ex vivo, mobilized leukapheresis product and peripheral blood Treg exhibited similar ability to suppress autologous CD4+ and CD8+ T cell proliferation. These observations indicate that leukapheresis products from combined GM-CSF- and G-CSF-mobilized individuals are a comparatively rich source of Treg and may circumvent long-term ex vivo expansion required for therapeutic application.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor , T-Lymphocytes, Regulatory , Animals , Granulocyte Colony-Stimulating Factor , Leukapheresis , Macaca mulatta , Transcription Factors
5.
Cell Immunol ; 351: 104087, 2020 05.
Article in English | MEDLINE | ID: mdl-32197811

ABSTRACT

Nonhuman primates (NHP) are important pre-clinical models for evaluation of the safety and efficacy of the most promising potential therapeutic advances in organ transplantation based on rodent studies. Although rare, dendritic cells (DC) play important roles in preservation of self tolerance and DC with immunoregulatory properties (regulatory DC; DCreg) can promote transplant tolerance in rodents when adoptively transferred to allograft recipients. NHP DCreg can be generated ex vivo from bone marrow precursors or blood monocytes of cynomolgus or rhesus macaques or baboons. NHP DCreg generated in the presence of anti-inflammatory factors that confer stability and resistance to maturation, subvert alloreactive T cell responses. When infused into rhesus renal allograft recipients before transplant, they safely prolong MHC mis-matched graft survival, associated with attenuation of anti-donor immune reactivity. In this concise review we describe the properties of NHP DCreg and discuss their influence on T cell responses, alloimmunity and organ transplant survival.


Subject(s)
Dendritic Cells/immunology , Disease Models, Animal , Kidney Transplantation , Transplantation Tolerance/immunology , Animals , Macaca fascicularis , Macaca mulatta , Papio , Primates , Transplantation, Homologous
6.
Xenotransplantation ; 26(4): e12516, 2019 07.
Article in English | MEDLINE | ID: mdl-30989742

ABSTRACT

Xenotransplantation research has made considerable progress in recent years, largely through the increasing availability of pigs with multiple genetic modifications. We suggest that a pig with nine genetic modifications (ie, currently available) will provide organs (initially kidneys and hearts) that would function for a clinically valuable period of time, for example, >12 months, after transplantation into patients with end-stage organ failure. The national regulatory authorities, however, will likely require evidence, based on in vitro and/or in vivo experimental data, to justify the inclusion of each individual genetic modification in the pig. We provide data both from our own experience and that of others on the advantages of pigs in which (a) all three known carbohydrate xenoantigens have been deleted (triple-knockout pigs), (b) two human complement-regulatory proteins (CD46, CD55) and two human coagulation-regulatory proteins (thrombomodulin, endothelial cell protein C receptor) are expressed, (c) the anti-apoptotic and "anti-inflammatory" molecule, human hemeoxygenase-1 is expressed, and (d) human CD47 is expressed to suppress elements of the macrophage and T-cell responses. Although many alternative genetic modifications could be made to an organ-source pig, we suggest that the genetic manipulations we identify above will all contribute to the success of the initial clinical pig kidney or heart transplants, and that the beneficial contribution of each individual manipulation is supported by considerable experimental evidence.


Subject(s)
Animals, Genetically Modified/genetics , Graft Rejection/prevention & control , Swine/genetics , Transplantation, Heterologous , Animals , Animals, Genetically Modified/immunology , CD47 Antigen/genetics , CD47 Antigen/immunology , CD55 Antigens/genetics , CD55 Antigens/immunology , Endothelial Protein C Receptor/genetics , Endothelial Protein C Receptor/immunology , Galactosyltransferases/deficiency , Galactosyltransferases/genetics , Galactosyltransferases/immunology , Gene Knock-In Techniques , Gene Knockout Techniques , Graft Rejection/immunology , Heme Oxygenase-1/genetics , Heme Oxygenase-1/immunology , Humans , Membrane Cofactor Protein/genetics , Membrane Cofactor Protein/immunology , Mixed Function Oxygenases/deficiency , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/immunology , N-Acetylgalactosaminyltransferases/deficiency , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/immunology , Swine/immunology , Thrombomodulin/genetics , Thrombomodulin/immunology
7.
Cell Immunol ; 329: 50-55, 2018 07.
Article in English | MEDLINE | ID: mdl-29803290

ABSTRACT

Putative monocytic myeloid-derived suppressor cells (mMDSC; lineage-HLA-DR-/lo) were generated in 7-day cultures from normal rhesus macaque bone marrow (BM) cells in GM-CSF and IL-6. Three subsets were identified based on their differential expression of CD14, CD33, CD34 and CD11b. Following flow sorting, assessment of the capacity of these subsets to suppress anti-CD3/CD28-stimulated CD4 and CD8 T cell proliferation revealed that the most potent population was CD14hiCD33-/loCD34loCD11bhi. These BM-derived mMDSC markedly increased the incidence of CD4+CD25+CD127-Foxp3+ regulatory T cells in responder T cell populations. They offer potential value in testing the therapeutic efficacy of immunoregulatory mMDSC for the promotion of tolerance in nonhuman primate transplant models.


Subject(s)
Bone Marrow Cells/cytology , Myeloid-Derived Suppressor Cells/physiology , T-Lymphocytes, Regulatory/physiology , Animals , Biomarkers , Bone Marrow/pathology , Bone Marrow/physiology , Flow Cytometry , Macaca mulatta , Male , Monocytes/physiology , Myeloid Cells/physiology , Myeloid-Derived Suppressor Cells/metabolism , Primary Cell Culture/methods , T-Lymphocyte Subsets/physiology
8.
Xenotransplantation ; 25(3): e12415, 2018 05.
Article in English | MEDLINE | ID: mdl-29913039

ABSTRACT

Regulatory T cells (Treg) are currently being evaluated in clinical allotransplantation for tolerance induction, with proven safety in humans with autoimmune diseases and graft-versus-host disease. A considerable amount of recent data suggests that additional factors may need to be validated, including the stability and commitment of newly discovered Treg subsets under inflammatory conditions, to further warrant safe and effective Treg-based therapeutic approaches. This review explores the opportunities and challenges of Treg-based cell therapy in xenotransplantation. The emerging new technologies for genetic modifications of the donor pig offer a major advantage for Treg therapy to improve xenograft protection. Particularly, the feasibility of (i) ex vivo expansion of donor (pig)-specific Treg for infusion, and (ii) development of Treg in situ for the life of the xenograft. Our understanding of the Treg biology and their role in xenograft protection, under the newly developed immunosuppressive protocols remains limited. The incidence of various Treg subpopulations in xenograft recipients and their suppressive efficacy across species barriers are largely unknown. Finally, deciphering the dynamics of Treg function, and their interaction with adaptive and innate immune cells are of critical importance to design safe, effective and clinically relevant Treg-based therapeutic approaches in xenotransplantation.


Subject(s)
Heterografts/immunology , Immunosuppressive Agents/pharmacology , T-Lymphocytes, Regulatory/immunology , Transplantation, Heterologous , Animals , Humans , Immunity, Innate/immunology , Inflammation/immunology , Transplantation, Heterologous/methods
10.
Curr Opin Organ Transplant ; 23(5): 538-545, 2018 10.
Article in English | MEDLINE | ID: mdl-30036199

ABSTRACT

PURPOSE OF REVIEW: There is currently increased focus on improved understanding of how dendritic cell tolerogenicity is determined and maintained, and on their therapeutic potential. We review recent progress in profiling of regulatory dendritic cells (DCreg), innovative approaches to enhancing dendritic cell tolerogenicity in situ, ex-vivo generation of DCreg and initial clinical testing of these cells in organ transplantation. RECENT FINDINGS: "Omics' studies indicate that the distinctive properties of DCreg are the result of a specific transcriptional program characterized by activation of tolerance-enhancing genes, rather than the retention of an immature state. In situ dendritic cell-directed targeting of nanovesicles bearing immune regulatory molecules can trigger in-vivo expansion of Ag-specific regulatory cells. Innovative approaches to ex-vivo modification of dendritic cells to enhance their regulatory function and capacity to migrate to secondary lymphoid organs has been described. Cross-dressing (with donor major histocompatibility complex molecules) of graft-infiltrating host dendritic cells that regulate antidonor T-cell responses has been implicated in "spontaneous' liver transplant tolerance. Clinical trials of DCreg therapy have begun in living donor renal and liver transplantation. SUMMARY: Further definition of molecules that can be targeted to promote the function and stability of DCreg in vivo may lead to standardization of DCreg manufacturing for therapeutic application.


Subject(s)
Dendritic Cells/immunology , Immune Tolerance/genetics , Humans
11.
Kidney Int ; 91(4): 790-796, 2017 04.
Article in English | MEDLINE | ID: mdl-27914702

ABSTRACT

There are >100,000 patients waiting for kidney transplants in the United States and a vast need worldwide. Xenotransplantation, in the form of the transplantation of kidneys from genetically engineered pigs, offers the possibility of overcoming the chronic shortage of deceased and living human donors. These genetic manipulations can take the form of (i) knockout of pig genes that are responsible for the expression of antigens against which the primate (human or nonhuman primate) has natural "preformed" antibodies that bind and initiate complement-mediated destruction or (ii) the insertion of human transgenes that provide protection against the human complement, coagulation, or inflammatory responses. Between 1989 and 2015, pig kidney graft survival in nonhuman primates increased from 23 days to almost 10 months. There appear to be no clinically significant physiological incompatibilities in renal function between pigs and primates. The organ-source pigs will be housed in a biosecure environment, and thus the risk of transferring an exogenous potentially pathogenic microorganism will be less than that after allotransplantation. Although the risk associated with porcine endogenous retroviruses is considered small, techniques are now available whereby they could potentially be excluded from the pig. The US Food and Drug Administration suggests that xenotransplantation should be restricted to "patients with serious or life-threatening diseases for whom adequately safe and effective alternative therapies are not available." These might include those with (i) a high degree of allosensitization to human leukocyte antigens or (ii) rapid recurrence of primary disease in previous allografts. The potential psychosocial, regulatory, and legal aspects of clinical xenotransplantation are briefly discussed.


Subject(s)
Kidney Transplantation/methods , Sus scrofa/genetics , Transplantation, Heterologous , Animals , Animals, Genetically Modified , Genotype , Graft Rejection/genetics , Graft Rejection/immunology , Graft Rejection/prevention & control , Graft Survival , Heterografts , History, 20th Century , History, 21st Century , Humans , Immunosuppressive Agents/therapeutic use , Kidney Transplantation/adverse effects , Kidney Transplantation/history , Phenotype , Risk Factors , Species Specificity , Sus scrofa/immunology , Tissue Donors/supply & distribution , Transplantation Tolerance , Transplantation, Heterologous/adverse effects , Transplantation, Heterologous/history , Treatment Outcome
12.
Xenotransplantation ; 24(4)2017 07.
Article in English | MEDLINE | ID: mdl-28547819

ABSTRACT

The results of the assay for measuring anti-non-Gal antibodies (which affect pig xenograft survival) in recipients are important. Serum incubation time and concentration may be important factors in the extent of antibody binding to the graft. The aim of this in vitro study was to determine the optimal incubation time and serum concentration for measuring anti-non-Gal antibody binding to porcine aortic endothelial cells (pAECs). Pooled human, naive, and sensitized baboon sera were incubated with wild-type, α1,3-galactosyltransferase gene-knockout (GTKO), and GTKO/human CD55 pAECs. IgM/IgG binding to pAECs after varying serum incubation times (0.5, 1, 2, and 3 hour) and concentrations (5, 10, 20, and 40 µL) was determined by flow cytometry. An increase in incubation time from 30 minutes to 2 hour was associated with increases in anti-non-Gal IgM/IgG binding to GTKO and GTKO/hCD55 pAECs of pooled human, naive and sensitized baboon sera (P<.05). Pooled human serum showed a significant increase in anti-non-Gal IgM (1.5 times) and a minimal increase in anti-non-Gal IgG antibody binding. IgM/IgG binding of sensitized baboon serum to GTKO pAECs after 2-hour incubation was 1.5 times and 2 times greater than after 30-minutes incubation, respectively, whereas naïve baboon sera showed minimal (non-significant) increase in anti-non-Gal IgM/IgG antibody binding. With 2-hour incubation, increasing the serum concentration from 5 µL to 20 µL significantly increased antibody binding to non-Gal antigens in pooled human and sensitized baboon serum. With naïve baboon serum, only IgG was significantly increased. Increasing the serum incubation time contributed to improve the sensitivity of detecting anti-non-Gal antibodies, without affecting cell viability in vitro.


Subject(s)
Endothelial Cells/metabolism , Immunoglobulin G/blood , Immunoglobulin M/blood , Animals , Animals, Genetically Modified/immunology , Antibodies, Heterophile/blood , Endothelial Cells/immunology , Gene Knockout Techniques , Graft Rejection/immunology , Graft Survival/immunology , Heterografts/immunology , Humans , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Swine , Time Factors , Transplantation, Heterologous/methods
13.
Xenotransplantation ; 24(2)2017 03.
Article in English | MEDLINE | ID: mdl-28294424

ABSTRACT

Inflammation is known to preclude tolerance after transplantation. We have previously shown that systemic inflammation in xenograft recipients (SIXR) precedes activation of coagulation in the absence of T cell responses. Accordingly, SIXR may amplify innate and adaptive immune responses against xenografts after pig-to-primate xenotransplantation, even with efficient immunosuppressive therapy. We evaluated the impact of anti-inflammatory agents on pro-inflammatory cytokines and chemokines in pig artery patch and heart xenograft recipients. Baboons received an artery patch (Group1, n=8) or heart (Group2, n=4) from genetically engineered pigs. All baboons received lymphodepletion with thymoglobulin (ATG) and costimulation blockade-based immunosuppression (anti-CD40 and/or CTLA4Ig). In Group1, baboons received either (i) no anti-inflammatory agents (n=2), (ii) cobra venom factor (CVF, n=2), (iii) α1-antitrypsin (AAT, n=2), or (iv) interleukin (IL)-6 receptor antagonist (IL-6RA, n=2). In Group2, all baboon received corticosteroids, either without (n=2) or with (n=2) IL-6RA. Serum IFN-γ, TNF-α, IL-1ß, IL-17, IL-6, IL-8, MCP-1, and sCD40L levels were measured by Luminex. Fibrinogen, D-dimers, and C-reactive protein (C-RP) were also measured. Recipient baboon T cell proliferation was evaluated by mixed lymphocyte reaction (MLR) before and after transplantation. Pig and baboon tissue factor (TF) mRNA levels in heart xenografts were measured by RT-PCR. In no recipient was a marked increase in T cell response to pig cells observed after transplantation. In Groups 1 and 2, post-transplantation levels of IFN-γ, TNF-α, IL-1ß, and IL-17 remained comparable to or lower than pre-transplant levels, except in one heart recipient that succumbed to CMV infection. In Group1, when no anti-inflammatory agent was administered, post-transplant levels of IL-6, IL-8, and MCP-1 were elevated. After CVF, IL-6, IL-8, and MCP-1 remained low. After IL-6RA, IL-6 and MCP-1 were elevated. After AAT, IL-8 was elevated. sCD40L became elevated intermittently in most recipients irrespective of the administered anti-inflammatory agent. In Group2, IL-6 was transiently elevated, particularly after IL-6RA administration. MCP-1 gradually increased by 2 months in Group2 recipients. sCD40L generally remained low except in one recipient. In Group1 and Group2 recipients, C-RP levels were elevated except after IL-6RA administration, while D-dimers were elevated regardless of administration of anti-inflammatory agent. In Group2, pig TF mRNA levels were increased in heart xenografts compared to naive pig hearts, irrespective of IL-6 receptor antagonist administration. Additionally, baboon TF mRNA levels were detectable in heart xenografts, but not in naive pig hearts. Some pro-inflammatory cytokines and chemokines are elevated in xenograft recipients, even with efficient T cell-directed immunosuppressive therapy. Persistent elevation of D-dimers, and individual cytokines and chemokines suggest a continuous inflammatory response, despite administration of anti-inflammatory agents. Systemic administration of combined anti-inflammatory agents as well as complement regulation may be essential to prevent SIXR after xenotransplantation.


Subject(s)
Graft Rejection/immunology , Graft Survival/immunology , Heart Transplantation , Heterografts/immunology , Inflammation/immunology , Transplantation, Heterologous , Animals , Animals, Genetically Modified , Graft Survival/drug effects , Heart Transplantation/methods , Humans , Immunosuppression Therapy/methods , Immunosuppressive Agents/pharmacology , Interleukin-17/metabolism , Papio , Swine , Transplantation, Heterologous/methods
14.
Xenotransplantation ; 24(2)2017 03.
Article in English | MEDLINE | ID: mdl-28303661

ABSTRACT

BACKGROUND: Genetically engineered pigs could provide a source of kidneys for clinical transplantation. The two longest kidney graft survivals reported to date have been 136 and 310 days, but graft survival >30 days has been unusual until recently. METHODS: Donor pigs (n=4) were on an α1,3-galactosyltransferase gene-knockout (GTKO)/human complement regulatory protein (CD46) background (GTKO/CD46). In addition, the pigs were transgenic for at least one human coagulation regulatory protein. Two baboons received a kidney from a six-gene pig (GroupA) and two from a three-gene pig (GroupB). Immunosuppressive therapy was identical in all four cases and consisted of anti-thymoglobulin (ATG)+anti-CD20mAb (induction) and anti-CD40mAb+rapamycin+corticosteroids (maintenance). Anti-TNF-α and anti-IL-6R mAbs were administered to reduce the inflammatory response. Baboons were followed by clinical/laboratory monitoring of immune/coagulation/inflammatory/physiological parameters. At biopsy or euthanasia, the grafts were examined by microscopy. RESULTS: The two GroupA baboons remained healthy with normal renal function >7 and >8 months, respectively, but then developed infectious complications. However, no features of a consumptive coagulopathy, eg, thrombocytopenia and reduction of fibrinogen, or of a protein-losing nephropathy were observed. There was no evidence of an elicited anti-pig antibody response, and histology of biopsies taken at approximately 4, 6, and 7 months and at necropsy showed no significant abnormalities. In contrast, both GroupB baboons developed features of a consumptive coagulopathy and required euthanasia on day 12. CONCLUSIONS: The combination of (i) a graft from a specific six-gene genetically modified pig, (ii) an effective immunosuppressive regimen, and (iii) anti-inflammatory therapy prevented immune injury, a protein-losing nephropathy, and coagulation dysfunction for >7 months. Although the number of experiments is very limited, our impression is that expression of human endothelial protein C receptor (±CD55) in the graft is important if coagulation dysregulation is to be avoided.


Subject(s)
Graft Rejection/immunology , Graft Survival/immunology , Kidney Transplantation , Kidney/surgery , Transplantation, Heterologous , Animals , Animals, Genetically Modified , Graft Rejection/genetics , Graft Survival/genetics , Humans , Immunosuppressive Agents/administration & dosage , Kidney/immunology , Kidney Transplantation/methods , Papio , Swine , Transplantation, Heterologous/methods , Transplants/drug effects , Transplants/immunology , Tumor Necrosis Factor-alpha/immunology
15.
Xenotransplantation ; 24(2)2017 03.
Article in English | MEDLINE | ID: mdl-28130881

ABSTRACT

BACKGROUND: Some patients with acute or acute-on-chronic hepatic failure die before a suitable human liver allograft becomes available. Encouraging results have been achieved in such patients by the transplantation of human hepatocyte progenitor cells from fetal liver tissue. The aim of the study was to explore survival of hepatocytes from genetically engineered pigs after direct injection into the spleen and other selected sites in immunosuppressed baboons to monitor the immune response and the metabolic function and survival of the transplanted hepatocytes. METHODS: Baboons (n=3) were recipients of GTKO/hCD46 pig hepatocytes. All three baboons received anti-thymocyte globulin (ATG) induction and tapering methylprednisolone. Baboon 1 received maintenance immunosuppressive therapy with tacrolimus and rapamycin. Baboons 2 and 3 received an anti-CD40mAb/rapamycin-based regimen that prevents sensitization to pig solid organ grafts. The baboons were euthanized 4 or 5 weeks after hepatocyte transplantation. The baboon immune response was monitored by the measurement of anti-non-Gal IgM and IgG antibodies (by flow cytometry) and CFSE-mixed lymphocyte reaction. Monitoring for hepatocyte survival and function was by (i) real-time PCR detection of porcine DNA, (ii) real-time PCR for porcine gene expression, and (iii) pig serum albumin levels (by ELISA). The sites of hepatocyte injection were examined microscopically. RESULTS: Detection of porcine DNA and porcine gene expression was minimal at all sites of hepatocyte injection. Serum levels of porcine albumen were very low-500-1000-fold lower than in baboons with orthotopic pig liver grafts, and approximately 5000-fold lower than in healthy pigs. No hepatocytes or infiltrating immune cells were seen at any of the injection sites. Two baboons (Baboons 1 and 3) demonstrated a significant increase in anti-pig IgM and an even greater increase in IgG, indicating sensitization to pig antigens. DISCUSSION AND CONCLUSIONS: As a result of this disappointing experience, the following points need to be considered. (i) Were the isolated pig hepatocytes functionally viable? (ii) Are pig hepatocytes more immunogenic than pig hearts, kidneys, artery patch grafts, or islets? (iii) Does injection of pig cells (antigens) into the spleen and/or lymph nodes stimulate a greater immune response than when pig tissues are grafted at other sites? (iv) Did the presence of the recipient's intact liver prevent survival and proliferation of pig hepatocytes? (v) Is pig CD47-primate SIRP-α compatibility essential? In conclusion, the transplantation of genetically engineered pig hepatocytes into multiple sites in immunosuppressed baboons was associated with very early graft failure. Considerable further study is required before clinical trials should be undertaken.


Subject(s)
Graft Survival/immunology , Hepatocytes/immunology , Transplantation, Heterologous , Animals , Animals, Genetically Modified , Antibodies/immunology , Antibody Formation/genetics , Antibody Formation/immunology , Antigens/immunology , Graft Survival/drug effects , Hepatocytes/transplantation , Immunosuppression Therapy/methods , Immunosuppressive Agents/pharmacology , Papio hamadryas/immunology , Swine , Transplantation, Heterologous/methods
16.
Xenotransplantation ; 23(2): 83-105, 2016 03.
Article in English | MEDLINE | ID: mdl-26813438

ABSTRACT

The immunologic barriers to successful xenotransplantation are related to the presence of natural anti-pig antibodies in humans and non-human primates that bind to antigens expressed on the transplanted pig organ (the most important of which is galactose-α1,3-galactose [Gal]), and activate the complement cascade, which results in rapid destruction of the graft, a process known as hyperacute rejection. High levels of elicited anti-pig IgG may develop if the adaptive immune response is not prevented by adequate immunosuppressive therapy, resulting in activation and injury of the vascular endothelium. The transplantation of organs and cells from pigs that do not express the important Gal antigen (α1,3-galactosyltransferase gene-knockout [GTKO] pigs) and express one or more human complement-regulatory proteins (hCRP, e.g., CD46, CD55), when combined with an effective costimulation blockade-based immunosuppressive regimen, prevents early antibody-mediated and cellular rejection. However, low levels of anti-non-Gal antibody and innate immune cells and/or platelets may initiate the development of a thrombotic microangiopathy in the graft that may be associated with a consumptive coagulopathy in the recipient. This pathogenic process is accentuated by the dysregulation of the coagulation-anticoagulation systems between pigs and primates. The expression in GTKO/hCRP pigs of a human coagulation-regulatory protein, for example, thrombomodulin, is increasingly being associated with prolonged pig graft survival in non-human primates. Initial clinical trials of islet and corneal xenotransplantation are already underway, and trials of pig kidney or heart transplantation are anticipated within the next few years.


Subject(s)
Graft Rejection/pathology , Graft Survival/immunology , Primates , Sus scrofa/immunology , Transplantation, Heterologous , Animals , Animals, Genetically Modified/immunology , Graft Rejection/immunology , Humans , Transplantation, Heterologous/methods
17.
Xenotransplantation ; 23(2): 137-50, 2016 03.
Article in English | MEDLINE | ID: mdl-26988899

ABSTRACT

BACKGROUND: The impact that the absence of expression of NeuGc in pigs might have on pig organ or cell transplantation in humans has been studied in vitro, but only using red blood cells (pRBCs) and peripheral blood mononuclear cells (pPBMCs) as the target cells for immune assays. We have extended this work in various in vitro models and now report our initial results. METHODS: The models we have used involve GTKO/hCD46 and GTKO/hCD46/NeuGcKO pig aortas and corneas, and pRBCs, pPBMCs, aortic endothelial cells (pAECs), corneal endothelial cells (pCECs), and isolated pancreatic islets. We have investigated the effect of the absence of NeuGc expression on (i) human IgM and IgG binding, (ii) the T-cell proliferative response, (iii) human platelet aggregation, and (iv) in an in vitro assay of the instant blood-mediated inflammatory reaction (IBMIR) following exposure of pig islets to human blood/serum. RESULTS: The lack of expression of NeuGc on some pig tissues (aortas, corneas) and cells (RBCs, PBMCs, AECs) significantly reduces the extent of human antibody binding. In contrast, the absence of NeuGc expression on some pig tissues (CECs, isolated islet cells) does not reduce human antibody binding, possibly due to their relatively low NeuGc expression level. The strength of the human T-cell proliferative response may also be marginally reduced, but is already weak to GTKO/hCD46 pAECs and islet cells. We also demonstrate that the absence of NeuGc expression on GTKO/hCD46 pAECs does not reduce human platelet aggregation, and nor does it significantly modify the IBMIR to pig islets. CONCLUSION: The absence of NeuGc on some solid organs from GTKO/hCD46/NeuGcKO pigs should reduce the human antibody response after clinical transplantation when compared to GTKO/hCD46 pig organs. However, the clinical benefit of using certain tissue (e.g., cornea, islets) from GTKO/hCD46/NeuGcKO pigs is questionable.


Subject(s)
Galactosyltransferases/metabolism , Membrane Cofactor Protein/metabolism , Neuraminic Acids/metabolism , Transplantation, Heterologous , Animals , Animals, Genetically Modified , Cells, Cultured , Endothelial Cells/immunology , Galactosyltransferases/deficiency , Humans , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/immunology , Membrane Cofactor Protein/genetics , Swine , Transplantation, Heterologous/methods
18.
Xenotransplantation ; 23(4): 293-9, 2016 07.
Article in English | MEDLINE | ID: mdl-27374212

ABSTRACT

BACKGROUND: It has been well documented that the level of serum/plasma free triiodothyronine (fT3) falls rapidly following brain death or during certain surgical procedures, for example, heart surgery carried out on cardiopulmonary bypass. The level in patients following cardiopulmonary bypass usually recovers within 2 days. METHODS: We have measured serum fT3 in healthy naïve baboons (n = 31), healthy naïve monkeys (n = 5), and after pig-to-baboon heterotopic heart xenotransplantation (xenoTx) (Group 1, n = 9), orthotopic liver xenoTx (Group 2, n = 10), artery patch xenoTx (Group 3, n = 9), and in monkey-to-monkey heterotopic heart alloTx (Group 4, n = 5). RESULTS: The mean level of fT3 in healthy naïve baboons was 3.1 ± 0.9 pg/ml and in healthy naïve monkeys was 2.6 ± 0.3 pg/ml. Following pig heart, liver, and artery patch xenoTx and monkey heart alloTx, there was an immediate rapid fall in fT3 level. Recovery of fT3 was more rapid in Groups 3 and 4 than in Groups 1 and 2. In Group 1, within 4 days fT3 had recovered, but only to the lower limit of normal range, where it remained throughout follow-up (for up to 42 days). In Group 2, no recovery was seen during the 7 days of follow-up. In immunosuppressed baboons with pig patch grafts that received IL-6R blockade (n = 2), the fT3 tended to rise higher than in those that received no IL-6R blockade (n = 6). CONCLUSIONS: Following operative procedures, there is a dramatic fall in serum fT3 levels. The persistent low level of fT3 after pig heart and liver xenoTx may be associated with a continuing inflammatory state. We suggest that consideration should be given to the replacement of T3 therapy to maintain normal fT3 levels, particularly in nonhuman primates undergoing orthotopic pig heart or liver xenoTx.


Subject(s)
Heterografts/metabolism , Thyroid Hormones/metabolism , Transplantation, Heterologous , Animals , Arteries/metabolism , Arteries/transplantation , Heart Transplantation , Liver Transplantation , Papio , Swine , Transplantation, Heterologous/methods , Triiodothyronine/blood
19.
Cell Immunol ; 295(1): 19-28, 2015 May.
Article in English | MEDLINE | ID: mdl-25732601

ABSTRACT

We expanded flow-sorted Foxp3(+) cynomolgus monkey regulatory T cells (Treg) >1000-fold after three rounds of stimulation with anti-CD3 mAb-loaded artificial antigen-presenting cells, rapamycin (first round only) and IL-2. The expanded Treg maintained their expression of Treg signature markers, CD25, CD27, CD39, Foxp3, Helios, and CTLA-4, as well as CXCR3, which plays an important role in T cell migration to sites of inflammation. In contrast to expanded effector T cells (Teff), expanded Treg produced minimal IFN-γ and IL-17 and no IL-2 and potently suppressed Teff proliferation. Following cryopreservation, thawed Treg were less viable than their freshly-expanded counterparts, although no significant changes in phenotype or suppressive ability were observed. Additional rounds of stimulation/expansion restored maximal viability. Furthermore, adoptively-transferred autologous Treg expanded from cryopreserved second round stocks and labeled with CFSE or VPD450 were detected in blood and secondary lymphoid tissues of normal or immunosuppressed recipients at least two months after their systemic infusion.


Subject(s)
Cell Culture Techniques/methods , Cell Proliferation , Cryopreservation/methods , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer/methods , Animals , Antigen-Presenting Cells/immunology , Cells, Cultured , Flow Cytometry , Fluoresceins/metabolism , Fluorescent Dyes/metabolism , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Immunophenotyping , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-7 Receptor alpha Subunit/immunology , Interleukin-7 Receptor alpha Subunit/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Macaca fascicularis , Succinimides/metabolism , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/transplantation , Time Factors , Transplantation, Autologous/methods
20.
Xenotransplantation ; 22(4): 239-48, 2015.
Article in English | MEDLINE | ID: mdl-25950141

ABSTRACT

Hepatocyte transplantation (Tx) is a potential therapy for certain diseases of the liver, including hepatic failure. However, there is a limited supply of human livers as a source of cells and, after isolation, human hepatocytes can be difficult to expand in culture, limiting the number available for Tx. Hepatocytes from other species, for example, the pig, have therefore emerged as a potential alternative source. We searched the literature through the end of 2014 to assess the current status of experimental research into hepatocyte xenoTx. The literature search identified 51 reports of in vivo cross-species Tx of hepatocytes in a variety of experimental models. Most studies investigated the Tx of human (n = 23) or pig (n = 19) hepatocytes. No studies explored hepatocytes from genetically engineered pigs. The spleen was the most common site of Tx (n = 23), followed by the liver (through the portal vein [n = 6]) and peritoneal cavity (n = 19). In 47 studies (92%), there was evidence of hepatocyte engraftment and function across a species barrier. The data provided by this literature search strengthen the hypothesis that xenoTx of hepatocytes is feasible and potentially successful as a clinical therapy for certain liver diseases, including hepatic failure. By excluding vascular structures, hepatocytes isolated from genetically engineered pig livers may address some of the immunological problems of xenoTx.


Subject(s)
Hepatocytes/transplantation , Transplantation, Heterologous/methods , Animals , Animals, Genetically Modified , Graft Survival , Heterografts , Humans , Models, Animal , Swine , Transplantation, Heterologous/trends
SELECTION OF CITATIONS
SEARCH DETAIL