Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Kidney Int ; 105(1): 84-98, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37839695

RESUMO

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.


Assuntos
Primatas , Linfócitos T Reguladores , Animais , Antígeno Ki-67/metabolismo , Rim , Aloenxertos , Células Mieloides , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo
2.
Am J Transplant ; 20(6): 1691-1702, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31883190

RESUMO

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.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos , Linfócitos T Reguladores , Animais , Fator Estimulador de Colônias de Granulócitos , Leucaférese , Macaca mulatta , Fatores de Transcrição
3.
Cell Immunol ; 351: 104087, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32197811

RESUMO

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.


Assuntos
Células Dendríticas/imunologia , Modelos Animais de Doenças , Transplante de Rim , Tolerância ao Transplante/imunologia , Animais , Macaca fascicularis , Macaca mulatta , Papio , Primatas , Transplante Homólogo
4.
Cell Immunol ; 329: 50-55, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29803290

RESUMO

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.


Assuntos
Células da Medula Óssea/citologia , Células Supressoras Mieloides/fisiologia , Linfócitos T Reguladores/fisiologia , Animais , Biomarcadores , Medula Óssea/patologia , Medula Óssea/fisiologia , Citometria de Fluxo , Macaca mulatta , Masculino , Monócitos/fisiologia , Células Mieloides/fisiologia , Células Supressoras Mieloides/metabolismo , Cultura Primária de Células/métodos , Subpopulações de Linfócitos T/fisiologia
5.
Xenotransplantation ; 25(3): e12415, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29913039

RESUMO

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.


Assuntos
Xenoenxertos/imunologia , Imunossupressores/farmacologia , Linfócitos T Reguladores/imunologia , Transplante Heterólogo , Animais , Humanos , Imunidade Inata/imunologia , Inflamação/imunologia , Transplante Heterólogo/métodos
7.
Curr Opin Organ Transplant ; 23(5): 538-545, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30036199

RESUMO

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.


Assuntos
Células Dendríticas/imunologia , Tolerância Imunológica/genética , Humanos
8.
Xenotransplantation ; 24(2)2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28294424

RESUMO

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.


Assuntos
Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/imunologia , Transplante de Coração , Xenoenxertos/imunologia , Inflamação/imunologia , Transplante Heterólogo , Animais , Animais Geneticamente Modificados , Sobrevivência de Enxerto/efeitos dos fármacos , Transplante de Coração/métodos , Humanos , Terapia de Imunossupressão/métodos , Imunossupressores/farmacologia , Interleucina-17/metabolismo , Papio , Suínos , Transplante Heterólogo/métodos
9.
Xenotransplantation ; 23(2): 83-105, 2016 03.
Artigo em Inglês | MEDLINE | ID: mdl-26813438

RESUMO

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.


Assuntos
Rejeição de Enxerto/patologia , Sobrevivência de Enxerto/imunologia , Primatas , Sus scrofa/imunologia , Transplante Heterólogo , Animais , Animais Geneticamente Modificados/imunologia , Rejeição de Enxerto/imunologia , Humanos , Transplante Heterólogo/métodos
10.
Xenotransplantation ; 23(2): 137-50, 2016 03.
Artigo em Inglês | MEDLINE | ID: mdl-26988899

RESUMO

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.


Assuntos
Galactosiltransferases/metabolismo , Proteína Cofatora de Membrana/metabolismo , Ácidos Neuramínicos/metabolismo , Transplante Heterólogo , Animais , Animais Geneticamente Modificados , Células Cultivadas , Células Endoteliais/imunologia , Galactosiltransferases/deficiência , Humanos , Leucócitos Mononucleares/imunologia , Ativação Linfocitária/imunologia , Proteína Cofatora de Membrana/genética , Suínos , Transplante Heterólogo/métodos
11.
Xenotransplantation ; 23(4): 293-9, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27374212

RESUMO

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.


Assuntos
Xenoenxertos/metabolismo , Hormônios Tireóideos/metabolismo , Transplante Heterólogo , Animais , Artérias/metabolismo , Artérias/transplante , Transplante de Coração , Transplante de Fígado , Papio , Suínos , Transplante Heterólogo/métodos , Tri-Iodotironina/sangue
12.
Cell Immunol ; 295(1): 19-28, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25732601

RESUMO

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.


Assuntos
Técnicas de Cultura de Células/métodos , Proliferação de Células , Criopreservação/métodos , Linfócitos T Reguladores/imunologia , Transferência Adotiva/métodos , Animais , Células Apresentadoras de Antígenos/imunologia , Células Cultivadas , Citometria de Fluxo , Fluoresceínas/metabolismo , Corantes Fluorescentes/metabolismo , Fatores de Transcrição Forkhead/imunologia , Fatores de Transcrição Forkhead/metabolismo , Imunofenotipagem , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-17/imunologia , Interleucina-17/metabolismo , Subunidade alfa de Receptor de Interleucina-2/imunologia , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Subunidade alfa de Receptor de Interleucina-7/imunologia , Subunidade alfa de Receptor de Interleucina-7/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Macaca fascicularis , Succinimidas/metabolismo , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/transplante , Fatores de Tempo , Transplante Autólogo/métodos
13.
Xenotransplantation ; 22(5): 399-405, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26292982

RESUMO

INTRODUCTION: In pig-to-baboon heart/artery patch transplantation models, adequate costimulation blockade prevents a T-cell response. After heart transplantation, coagulation dysfunction (thrombocytopenia, reduced fibrinogen, increased D-dimer) and inflammation (increased C-reactive protein [CRP]) develop. We evaluated whether coagulation dysfunction and/or inflammation can be detected following pig artery patch transplantation. METHODS: Baboons received heart (n = 8) or artery patch (n = 16) transplants from genetically engineered pigs and a costimulation blockade-based regimen. Heart grafts functioned for 15-130 days. Artery recipients were euthanized after 28-84 days. Platelet counts, fibrinogen, D-dimer, and CRP were measured. RESULTS: Thrombocytopenia and reduced fibrinogen developed only in recipients of hearts not expressing a coagulation-regulatory protein (n = 4), but not in other heart or patch recipients. However, in heart recipients (n = 8), there were sustained increases in D-dimer (<0.5 to 1.9 ug/ml [P < 0.01]) and CRP (0.26-2.2 mg/dl [P < 0.01]). In recipients of artery patches, there were also sustained increases in D-dimer (<0.5 to 1.4 ug/ml [P < 0.01]) and CRP (0.26 to 1.5 mg/dl [P < 0.001]). An IL-6R antagonist suppressed the increase in CRP, but not D-dimer. CONCLUSION: The pig artery patch model has proved valuable for determining immunosuppressive regimens that prevent sensitization to pig antigens. This model also provides information on the sustained systemic inflammation in xenograft recipients (SIXR). An IL-6R antagonist may help suppress this response.


Assuntos
Artérias/transplante , Rejeição de Enxerto/imunologia , Transplante de Coração , Inflamação/imunologia , Complicações Pós-Operatórias/imunologia , Transplante Heterólogo , Animais , Animais Geneticamente Modificados , Rejeição de Enxerto/prevenção & controle , Inflamação/etiologia , Inflamação/prevenção & controle , Papio , Complicações Pós-Operatórias/prevenção & controle , Suínos
14.
Xenotransplantation ; 22(1): 32-47, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25209710

RESUMO

BACKGROUND: Dysregulation of coagulation is considered a major barrier against successful pig organ xenotransplantation in non-human primates. Inflammation is known to promote activation of coagulation. The role of pro-inflammatory factors as well as the relationship between inflammation and activation of coagulation in xenograft recipients is poorly understood. METHODS: Baboons received kidney (n=3), heart (n=4), or artery patch (n=8) xenografts from α1,3-galactosyltransferase gene-knockout (GTKO) pigs or GTKO pigs additionally transgenic for human complement-regulatory protein CD46 (GTKO/CD46). Immunosuppression (IS) was based on either CTLA4Ig or anti-CD154 costimulation blockade. Three artery patch recipients did not receive IS. Pro-inflammatory cytokines, chemokines, and coagulation parameters were evaluated in the circulation after transplantation. In artery patch recipients, monocytes and dendritic cells (DC) were monitored in peripheral blood. Expression of tissue factor (TF) and CD40 on monocytes and DC were assessed by flow cytometry. C-reactive protein (C-RP) levels in the blood and C-RP deposition in xenografts as well as native organs were evaluated. Baboon and pig C-RP mRNA in heart and kidney xenografts were evaluated. RESULTS: In heart and kidney xenograft recipients, the levels of INFγ, TNF-α, IL-12, and IL-8 were not significantly higher after transplantation. However, MCP-1 and IL-6 levels were significantly higher after transplantation, particularly in kidney recipients. Elevated C-RP levels preceded activation of coagulation in heart and kidney recipients, where high levels of C-RP were maintained until the time of euthanasia in both heart and kidney recipients. In artery patch recipients, INFγ, TNF-α, IL-12, IL-8, and MCP-1 were elevated with no IS, while IL-6 was not. With IS, INFγ, TNF-α, IL-12, IL-8, and MCP-1 were reduced, but IL-6 was elevated. Elevated IL-6 levels were observed as early as 2 weeks in artery patch recipients. While IS was associated with reduced thrombin activation, fibrinogen and C-RP levels were increased when IS was given. There was a significant positive correlation between C-RP, IL-6, and fibrinogen levels. Additionally, absolute numbers of monocytes were significantly increased when IS was given, but not without IS. This was associated with increased CD40 and TF expression on CD14+ monocytes and lineage(neg) CD11c+ DC, with increased differentiation of the pro-inflammatory CD14+ CD11c+ monocyte population. At the time of euthanasia, C-RP deposition in kidney and heart xenografts, C-RP positive cells in artery patch xenograft and native lungs were detected. Finally, high levels of both pig and baboon C-RP mRNA were detected in heart and kidney xenografts. CONCLUSIONS: Inflammatory responses precede activation of coagulation after organ xenotransplantation. Early upregulation of C-RP and IL-6 levels may amplify activation of coagulation through upregulation of TF on innate immune cells. Prevention of systemic inflammation in xenograft recipients (SIXR) may be required to prevent dysregulation of coagulation and avoid excessive IS after xenotransplantation.


Assuntos
Coagulação Sanguínea , Xenoenxertos/patologia , Inflamação/etiologia , Complicações Pós-Operatórias/etiologia , Animais , Animais Geneticamente Modificados , Artérias/transplante , Fatores de Coagulação Sanguínea/análise , Proteína C-Reativa/biossíntese , Proteína C-Reativa/genética , Quimiocinas/sangue , Citocinas/sangue , Células Dendríticas/imunologia , Galactosiltransferases/deficiência , Galactosiltransferases/genética , Técnicas de Inativação de Genes , Transplante de Coração , Xenoenxertos/imunologia , Humanos , Imunossupressores , Inflamação/sangue , Inflamação/genética , Transplante de Rim , Proteína Cofatora de Membrana/genética , Proteína Cofatora de Membrana/imunologia , Monócitos/imunologia , Papio , Complicações Pós-Operatórias/sangue , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Sus scrofa , Suínos , Tromboplastina/biossíntese , Tromboplastina/genética
15.
Xenotransplantation ; 22(4): 310-6, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26174749

RESUMO

We describe the incidence of early graft failure (EGF, defined as loss of function from any cause within 3 days after transplant) in a large cohort of GalTKO pig organs transplanted into baboons in three centers, and the effect of additional expression of a human complement pathway-regulatory protein, CD46 or CD55 (GalTKO.hCPRP). Baboon recipients of life-supporting GalTKO kidney (n = 7) or heterotopic heart (n = 14) grafts received either no immunosuppression (n = 4), or one of several partial or full immunosuppressive regimens (n = 17). Fourteen additional baboons received a GalTKO.hCPRP kidney (n = 5) or heart (n = 9) and similar treatment regimens. Immunologic, pathologic, and coagulation parameters were measured at frequent intervals. EGF of GalTKO organs occurred in 9/21 baboons (43%). hCPRP expression reduced the GalTKO EGF incidence to 7% (1/14; P < 0.01 vs. GalTKO alone). At 30 mins, complement deposits were more intense in organs in which EGF developed (P < 0.005). The intensity of peri-transplant platelet activation (as ß-thromboglobulin release) correlated with EGF, as did the cumulative coagulation score (P < 0.01). We conclude that (i) the transgenic expression of a hCPRP on the vascular endothelium of a GalTKO pig reduces the incidence of EGF and reduces complement deposition, (ii) complement deposition and platelet activation correlate with early GalTKO organ failure, and (iii) the expression of a hCPRP reduces EGF but does not prevent systemic coagulation activation. Additional strategies will be required to control coagulation activation.


Assuntos
Antígenos CD55/imunologia , Galactosiltransferases/deficiência , Rejeição de Enxerto/prevenção & controle , Proteína Cofatora de Membrana/imunologia , Transplante Heterólogo/métodos , Animais , Animais Geneticamente Modificados , Antígenos CD55/genética , Ativação do Complemento , Dissacarídeos/imunologia , Galactosiltransferases/genética , Galactosiltransferases/imunologia , Técnicas de Inativação de Genes , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/imunologia , Transplante de Coração/efeitos adversos , Transplante de Coração/métodos , Humanos , Transplante de Rim/efeitos adversos , Transplante de Rim/métodos , Proteína Cofatora de Membrana/genética , Papio , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Suínos , Transplante Heterólogo/efeitos adversos
16.
Xenotransplantation ; 21(3): 201-20, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24571124

RESUMO

Xenotransplantation could resolve the increasing discrepancy between the availability of deceased human donor organs and the demand for transplantation. Most advances in this field have resulted from the introduction of genetically engineered pigs, e.g., α1,3-galactosyltransferase gene-knockout (GTKO) pigs transgenic for one or more human complement-regulatory proteins (e.g., CD55, CD46, CD59). Failure of these grafts has not been associated with the classical features of acute humoral xenograft rejection, but with the development of thrombotic microangiopathy in the graft and/or consumptive coagulopathy in the recipient. Although the precise mechanisms of coagulation dysregulation remain unclear, molecular incompatibilities between primate coagulation factors and pig natural anticoagulants exacerbate the thrombotic state within the xenograft vasculature. Platelets play a crucial role in thrombosis and contribute to the coagulation disorder in xenotransplantation. They are therefore important targets if this barrier is to be overcome. Further genetic manipulation of the organ-source pigs, such as pigs that express one or more coagulation-regulatory genes (e.g., thrombomodulin, endothelial protein C receptor, tissue factor pathway inhibitor, CD39), is anticipated to inhibit platelet activation and the generation of thrombus. In addition, adjunctive pharmacologic anti-platelet therapy may be required. The genetic manipulations that are currently being tested are reviewed, as are the potential pharmacologic agents that may prove beneficial.


Assuntos
Transtornos da Coagulação Sanguínea/etiologia , Plaquetas/fisiologia , Complicações Pós-Operatórias/etiologia , Transplante Heterólogo , Animais , Anticoagulantes/uso terapêutico , Transtornos da Coagulação Sanguínea/sangue , Transtornos da Coagulação Sanguínea/terapia , Engenharia Genética , Humanos , Ativação Plaquetária , Inibidores da Agregação Plaquetária/uso terapêutico , Complicações Pós-Operatórias/sangue , Complicações Pós-Operatórias/terapia , Suínos
17.
Xenotransplantation ; 21(1): 72-83, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24188473

RESUMO

BACKGROUND: Coagulation disorders remain barriers to successful pig-to-primate organ xenotransplantation. In vitro, we investigated the impact of pig genetic modifications on human platelet aggregation in response to pig aortic endothelial cells (pAEC). METHODS: In comparison to human (h)AEC and wild-type (WT) pAEC, the expression of human complement- (CD46, CD55) or coagulation (thrombomodulin [TBM], endothelial protein C receptor [EPCR]) -regulatory proteins on pAEC from WT or α1,3-galactosyltransferase gene-knockout (GTKO) pigs was studied by flow cytometry. Using platelet-aggregometry, human whole blood platelet aggregation was evaluated after co-incubation with various AEC. Further, the inhibitory effect on aggregation of heparin, low molecular weight heparin, and hirudin was assessed. RESULTS: Heparin, low molecular weight heparin and hirudin almost completely prevented platelet aggregation induced by WT pAEC. The level of expression of human CD46, CD55, TBM and EPCR on pAEC was comparable to that on hAEC. Platelet aggregation induced by all genetically modified pAEC was significantly less (P < 0.05) than that by WT pAEC (which was 54%). GTKO/CD46/TBM pAEC induced the least platelet aggregation (27%)-a reduction of almost 50%-but this remained significantly greater (P < 0.01) than aggregation induced by hAEC (4%). There was significant positive correlation between reduction of aggregation and TBM or EPCR expression on pAEC (r = 0.89 and r = 0.86, respectively; P < 0.05). Platelet aggregation induced by GTKO/CD46/TBM pAEC in the presence of hirudin (1 IU/ml) was comparable to platelet aggregation induced by hAEC. CONCLUSIONS: Genetic modification of pAEC is associated with significant reduction of human platelet aggregation in vitro. With concomitant thrombin inhibition, platelet aggregation was comparable to that stimulated by hAEC.


Assuntos
Células Endoteliais/efeitos dos fármacos , Galactosiltransferases/genética , Agregação Plaquetária/efeitos dos fármacos , Trombina/farmacologia , Transplante Heterólogo , Animais , Animais Geneticamente Modificados , Coagulação Sanguínea/genética , Plaquetas/imunologia , Técnicas de Inativação de Genes , Humanos , Sus scrofa , Suínos
18.
Xenotransplantation ; 21(1): 16-24, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25405250

RESUMO

Development of coagulation disorders remains a major challenge in pig-to-primate organ xenotransplantation. Our previous studies demonstrated that porcine aortic endothelial cells (pAEC) activate human platelets to express tissue factor (TF). In this study, we investigated the molecular interaction between human platelets and pAEC to identify possible targets for further genetic modification and/or systemic therapy. Human platelets were incubated with pAEC from wild-type (WT), α1,3-galactosyltransferase gene-knockout (GTKO), and GTKO pigs expressing human CD46, after which the platelets were analyzed for TF expression, TF mRNA level and TF function. pAEC were analyzed for von Willebrand factor (vWF) expression and mRNA level as well. Neutralizing antibodies for P-selectin and P-selectin glycoprotein ligand-1 (PSGL-1) were used to block the molecular interaction between platelets and pAEC. GTKO and GTKO/CD46 pAEC-activated human platelets to induce human TF activity equivalently to WT pAEC. Simultaneously, after incubation with pAEC, platelets co-expressed TF and P-selectin. TF expression was blocked when pAEC and platelets were pre-incubated with anti-human P-selectin or anti-human PSGL-1 antibodies, but not by anti-porcine P-selectin antibody. Activated pAEC up-regulated TF on platelets through the interaction of porcine vWF with the human GPIb receptor. Up-regulation of TF on human platelets by GTKO and GTKO/CD46 pAEC was comparable to that by WT pAEC, which is associated with concomitant expression of P-selectin and PSGL-1, forming an auto-augmented loop of pAEC and platelet activation. Blocking of P-selectin and PSGL-1 interaction may be required to prevent up-regulation of recipient TF in vivo after organ xenotransplantation.


Assuntos
Plaquetas/imunologia , Células Endoteliais/efeitos dos fármacos , Glicoproteínas de Membrana/metabolismo , Selectina-P/metabolismo , Transplante Heterólogo , Animais , Animais Geneticamente Modificados , Células Endoteliais/citologia , Humanos , Ativação Plaquetária/fisiologia , Suínos , Tromboplastina/metabolismo
19.
Xenotransplantation ; 21(5): 397-419, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25176336

RESUMO

BACKGROUND: The pig-to-non-human primate model is the standard choice for in vivo studies of organ and cell xenotransplantation. In 1998, Lambrigts and his colleagues surveyed the entire world literature and reported all experimental studies in this model. With the increasing number of genetically engineered pigs that have become available during the past few years, this model is being utilized ever more frequently. METHODS: We have now reviewed the literature again and have compiled the data we have been able to find for the period January 1, 1998 to December 31, 2013, a period of 16 yr. RESULTS: The data are presented for transplants of the heart (heterotopic and orthotopic), kidney, liver, lung, islets, neuronal cells, hepatocytes, corneas, artery patches, and skin. Heart, kidney, and, particularly, islet xenograft survival have increased significantly since 1998. DISCUSSION: The reasons for this are briefly discussed. A comment on the limitations of the model has been made, particularly with regard to those that will affect progression of xenotransplantation toward the clinic.


Assuntos
Transplante de Células/métodos , Modelos Animais , Transplante de Órgãos/métodos , Primatas , Suínos , Transplante Heterólogo/métodos , Animais
20.
Cell Immunol ; 285(1-2): 23-30, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24044963

RESUMO

Mesenchymal stromal cells (MSC) obtained from α1,3-galactosyltransferase gene knock-out pigs transgenic for the human complement-regulatory protein CD46 (GTKO/CD46 pMSC) suppress in vitro human anti-pig cellular responses as efficiently as allogeneic human MSC. We investigated the immunoregulatory effects of GTKO/CD46 pMSC on human CD4(+) and CD8(+) T cell proliferation in response to pig aortic endothelial cells (pAEC). pMSC efficiently suppressed T cell proliferation, which was associated with downregulation of granzyme B expression. No induction of CD4(+)CD25(+)Foxp3(hi) regulatory T cells or T cell apoptosis was documented. In correlation with T cell proliferation, CD25 expression was upregulated on T cells in response to pAEC but not to pMSC. In contrast, CD69 expression was upregulated on T cells in response to both pMSC and pAEC, which was associated with a significant increase in the phosphorylation of STAT5. GTKO/CD46 pMSC possibly regulate human T cell responses through modulation of CD69 expression and STAT5 signaling.


Assuntos
Antígenos CD/metabolismo , Antígenos de Diferenciação de Linfócitos T/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Lectinas Tipo C/metabolismo , Proteína Cofatora de Membrana/metabolismo , Células-Tronco Mesenquimais/imunologia , Animais , Animais Geneticamente Modificados , Antígenos CD/biossíntese , Antígenos de Diferenciação de Linfócitos T/biossíntese , Apoptose/imunologia , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Células Endoteliais , Fatores de Transcrição Forkhead/metabolismo , Galactosiltransferases/genética , Granzimas/biossíntese , Humanos , Integrina beta3/biossíntese , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Lectinas Tipo C/biossíntese , Ativação Linfocitária/imunologia , Proteína Cofatora de Membrana/genética , Proteína Cofatora de Membrana/imunologia , Fosforilação , Fator de Transcrição STAT5/metabolismo , Suínos , Transplante Heterólogo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA