Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cell Transplant ; 17(6): 713-20, 2008.
Article in English | MEDLINE | ID: mdl-18819259

ABSTRACT

The mechanisms mediating T-cell depletion plus 15-deoxyspergualin (DSG)-induced prolonged allograft survival or tolerance are uncertain. The purpose of this study is to evaluate the role of IL-4 and IL-10 in prolonged allograft survival induced by T-cell depletion plus DSG. MHC mismatched skin allograft transplantation was performed, using wild-type and three separate knockout (i.e., IL-4-/-, Stat6-/-, or IL-1-/ -) mice as recipients. Induction therapy consisted of T-cell depletion and or brief course of DSG. The data demonstrate that monotherapy with T-cell-depleting mAbs or DSG prolonged skin allograft survival, compared to controls, in wild-type Balb/c recipients [median survival time (MST) = 25 and 21 vs. 10 days, p < 0.007]. T-cell depletion plus DSG further augmented skin allograft survival in wild-type animals relative to monotherapy (MST = 35 days vs. 25 and 21 days, p < 0.006 vs. mAbs or DSG only), and was equally effective in IL-4-/- and Stat6-/- recipients. In contrast, combined therapy was no better than monotherapy in IL-10-/- animals (p > 0.05). Furthermore, skin allograft survival after combined therapy was shorter in IL-10-/- versus wild-type recipients (MST 20 and 41 days, respectively, p < 0.001). IL-4-mediated signaling through Stat6 is dispensable for prolonged allograft survival induced by T-cell depletion plus DSG. In contrast, IL-10 appears to be important for prolonged allograft survival induced by combined therapy in this model.


Subject(s)
Graft Survival , Interleukin-10/immunology , Interleukin-4/immunology , Lymphocyte Depletion , Skin Transplantation/immunology , T-Lymphocytes/immunology , Animals , Graft Enhancement, Immunologic , Graft Survival/drug effects , Graft Survival/immunology , Guanidines/pharmacology , Immunosuppressive Agents/pharmacology , Interleukin-10/genetics , Interleukin-10/pharmacology , Interleukin-4/genetics , Interleukin-4/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/immunology , T-Lymphocytes/drug effects
2.
Immunol Res ; 40(1): 95, 2008.
Article in English | MEDLINE | ID: mdl-18340653
4.
J Med Primatol ; 37(1): 1-11, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18199066

ABSTRACT

BACKGROUND: Natural killer T cells (NKT) possess dual functions of innate and adaptive immune systems, controlling viral infections and regulating autoimmune diseases. Non-human primates (NHP) are penultimate models for advancing therapeutic immunoregulatory strategies for translational application in humans, though, little is known about NHP NKT cells. Here we characterized rhesus macaque NKT cells ex vivo. METHODS: The frequency, phenotype and intracellular cytokine production of V alpha 24+ 6B11+ invariant NKT (iNKT) cells were analyzed by multi-color flow cytometry. V alpha 24J alpha Q mRNA expression was analyzed by real-time RT-PCR. RESULTS: The frequencies of peripheral blood (PB) and spleen V alpha 24+ 6B11+ iNKT cells were not significantly different. The iNKT cell subset in spleen was significantly increased for CD4+ CD8+ and CD3+ CD56+ co-expression as well as intracellular interleukin-4 production, which was rarely observed in circulating PB. CONCLUSION: Spleen iNKT cells in rhesus macaques are Th2 biased and display phenotypically and functionally distinct profiles from their PB counterpart.


Subject(s)
Killer Cells, Natural/cytology , Macaca mulatta/immunology , Spleen/immunology , Animals , Cytokines/metabolism , DNA Primers/genetics , Flow Cytometry , Immunophenotyping , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spleen/cytology , Statistics, Nonparametric
5.
Protein Eng Des Sel ; 20(9): 425-32, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17693455

ABSTRACT

T-cell depleting anti-CD3 immunotoxins have utility in non-human primate models of transplantation tolerance and autoimmune disease therapy. We recently reported that an affinity matured single-chain (scFv) anti-monkey CD3 antibody, C207, had increased binding to T-cells and increased bioactivity in a diphtheria toxin (DT)-based biscFv immunotoxin compared with the parental antibody, FN18. However, FN18 scFvs and their mutant derivatives such as C207 did not exhibit robust bivalent character in the biscFv format. We now report that C207 in a diabody format exhibits a 7-fold increase in binding to T-cells over scFv (C207) indicating considerable divalent character for the diabody. This construct was formed by reducing the V(L)/V(H) linker to five residues and was secreted from Pichia pastoris as the non-covalent dimer. An immunotoxin based on this diabody format was secreted as a non-covalent dimer but was devoid of bioactivity and failed to bind T-cells, suggesting steric hindrance from the two large closely positioned truncated DT moieties. We constructed a single-chain diabody immunotoxin by fusing to the truncated DT C-terminus L1-VL-L1-VH-L2-VL-L1-VH where L1 is a five-residue linker and L2 is the longer (G4S)3 linker permitting interactions between the distal and proximal VL/VH domains. This 'fold-back' immunotoxin was secreted predominantly as the monomer and exhibited a 5- to 7-fold increase in bioactivity over DT390biscFv(C207) and depleted monkey T-cells in vivo.


Subject(s)
CD3 Complex/chemistry , Diphtheria Toxin/chemistry , Immunotoxins/chemistry , Protein Engineering/methods , Animals , Dimerization , Haplorhini , Immunoglobulin Fragments/chemistry , Pichia/metabolism , Plasmids/metabolism , Recombinant Fusion Proteins/chemistry , T-Lymphocytes/metabolism , Time Factors
6.
Hum Immunol ; 68(6): 478-90, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17509447

ABSTRACT

CD4(+)CD25(+) regulatory T cells (Tregs) play an important role in allograft and self-tolerance and thus have potential therapeutic application in transplantation, autoimmunity, and allergy. Although nonhuman primate (NHP) provide the most accepted preclinical models for translational studies in allograft tolerance and infectious diseases, CD4(+)CD25(+) Tregs have been rarely studied in NHP. The low frequencies of Tregs in peripheral blood will likely necessitate ex vivo expansion to enable Tregs adaptive immune therapy in NHP and humans. Tregs were isolated by magnetic and flow sorting and then stimulated weekly with antirhesus CD3 clone FN18 and antihuman CD28-coated Dynal beads plus 100 U/ml rhIL-2. Under these conditions, the Tregs were expanded 300- to 2000-fold in 4 weeks. Expanded CD4(+)CD25(+) Tregs expressed high to moderate levels of FOXP3 as well as CD95, CD62L, CD69, and CCR7 surface antigens. Expanded rhesus Tregs were anergic and suppressed the proliferation of autologous peripheral blood mononuclear cells (PBMC) in a dose-dependent fashion, and the suppression was partially reversed by anti-transforming growth factor (TGF)-beta1 neutralizing antibody (Ab). These results demonstrate that rhesus macaque suppressive regulatory CD4(+)CD25(+)FOXP3(+) Tregs can be efficiently expanded in vitro under rhesus-specific stimulation, which enables preclinical testing of Treg therapy in the NHP model.


Subject(s)
CD4 Antigens/immunology , Interleukin-2 Receptor alpha Subunit/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Monoclonal/chemistry , CD28 Antigens/immunology , Diphtheria Toxin/chemistry , Humans , Leukocytes, Mononuclear , Macaca mulatta , Microspheres , Models, Animal , Recombinant Fusion Proteins/chemistry , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism
9.
J Immunol ; 175(12): 8060-8, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16339543

ABSTRACT

Regulatory T cells (Tregs) are implicated in immune tolerance and are variably dependent on IL-10 for in vivo function. Brief peritransplant treatment of multiple nonhuman primates (NHP) with anti-CD3 immunotoxin and deoxyspergualin has induced stable (5-10 years) rejection-free tolerance to MHC-mismatched allografts, which associated with sustained elevations in serum IL-10. In this study, we demonstrate that resting and activated PBMC from long-term tolerant NHP recipients are biased to secrete high levels of IL-10, compared with normal NHP PBMC. Although IL-10-producing CD4+ Tregs (type 1 regulatory cells (TR1)/IL-10 Tregs) were undetectable (<0.5%) in normal rhesus monkeys, 7.5 +/- 1.7% of circulating CD4+ T cells of tolerant rhesus recipients expressed IL-10. In addition to this >15-fold increase in Tr1/IL-10 Tregs, the tolerant monkeys exhibited a nearly 3-fold increase in CD4+CD25+ Tregs, 8.1 +/- 3.0% of CD4 T cells vs 2.8 +/- 1.4% in normal cohorts (p < 0.02). The frequency of CD4+CD25+IL-10+ cells was elevated 5-fold in tolerant vs normal NHP (1.8 +/- 0.9% vs 0.4 +/- 0.2%). Rhesus CD4+CD25+ Tregs exhibited a memory phenotype, and expressed high levels of Foxp3 and CTLA-4 compared with CD4+CD25- T cells. Also, NHP CD4+CD25+ Tregs proliferated poorly after activation and suppressed proliferation of CD4+CD25- effector T cells, exhibiting regulatory properties similar to rodent and human CD4+CD25+ Tregs. Of note, depletion of CD4+CD25+ Tregs restored indirect pathway antidonor responses in tolerant NHP. Our study demonstrates an expanded presence of Treg populations in tolerant NHP recipients, suggesting that these adaptations may be involved in maintenance of stable tolerance.


Subject(s)
Guanidines/pharmacology , Immune Tolerance , Immunotoxins/pharmacology , T-Lymphocytes, Regulatory/immunology , Transplantation Immunology , Animals , Antibodies, Monoclonal/pharmacology , CD3 Complex/immunology , CD4 Lymphocyte Count , Immunologic Memory , Immunosuppressive Agents/pharmacology , Interleukin-10/biosynthesis , Macaca mulatta , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism
10.
Cell Immunol ; 223(2): 103-12, 2003 Jun.
Article in English | MEDLINE | ID: mdl-14527508

ABSTRACT

Peritransplant treatment with anti-CD3 immunotoxin plus deoxyspergualin induces tolerance to kidney allografts in most rhesus macaque recipients. Tolerant recipients maintain normal function for years without evidence of chronic rejection. Indirect alloantigen presentation is implicated in chronic rejection. Accordingly, we determined if anti-CD3 immunotoxin plus deoxyspergualin induced rejection-free tolerance associates with suppression of anti-donor indirect pathway responses. Tolerant recipients exhibited an early decrease in direct anti-donor responses with recovery to baseline levels by 3 years posttransplantation. In contrast, tolerant monkeys were unresponsive to donor antigens presented by the indirect pathway. Recipients that rejected their allografts retained vigorous direct and indirect anti-donor responses. Therefore, following temporary donor-specific hyporesponsiveness, direct responses recover in tolerant recipients >1.5 years after transplantation. However, tolerant recipients tested at 1.9-4 years posttransplant are specifically unresponsive to donor antigens presented by the indirect pathway. Thus, the rejection-free state of tolerant recipients may depend on mechanisms regulating indirect pathway responsiveness.


Subject(s)
CD3 Complex/immunology , Guanidines/pharmacology , Immune Tolerance/drug effects , Immunosuppressive Agents/pharmacology , Kidney Transplantation/immunology , Transplantation Conditioning , Animals , Graft Survival/immunology , Histocompatibility , Immunotoxins/pharmacology , Leukocytes, Mononuclear/immunology , Lymphocyte Culture Test, Mixed , Macaca mulatta , Male , Transplantation, Homologous/immunology
11.
Transpl Immunol ; 11(3-4): 335-44, 2003.
Article in English | MEDLINE | ID: mdl-12967786

ABSTRACT

Brief treatment of rhesus macaques with immunotoxin plus 15-deoxyspergualin has yielded exceptional numbers (54%) of stable tolerant kidney allograft recipients, surviving over 6 years without rejection or immunosuppression. An early increase in IL-10 and reduction in IFNgamma distinguished recipients that subsequently became tolerant. Furthermore, analysis suggested that this immune switch was programmed within hours of transplantation. Administering deoxyspergualin within 5 h of surgery gave a higher incidence of tolerance (76%) compared to administration >5 h before or after surgery (11%, P<0.01). Deoxyspergualin inhibits nuclear translocation of activated NF-kappaB through heat shock proteins. Lymph node biopsies from tolerant recipients showed significant reductions in cytoplasmic expression of Hsp70 and RelB and almost complete inhibition of nuclear translocation of both. The early timing effect of deoxyspergualin suggests a crucial limitation to induction of stable tolerance is activation of Hsp-dependent innate responses to damage by ischemia-reperfusion. This was supported by studies in murine kidney reperfusion injury, where deoxyspergualin given 5 h before reperfusion protected renal function and reduced levels of IL-6 and IL-12. The narrow timing window for initiating deoxyspergualin treatment suggests the innate immune system is poised to defeat allograft tolerance induction, so effective blockade of NF-kappaB-mediated innate immunity must be in place early, to enable development of a tolerogenic environment.


Subject(s)
Guanidines/therapeutic use , Immunotoxins/therapeutic use , Macaca mulatta/immunology , Transplantation Tolerance , Animals , Cytokines/antagonists & inhibitors , Humans , Immune Tolerance , Immunity, Innate/drug effects , Kidney Transplantation/immunology , Male , Mice , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control
12.
J Immunol ; 171(6): 2904-11, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12960313

ABSTRACT

Natural killer T cells are immunoregulatory cells, which have important roles in tolerance and autoimmunity, as demonstrated primarily in mice and humans. In this study, we define the phenotype and function of Valpha24(+) T cells derived from the spleens of rhesus macaques, a species increasingly used in models of immune tolerance. Valpha24(+) cells were isolated and expanded with monocyte-derived immature dendritic cells in the presence of alpha-galactosylceramide, IL-2, and IL-15. Rhesus NKT cells were stained with mAbs against both Valpha24 and the invariant complementarity-determining region 3 epitope of the human Valpha24/JalphaQ TCR. The cells were CD4, CD8 double negative and expressed CD56. Rhesus NKT cells also exhibited moderate to high expression of CD95, CD45RO, CD11a, and beta(7) integrin, but did not express CD45 RA, CD62L, CCR7, CD28, and other activation, costimulatory molecules (CD69 and CD40L). By intracellular staining, >90% of unstimulated rhesus NKT cells expressed IL-10, but not IFN-gamma. However, the latter was strongly expressed after stimulation. Rhesus NKT secreted large amounts of TGF-beta, IL-13, and IL-6, and modest levels of IFN-gamma, whereas IL-10 secretion was negligible and no detectable IL-4 was observed either intracellularly or in culture supernatants. Functionally, the NKT cells and their supernatants suppressed T cell proliferation in allogeneic MLR. We conclude that long-term cultured rhesus macaque spleen-derived Valpha24(+) T cells are semi-invariant double-negative cells with effector memory phenotype. These cells are semianergic, polarized to a uniquely Th3 > T regulatory-1 regulatory cell phenotype, and have regulatory/suppressive function in vitro.


Subject(s)
Immunophenotyping , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Spleen/immunology , Spleen/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Biomarkers , CD3 Complex/biosynthesis , CD4 Antigens/biosynthesis , CD8 Antigens/biosynthesis , Cells, Cultured , Clonal Anergy , Complementarity Determining Regions/biosynthesis , Cytokines/metabolism , Immunologic Memory , Lymphocyte Activation/immunology , Lymphocyte Culture Test, Mixed , Macaca mulatta , Male , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Spleen/cytology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/immunology , Time Factors
13.
Transplantation ; 76(1): 237-43, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12865816

ABSTRACT

BACKGROUND: Donor bone marrow infusion has long been used to enhance graft survival or induce tolerance in T cell depleted solid organ allograft recipients. However, the mechanisms through which bone marrow cells affect tolerance remain obscure. We studied the affect of allogeneic bone marrow cells on the activation of allospecific T cells in vitro. METHODS: Carboxyfluorescein-diacetate succinimidyl ester-labeled CBA/Ca strain CD8+ splenocytes, bearing T-cell receptor alpha and beta transgenes from the BM3.3 T-cell clone specific for the major histocompatibility complex class I antigen Kb, were placed in culture with irradiated C57BL/6J stimulator cells in the presence of increasing numbers of C57BL/6J or Balb/cJ bone marrow cells for 1 to 3 days. Responder cells were individually analyzed for proliferative history, expression of activation-associated antigens, and intracellular cytokine production. RESULTS: Allogeneic bone marrow cells exert a dose-dependent inhibitory effect on proliferation of allospecific CD8+ T cells in mixed lymphocyte culture. However, the inhibited T-cell subpopulations show physiologic changes associate with the early stages of T-cell activation, including expression of CD69 and early decrease of surface T-cell expression. Unlike cells not co-cultured with bone marrow, these cells fail to reexpress the T-cell receptor (TCR) by 72 hr of culture. The observed inhibitory effect is also associated with a decrease in the proportion of CD8+ cells expressing interleukin-2 and interferon-gamma. CONCLUSIONS: Collectively, these results suggest that peripheral allospecific T cells undergo the initial stages of activation on exposure to antigen in the presence of bone marrow cells, but the cell cycle is arrested and TCR reexpression is inhibited. We speculate that bone marrow cells effect this inhibition through a receptor-ligand interaction that modulates the transmembrane signal pathway for the TCR.


Subject(s)
Bone Marrow Transplantation/immunology , Immune Tolerance , Lymphocyte Activation , T-Lymphocytes/immunology , Animals , Bone Marrow Cells/immunology , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Transplantation, Homologous/immunology
14.
Curr Opin Investig Drugs ; 4(5): 530-5, 2003 May.
Article in English | MEDLINE | ID: mdl-12833645

ABSTRACT

The immunosuppressive regimens currently in use have been responsible for major improvements in transplanted organ acceptance, but long-term survival can be compromised by drug toxicity and/or chronic immune deficiency. The ultimate goal for transplantation is tolerance, defined as durable, donor-specific allograft acceptance in the absence of long-term immunosuppression. This review describes current experimental strategies for tolerance induction in primate models that are poised for clinical application.


Subject(s)
Graft Rejection/drug therapy , Graft Rejection/immunology , Transplantation Tolerance/drug effects , Transplantation Tolerance/immunology , Animals , Humans , Immunosuppressive Agents/therapeutic use
15.
Am J Transplant ; 3(2): 128-38, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12603208

ABSTRACT

Pancreatic islet transplantation (PIT) is an attractive alternative for type 1 diabetic patients. PIT is not yet an effective clinical reality due in part to early loss of functional islet mass. In addition, current immunosuppressive drugs have toxic effects on islets and increase the risk of morbidity and mortality. Precise and durable alpha- and beta-cell function is essential for the success of PIT. Therefore, it is important to establish whether PIT can produce adequate long-term metabolic control, especially in the absence of chronic immunosuppressive therapy (CIT). In the present study, the stability of functional alpha- and beta-cell mass and metabolic function was assessed in streptozotocin (STZ)-induced diabetic primates following PIT in the absence of CIT. Diabetes was induced in rhesus macaques with STZ, 140 mg/kg. Hyperglycemia was reversed rapidly by PIT coupled with a 14-day tolerance induction protocol based on F(Ab)2-IT and DSG (n = 7). Two diabetic animals received the tolerance induction protocol without PIT. Acute rejection was presented in three animals at 70, 353 and 353 days post transplant in the tolerance induction protocol, whereas the controls [F(Ab)2-IT or DSG alone] showed early 10-day function but all lost islet function by days 15-70. One recipient [F(Ab)2-IT or DSG] died euglycemic after a surgical procedure on day 187. At 2 years, three animals studied had a normal FIM evaluated by oral glucose tolerance test, mixed meal test, acute insulin response to glucose, glucose disposal rate, and hyperinsulinemic hypoglycemic clamp. PIT in STZ-induced diabetic primates resulted in restoration of normal alpha- and beta-cell function. Operational tolerance induction was achieved with only peritransplant administration of F(Ab)2-IT and DSG sparing the animals from chronic exposure of diabetogenic immunosuppressive drugs. These results offer an exciting new potential for treatment of type 1 diabetes mellitus.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Experimental/surgery , Islets of Langerhans Transplantation/physiology , Transplantation Tolerance/physiology , Animals , Biomarkers/blood , Blood Glucose/metabolism , Glucose Tolerance Test , Graft Rejection/blood , Guanidines/therapeutic use , Immunosuppressive Agents/therapeutic use , Macaca mulatta , Male , Reference Values , Treatment Outcome
16.
Immunol Res ; 26(1-3): 143-52, 2002.
Article in English | MEDLINE | ID: mdl-12403353

ABSTRACT

Although contemporary immunosuppressive regimens are responsible for major improvements in allograft acceptance, there are indications that long-term survival may be compromised through drug toxicity and/or chronic immune deficiency. The ultimate goal for transplantation is tolerance, defined as durable, donor-specific allograft acceptance in the absence of long-term immunosuppression. This article reviews the nonhuman primate STEALTH model of tolerance recently developed by the transplant immunobiology group at University of Alabama at Birmingham. The STEALTH model was designed for future application to human transplantation and comprises a concise peritransplant treatment strategy of only 2 wk. Tolerance is induced by depletion of T cells, with concomitant inhibition of nuclear factor-kappaB/RelB-dependent proinflammatory signaling. This treatment has resulted in an unprecedented frequency of kidney allograft survival (62.5% at 3 yr), with some primate recipients remaining in good health more than 6 yr posttransplant, in the complete absence of chronic pharmacologic immunosuppression.


Subject(s)
Immune Tolerance , Transplantation Immunology , Animals , Antigen-Presenting Cells/immunology , B-Lymphocytes/immunology , Cytokines/biosynthesis , Graft Rejection , Graft Survival , Guanidines/pharmacology , Humans , Immunosuppressive Agents/pharmacology , Lymphocyte Depletion , Macaca , Models, Animal , Models, Immunological , T-Lymphocytes/immunology , Transplantation, Homologous
17.
Immunol Res ; 26(1-3): 289-96, 2002.
Article in English | MEDLINE | ID: mdl-12403366

ABSTRACT

Isolated islet transplantation is poised for clinical application to treat insulin-dependent diabetes. Unlike exogenous insulin therapy, islet transplantation has promise for preventing and/or reversing the dismal secondary complications of diabetes. Islet transplants are arguably the most unique type of allografts, and we discuss their properties, limitations, and potential in this overview. The induction of immunologic tolerance to allow islet grafts to endure and prevail, without the hardship of chronic immunosuppressive therapy, is a major goal in this field. In this context, we discuss our successful results in preclinical models of primate allogeneic and xenogeneic islet graft tolerance.


Subject(s)
Islets of Langerhans Transplantation/trends , Animals , Apoptosis , Autoimmunity , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/surgery , Humans , Immune Tolerance , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Islets of Langerhans Transplantation/immunology , Primates , Transplantation, Heterologous , Transplantation, Homologous
18.
Immunol Res ; 26(1-3): 297-302, 2002.
Article in English | MEDLINE | ID: mdl-12403367

ABSTRACT

T cell depletion plus donor bone marrow cell (BMC) infusion induces long-term kidney allograft survival in a limited number of rhesus macaque recipients. Therefore, there is a need to enhance the tolerogenic activity of donor BMCs. The tolerogenic effect of donor BMCs is ascribed to a veto activity, mediated by a CD8+ subset that upregulates immunoregulatory effector molecules, transforming growth factor-beta1 (TGF-beta), and FasL, after interaction with donor-reactive cytotoxic T lymphocyte precursors (CTLp), leading to clonal inactivation/deletion of donor-reactive CTLp. Of note, the receptors for TGF-beta1- and FasL-induced signal transduction are upregulated in activated T cells. Since mature dendritic cells (DCs) are exceptionally efficient activators of T cells, we postulated that mature DCs modified to overexpress TGF-beta1 and FasL might exert potent veto (i.e., inactivating/deleting) activity independent of CD8 expression. A fusion protein comprising antihuman CD40 single-chain antibody and soluble coxsackie-adenovirus receptor enabled high-efficiency transduction of rhesus monocyte-derived DCs (Rh MDDCs) by recombinant adenovirus (Ad). Mature Rh MDDCs transduced with Ad encoding active TGF-beta1 retained a mature phenotype yet exhibited potent alloantigen-specific cellular immunosuppression. Such modified MDDCs have the potential to promote tolerance induction to allografts in vivo.


Subject(s)
Dendritic Cells/immunology , Immune Tolerance/genetics , Animals , Antigens/metabolism , Fas Ligand Protein , Genetic Engineering , Immunity, Cellular , Kidney Transplantation/immunology , Macaca mulatta , Membrane Glycoproteins/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1 , Transplantation, Homologous
19.
Transplantation ; 74(5): 629-37, 2002 Sep 15.
Article in English | MEDLINE | ID: mdl-12352878

ABSTRACT

BACKGROUND: The tolerogenic activity of allogeneic bone marrow cells (BMCs) associates with functional inactivation of alloreactive T cells and has been attributed to a veto effect. Studies in mice and rhesus monkeys indicated that the CD8alpha molecule expressed on a subpopulation of allogeneic BMCs is necessary to induce signal transduction within the BMCs to increase veto effector molecules such as transforming growth factor (TGF)-beta1. In vitro activation of alloreactive cytotoxic T-lymphocyte precursor enhances their susceptibility to veto-mediated functional inactivation by specific alloantigen-bearing BMCs. Accordingly, we examined a hypothesis that mature rhesus monkey (Rh) monocyte-derived dendritic cells (MDDCs) modified by gene transfer to over-express active TGF-beta1 might mediate veto activity without the need to express CD8alpha. METHODS: Rh MDDCs were modified by recombinant adenovirus (Ad) transduction and characterized by phenotype and functional studies. RESULTS: Rh MDDC transduction with Ad vectors using conventional methods was remarkably inefficient. However, a single-chain anti-CD40/soluble Coxsackie and adenovirus receptor-fusion protein (G28/sCAR) permitted high-efficiency transduction of Rh MDDCs by retargeting Ad to Rh MDDC CD40. Mature Rh MDDCs that were transduced to overexpress active TGF-beta1 (AdTGF-beta1 Rh MDDC) significantly suppressed alloimmune responses in [ H]thymidine uptake mixed leukocyte reaction assays. We showed by the carboxyfluorescein succinimidyl ester dilution method that allogeneic mature AdTGF-beta1 Rh MDDCs inhibited proliferation of CD4 and CD8 responder T cells. Notably, AdTGF-beta1 Rh MDDC abrogated alloimmune responses induced by control AdGFP Rh MDDC in an antigen-specific manner. CONCLUSIONS: These results suggest that nonhuman primate mature MDDCs can be genetically engineered to function as alloantigen-specific cellular immunosuppressants, an approach that has potential to facilitate induction of allograft tolerance in vivo.


Subject(s)
Dendritic Cells/immunology , Monocytes/immunology , Transforming Growth Factor beta/genetics , Adenoviridae , Animals , Antibodies, Monoclonal , Antigens, CD/immunology , B7-1 Antigen/immunology , B7-2 Antigen , CD40 Antigens/immunology , Flow Cytometry , Genes, Reporter , Green Fluorescent Proteins , HLA-DR Antigens/immunology , Lipopolysaccharide Receptors/immunology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Lymphocyte Culture Test, Mixed , Macaca mulatta , Membrane Glycoproteins/immunology , Transduction, Genetic , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
20.
Hum Immunol ; 63(3): 164-75, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11872234

ABSTRACT

Natural killer (NK) T cells are restricted by CD1d and play an important role in the rejection of malignant tumors, but how kill these tumors is unclear. To investigate this, we cultured Valpha24+CD4+ NK T cells in human umbilical cord blood, which was enriched by immunomagnetic beads. In short-term (4 h) cytotoxicity assays, the NK T cells could kill only those targets expressing CD1d. In longer cytotoxicity assays (20 h), however, the NK T cells were able to kill all the tumors, regardless of CD1d expression. When each of the perforin, Fas-FasL, and TNF-alpha cytotoxic mechanisms were blocked, it was apparent that perforin killing dominated in both the short- and long-term assays. In the short-term assay, perforin killing required that the targets expressed CD1d, but killing was more efficient if Fas was present because then the Fas-FasL mechanism was also used. Thus, cells that lacked Fas and CD1d and were not killed in the 4-h assay, were instead lysed in 20-h assay through a combination of perforin and TNF-alpha killing. NK T cells can kill tumor targets by perforin, Fas-FasL, and TNF-alpha mechanisms. TNF-alpha killing requires longer contact between effectors and targets, suggesting that TNF-alpha acts by enhancing perforin killing.


Subject(s)
Antigens, CD1/immunology , Cytotoxicity, Immunologic , Fetal Blood/immunology , Killer Cells, Natural/immunology , Membrane Glycoproteins/immunology , Neoplasms/immunology , Antigens, CD1/metabolism , Antigens, CD1d , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cytokines/analysis , Cytokines/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Fas Ligand Protein , Female , Humans , Interferon-gamma/analysis , Killer Cells, Natural/metabolism , Lymphocyte Culture Test, Mixed , Membrane Glycoproteins/metabolism , Neoplasms/metabolism , Perforin , Pore Forming Cytotoxic Proteins , Tumor Necrosis Factor-alpha/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...