Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
1.
BMC Immunol ; 21(1): 19, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32299357

ABSTRACT

BACKGROUND: Miscellaneous memory cell populations that exist before organ transplantation are crucial barriers to transplantation. In the present study, we used a skin-primed heart transplantation model in mouse to evaluate the abilities of Thalidomide (TD), alone or in combination with co-stimulatory blockade, using monoclonal antibodies (mAbs) against memory T cells and alloantibodies to prolong the second cardiac survival. RESULTS: In the skin-primed heart transplantation model, TD combined with mAbs significantly prolonged the second cardiac survival, accompanied by inhibition of memory CD8+ T cells. This combined treatment enhanced the CD4+Foxp3+ regulatory T cells ratio in the spleen, restrained the infiltration of lymphocytes into the allograft, and suppressed the allo-response of spleen T cells in the recipient. The levels of allo-antibodies also decreased in the recipient serum. In addition, we detected low levels of the constitutions of the lytic machinery of cytotoxic cells, which cause allograft damage. CONCLUSIONS: Our study indicated a potential synergistic action of TD in combination with with mAbs to suppress the function of memory T cells and increase the survival of second allografts in alloantigen-primed mice.


Subject(s)
Graft Rejection/drug therapy , Graft Survival/drug effects , Heart/drug effects , Isoantigens/pharmacology , Thalidomide/pharmacology , Allografts/drug effects , Animals , Antibodies, Monoclonal/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Female , Forkhead Transcription Factors/metabolism , Graft Rejection/metabolism , Heart Transplantation/methods , Immunologic Memory/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Spleen/drug effects , T-Lymphocytes, Regulatory/drug effects , Transplantation, Homologous/methods
2.
Fish Shellfish Immunol ; 40(1): 109-19, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24973517

ABSTRACT

Estrogens are recognized as modulators of immune responses in mammals and teleosts. While it is known that the effects of estrogens are mediated via leukocyte-specific estrogen receptors (ERs) in humans and mice, leucocyte-specific estrogen receptor expression and the effects of estrogens on this cell population is less explored and poorly understood in teleosts. Here in, we verify that channel catfish (Ictalurus punctaus) leukocytes express ERα and ERß2. Transcripts of these isoforms were detected in tissue-associated leukocyte populations by PCR, but ERß2 was rarely detected in PBLs. Expression of these receptors was temporally regulated in PBLs following polyclonal activation by concanavalin A, lipopolysaccharide or alloantigen based on evaluation by quantitative and end-point PCR. Examination of long-term leukocyte cell lines demonstrated that these receptors are differentially expressed depending on leukocyte lineage and phenotype. Expression of ERs was also temporally dynamic in some leukocyte lineages and may reflect stage of cell maturity. Estrogens affect the responsiveness of channel catfish peripheral blood leukocytes (PBLs) to mitogens in vitro. Similarly, bactericidal activity and phorbol 12-myristate 13-acetate induced respiratory burst was modulated by 17ß-estradiol. These actions were blocked by the pure ER antagonist ICI 182780 indicating that response is, in part, mediated via ERα. In summary, estrogen receptors are expressed in channel catfish leukocytes and participate in the regulation of the immune response. This is the first time leukocyte lineage expression has been reported in teleost cell lines.


Subject(s)
Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Estrogens/metabolism , Fish Proteins/genetics , Gene Expression Regulation , Ictaluridae/genetics , Leukocytes/immunology , Animals , Cell Line , Cell Proliferation , Concanavalin A/pharmacology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Fish Proteins/metabolism , Ictaluridae/immunology , Isoantigens/pharmacology , Leukocytes/metabolism , Lipopolysaccharides/pharmacology , Mitogens/pharmacology , Real-Time Polymerase Chain Reaction/veterinary
3.
Viral Immunol ; 27(6): 305-15, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24844121

ABSTRACT

CD8+ T-cells of asymptomatic HIV-1 carriers (AC) suppress human immunodeficiency virus type 1 (HIV-1) replication in a class I major histocompatibility complex (MHC-I)-restricted and -unrestricted manner. In order to investigate the mechanism of MHC-I-unrestricted CD8+ T-cell-mediated HIV-1 suppression, we previously established allo-antigen stimulated CD8+T-cells from HIV-1-uninfected donors. These allo-antigen stimulated CD8+ T-cells suppressed HIV-1 replication in acutely infected autologous CD4+ T-cells when directly co-cultured. To elucidate the mechanism of HIV-1 replication suppression, we analyzed DNA-binding activity and phosphorylation of transcriptional factors associated with HIV-1 replication by electrophoresis mobility shift assay and Western blotting. When CD4+ T-cells were cultured with allo-antigen stimulated CD8+ T-cells, the reduction of NF-κB and Ets-1 DNA-binding activity was observed. Nuclear localization of NF-κB p65 and Ets-1 was suppressed in CD4+ T-cells. Although NF-κB p65 and Ets-1 are known to be regulated by protein kinase A (PKA), no difference was observed in the expression and phosphorylation of the PKA catalytic subunit in CD4+ T-cells cultured with PHA-treated CD8+ T-cells or allo-antigen stimulated CD8+ T-cells. Cyclic AMP is also known to enter through gap junctions, but the suppression of HIV-1 replication mediated by allo-antigen stimulated CD8+ T-cells was not affected by the gap junction inhibitor. The nuclear transport of phosphorylated NF-κB p65 (Ser276) was inhibited only in CD4+ T-cells cultured with allo-antigen stimulated CD8+ T-cells. Our results indicate that allo-antigen stimulated CD8+ T-cells suppress the transcriptional activity of NF-κB p65 or Ets-1 in an antigen-nonspecific manner, and inhibit the nuclear transport of phosphorylated NF-κB p65 (Ser276).


Subject(s)
CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , HIV-1/physiology , Isoantigens/pharmacology , NF-kappa B/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Flow Cytometry , Gene Expression Regulation, Viral , HIV-1/immunology , Humans , NF-kappa B/genetics , Phosphorylation , Protein Binding , Protein Transport , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Virus Replication , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Mol Biol Rep ; 41(5): 3425-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24510407

ABSTRACT

Liver-specific protein F is commonly used in liver transplantation studies for its allograft immunogenicity. The objective of this study was to investigate immune tolerance induced by protein F in liver transplantation in rats. Healthy inbred male Wistar and Sprague-Dawley (SD) rats were used in this study. The transplant recipient rats were randomly divided into three groups. The SD rats transplanted with liver tissues from SD rats or Wistar rats were defined as intragraft control group (Group A) or acute reaction group (Group B), respectively. The SD rats that received thymic administration of 4 mg protein F 1 week prior to transplantation with livers from Wistar rats were defined as protein F interference group (Group C). Kamada's two-cuff technique was utilized in the liver transplantation surgeries. The postoperative general condition, transplantation survival time, pathological examination, and serum IFN-γ level (quantified by ELISA) were recorded and compared to evaluate the immune response and outcomes in the recipient rats after liver transplantation. Group A rats exhibited good postoperative condition and prolonged survival (median survival time was 92 days). In contrast, Group B rats lost body weight rapidly after liver transplantation, and died starting at day 12 (median survival time was 15 days). Compared to Group B, Group C rats showed significantly longer survival (medium survival time was 71 days). Our findings indicate that protein F is an important transplantation antigen with allograft immunogenicity, which could successfully induce immune tolerance in liver transplantation.


Subject(s)
Immune Tolerance , Isoantigens/immunology , Liver Transplantation , Transplantation Immunology , Animals , Immune Tolerance/drug effects , Interferon-gamma/blood , Isoantigens/pharmacology , Liver/pathology , Male , Models, Animal , Rats , Transplantation, Homologous
5.
Cell Transplant ; 21(2-3): 581-90, 2012.
Article in English | MEDLINE | ID: mdl-22793067

ABSTRACT

Bone marrow-derived mesenchymal stem cells (MSCs) could exert a potent immunosuppressive effect, and therefore may have a therapeutic potential in T-cell-dependent pathologies. In the present study, we aimed to determine whether MSCs could be used to control graft-versus-host disease (GvHD), a major cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (HSCT). MSCs were isolated from Lewis rat bone morrow and then cultured in 10% FBS DMEM at 37°C for 4 weeks. The enriched conventional MSCs and macrophages were purified by auto-MACS. Cloned MSCs were obtained by cloning using the limiting dilution method and expanded up to more than 6 months. The identity of MSCs was confirmed by their typical spindle-shaped morphology and immunophenotypic criteria, based on the absence of expression of CD45 and CD11b/c molecules. Both types of MSCs were also tested for their ability to differentiate into adipocytes. We showed that MSCs, like macrophages, exhibit immunomodulatory properties capable of inhibiting T-cell proliferation stimulated by alloantigens, anti-CD3e/CD28 mAbs, and ConA in a dose-dependent manner in vitro. After performing adoptive transfer, MSCs suppressed systemic Lewis to (Lewis × DA)F1 rat GvHD. In contrast to the immunosuppressive activities of conventional MSCs, the cloned MSCs enhanced T-cell proliferation in vitro and yielded no clinical benefit in regard to the incidence or severity of GvHD. Therefore, these rat models have shown intriguing differences in the suppression effects of lymphocyte proliferation and GvHD prevention between short-term cultured conventional MSCs and cloned MSCs.


Subject(s)
Bone Marrow Cells/cytology , Graft vs Host Disease/prevention & control , Mesenchymal Stem Cells/cytology , T-Lymphocytes/cytology , Adipocytes/cytology , Animals , CD11 Antigens/metabolism , Cell Differentiation , Cell Proliferation/drug effects , Cell- and Tissue-Based Therapy , Cells, Cultured , Disease Models, Animal , Gamma Rays , Graft vs Host Disease/immunology , Graft vs Host Disease/mortality , Hematopoietic Stem Cell Transplantation , Immunophenotyping , Immunosuppression Therapy , Isoantigens/pharmacology , Leukocyte Common Antigens/metabolism , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/pathology , Rats , Rats, Inbred Lew , Survival Rate , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
6.
Med Hypotheses ; 76(3): 441-4, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21134721

ABSTRACT

Topical immune suppression is an attractive and practical therapeutic option to prolong survival time of allografts, before the appearance of new agent with higher immunosuppressive efficacy and lower undesirable side effects. The initiation of rejection and outcome of allografts is principally mediated by alloantigen reactive T cells. The activation of T cells requires at least two signals, first is T-cell receptor signal and second is costimulatory signal. T cells that encounter antigen without the appropriate costimulatory signal become anergy or tolerance. Migration of alloantigen-bearing dendritic cells into the T-cell zone of secondary lymphoid tissues, which are essential for primary alloimmune responses, effectively induces T-cell activation and expansion with the presence of two signals. Draining lymph nodes are the promising targets for topical immune suppression, as disrupting lymphatic drainage from the transplanted graft to lymph nodes prevented rejection of skin allografts and lymphadenectomy prolong the survival time of skin and corneal allografts in experimental animals. Therefore, we hypothesize that inhibition of T cell costimulatory pathways in draining lymph nodes could impair the alloantigen-specific immune response and reduce systemic immunosuppressive drugs dose for allografts survival. Further investigations are required to identify most efficient way for draining lymph nodes transfer of costimulatory molecule gene or topical drug administration of costimulatory inhibitors to draining lymph nodes.


Subject(s)
Graft Rejection , Interleukin-2/pharmacology , Lymph Nodes/immunology , Models, Immunological , T-Lymphocytes , Administration, Topical , Animals , B7-2 Antigen/genetics , Dendritic Cells/immunology , Graft Rejection/immunology , Immune Tolerance/immunology , Immunosuppressive Agents/immunology , Immunosuppressive Agents/pharmacology , Interleukin-2/immunology , Isoantigens/immunology , Isoantigens/pharmacology , Lymph Nodes/pathology , Lymphocyte Activation/immunology , Mice , Mice, Nude , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Transplantation, Homologous
7.
Transpl Immunol ; 22(3-4): 150-6, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19879946

ABSTRACT

The aim of our study was to investigate the allogeneic influence inside and outside the liver in vitro following donor-specific cell injection (DSI). DA rats (RT1a) were used as donors and WS rats (RT1k) as recipients. WS were sensitized with DA spleen cells, followed 24h later by total hepatectomy. The liver was transplanted into another WS (sensitized liver-grafted; SL-Grafted). The hepatectomized WS underwent liver transplantation from a naive WS (sensitized liver-removed; SL-Removed). Alloantigens accumulated in the liver in SL-Grafted and in the extrahepatic tissue/organ(s) in SL-Removed. DA hearts were transplanted 10days after antigen administration. To analyze the immune responses, we measured Th1/Th2 cytokine profiles, and perforin mRNA in various organs, allogeneic mixed lymphocyte reaction (MLR), and donor-specific immunoglobulin. Th1 cytokine levels in the liver of SL-Grafted and in spleen of SL-Removed were highly and rapidly upregulated but decreased thereafter. IFN-gamma and perforin mRNAs were significantly higher in SL-Grafted and lower in SL-Removed. MLR was significantly higher in SL-Grafted than SL-Removed and controls. There was no significant difference in the donor-specific immunoglobulin level. Our findings suggest that liver and other organs may behave differently to alloantigen, suggesting the importance of an early Th1 reaction in the liver and spleen.


Subject(s)
Graft Rejection/immunology , Isoantigens/pharmacology , Liver/metabolism , Th1 Cells/metabolism , Th2 Cells/metabolism , Animals , Cell Proliferation/drug effects , Cell Transplantation , Cytokines/metabolism , Graft Rejection/pathology , Heart Transplantation , Immunomodulation , Isoantigens/administration & dosage , Isoantigens/immunology , Liver/immunology , Liver/pathology , Liver/surgery , Liver Transplantation , Lymphocyte Culture Test, Mixed , Male , Perforin/genetics , Perforin/metabolism , Rats , Rats, Wistar , Spleen/immunology , Spleen/metabolism , Spleen/pathology , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/pathology , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/pathology
8.
Transplantation ; 86(12): 1830-6, 2008 Dec 27.
Article in English | MEDLINE | ID: mdl-19104430

ABSTRACT

BACKGROUND: Immunological unresponsiveness of T cells to alloantigen can be induced by intrathymic injection of donor-specific antigen in small-animal models. Intrathymic tolerance to vascularized grafts in large animals has not previously been reported. METHODS: Thirty-two dogs were allocated into dog leukocyte antigen DP locus allele (class II)-matched donor-recipient pairs. Female recipients were paired with male donors. Tissue typing was based on restriction fragment length polymorphism. Recipients were given 18 Gy total lymphoid irradiation in 16 fractions (1.125 Gy each) over 4 weeks. Thoracotomy after the 6th fraction permitted perithymic (n=4) or intrathymic (n=4) injection of donor bone marrow (BM) or intrathymic injection of saline (n=5). Another group received intravenous peripheral BM infusion (n=3). Fifty days postthoracotomy recipients underwent bilateral nephrectomy and donor-specific kidney transplantation. Acute rejection, suspected when serum creatinine was more than 600 mumol/L or urea was more than 40 mmol/L, was confirmed histologically. Full-thickness skin grafts followed more than 100 days posttransplantation. Tissue samples were taken for Y-chromosome polymerase chain reaction. RESULTS: One intrathymic (25%) and three perithymic (75%) BM recipients developed tolerance to renal allografts. Three intrathymic BM recipients rejected after 27, 32, and 54 days and one perithymic BM recipient rejected after 42 days. All recipients given peripheral BM or saline had rejected by 29 and 38 days, respectively. All recipients surviving more than 100 days posttransplantation, accepted donor specific and rejected dog leukocyte antigen-DP locus allele (class II) identical third-party skin grafts. Polymerase chain reaction detected intrathymic but not hematopoietic chimerism in sex-mismatched pairs. CONCLUSIONS: Fractionated total lymphoid irradiation and perithymic or intrathymic donor-specific BM induced tolerance to renal and skin allografts without inducing hematopoietic chimerism.


Subject(s)
Histocompatibility Testing/methods , Immune Tolerance , Isoantigens/pharmacology , Kidney Transplantation/immunology , Lymphatic System/radiation effects , T-Lymphocytes/immunology , Animals , Bone Marrow Cells/pathology , Bone Marrow Cells/physiology , Bone Marrow Transplantation , Computer Simulation , Dogs , Female , Histocompatibility Antigens Class II/immunology , Lymphatic System/immunology , Male , Models, Animal , Nephrectomy , Polymorphism, Restriction Fragment Length , Skin Transplantation/immunology
9.
J Immunol ; 181(11): 7499-506, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19017939

ABSTRACT

We have previously shown that HLA-DQA1, a peptide derived from a highly conserved region of MHC class II, prevents alloreactive T cell priming and effector function in vivo, although underlying mechanisms are obscure. In this study, we demonstrate that 28% of mice treated with HLA-DQA1 combined with low-dose rapamycin achieved permanent engraftment of fully MHC-disparate islet allografts and significantly prolonged survival in the remaining animals (log rank, p < 0.001). Immunohistologic examination of the grafts from HLA-DQA1/rapamycin-treated animals revealed up-regulated expression of TGF-ss and FoxP3. In vivo administration of blocking anti-TGF-ss or depleting anti-CD25 mAb augmented T cell alloimmunity and prevented the long-term engraft induced by HLA-DQA1. In vitro experiments further showed that HLA-DQA1 induced differentiation of CD4(+) T cells into CD4(+)CD25(+)FoxP3(+) regulatory T cells. Together, these data provide the first demonstration that HLA-DQA1, a MHC class II-derived peptide, can prolong allograft survival via a TGF-beta and regulatory T cell-dependent mechanisms.


Subject(s)
Graft Survival/drug effects , HLA-DQ Antigens/pharmacology , Islets of Langerhans Transplantation , Islets of Langerhans/immunology , Isoantigens/pharmacology , Peptides/pharmacology , T-Lymphocytes, Regulatory/immunology , Animals , Antibiotics, Antineoplastic/pharmacology , Antibodies, Monoclonal/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Forkhead Transcription Factors/immunology , Graft Survival/immunology , HLA-DQ alpha-Chains , Humans , Interleukin-2 Receptor alpha Subunit/antagonists & inhibitors , Interleukin-2 Receptor alpha Subunit/immunology , Mice , Mice, Inbred BALB C , Sirolimus/pharmacology , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/immunology , Transplantation Immunology/drug effects , Transplantation, Homologous , Up-Regulation/drug effects , Up-Regulation/immunology
10.
J Heart Lung Transplant ; 27(4): 362-71, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18374871

ABSTRACT

BACKGROUND: It is well known that CD4(+)CD25(+) regulatory T (Treg) cells play a central role in the suppression of autoimmunity, inflammation and allograft rejection. Therefore, therapeutic agents that capable of enhancing the number and activity of this T-cell subset are highly desirable. METHODS: The present study was designed to investigate the effects of superagonistic CD28-specific monoclonal antibody (supCD28 MAb) on preferentially expanded rat naturally occurring CD4(+)CD25(+) Treg (nTreg) cells and its applicability in cardiac transplantation. RESULTS: A single administration of supCD28 MAb preferentially proliferated nTreg cells. The increase of Foxp3 expression and polarization toward a Th2 cytokine profile correlated with decreased production of interferon-gamma and increased production of interleukin-4 and -10 in the expanded CD4(+)CD25(+) Treg subset, which was capable of suppressing CD4(+)CD25(-) T-cell proliferation after purification. Furthermore, supCD28 MAb administration revealed that nTreg cells were preferentially proliferating in vivo and recruited into the grafts, resulting in significant prolongation of full MHC-mismatch cardiac graft survival. CONCLUSIONS: Our data demonstrate that supCD28 MAb targets expansion of nTreg cells in vivo and maintains and enhances their regulatory functions, which represents a major advance toward the therapeutic use of polyclonally activated Treg cells as cellular therapy for treatment of allograft rejection.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD28 Antigens/immunology , Forkhead Transcription Factors/metabolism , Graft Rejection/prevention & control , Heart Transplantation , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Animals , Blood Cells/metabolism , Cell Proliferation/drug effects , Concanavalin A/pharmacology , Graft Survival/drug effects , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukins/metabolism , Isoantigens/pharmacology , Male , Myocardium/metabolism , Myocardium/pathology , Postoperative Period , Rats , Rats, Inbred Strains , T-Lymphocytes, Regulatory/drug effects , Time Factors , Transplantation, Heterotopic
11.
Transplantation ; 85(2): 237-46, 2008 Jan 27.
Article in English | MEDLINE | ID: mdl-18212629

ABSTRACT

BACKGROUND: We showed recently that limb allograft survival could be enhanced by administration of alloantigen (Ag)-pulsed immature dendritic cells (DC) after transplantation. Since indefinite graft survival was not achieved, we have further modified the DC by pharmacologic (rapamycin; Rapa) conditioning and ascertained their influence on graft survival, without continued immunosuppressive therapy. METHODS: We compared the ability of donor Ag-pulsed, Rapa-conditioned rat myeloid DC (Rapa DC) and control DC (CTR DC) to inhibit alloreactive T-cell responses after limb transplantation in antilymphocyte serum (ALS)-treated recipients given a short postoperative course of cyclosporine (CsA). RESULTS: Both DC populations expressed similar levels of major histocompatibility complex (MHC) II, CD40 and CD54, but Rapa DC expressed lower CD86. After toll-like receptor activation, both populations produced minimal interleukin (IL)-12p70, but Rapa DC secreted lower levels of IL-6 and IL-10. The capacity of DCs to stimulate T-cell proliferation in mixed leukocyte reactions was very low. Pulsing of the DC with donor Ag did not alter their phenotype or function. Interestingly, posttransplant administration of donor Ag-pulsed Rapa DC to rats given perioperative ALS and 21 days CsA significantly delayed graft rejection and promoted long-term (>125 days) graft survival. AlloAg-pulsed Rapa DC induced T-cell hyporesponsiveness and promoted the generation of IL-10-secreting CD4 T cells upon ex vivo challenge. CONCLUSIONS: Infusion of donor Ag-pulsed, Rapa-conditioned DC after composite tissue transplantation can prevent rejection of the grafts, including skin, across a full MHC mismatch and in the absence of continued immunosuppressive therapy.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/transplantation , Graft Survival , Hindlimb/transplantation , Isoantigens/pharmacology , Transplantation, Homologous/physiology , Animals , Bone Marrow Cells/immunology , Immunosuppression Therapy , Immunosuppressive Agents/pharmacology , Interferon-gamma/metabolism , Male , Rats , Rats, Inbred Lew , Rats, Inbred WF , Rats, Sprague-Dawley , Sirolimus/pharmacology , Transplantation Chimera
12.
Cell Immunol ; 248(1): 31-43, 2007 Jul.
Article in English | MEDLINE | ID: mdl-18023633

ABSTRACT

The inherent ability of the host immune system to distinguish between self- and non-self forms the basis of allorecognition. T lymphocytes constitute the most important effector arm of allorecognition. Here we describe the fundamentals of direct and indirect pathways by which allopeptides are presented to effector T cells. The nature of allopeptides presented along with tolerogenic strategies like altered peptide ligands and intra- or extra-thymic allopeptide inoculation are discussed. In addition, we speculate on the potential of regulatory T cells to modulate alloimmune responses.


Subject(s)
Antigen Presentation , Graft Rejection/immunology , Graft Survival/immunology , Immune Tolerance , Isoantigens/immunology , Lymphocyte Activation/immunology , Peptides/immunology , Animals , Graft Rejection/prevention & control , Heart Transplantation , Histocompatibility Antigens , Humans , Immunosuppressive Agents/immunology , Immunosuppressive Agents/pharmacology , Isoantigens/pharmacology , Kidney Transplantation , Lymphocyte Activation/drug effects , Mice , Models, Animal , Peptides/chemical synthesis , Peptides/pharmacology , Rats , Rats, Inbred Lew , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Transplantation Immunology , Transplantation, Homologous
13.
Am J Transplant ; 7(12): 2683-92, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17924994

ABSTRACT

Programmed death-1 (PD-1) is a recently identified coinhibitory molecule that belongs to the CD28 superfamily. PD-1 has two ligands PD-L1 and PD-L2. There is some evidence that PD-L1 and PD-L2 serve distinct functions, but their exact function in alloimmunity remains unclear. In the present study, we used a GVHD-like model that allows detailed analyses of T-cell activation at a single cell level in vivo to examine the role of PD-1/PD-L1 and PD-1/PD-L2 interactions in regulating proliferation of CD4(+) and CD8(+) T cells in response to alloantigen stimulation. We found that both CD4(+) and CD8(+) T cells proliferated vigorously in vivo and that PD-L1 and PD-L2 exhibit strikingly different effect on T-cell proliferation. While blocking PD-L1 did not affect the in vivo proliferation of CD4(+) and CD8(+) T cells regardless of CD28 costimulation, blocking PD-L2 resulted in a marked increase in the responder frequency of CD8(+) T-cells in vivo. The effect of PD-L2 on the CD8(+) T-cell proliferation is regulated by CD28 costimulation and by the CD4(+) T cells. We conclude that PD-L1 and PD-L2 function differently in regulating alloreactive T-cell activation in vivo, and PD-L2 is predominant in this model in limiting alloreactive CD8(+) T-cell proliferation.


Subject(s)
B7-1 Antigen/metabolism , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Graft vs Host Disease/metabolism , Graft vs Host Disease/pathology , Membrane Glycoproteins/metabolism , Peptides/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , B7-1 Antigen/immunology , B7-H1 Antigen , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Disease Models, Animal , Graft vs Host Disease/immunology , Isoantigens/immunology , Isoantigens/pharmacology , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Peptides/immunology , Programmed Cell Death 1 Ligand 2 Protein , Signal Transduction/drug effects , Signal Transduction/physiology
14.
Transplantation ; 84(3): 407-14, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17700168

ABSTRACT

BACKGROUND: There is great interest in the recently developed immunosuppressant NK026680, which is a derivative of triazolopyrimidine. Its unique chemical structure and action mechanism are completely different from those of conventional immunosuppressants. METHODS: The present study was designed to investigate the effects of NK026680 on rat bone-marrow-derived dendritic cell (BMDC) differentiation and maturation in an in vitro culture system and its applicability in liver transplantation. RESULTS: NK026680 inhibited T-cell proliferation stimulated by alloantigen in a dose-dependent manner, but did not inhibit concanavalin A. The populations of OX6+CD161a cells and CD86+CD161a cells were suppressed in NK026680-treated dendritic cells (DCs). Exposure of DCs to NK026680 downregulated the interleukin (IL)-12 (p40, p35), interferon-gamma mRNA expression and upregulated IL-10, transforming growth factor-beta, in which impaired the ability of DC to stimulate T cell proliferation. Furthermore, oral administration of NK026680 for 14 days significantly prolonged liver allograft survival and limitation of T-cell responses and polarization toward a Th2 cytokine profile. CONCLUSIONS: These results demonstrate that NK026680 may have therapeutic potential for preventing allo-rejection in organ transplantation, acting at the step of immune response through inhibiting BMDC differentiation and maturation into potent antigen-presenting cells.


Subject(s)
Dendritic Cells/drug effects , Dendritic Cells/physiology , Graft Survival/physiology , Immunosuppressive Agents/pharmacology , Liver Transplantation/physiology , Pyrimidines/pharmacology , Triazoles/pharmacology , Animals , B7-2 Antigen/genetics , B7-2 Antigen/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Concanavalin A/pharmacology , Dendritic Cells/cytology , Dose-Response Relationship, Drug , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Isoantigens/pharmacology , Liver Transplantation/pathology , Male , Mitogens/pharmacology , Rats , Rats, Inbred Lew , T-Lymphocytes/drug effects , T-Lymphocytes/pathology , T-Lymphocytes/physiology
15.
J Immunol ; 179(1): 45-52, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17579020

ABSTRACT

Ag presentation is a key step in the initiation of adaptive immune responses that depends on the expression of MHC Ags and costimulatory molecules. Immune-enhancing CpG and non-CPG oligodeoxynucleotides (ODNs) stimulate Ag presentation by stimulating the expression of these molecules and by promoting dendritic cell maturation. In this report, we identify immunoregulatory orthophosphorothioate non-CpG molecules, referred to as regulatory ODNs (rODNs), by their ability to inhibit allogeneic monocyte-stimulated T cell responses and down-regulate HLA-DR in human primary monocytes. The rODNs promoted the survival of macrophages and were able to activate IL-8 secretion through a chloroquine-resistant pathway. Messenger RNAs for HLA-DR alpha and beta and the MHC CIITA were reduced by rODNs but not by stimulatory CpG ODN2006 and non-CpG ODN2006a. CIITA transcription in monocytes was controlled primarily by promoter III and not by promoter I or IV. rODNs blocked promoter III-directed transcription of CIITA in these cells. Under conditions that induced dendritic cell differentiation, rODNs also reduced HLA-DR expression. The activity of rODNs is phosphorothioate chemistry and G stretch dependent but TLR9 independent. G tetrads were detected by circular dichroism in active rODNs and associated with high m.w. multimers on nondenaturing gels. Heat treatment of rODNs disrupted G tetrads, the high m.w. aggregates, and the HLA-DR inhibitory activity of the ODNs. The inhibition of immune responses by regulatory oligodeoxynucleotides may be useful for the treatment of immune-mediated disorders including autoimmune diseases and graft rejection.


Subject(s)
Adjuvants, Immunologic/pharmacology , Down-Regulation/immunology , Monocytes/immunology , Monocytes/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Oligodeoxyribonucleotides/pharmacology , Promoter Regions, Genetic/immunology , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Cell Proliferation , Cells, Cultured , Growth Inhibitors/physiology , HLA-DR Antigens/biosynthesis , HLA-DR Antigens/metabolism , HLA-DR alpha-Chains , Humans , Isoantigens/pharmacology , Nuclear Proteins/biosynthesis , Promoter Regions, Genetic/genetics , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Trans-Activators/biosynthesis , Transcription, Genetic/immunology
16.
J Immunol ; 178(4): 2221-8, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17277127

ABSTRACT

Alloantibody is an important effector mechanism for allograft rejection. In this study, we tested the hypothesis that regulatory T cells with indirect allospecificity can prevent humoral rejection by using a rat transplant model in which acute rejection of MHC class I-disparate PVG.R8 heart grafts by PVG.RT1(u) recipients is mediated by alloantibody and is dependent upon help from CD4 T cells that can recognize the disparate MHC alloantigen only via the indirect pathway. Pretransplant treatment of PVG.RT1(u) recipients with anti-CD4 mAb plus donor-specific transfusion abrogated alloantibody production and prolonged PVG.R8 graft survival indefinitely. Naive syngeneic splenocytes injected into tolerant animals did not effect heart graft rejection, suggesting the presence of regulatory mechanisms. Adoptive transfer experiments into CD4 T cell-reconstituted, congenitally athymic recipients confirmed that regulation was mediated by CD4 T cells and was alloantigen-specific. CD4 T cell regulation could be broken in tolerant animals either by immunizing with an immunodominant linear allopeptide or by depleting tolerant CD4 T cells, but surprisingly this resulted in neither alloantibody generation nor graft rejection. These findings demonstrate that anti-CD4 plus donor-specific transfusion treatment results in the development of CD4 regulatory T cells that recognize alloantigens via the indirect pathway and act in an Ag-specific manner to prevent alloantibody-mediated rejection. Their development is associated with intrinsic tolerance within the alloantigen-specific B cell compartment that persists after T cell help is made available.


Subject(s)
Antibody Specificity/immunology , Graft Rejection/immunology , Heart Transplantation/immunology , Isoantibodies/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibody Formation/drug effects , Antibody Formation/immunology , Antibody Specificity/drug effects , Antigen Presentation/drug effects , Antigen Presentation/immunology , B-Lymphocytes/immunology , CD4 Antigens/immunology , Graft Rejection/prevention & control , Immune Tolerance/drug effects , Immune Tolerance/immunology , Isoantigens/immunology , Isoantigens/pharmacology , Lymphocyte Depletion , Peptides/immunology , Peptides/pharmacology , Rats , Rats, Inbred Lew , T-Lymphocytes, Regulatory/transplantation , Transplantation, Homologous
17.
Cell Death Differ ; 14(3): 480-8, 2007 Mar.
Article in English | MEDLINE | ID: mdl-16902496

ABSTRACT

Genetically modified antigen-presenting cells (APC) represent an attractive strategy for in vitro immunomodulation. In the human system, APC expressing HLA-A1 and a membrane-bound form of CD95L (m-CD95L) were used for selective depletion of HLA-A1-specific T cells. In short-term assays, m-CD95L-expressing APC-induced apoptosis in activated T cells and the constitutive presence of m-CD95L and HLA-A1 expressing APC in long-term T cell cultures prevented the expansion of CD4(+) and CD8(+) HLA-A1-specific T cells and the development of HLA-A1-specific cytotoxicity. However, immunity towards third party, viral and bacterial antigens was maintained and T cells spared from depletion could be induced to develop cytotoxicity towards unrelated antigens. Interestingly, inhibition of HLA-A1-specific T cell response absolutely requires the coexpression of m-CD95L and HLA-A1 antigen on the same APC. Thus, m-CD95L expressing APC might be used in clinical settings to obtain tolerance induction in allogeneic transplantation systems or autoimmune diseases.


Subject(s)
Antigen-Presenting Cells/metabolism , Antigens, Viral/immunology , Cell Membrane/metabolism , Fas Ligand Protein/metabolism , Immunity, Cellular , Isoantigens/immunology , T-Lymphocytes/immunology , Antigen Presentation , Antigen-Presenting Cells/immunology , Antigens, Bacterial/immunology , Cells, Cultured , Cytotoxicity, Immunologic , HLA-A1 Antigen/genetics , HLA-A1 Antigen/metabolism , Herpesvirus 4, Human/immunology , Humans , Immunity, Active , Isoantigens/pharmacology , Jurkat Cells , Lymphocyte Activation , T-Lymphocytes/virology , Transfection
18.
Med Oncol ; 23(2): 273-82, 2006.
Article in English | MEDLINE | ID: mdl-16720928

ABSTRACT

A variety of approaches have been used to deliver tumor-associated antigens (TAA) in conjunction with dendritic cells (DC) as cellular adjuvants. DC derived from monocytic precursors have been pulsed with whole tumor antigen using a variety of strategies and have been demonstrated to induce CD4+ and CD8+ antitumor responses. In the present study, monocyte-derived DC have been pulsed with lysate from an allogeneic melanoma cell line, A-375, and used to repeatedly stimulate T cells. The resultant T cells were examined for cytotoxic activity against A-375 targets as well as the HLA A2-positive melanoma cell line DFW. Uptake of FITC-labeled melanoma lysate by DC established that lysate of melanoma cells was efficiently endocytosed. Stimulation with lysate-pulsed DC resulted in strong proliferative responses by T cells, which could be inhibited by antibodies against both MHC class I and class II. T cells stimulated in vitro with lysate-pulsed DC demonstrated potent cytotoxicity against the melanoma targets which were blocked by antibodies against MHC class I. Lysate-pulsed DC also elicited IFN-gamma secretion by T cells as measured in an ELISPOT assay. We have also examined the ability of lysate-pulsed DC to present melanoma-associated antigens to T cells. ELISPOT assays with synthetic peptides of melanoma-associated antigens, such as gp100, mage1, NY-ESO, and MART-1, revealed that lysate-pulsed DC could stimulate T cells in an antigen-specific manner. The results demonstrate that lysate from allogeneic tumor cells may be used as a source of antigens to stimulate tumor-specific T cells in melanoma.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Dendritic Cells/immunology , Lymphocyte Activation/immunology , Melanoma/immunology , T-Lymphocytes/immunology , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/pharmacology , Cell Line, Tumor , Coculture Techniques , Humans , Isoantigens/chemistry , Isoantigens/immunology , Isoantigens/pharmacology , K562 Cells , Melanoma/chemistry , Monocytes
19.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 22(1): 11-3, 17, 2006 Jan.
Article in Chinese | MEDLINE | ID: mdl-16388734

ABSTRACT

AIM: To explore the immunosuppressive effects of a novel HLA-derived peptide, RDP1258, on rat splenocyte proliferation in vivo and its underlying mechanisms. METHODS: RDP1258 was synthesized through artificial solid-phase synthesis method. Its effects on proliferative reaction of rat splenocytes in vivo were observed by (3)H-TDR incorporation method. And its effects on the heme oxygenase(HO) enzyme activity were analyzed by enzyme chemistry method. RESULTS: The results showed that the synthetic HLA-derived peptide RDP1258 obviously inhibited the proliferation of rat splenocytes stimulated by isoantigen and mitogen. In vivo administration of RDP1258 peptide to rats resulted in upregulation of splenic HO activity and rapid upregulation of HO-1 protein expression. CONCLUSION: HO-1 might participate in RDP1258 inhibiting the proliferation of rat splenocytes induced by mitogen and isoantigen in vivo.


Subject(s)
Cell Proliferation/drug effects , Oligopeptides/pharmacology , Spleen/cytology , Animals , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/metabolism , Immunohistochemistry , Isoantigens/pharmacology , Mitogens/pharmacology , Rats , Rats, Inbred F344 , Spleen/metabolism
20.
J Leukoc Biol ; 79(1): 46-58, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16380601

ABSTRACT

The expression of CD33, a restricted leukocyte antigen considered specific for myeloid lineage, has been studied extensively on lymphoid cells. We demonstrated that wide subsets of mitogen- or alloantigen-activated human T and natural killer (NK) cells express CD33 at protein and nucleic acid levels. CD33+ and CD33- T and NK cell populations showed identical surface expression of activation markers such as CD25, CD28, CD38, CD45RO, or CD95. Myeloid and lymphoid CD33 cDNA were identical. However, lymphoid CD33 protein had lower molecular weight, suggesting cell type-specific, post-translational modifications. Additionally, reverse transcriptase-polymerase chain reaction and Northern blot analysis showed an unknown CD33 isoform (CD33m) expressed on all CD33+ cell lines or T cell clones tested. CD33m was identical to CD33 (CD33M) in the signal peptide, the immunoglobulin (Ig) domain C2, the transmembrane, and the cytoplasmic regions but lacked the extracellular ligand-binding variable Ig-like domain encoded by the second exon. CD33m mRNA was mostly detected on NKL and myeloid cell lines but poorly expressed on B cell lines and T lymphocytes. The CD33m extracellular portion was successfully expressed as a soluble fusion protein on transfected human cells, suggesting a functional role on cell membranes. Cross-linking of CD33 diminished the cytotoxic activity of NKL cells against K562 and P815 target cells, working as an inhibitory receptor on NK cells. These data demonstrate that CD33 expression is not restricted to the myeloid lineage and could exist as two different splicing variants, which could play an important role in the regulation of human lymphoid and myeloid cells.


Subject(s)
Alternative Splicing/immunology , Antigens, CD/biosynthesis , Antigens, Differentiation, Myelomonocytic/biosynthesis , Gene Expression Regulation/immunology , Killer Cells, Natural/immunology , T-Lymphocytes/immunology , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Cell Membrane/immunology , Exons/immunology , Gene Expression Regulation/drug effects , HL-60 Cells , Humans , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Isoantigens/immunology , Isoantigens/pharmacology , K562 Cells , Killer Cells, Natural/cytology , Ligands , Mitogens/immunology , Mitogens/pharmacology , Organ Specificity/immunology , Protein Binding/immunology , Protein Isoforms/biosynthesis , Protein Isoforms/immunology , Protein Modification, Translational/drug effects , Protein Modification, Translational/immunology , Protein Structure, Tertiary , RNA, Messenger/biosynthesis , RNA, Messenger/immunology , Sialic Acid Binding Ig-like Lectin 3 , T-Lymphocytes/cytology , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...