Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Am J Transplant ; 17(6): 1476-1489, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28009481

ABSTRACT

Systemic administration of autologous regulatory dendritic cells (DCreg; unpulsed or pulsed with donor antigen [Ag]), prolongs allograft survival and promotes transplant tolerance in rodents. Here, we demonstrate that nonhuman primate (NHP) monocyte-derived DCreg preloaded with cell membrane vesicles from allogeneic peripheral blood mononuclear cells induce T cell hyporesponsiveness to donor alloantigen (alloAg) in vitro. These donor alloAg-pulsed autologous DCreg (1.4-3.6 × 106 /kg) were administered intravenously, 1 day before MHC-mismatched renal transplantation to rhesus monkeys treated with costimulation blockade (cytotoxic T lymphocyte Ag 4 immunoglobulin [CTLA4] Ig) and tapered rapamycin. Prolongation of graft median survival time from 39.5 days (no DCreg infusion; n = 6 historical controls) and 29 days with control unpulsed DCreg (n = 2), to 56 days with donor Ag-pulsed DCreg (n = 5) was associated with evidence of modulated host CD4+ and CD8+ T cell responses to donor Ag and attenuation of systemic IL-17 production. Circulating anti-donor antibody (Ab) was not detected until CTLA4 Ig withdrawal. One monkey treated with donor Ag-pulsed DCreg rejected its graft in association with progressively elevated anti-donor Ab, 525 days posttransplant (160 days after withdrawal of immunosuppression). These findings indicate a modest but not statistically significant beneficial effect of donor Ag-pulsed autologous DCreg infusion on NHP graft survival when administered with a minimal immunosuppressive drug regimen.


Subject(s)
Dendritic Cells/immunology , Graft Survival/immunology , Isoantigens/immunology , Kidney Failure, Chronic/surgery , Kidney Transplantation , T-Lymphocytes/immunology , Tissue Donors , Animals , Leukocytes, Mononuclear , Macaca mulatta , Male , Transplantation Tolerance , Transplantation, Homologous
3.
Am J Transplant ; 15(6): 1472-4, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25846743

ABSTRACT

Increasing evidence indicates the existence of a complex cross-regulation between the most important biosensors of the human body: The immune and nervous systems. Cytokines control body temperature and trigger autoimmune disorders in the central nervous system, whereas neuropeptides released in peripheral tissues and lymphoid organs modulate inflammatory (innate) and adaptive immune responses. Surprisingly, the effects of nerve fibers and the antidromic release of its pro-inflammatory neuropeptides on the leukocytes of the immune system that mediate graft rejection are practically unknown. In the transplantation field, such area of research remains practically unexplored. A recent study by Riol-Blanco et al has revealed new details on how nociceptive nerves regulate the pro-inflammatory function of leukocytes in peripheral tissues. Although the mechanism(s) by which neuroinflammation affects the immune response against the allograft remains unknown, recent data suggest that this new area of research is worth exploring for potential development of novel complementary therapies for prevention/treatment of graft rejection.


Subject(s)
Biomedical Research/trends , Immune System/physiology , Nociceptors/physiology , Organ Transplantation , Allografts/immunology , Allografts/physiology , Animals , Cytokines/physiology , Graft Rejection/physiopathology , Graft Rejection/prevention & control , Humans , Models, Animal
5.
Am J Transplant ; 13(8): 1989-2005, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23758811

ABSTRACT

We examined the influence of regulatory dendritic cells (DCreg), generated from cytokine-mobilized donor blood monocytes in vitamin D3 and IL-10, on renal allograft survival in a clinically relevant rhesus macaque model. DCreg expressed low MHC class II and costimulatory molecules, but comparatively high levels of programmed death ligand-1 (B7-H1), and were resistant to pro-inflammatory cytokine-induced maturation. They were infused intravenously (3.5-10 × 10(6) /kg), together with the B7-CD28 costimulation blocking agent CTLA4Ig, 7 days before renal transplantation. CTLA4Ig was given for up to 8 weeks and rapamycin, started on Day -2, was maintained with tapering of blood levels until full withdrawal at 6 months. Median graft survival time was 39.5 days in control monkeys (no DC infusion; n = 6) and 113.5 days (p < 0.05) in DCreg-treated animals (n = 6). No adverse events were associated with DCreg infusion, and there was no evidence of induction of host sensitization based on circulating donor-specific alloantibody levels. Immunologic monitoring also revealed regulation of donor-reactive memory CD95(+) T cells and reduced memory/regulatory T cell ratios in DCreg-treated monkeys compared with controls. Termination allograft histology showed moderate combined T cell- and Ab-mediated rejection in both groups. These findings justify further preclinical evaluation of DCreg therapy and their therapeutic potential in organ transplantation.


Subject(s)
Dendritic Cells/transplantation , Graft Survival/immunology , Immune Tolerance/immunology , Immunologic Memory/immunology , Kidney Diseases/prevention & control , Kidney Transplantation/immunology , T-Lymphocytes, Regulatory/immunology , Abatacept , Animals , Combined Modality Therapy , Dendritic Cells/cytology , Dendritic Cells/immunology , Immunoconjugates/immunology , Immunosuppressive Agents/therapeutic use , Kidney Diseases/immunology , Macaca mulatta , Male , Sirolimus/therapeutic use , Transplantation, Homologous
7.
Am J Transplant ; 12(6): 1398-408, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22500950

ABSTRACT

A critical goal in transplantation is the achievement of donor-specific tolerance, minimizing the use of immunosuppressants. Dendritic cells (DCs) are antigen (Ag) presenting cells (APCs) with capability to promote immunity or tolerance. The immune-regulatory properties of DCs have been exploited for generation of tolerogenic/immunosuppressive (IS) DCs that, when transfer systemically, prolong allograft survival in murine models. Surprisingly, the in vivo mechanisms of therapies based on (donor- or recipient-derived) ISDCs in transplantation remain unknown, given that previous studies investigated their effects in vitro, or ex vivo after transplantation. Since once injected, ISDCs are short-lived and transfer Ag to recipient APCs, we assessed the role of recipient DCs by depleting them at the time of ISDC-therapy in a mouse model of cardiac transplantation. The results indicate that, contrary to the accepted paradigm, systemically administered ISDCs reduce the alloresponse and prolong allograft survival, not by themselves, but through conventional DCs (cDCs) of the recipient. These findings raise doubts on the advantages of the currently used ISDC-therapies, since the immune-regulatory properties of the injected ISDC do not seem to be functionally relevant in vivo, and the quiescent/pro-tolerogenic status of cDCs may be compromised in patients with end-stage diseases that require transplantation.


Subject(s)
Dendritic Cells , Transplantation , Adoptive Transfer , Animals , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunosuppressive Agents/administration & dosage , Mice , Mice, Inbred Strains
8.
Mol Hum Reprod ; 18(8): 417-24, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22383544

ABSTRACT

The largest gene cluster of human microRNAs (miRNAs), the chromosome 19 miRNA cluster (C19MC), is exclusively expressed in the placenta and in undifferentiated cells. The precise expression pattern and function of C19MC members are unknown. We sought to profile the relative expression of C19MC miRNAs in primary human trophoblast (PHT) cells and exosomes. Using high-throughput profiling, confirmed by PCR, we found that C19MC miRNAs are among the most abundant miRNAs in term human trophoblasts. Hypoxic stress selectively reduced miR-520c-3p expression at certain time-points with no effect on other C19MC miRNAs. Similarly, differentiation in vitro had a negligible effect on C19MC miRNAs. We found that C19MC miRNAs are the predominant miRNA species expressed in exosomes released from PHT, resembling the profile of trophoblastic cellular miRNA. Predictably, we detected the similar levels of circulating C19MC miRNAs in the serum of healthy pregnant women at term and in women with pregnancies complicated by fetal growth restriction. Our data define the relative expression levels of C19MC miRNAs in trophoblasts and exosomes, and suggest that C19MC miRNAs function in placental-maternal signaling.


Subject(s)
Chromosomes, Human, Pair 19/genetics , Exosomes/metabolism , MicroRNAs/biosynthesis , MicroRNAs/genetics , Trophoblasts/metabolism , Adult , Cell Differentiation , Cells, Cultured , Female , Fetal Growth Retardation/genetics , Humans , MicroRNAs/blood , Placenta/cytology , Pregnancy , Pregnancy Complications/genetics , Pregnancy Trimester, Third
9.
Am J Transplant ; 7(5): 1215-23, 2007 May.
Article in English | MEDLINE | ID: mdl-17331111

ABSTRACT

Posttransplantation lymphoproliferative disorders (PTLD) are life-threatening complications of solid organ transplantation, triggered by EBV infection in chronically immunosuppressed (IS) patients. Our goal is to establish DC-based protocols for adoptive immunotherapy of refractory PTLD, while understanding how the immunosuppressive drug environment may subvert DC-EBV-specific T cell interactions. Type-1 CD8(+) T cells are critical for efficient immune surveillance and control of EBV infection, whereas type-2 or Treg/type-3 responses may provide an environment conductive to disease progression. We have recently reported that chronic IS inhibits DC function in transplant patients. Here, we have analyzed the comparative ability of mature, type-1 polarized DCs (i.e. DC1) generated from quiescent transplant patients or healthy controls, to boost type-1 EBV-specific CD8(+) T cells in vitro. Our results show that unlike healthy controls, where DC1 loaded with MHC class I EBV peptides preferentially reactivate specific type-1 CD8(+) T cells, DC1 generated from transplant patients reactivate EBV-specific CD8(+) T cells that produce both IFN-gamma and IL-10, up-regulate FOXP3 mRNA, and suppress noncognate CD4(+) T-cell proliferation via cell-cell contact. These data support a novel regulatory pathway for anti-EBV T-cell-mediated responses in IS transplant patients, with implications for the design of adoptive immunotherapies in this setting.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Herpesvirus 4, Human/immunology , Lymphocyte Activation/physiology , T-Lymphocytes, Regulatory/physiology , Transplantation Immunology/physiology , Antibodies, Viral/metabolism , Apoptosis/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Case-Control Studies , Cell Communication/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/physiology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Immunosuppressive Agents/pharmacology , Immunotherapy, Adoptive , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Lymphocyte Activation/immunology , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/prevention & control , Organ Transplantation/adverse effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transplantation Immunology/immunology
10.
Am J Transplant ; 6(6): 1297-311, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16686754

ABSTRACT

Tolerance induction against donor allo-antigens (allo-Ag) remains one of the most challenging aspects of transplant immunology. The ability of dendritic cells (DC) to participate in immunity and tolerance makes them an excellent tool for tolerance induction. Here, we employed the immunosuppressive properties of apoptotic cells to deliver simultaneously an inhibitory signal and donor allo-Ag to recipient DC for treatment of allograft rejection. DC that captured apoptotic cells remained immature and activated deficiently anti-donor CD4(+) T cells that were unable to upregulate T-cell activation markers, to secrete IL-2 and IFN-gamma and to survive under homeostatic conditions due to low expression of Bcl-X(L), IL-7R and IL-15R. Administration of donor apoptotic cells decreased the systemic anti-donor T- and B-cell response and prolonged cardiac allograft survival in mice. The effect was donor specific and required the interaction of donor apoptotic cells with recipient quiescent CD8alpha(+) DC. When combined with CD40-CD154-blockade, administration of donor apoptotic cells resulted in indefinite graft survival mediated by generation of regulatory T cells. The use of the inhibitory effects of apoptotic cells on the anti-donor response provides a new approach to treat transplant rejection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Graft Survival/immunology , Heart Transplantation/immunology , Lymphocyte Depletion , Lymphocyte Transfusion , Animals , Apoptosis/radiation effects , Isoantibodies/blood , Lymph Nodes/immunology , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Models, Animal , Spleen/immunology , Transplantation, Homologous/immunology , Ultraviolet Rays
11.
Am J Transplant ; 6(2): 254-61, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16426309

ABSTRACT

Dendritic cells (DC) are professional antigen (Ag) presenting cells (APC) that trigger the anti-donor T-cell response that causes allograft rejection. During the past decade several laboratories have employed in vitro generated DC with tolerogenic potential for prolongation of allograft survival. This minireview describes the development of a second-generation of DC-based strategies for transplantation tolerance based on the delivery in situ of donor allogeneic (allo)-Ag to quiescent DC of graft recipients by means of donor-derived apoptotic cells or exosomes. Donor leukocytes in early apoptosis are rich in allo-Ag, are internalized efficiently by recipient DC in vivo and deliver immunosuppressive signals to DC. Administration (i.v.) of donor apoptotic leukocytes prolongs bone marrow engraftment and cardiac allografts survival in mice by exerting a profound down-regulatory effect on the anti-donor T-cell response. Exosomes are nanovesicles (<100 nm) produced by different cell types, including APC. DC-derived exosomes are rich in major histocompatibility complex (MHC) molecules that can be employed to target DC in situ. Once i.v. injected, exosomes carrying donor MHC molecules are captured by recipient's DC and prolong allograft survival in rodents. The use of the regulatory functions of apoptotic cells and exosomes may be useful tools to develop new strategies for transplantation tolerance.


Subject(s)
Apoptosis/immunology , Dendritic Cells/immunology , Dendritic Cells/transplantation , Transplantation Immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/transplantation , Humans , Mice , Models, Animal , Models, Immunological
12.
Gene Ther ; 10(2): 143-52, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12571643

ABSTRACT

C57BL/10 (B10; H2(b)) bone marrow-derived myeloid dendritic cells (DC) propagated in GM-CSF + IL-4 were transduced with r adenoviral (Ad) vectors encoding either control neomycin-resistance gene (Ad-Neo) or murine IL-4 (Ad-IL-4) on day 5 of culture following CD11c immunomagnetic bead purification. Both Ad-Neo- and Ad-IL-4-transduced DC displayed upregulated surface MHC class II and costimulatory molecules (CD40, CD80, CD86). Ad-IL-4 DC secreted higher levels of bioactive IL-12p70 after CD40 ligation or LPS stimulation than either Ad-Neo or unmodified DC. Only Ad-IL-4 DC produced IL-12p70 in primary MLR, in which they induced augmented proliferative responses of naïve allogeneic C3H/HeJ (C3H; H2(k)) T-cells. Compared with Ad-Neo DC, Ad-IL-4 DC were also more effective in priming naïve allogeneic recipients to exhibit specifically enhanced anti-donor T-cell proliferative and CTL responses. T-cells primed in vivo 7 days previously with Ad-IL-4 DC displayed enhanced secretion of Th2 (IL-4, IL-10) but also higher Th1 cytokine (IFNgamma) production following ex vivo challenge with donor alloAg. Moreover, pretreatment of vascularized heart graft recipients with i.v. Ad-IL-4 DC, 1 week before transplant, significantly accelerated rejection and antagonized the therapeutic effect of anti-CD40L (CD154) mAb. These data contrast markedly with recently reported inhibitory effects of autologous Ad-IL-4 DC on autoimmune inflammatory disease.


Subject(s)
Dendritic Cells/immunology , Genetic Therapy/methods , Graft Rejection/immunology , Interleukin-12/metabolism , Interleukin-4/genetics , Animals , Autoimmune Diseases/immunology , CD40 Antigens/immunology , Genetic Engineering , Heart Transplantation , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , T-Lymphocytes/immunology , Transduction, Genetic/methods , Transplantation, Homologous
13.
Nat Immunol ; 2(12): 1151-8, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11702065

ABSTRACT

Epidermal Langerhans cells (LCs) show extraordinary immunostimulatory capacity and play a key role in the initiation and regulation of immune responses. Studies of LC biology are currently the focus of efforts to engineer immune responses and to better understand the immunopathology of cutaneous diseases. Here we identified and characterized a population of LC precursors that were resident in human skin. These immediate precursors expressed CD14, langerin and functional CCR6. When cultured with transforming growth factor-beta1 alone, they had the potential to differentiate into epidermal LCs; when cultured in the presence of granulocyte macrophage-colony-stimulating factor and interleukin 4 they differentiated into functionally mature dendritic cells. Identification and characterization of these LC precursors provided insight into LC biology and the mechanism(s) through which LCs repopulate the epidermis.


Subject(s)
Dermis/cytology , Langerhans Cells/immunology , Langerhans Cells/ultrastructure , Lipopolysaccharide Receptors/analysis , Skin/immunology , Antigens/metabolism , Cell Differentiation , Cell Division , Cells, Cultured , Chemokine CCL20 , Chemokines, CC/pharmacology , Chemotaxis , Cytokines/pharmacology , Endocytosis , Humans , Immunohistochemistry , Immunophenotyping , Langerhans Cells/classification , Lipopolysaccharide Receptors/immunology , Lymphocyte Activation , Macrophage Inflammatory Proteins/pharmacology , Receptors, CCR6 , Receptors, Chemokine/biosynthesis , Stem Cells/drug effects , Stem Cells/immunology , Stem Cells/ultrastructure
14.
Semin Immunol ; 13(5): 323-35, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11502167

ABSTRACT

Dendritic cells (DC) are professional antigen (Ag)-presenting cells considered traditionally as the passenger leukocytes that, after migration from transplanted tissues, stimulate allospecific naive T cell responses and trigger acute rejection. However, there is recent evidence that, besides their role in central T lymphocyte deletion in the thymus, DC perform a crucial function to induce/maintain peripheral T cell tolerance. This paper outlines conceptual models that try to explain how DC may induce/maintain tolerance. It also considers how such ideas have been implemented recently in an effort to generate tolerogenic DC to induce donor Ag-specific tolerance/ immunosuppression and prolonged allograft survival. These approaches include genetic engineering of donor- or recipient-derived DC to express molecules capable of promoting tolerance to alloAg.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/transplantation , Immune Tolerance/immunology , Animals , Humans
15.
Blood ; 98(5): 1512-23, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11520802

ABSTRACT

Although it is known that dendritic cells (DCs) produce cytokines, there is little information about how cytokine synthesis is regulated during DC development. A range of cytokine mRNA/proteins was analyzed in immature (CD86-) or mature (CD86+) murine bone marrow (BM)- derived DCs. Highly purified, flow-sorted, immature DCs exhibited higher amounts of interleukin-1alpha (IL-1alpha), IL-1beta, tumor necrosis factor-alpha (TNF-alpha), transforming growth factor beta1 (TGF-beta1), and macrophage migration inhibitory factor (MIF) mRNA/protein than mature DCs. After differentiation, DC up-regulated the levels of IL-6 and IL-15 mRNA/protein and synthesized de novo mRNA/protein for IL-12p35, IL-12p40, and IL-18. Although immature BM-derived DCs did not stimulate naive allogeneic T cells, mature DCs elicited a mixed population of T helper (Th) 1 (mainly) and Th2 cells in 3d-mixed leukocyte reactions. CD86+ BM DCs switched to different cytokine patterns according to whether they were terminally differentiated by lipopolysaccharide (LPS) or CD40 ligation. Although both stimuli increased IL-6, IL-12p40, IL-15, and TNF-alpha mRNA/protein levels, only LPS up-regulated transcription of IL-1alpha, IL-1beta, IL-12p35, and MIF genes. Although LPS and CD40 cross-linking increased the T-cell allostimulatory function of BM DCs, only LPS stimulation shifted the balance of naive Th differentiation to Th1 cells, a mechanism dependent on the up-regulation of IL-12p35 and not of IL-23. These results demonstrate that, depending on the stimuli used to terminally mature BM DCs, DCs synthesize a different pattern of cytokines and exhibit distinct Th cell-driving potential.


Subject(s)
CD40 Antigens/physiology , Cytokines/biosynthesis , Dendritic Cells/metabolism , Gene Expression Regulation/drug effects , Lipopolysaccharides/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antigens, CD/biosynthesis , B7-2 Antigen , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , CD40 Antigens/immunology , Cell Differentiation/drug effects , Cells, Cultured/metabolism , Dendritic Cells/drug effects , Endocytosis , Gene Expression Profiling , Immunophenotyping , Interleukin-12/physiology , Interleukin-23 , Interleukin-23 Subunit p19 , Interleukins/biosynthesis , Interleukins/genetics , Interleukins/physiology , Lymphocyte Culture Test, Mixed , Macrophage Migration-Inhibitory Factors/biosynthesis , Macrophage Migration-Inhibitory Factors/genetics , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , RNA, Messenger/biosynthesis , Specific Pathogen-Free Organisms , Th1 Cells/immunology , Th2 Cells/immunology , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
16.
Trends Immunol ; 22(8): 437-42, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11473833

ABSTRACT

Dendritic cells (DCs) are professional antigen-presenting cells (APCs) that play crucial roles as initiators and modulators of adaptive immune responses. Although DC-based vaccines have been utilized successfully to generate cytolytic T-cell activity against tumor antigens (Ags), evidence has accumulated that DCs also have potent capabilities to tolerize T cells in an Ag-specific manner. DCs cultured in the laboratory can suppress auto- or alloimmunity. Current and prospective strategies to promote this inherent tolerogenic potential of DCs might prove to be important for the therapy of transplant rejection and autoimmune diseases.


Subject(s)
Dendritic Cells/immunology , Immune Tolerance , Immunosuppression Therapy/methods , Animals , Autoimmune Diseases/therapy , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Dendritic Cells/cytology , Graft Rejection/therapy , Humans , Immune Tolerance/genetics
17.
J Immunol ; 166(12): 7053-62, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11390449

ABSTRACT

Aspirin is the most commonly used analgesic and antiinflammatory agent. In this study, at physiological concentrations, it profoundly inhibited CD40, CD80, CD86, and MHC class II expression on murine, GM-CSF + IL-4 stimulated, bone marrow-derived myeloid dendritic cells (DC). CD11c and MHC class I expression were unaffected. The inhibitory action was dose dependent and was evident at concentrations higher than those necessary to inhibit PG synthesis. Experiments with indomethacin revealed that the effects of aspirin on DC maturation were cyclooxygenase independent. Nuclear extracts of purified, aspirin-treated DC revealed a decreased NF-kappaB DNA-binding activity, whereas Ab supershift analysis indicated that aspirin targeted primarily NF-kappaB p50. Unexpectedly, aspirin promoted the generation of CD11c+ DC, due to apparent suppression of granulocyte development. The morphological and ultrastructural appearance of aspirin-treated cells was consistent with immaturity. Aspirin-treated DC were highly efficient at Ag capture, via both mannose receptor-mediated endocytosis and macropinocytosis. By contrast, they were poor stimulators of naive allogeneic T cell proliferation and induced lower levels of IL-2 in responding T cells. They also exhibited impaired IL-12 expression and did not produce IL-10 after LPS stimulation. Assessment of the in vivo function of aspirin-treated DC, pulsed with the hapten trinitrobenzenesulfonic acid, revealed an inability to induce normal cell-mediated contact hypersensitivity, despite the ability of the cells to migrate to T cell areas of draining lymphoid tissue. These data provide new insight into the immunopharmacology of aspirin and suggest a novel approach to the manipulation of DC for therapeutic application.


Subject(s)
Aspirin/pharmacology , Dendritic Cells/immunology , Growth Inhibitors/pharmacology , Immunosuppressive Agents/pharmacology , Lymphocyte Activation/drug effects , Myeloid Cells/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/enzymology , Bone Marrow Cells/immunology , Bone Marrow Transplantation , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Movement/drug effects , Cell Movement/immunology , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Dendritic Cells/drug effects , Dendritic Cells/enzymology , Dendritic Cells/transplantation , Dermatitis, Contact/immunology , Dose-Response Relationship, Drug , Endocytosis/drug effects , Endocytosis/immunology , Immunity, Cellular/drug effects , Immunophenotyping , Injections, Subcutaneous , Integrin alphaXbeta2/biosynthesis , Interleukin-10/antagonists & inhibitors , Interleukin-10/metabolism , Interleukin-12/antagonists & inhibitors , Interleukin-12/biosynthesis , Interleukin-2/antagonists & inhibitors , Interleukin-2/biosynthesis , Lymphocyte Culture Test, Mixed , Lymphoid Tissue/immunology , Lymphoid Tissue/pathology , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Cells/drug effects , Myeloid Cells/enzymology , Myeloid Cells/transplantation , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , NF-kappa B p50 Subunit , Prostaglandin-Endoperoxide Synthases/physiology , Signal Transduction/drug effects , Signal Transduction/immunology
18.
J Immunol ; 166(12): 7136-43, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11390459

ABSTRACT

The immunosuppressive and anti-inflammatory cytokine IL-10 inhibits the phenotypic and functional maturation of dendritic cells (DC) and has been reported to confer tolerogenic properties on these important professional APC. Here, we exposed murine bone marrow-derived myeloid DC to either mouse (m) or viral (v) IL-10 early during their in vitro generation in response to GM-CSF and IL-4. Both mIL-10 and vIL-10 down-regulated the expression of CCR7 mRNA determined by RT-PCR, while mIL-10 up-regulated the expression of CCR5 transcripts. These changes in CCR7 and CCR5 expression were associated with inhibition and augmentation, respectively, of DC chemotaxis toward their respective agonists, macrophage inflammatory proteins 3beta and 1alpha, while in vivo homing of DC from peripheral s.c. sites to secondary lymphoid tissue of syngeneic or allogeneic recipients was significantly impaired. Anti-mIL-10R mAb reversed the effects of mIL-10 on CCR expression and restored DC homing ability. Retroviral transduction of mIL-10- and vIL-10-treated DC to overexpress transgenic CCR7 partially restored the cells' lymphoid tissue homing ability in allogeneic recipients. However, CCR7 gene transfer did not reinstate the capacity of IL-10-treated DC to prime host naive T cells for ex vivo proliferative responses or Th1 cytokine (IFN-gamma) production in response to rechallenge with (donor) alloantigen. These findings suggest that in addition to their capacity to subvert DC maturation/function and confer tolerogenic potential on these cells, mIL-10 and vIL-10 regulate DC migratory responses via modulation of CCR expression.


Subject(s)
Adjuvants, Immunologic/physiology , Cell Movement/immunology , Chemotaxis/immunology , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Herpesvirus 4, Human/immunology , Interleukin-10/physiology , Receptors, CCR5/biosynthesis , Receptors, Chemokine/antagonists & inhibitors , Receptors, Chemokine/biosynthesis , Viral Proteins/physiology , Animals , Cell Differentiation/immunology , Cells, Cultured , Chemokine CCL19 , Chemokines, CC/physiology , Dendritic Cells/cytology , Dendritic Cells/immunology , Down-Regulation/immunology , Gene Transfer Techniques , Interleukin-10/metabolism , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Myeloid Cells/cytology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/transplantation , Receptors, CCR5/physiology , Receptors, CCR7 , Receptors, Chemokine/genetics , Receptors, Chemokine/physiology , Receptors, Interleukin/antagonists & inhibitors , Receptors, Interleukin-10
19.
Curr Opin Nephrol Hypertens ; 9(6): 607-13, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11128422

ABSTRACT

Graft-derived 'passenger' dendritic cells have been classically considered as the instigators of acute organ rejection. However, recent advances have revealed that dendritic cells are also involved in the induction/maintenance of peripheral tolerance. This paper briefly reviews the most recent knowledge of the role of donor and recipient dendritic cells during the immune response against allografts, and of the clinical potential of 'tolerogenic' dendritic cells.


Subject(s)
Dendritic Cells/physiology , Transplantation Immunology/physiology , Animals , Antibody Formation/physiology , Chimera , Dendritic Cells/transplantation , Graft Rejection/prevention & control , Graft Survival , Humans , Immune Tolerance/physiology , Transplantation, Homologous/immunology
20.
Mol Ther ; 2(6): 579-87, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11124058

ABSTRACT

To achieve transient transgenesis within specific areas or cell populations in the adult central nervous system (CNS), we have developed a dual adenoviral vector system encoding for cell-type-specific and regulatable transcription units. To achieve combined cell-type-specific transcriptional targeting and inducible expression, we have engineered the expression of the tetracycline-dependent transcriptional elements (1) to be under the transcriptional control of either the astrocyte-specific, glial fibrillary acidic protein (GFAP) (2) or the neuronal specific enolase (NSE) promoter (3) within a dual adenoviral vector system. Cell-type specificity, inducibility, and levels of transgene expression were characterized in vitro in cell lines, and primary neocortical cultures and in the central nervous system (CNS) in vivo, and compared to a powerful pancellular beta-actin/CMV promoter. We demonstrate that the GFAP promoter is able to restrict tetracycline-dependent transgene expression to glial cells in cell lines, primary cultures, and in the CNS in vivo. However, although the NSE promoter did not show neuronal restricted transgene expression in vitro, it did so in the CNS in vivo. Our dual viral system also has provided evidence that an excess of transactivator is needed to achieve maximal transgene expression. Administration of doxycycline completely abrogated transgene expression both in vitro and in vivo. Consequently, our strategy demonstrates that combined cell-type specificity and simultaneous regulation of transgene expression can be obtained in the brain using adenoviral vectors.


Subject(s)
Adenoviridae/genetics , Gene Expression Regulation/genetics , Gene Targeting , Transcription, Genetic/genetics , Transgenes , Animals , Animals, Genetically Modified , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/genetics , Male , Mice , Rats , Rats, Sprague-Dawley , Tetracycline/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...