Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Cells ; 9(4)2020 04 23.
Article in English | MEDLINE | ID: mdl-32340348

ABSTRACT

Mesenchymal stromal cells (MSCs) can generate immunological tolerance due to their regulatory activity in many immune cells. Extracellular vesicles (EVs) release is a pivotal mechanism by which MSCs exert their actions. In this study, we evaluate whether mesenchymal stromal cell extracellular vesicles (MSC-EVs) can modulate T cell response. MSCs were expanded and EVs were obtained by differential ultracentrifugation of the supernatant. The incorporation of MSC-EVs by T cells was detected by confocal microscopy. Expression of surface markers was detected by flow cytometry or CytoFLEX and cytokines were detected by RT-PCR, FACS and confocal microscopy and a miRNA PCR array was performed. We demonstrated that MSC-EVs were incorporated by lymphocytes in vitro and decreased T cell proliferation and Th1 differentiation. Interestingly, in Th1 polarization, MSC-EVs increased Foxp3 expression and generated a subpopulation of IFN-γ+/Foxp3+T cells with suppressive capacity. A differential expression profile of miRNAs in MSC-EVs-treated Th1 cells was seen, and also a modulation of one of their target genes, TGFbR2. MSC-EVs altered the metabolism of Th1-differentiated T cells, suggesting the involvement of the TGF-ß pathway in this metabolic modulation. The addition of MSC-EVs in vivo, in an OVA immunization model, generated cells Foxp3+. Thus, our findings suggest that MSC-EVs are able to specifically modulate activated T cells at an alternative regulatory profile by miRNAs and metabolism shifting.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , Cell Differentiation/genetics , Cell Proliferation/genetics , Extracellular Vesicles/ultrastructure , Forkhead Transcription Factors/metabolism , Glycolysis , Membrane Potential, Mitochondrial , Mesenchymal Stem Cells/ultrastructure , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Signal Transduction/genetics , T-Lymphocytes, Regulatory/cytology
2.
Cells ; 9(4): 1059, 2020.
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: but-ib17629

ABSTRACT

Mesenchymal stromal cells (MSCs) can generate immunological tolerance due to their regulatory activity in many immune cells. Extracellular vesicles (EVs) release is a pivotal mechanism by which MSCs exert their actions. In this study, we evaluate whether mesenchymal stromal cell extracellular vesicles (MSC-EVs) can modulate T cell response. MSCs were expanded and EVs were obtained by differential ultracentrifugation of the supernatant. The incorporation of MSC-EVs by T cells was detected by confocal microscopy. Expression of surface markers was detected by flow cytometry or CytoFLEX and cytokines were detected by RT-PCR, FACS and confocal microscopy and a miRNA PCR array was performed. We demonstrated that MSC-EVs were incorporated by lymphocytes in vitro and decreased T cell proliferation and Th1 differentiation. Interestingly, in Th1 polarization, MSC-EVs increased Foxp3 expression and generated a subpopulation of IFN-gama+/Foxp3+T cells with suppressive capacity. A differential expression profile of miRNAs in MSC-EVs-treated Th1 cells was seen, and also a modulation of one of their target genes, TGFbR2. MSC-EVs altered the metabolism of Th1-differentiated T cells, suggesting the involvement of the TGF-ß pathway in this metabolic modulation. The addition of MSC-EVs in vivo, in an OVA immunization model, generated cells Foxp3+. Thus, our findings suggest that MSC-EVs are able to specifically modulate activated T cells at an alternative regulatory profile by miRNAs and metabolism shifting

3.
Cells, v. 9, n. 4, 1059, abr. 2020
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: bud-3020

ABSTRACT

Mesenchymal stromal cells (MSCs) can generate immunological tolerance due to their regulatory activity in many immune cells. Extracellular vesicles (EVs) release is a pivotal mechanism by which MSCs exert their actions. In this study, we evaluate whether mesenchymal stromal cell extracellular vesicles (MSC-EVs) can modulate T cell response. MSCs were expanded and EVs were obtained by differential ultracentrifugation of the supernatant. The incorporation of MSC-EVs by T cells was detected by confocal microscopy. Expression of surface markers was detected by flow cytometry or CytoFLEX and cytokines were detected by RT-PCR, FACS and confocal microscopy and a miRNA PCR array was performed. We demonstrated that MSC-EVs were incorporated by lymphocytes in vitro and decreased T cell proliferation and Th1 differentiation. Interestingly, in Th1 polarization, MSC-EVs increased Foxp3 expression and generated a subpopulation of IFN-gama+/Foxp3+T cells with suppressive capacity. A differential expression profile of miRNAs in MSC-EVs-treated Th1 cells was seen, and also a modulation of one of their target genes, TGFbR2. MSC-EVs altered the metabolism of Th1-differentiated T cells, suggesting the involvement of the TGF-ß pathway in this metabolic modulation. The addition of MSC-EVs in vivo, in an OVA immunization model, generated cells Foxp3+. Thus, our findings suggest that MSC-EVs are able to specifically modulate activated T cells at an alternative regulatory profile by miRNAs and metabolism shifting

4.
Front Immunol ; 8: 881, 2017.
Article in English | MEDLINE | ID: mdl-28824619

ABSTRACT

Mesenchymal stromal cells (MSCs) are multipotent cells with abilities to exert immunosuppressive response promoting tissue repair. Studies have shown that MSCs can secrete extracellular vesicles (MVs-MSCs) with similar regulatory functions to the parental cells. Furthermore, strong evidence suggesting that MVs-MSCs can modulate several immune cells (i.e., Th1, Th17, and Foxp3+ T cells). However, their precise effect on macrophages (Mϕs) remains unexplored. We investigated the immunoregulatory effect of MVs-MSCs on activated M1-Mϕs in vitro and in vivo using differentiated bone marrow Mϕs and an acute experimental model of thioglycollate-induced peritonitis, respectively. We observed that MVs-MSCs shared surface molecules with MSCs (CD44, CD105, CD90, CD73) and expressed classical microvesicle markers (Annexin V and CD9). The in vitro treatment with MVs-MSCs exerted a regulatory-like phenotype in M1-Mϕs, which showed higher CD206 level and reduced CCR7 expression. This was associated with decreased levels of inflammatory molecules (IL-1ß, IL-6, nitric oxide) and increased immunoregulatory markers (IL-10 and Arginase) in M1-Mϕs. In addition, we detected that MVs-MSCs promoted the downregulation of inflammatory miRNAs (miR-155 and miR-21), as well as, upregulated its predicted target gene SOCS3 in activated M1-Mϕs. In vivo MVs-MSCs treatment reduced the Mϕs infiltrate in the peritoneal cavity inducing a M2-like regulatory phenotype in peritoneal Mϕs (higher arginase activity and reduced expression of CD86, iNOS, IFN-γ, IL-1ß, TNF-α, IL-1α, and IL-6 molecules). This in vivo immunomodulatory effect of MVs-MSCs on M1-Mϕs was partially associated with the upregulation of CX3CR1 in F4/80+/Ly6C+/CCR2+ Mϕs subsets. In summary, our findings indicate that MVs-MSCs can modulate an internal program in activated Mϕs establishing an alternative regulatory-like phenotype.

5.
J Immunol ; 191(5): 2818-28, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23913968

ABSTRACT

In this single-center matched-cohort study, we evaluated the phenotype of repopulating B cells and its correlation with donor-specific anti-HLA Ab development and long-term graft function in 16 renal transplant recipients and 32 age- and gender-matched controls induced with alemtuzumab or basiliximab (Bas)/low-dose rabbit anti-thymocyte globulin (rATG), respectively. Alemtuzumab, but not Bas/rATG, profoundly depleted peripheral B cells in the first 2 mo posttransplantation. Early posttransplant, naive B cells were significantly depleted, whereas Ag-experienced and memory B cells were partially spared. Transitional B cells transiently increased 2 mo posttransplant. At month 6 posttransplant, pregerminal center B cells emerged, a process promoted by increased BAFF serum levels. Thereafter, B cell counts increased progressively, mainly due to expansion of naive B cells. Conversely, Bas/rATG did not modify the B cell phenotype throughout the follow-up period. Alemtuzumab was associated with a higher incidence of de novo DSA compared with Bas/rATG. DSA development was predicted by changes in the B cell compartment and correlated with worse long-term graft function. Thus, alemtuzumab-induced B cell depletion/reconstitution may promote chronic humoral responses against the graft.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal/immunology , B-Lymphocytes/immunology , Graft Rejection/immunology , Histocompatibility Antigens/immunology , Immunosuppressive Agents/immunology , Kidney Transplantation/immunology , Recombinant Fusion Proteins/immunology , Adult , Alemtuzumab , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Antilymphocyte Serum/immunology , Antilymphocyte Serum/therapeutic use , B-Lymphocytes/drug effects , Basiliximab , Cohort Studies , Female , Humans , Immunosuppressive Agents/therapeutic use , Lymphocyte Depletion/methods , Male , Middle Aged , Rabbits , Recombinant Fusion Proteins/therapeutic use
6.
Transpl Int ; 26(9): 867-78, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23738760

ABSTRACT

Bone marrow-derived mesenchymal stromal cells (MSC) have emerged as useful cell population for immunomodulation therapy in transplantation. Moving this concept towards clinical application, however, should be critically assessed by a tailor-made step-wise approach. Here, we report results of the second step of the multistep MSC-based clinical protocol in kidney transplantation. We examined in two living-related kidney transplant recipients whether: (i) pre-transplant (DAY-1) infusion of autologous MSC protected from the development of acute graft dysfunction previously reported in patients given MSC post-transplant, (ii) avoiding basiliximab in the induction regimen improved the MSC-induced Treg expansion previously reported with therapy including this anti-CD25-antibody. In patient 3, MSC treatment was uneventful and graft function remained normal during 1 year follow-up. In patient 4, acute cellular rejection occurred 2 weeks post-transplant. Both patients had excellent graft function at the last observation. Circulating memory CD8(+) T cells and donor-specific CD8(+) T-cell cytolytic response were reduced in MSC-treated patients, not in transplant controls not given MSC. CD4(+) FoxP3(+) Treg expansion was comparable in MSC-treated patients with or without basiliximab induction. Thus, pre-transplant MSC no longer negatively affect kidney graft at least to the point of impairing graft function, and maintained MSC-immunomodulatory properties. Induction therapy without basiliximab does not offer any advantage on CD4(+) FoxP3(+) Treg expansion (ClinicalTrials.gov number: NCT 00752479).


Subject(s)
Graft Rejection/prevention & control , Kidney Transplantation/methods , Mesenchymal Stem Cell Transplantation/methods , Transplantation Immunology , Adult , Antibodies, Monoclonal/adverse effects , Antilymphocyte Serum/therapeutic use , Basiliximab , Clinical Protocols , Female , Humans , Immunosuppressive Agents/adverse effects , Kidney Failure, Chronic/surgery , Male , Recombinant Fusion Proteins/adverse effects , T-Lymphocytes, Regulatory/immunology
7.
Clin J Am Soc Nephrol ; 6(2): 412-22, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20930086

ABSTRACT

BACKGROUND AND OBJECTIVES: Mesenchymal stromal cells (MSCs) abrogate alloimmune response in vitro, suggesting a novel cell-based approach in transplantation. Moving this concept toward clinical application in organ transplantation should be critically assessed. DESIGN, SETTING, PARTICIPANTS & MEASUREMENTS: A safety and clinical feasibility study (ClinicalTrials.gov, NCT00752479) of autologous MSC infusion was conducted in two recipients of kidneys from living-related donors. Patients were given T cell-depleting induction therapy and maintenance immunosuppression with cyclosporine and mycophenolate mofetil. On day 7 posttransplant, MSCs were administered intravenously. Clinical and immunomonitoring of MSC-treated patients was performed up to day 360 postsurgery. RESULTS: Serum creatinine levels increased 7 to 14 days after cell infusion in both MSC-treated patients. A graft biopsy in patient 2 excluded acute graft rejection, but showed a focal inflammatory infiltrate, mostly granulocytes. In patient 1 protocol biopsy at 1-year posttransplant showed a normal graft. Both MSC-treated patients are in good health with stable graft function. A progressive increase of the percentage of CD4+CD25highFoxP3+CD127- Treg and a marked inhibition of memory CD45RO+RA-CD8+ T cell expansion were observed posttransplant. Patient T cells showed a profound reduction of CD8+ T cell activity. CONCLUSIONS: Findings from this study in the two patients show that MSC infusion in kidney transplant recipients is feasible, allows enlargement of Treg in the peripheral blood, and controls memory CD8+ T cell function. Future clinical trials with MSCs to look with the greatest care for unwanted side effects is advised.


Subject(s)
Kidney Failure, Chronic/surgery , Kidney Transplantation , Mesenchymal Stem Cell Transplantation , Adult , Biomarkers/blood , Biopsy , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Creatinine/blood , Drug Therapy, Combination , Feasibility Studies , Humans , Immunohistochemistry , Immunologic Memory , Immunophenotyping , Immunosuppressive Agents/therapeutic use , Kidney Transplantation/adverse effects , Kidney Transplantation/immunology , Living Donors , Male , Mesenchymal Stem Cell Transplantation/adverse effects , Pilot Projects , T-Lymphocytes, Regulatory/immunology , Time Factors , Transplantation Conditioning , Transplantation, Autologous , Treatment Outcome , Young Adult
8.
J Immunol ; 183(7): 4249-60, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19734209

ABSTRACT

Members of the TLR/IL-1R superfamily mediate ischemia/reperfusion injury and initiate immune response in transplanted organs. In this study, we tested the hypothesis that Toll-IL-1R8 (TIR8), a negative regulator of TLR/IL-1R highly expressed in the kidney, modulates immune cell activation underlying kidney rejection. In a mouse model of fully mismatched kidney allotransplantation in which the graft is spontaneously accepted, intragraft Tir8 expression was enhanced compared with naive kidneys. Targeted deletion of Tir8 in the graft exerted a powerful antitolerogenic action leading to acute rejection. Similarly, in a mouse model of kidney graft acceptance induced by costimulation blockade, most Tir8(-/-) grafts were acutely rejected. Despite similar levels of TLR4, IL-1R, and their ligands, the posttransplant ischemia/reperfusion-induced inflammatory response was more severe in Tir8(-/-) than in Tir8(+/+) grafts and was followed by expansion and maturation of resident dendritic cell precursors. In vitro, Tir8(-/-) dendritic cell precursors acquired higher allostimulatory activity and released more IL-6 upon stimulation with a TLR4 ligand and TNF-alpha than Tir8(+/+) cells, which may explain the increased frequency of antidonor-reactive T cells and the block of regulatory T cell formation in recipients of a Tir8(-/-) kidney. Thus, TIR8 acts locally as a key regulator of allogeneic immune response in the kidney. Tir8 expression and/or signaling in donor tissue are envisaged as a novel target for control of innate immunity and amelioration of graft survival.


Subject(s)
Down-Regulation/immunology , Graft Rejection/immunology , Kidney Transplantation/immunology , Membrane Glycoproteins/physiology , Receptors, Interleukin-1/physiology , Acute Disease , Animals , Disease Models, Animal , Down-Regulation/genetics , Graft Rejection/genetics , Graft Rejection/prevention & control , Immune Tolerance/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Membrane Glycoproteins/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred DBA , Mice, Knockout , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/genetics , Reperfusion Injury/genetics , Reperfusion Injury/immunology
9.
J Immunol ; 181(6): 3933-46, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18768848

ABSTRACT

In this study, we investigated whether mesenchymal stem cells (MSC) had immunomodulatory properties in solid organ allotransplantation, using a semiallogeneic heart transplant mouse model, and studied the mechanism(s) underlying MSC tolerogenic effects. Either single (portal vein, day -7) or double (portal vein, day -7 and tail vein, day -1) pretransplant infusions of donor-derived B6C3 MSC in B6 recipients induced a profound T cell hyporesponsiveness and prolonged B6C3 cardiac allograft survival. The protolerogenic effect was abrogated when donor-derived MSC were injected together with B6C3 hematopoietic stem cells (HSC), suggesting that HSC negatively impact MSC immunomodulatory properties. Both the induction (pretransplant) and the maintenance phase (>100 days posttransplant) of donor-derived MSC-induced tolerance were associated with CD4(+)CD25(+)Foxp3(+) Treg expansion and impaired anti-donor Th1 activity. MSC-induced regulatory T cells (Treg) were donor-specific since adoptive transfer of splenocytes from tolerant mice prevented the rejection of fully MHC-mismatched donor-specific secondary allografts but not of third-party grafts. In addition, infusion of recipient-derived B6 MSC tolerized a semiallogeneic B6C3 cardiac allograft, but not a fully MHC-mismatched BALB/c graft, and expanded Treg. A double i.v. pretransplant infusion of recipient-derived MSC had the same tolerogenic effect as the combined intraportal/i.v. MSC infusions, which makes the tolerogenic protocol applicable in a clinical setting. In contrast, single MSC infusions given either peritransplant or 1 day after transplant were less effective. Altogether these findings indicate that MSC immunomodulatory properties require HSC removal, partial sharing of MHC Ags between the donor and the recipient and pretransplant infusion, and are associated with expansion of donor-specific Treg.


Subject(s)
Cell Differentiation/immunology , Graft Survival/immunology , Heart Transplantation/immunology , Mesenchymal Stem Cell Transplantation , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Transplantation Conditioning , Transplantation Tolerance/immunology , Animals , Bone Marrow Cells/immunology , Female , Heart Transplantation/methods , Male , Mesenchymal Stem Cell Transplantation/methods , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Transplantation Conditioning/methods , Transplantation, Heterotopic , Transplantation, Homologous , Transplantation, Isogeneic
10.
Arthritis Rheum ; 56(5): 1629-37, 2007 May.
Article in English | MEDLINE | ID: mdl-17469145

ABSTRACT

OBJECTIVE: To examine whether the cyclin-dependent kinase (CDK) inhibitor seliciclib ameliorates autoimmune nephritis in (NZB x NZW)F(1) mice. METHODS: In experiment 1, NZB x NZW mice received seliciclib (100 mg/kg or 200 mg/kg) or vehicle by gavage, beginning at age 2 months and ending at 8 months of age. In experiment 2, seliciclib (200 mg/kg) was administered alone or combined with low-dose methylprednisolone, starting at age 5 months, when immune complex deposition in the kidney had already occurred. Animals were followed up until all vehicle-treated mice died. In 2 additional groups of NZB x NZW mice treated with seliciclib or vehicle from 2 months of age until 5 months of age, splenocytes were isolated and tested ex vivo for T cell and B cell activity. RESULTS: Seliciclib, given at an early phase of disease, prolonged survival, delayed the onset of proteinuria and renal function impairment, and protected the kidney against glomerular hypercellularity, tubulointerstitial damage, and inflammation. Combining seliciclib with low-dose methylprednisolone in mice with established disease extended the lifespan and limited proteinuria and renal damage more than treatment with either agent alone. Seliciclib limited immunologic signs of disease, reducing glomerular IgG and C3 deposits and levels of serum anti-DNA antibodies. Moreover, it inhibited ex vivo T cell and B cell proliferative responses to polyclonal stimuli. T cell production of interferon-gamma and interleukin-10 and B cell release of IgG2a were reduced by treatment with seliciclib. CONCLUSION: These findings suggest that CDK activity may be a useful target in the treatment of systemic lupus erythematosus. A direct immunomodulatory action of seliciclib on T cells and B cells may be one of the mechanisms underlying the beneficial effects.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Glomerulonephritis/drug therapy , Longevity/drug effects , Lupus Erythematosus, Systemic/drug therapy , Protein Kinase Inhibitors/therapeutic use , Purines/therapeutic use , Animals , Anti-Inflammatory Agents/therapeutic use , Autoimmunity/drug effects , Autoimmunity/physiology , B-Lymphocytes/drug effects , B-Lymphocytes/pathology , Cell Proliferation/drug effects , Cyclin-Dependent Kinases/physiology , Disease Models, Animal , Drug Therapy, Combination , Female , Glomerulonephritis/etiology , Glomerulonephritis/physiopathology , Longevity/physiology , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/physiopathology , Methylprednisolone/therapeutic use , Mice , Mice, Inbred NZB , Mice, Inbred Strains , Protein Kinase Inhibitors/pharmacology , Proteinuria/drug therapy , Proteinuria/etiology , Proteinuria/physiopathology , Purines/pharmacology , Roscovitine , T-Lymphocytes/drug effects , T-Lymphocytes/pathology
11.
Transplantation ; 83(4): 474-84, 2007 Feb 27.
Article in English | MEDLINE | ID: mdl-17318081

ABSTRACT

BACKGROUND: We previously documented that rat bone marrow-derived dendritic cells (DCs), transfected with an adenovirus encoding a dominant negative form of IKK2 (dnIKK2), have impaired allostimulatory capacity and generate CD4 T cells with regulatory function. Here we investigate the potency, the phenotype, and the mechanism of action of dnIKK2-DC-induced regulatory cells and we evaluated their tolerogenic properties in vivo. METHODS: Brown Norway (BN) transfected dnIKK2-DCs were cultured with Lewis (LW) lymphocytes in primary mixed lymphocyte reaction (MLR). CD4 T cells were purified from primary MLR and incubated in secondary coculture MLR with LW lymphocytes. Phenotypic characterization was performed by fluorescence-activated cell sorting and real-time polymerase chain reaction. The tolerogenic potential of CD4 T cells pre-exposed to dnIKK2-DCs was evaluated in vivo in a model of kidney allotransplantation. RESULTS: CD4 T cells pre-exposed to dnIKK2-DCs were CD4CD25 and expressed interleukin (IL)-10, transforming growth factor-beta, interferon-gamma, IL-2, and inducible nitric oxide synthase (iNOS). These cells (dnIKK2-Treg), cocultured (at up to 1:10 ratio) with a primary MLR, suppressed T-cell proliferation to alloantigens. The regulatory effect was cell-to-cell contact-independent since it was also observed in a transwell system. A nitric oxide synthase inhibitor significantly reverted dnIKK2-Treg-mediated suppression, whereas neutralizing antibodies to IL-10 and TGF-beta had no significant effect. DnIKK2-Treg given in vivo to LW rats prolonged the survival of a kidney allograft from BN rats (the donor rat strain used for generating DCs). CONCLUSIONS: DnIKK2-Treg is a unique population of CD4CD25 T cells expressing high levels of iNOS. These cells potently inhibit T-cell response in vitro and induce prolongation of kidney allograft survival in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Dendritic Cells/metabolism , I-kappa B Kinase/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Nitric Oxide Synthase Type II/metabolism , Animals , Cells, Cultured , Coculture Techniques , Graft Survival , I-kappa B Kinase/genetics , Immune Tolerance , Kidney Transplantation/immunology , Phenotype , Rats , Solubility , Transplantation, Homologous/immunology
12.
Transplantation ; 79(9): 1034-9, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15880039

ABSTRACT

BACKGROUND: It was suggested that maintenance of tolerance to organ transplantation may depend on the formation of T regulatory cells. METHODS: Lewis (LW) rats were made tolerant to a Brown Norway kidney by pretransplant donor peripheral blood mononuclear cells (PBMC) infusion. At greater than 90 days after transplantation, lymph node cells (LN) and graft-infiltrating leukocytes (GIL) alloreactivity was tested in mixed lymphocyte reaction (MLR), coculture, and transwell experiments. GIL phenotype was analyzed by FACS. mRNA expression of cytokines and other markers was analyzed on CD4+ T cells from LN. The tolerogenic potential of tolerant cells in vivo was evaluated by adoptive transfer. RESULTS: Tolerant LN cells showed a reduced proliferation against donor stimulators but a normal anti-third-party alloreactivity. In coculture, these cells inhibited antidonor but not antithird-party reactivity of naïve LN cells. Interleukin (IL)-10 and FasL mRNA expression was up-regulated in tolerant CD4+ T cells, but an anti-IL-10 monoclonal antibody (mAb) only partially reversed their inhibitory effect. Immunoregulatory activity was concentrated in the CD4+ CD25+ T-cell subset. In a transwell system, tolerant T cells inhibited a naïve MLR to a lesser extent than in a standard coculture. Regulatory cells transferred tolerance after infusion into naïve LW recipients. CD4+ T cells isolated from tolerized grafts were hyporesponsive to donor stimulators and suppressed a naïve MLR against donor antigens. CONCLUSIONS: Donor-specific regulatory T cells play a role in tolerance induction by donor PBMC infusion. Regulatory activity is concentrated in the CD4+ CD25+ subset and requires cell-to-cell contact. Regulatory CD4+ T cells accumulate in tolerized kidney grafts where they could exert a protective function against host immune response.


Subject(s)
Lymphocyte Transfusion , T-Lymphocyte Subsets/immunology , Transplantation Tolerance/physiology , Animals , CD4-Positive T-Lymphocytes/immunology , Graft Rejection/immunology , Humans , Kidney Transplantation/immunology , Lymph Nodes/immunology , Lymphocyte Culture Test, Mixed , Male , Models, Animal , Polymerase Chain Reaction , Rats , Rats, Inbred BN , Rats, Inbred Lew , Transplantation, Homologous/immunology
13.
Transplantation ; 79(9): 1056-61, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15880043

ABSTRACT

BACKGROUND: Immature dendritic cells (DC), characterized by low expression of both major histocompatibility complex class II antigens and co-stimulatory molecules, can be instrumental in the induction of peripheral tolerance. Because nuclear factor (NF)-kappa B is central to DC maturation, the authors engineered DC with an adenoviral vector (Adv) encoding for a kinase-defective dominant negative form of IKK2 (dnIKK2) to block NF-kappa B activation and inhibit DC maturation. METHODS: DC were obtained by culturing bone marrow from Brown Norway (BN) rats with granulocyte-macrophage colony-stimulating factor and interleukin-4 for 11 days. To block NF-kappa B activation, at day 9, cells were transfected with AdV-dnIKK2. At day 11, cells were used as stimulators in primary mixed leukocyte reaction (MLR) with naive Lewis rat lymphocytes as responders. CD4+ T cells were purified from primary MLR and tested in secondary MLR with allogeneic mature DC and in co-culture MLR with naive lymphocytes. The tolerogenic potential of dnIKK2-DC was evaluated in vivo in a model of rat kidney allotransplantation. RESULTS: DnIKK2-DC were immature and lacked any allostimulatory activity. T cells preexposed to allogeneic dnIKK2-DC were hyporesponsive to a secondary stimulation with mature DC and acquired potent regulatory properties, inhibiting naive T-cell proliferation toward allogeneic stimuli. Pretransplant infusion of allogeneic donor dnIKK2-DC prolonged the survival of a kidney allograft from the same allogeneic donor, without the need for immunosuppressive therapy. CONCLUSIONS: Allogeneic DC, rendered immature by dnIKK2 transfection, induce in vitro differentiation of naive T cells into CD4+ T-regulatory cells, effective at low ratios with target cells, rendering them applicable for cellular therapy of immune-mediated abnormalities and for preventing transplant rejection.


Subject(s)
Bone Marrow Transplantation/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Kidney Transplantation/immunology , Protein Serine-Threonine Kinases/genetics , Adenoviridae , Animals , Bone Marrow Cells/immunology , Cell Differentiation , Genetic Engineering/methods , Genetic Vectors , I-kappa B Kinase , Lymphocyte Culture Test, Mixed , Models, Animal , Rats , Rats, Inbred BN , Rats, Inbred Lew , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transfection/methods , Transplantation, Homologous
14.
J Am Soc Nephrol ; 12(12): 2815-2826, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11729252

ABSTRACT

This study found that pretransplant infusion of donor peripheral blood leukocytes, either total leukocytes (peripheral blood leukocytes) or peripheral blood mononuclear cells (PBMC), under appropriate immunomodulating conditions was more effective than donor bone marrow (BM) in prolonging the survival of rats that received kidney grafts. A higher percentage of MHCII(+) cells was found in donor PBMC than in BM cells, and depletion of MHCII(+) cells from donor PBMC abolished their tolerogenic potential. By the analysis of microchimerism in rats infused with donor cells and killed at different time points thereafter, the better tolerogenic potential of leukocyte infusion related to a higher capability of these cells to engraft the recipient thymus. PCR analysis on OX6-immunopurified cells revealed the presence of donor MHCII(+) cells in the thymus of these animals. The role of intrathymic microchimerism was reinforced by findings that thymectomy at the time of transplant prevented tolerance induction by donor leukocytes. Donor DNA was found in the thymus of most long-term graft animals that survived, but in none of those that rejected their grafts. The presence of intrathymic microchimerism correlated with graft survival, and microchimerism in other tissues was irrelevant. PCR analysis of DNA from thymic cell subpopulations revealed the presence of donor MHCII(+) cells in the thymus of long-term surviving animals. Thus, in rats, donor leukocyte infusion is better than donor BM for inducing graft tolerance, defined by long-term graft survival, donor-specific T cell hyporesponsiveness, and reduced interferon gamma production. This effect appears to occur through migration of donor MHCII(+) cells in the host thymus.


Subject(s)
Kidney Transplantation , Thymus Gland/physiology , Transplantation Chimera , Transplantation Conditioning , Animals , Bone Marrow Cells/immunology , Bone Marrow Transplantation , Graft Survival , Histocompatibility Antigens Class II/analysis , Lymphocyte Culture Test, Mixed , Lymphocyte Transfusion , Male , Monocytes/immunology , Monocytes/transplantation , Rats , Rats, Inbred BN , Rats, Inbred Lew , Rats, Inbred WF , Thymectomy , Tissue Donors , Transplantation Tolerance , Transplantation, Homologous , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...