Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 207
Filter
1.
Nat Commun ; 13(1): 3121, 2022 06 03.
Article in English | MEDLINE | ID: mdl-35660734

ABSTRACT

Regulatory B cells (Breg) are a heterogenous population with immune-modulating functions. The rarity of human IL-10+ Breg makes translational studies difficult. Here we report ex vivo expansion of human B cells with in vivo regulatory function (expBreg). CD154-stimulation of human CD19+ B cells drives >900-fold expansion of IL-10+ B cells that is maintained in culture for 14 days. Whilst expBreg-mediated suppressive function is partially dependent on IL-10 expression, CRISPR-mediated gene deletions demonstrate predominant roles for TIM-1 and CD154. TIM-1 regulates STAT3 signalling and modulates downstream suppressive function. In a clinically relevant humanised mouse model of skin transplantation, expBreg prolongs human allograft survival. Meanwhile, CD19+CD73-CD25+CD71+TIM-1+CD154+ Breg cells are enriched in the peripheral blood of human donors with cutaneous squamous cell carcinoma (SCC). TIM-1+ and pSTAT3+ B cells are also identified in B cell clusters within histological sections of human cutaneous SCC tumours. Our findings thus provide insights on Breg homoeostasis and present possible targets for Breg-related therapies.


Subject(s)
B-Lymphocytes, Regulatory , Carcinoma, Squamous Cell , Skin Neoplasms , Animals , Antigens, CD19/metabolism , Carcinoma, Squamous Cell/metabolism , Humans , Interleukin-10/metabolism , Lymphocyte Activation , Mice , STAT3 Transcription Factor/metabolism , Skin Neoplasms/metabolism
2.
Am J Transplant ; 16(5): 1394-407, 2016 05.
Article in English | MEDLINE | ID: mdl-26603026

ABSTRACT

Assays designed to select transplant recipients for immunosuppression withdrawal have met with limited success, perhaps because they measure events downstream of T cell-alloantigen interactions. Using in vitro time-lapse microscopy in a mouse transplant model, we investigated whether transplant outcome would result in changes in the proportion of CD4(+) T cells forming prolonged interactions with donor dendritic cells. By blocking CD4-MHC class II and CD28-B7 interactions, we defined immunologically relevant interactions as those ≥500 s. Using this threshold, T cell-dendritic cell (T-DC) interactions were examined in rejection, tolerance and T cell control mediated by regulatory T cells. The frequency of T-DC contacts ≥500 s increased with T cells from mice during acute rejection and decreased with T cells from mice rendered unresponsive to alloantigen. Regulatory T cells reduced prolonged T-DC contacts. Importantly, this effect was replicated with human polyclonally expanded naturally occurring regulatory T cells, which we have previously shown can control rejection of human tissues in humanized mouse models. Finally, in a proof-of-concept translational context, we were able to visualize differential allogeneic immune synapse formation in polyclonal CD4(+) T cells using high-throughput imaging flow cytometry.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Graft Rejection/diagnosis , Heart Transplantation/adverse effects , Immunological Synapses/immunology , Isoantigens/immunology , T-Lymphocytes, Regulatory/immunology , Time-Lapse Imaging/methods , Animals , Dendritic Cells/immunology , Graft Rejection/etiology , Graft Survival/immunology , Immune Tolerance/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA
3.
Am J Transplant ; 14(11): 2617-22, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25250867

ABSTRACT

Malignancy is an important cause of death in transplant recipients. Cutaneous squamous cell carcinoma (cSCC) causes significant morbidity and mortality as 30% of transplant recipients will develop cSCC within 10 years of transplantation. Previously we have shown that high numbers of regulatory T cells (Tregs) are associated with the development of cSCC in kidney transplant recipients (KTRs). Demethylation analysis of the Treg-specific demethylated region (TSDR) provides a more accurate association with cSCC risk after transplantation. Age, gender and duration of immunosuppression matched KTRs with (n=32) and without (n=27) cSCC, were re-analyzed for putative clinical and immunological markers of cancer risk. The proportion of FOXP3+ CD4+ cells was higher in the population with a previous SCC. Major T cell subsets remained stable over time; although B cell, CD8 and CD4 subpopulations demonstrated age-related changes. TSDR methylation analysis allowed clarification of Treg numbers, enhancing the association of high Treg levels in KTRs with cSCC compared to the cSCC-free cohort. These data validate and expand on previous findings in long-term KTRs, and show that immune markers remain stable over time. TSDR demethylation analysis provides a more accurate biomarker of cancer posttransplantation.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Squamous Cell/blood , DNA Methylation , Transplantation/adverse effects , Adult , Aged , Aged, 80 and over , Female , Humans , Immunosuppressive Agents/administration & dosage , Male , Middle Aged
4.
Am J Transplant ; 14(4): 779-87, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24580828

ABSTRACT

The impact of donor-specific HLA alloantibodies (DSA) on short- and long-term liver transplant outcome is not clearly defined. While it is clear that not all levels of allosensitization produce overt clinical injury, and that liver allografts possess some degree of alloantibody resistance, alloantibody-mediated adverse consequences are increasingly being recognized. To better define the current state of this topic, we assembled experts to provide insights, explore controversies and develop recommendations for future research on the consequences of DSA in liver transplantation. This article summarizes the proceedings of this inaugural meeting. Several insights emerged. Acute antibody-mediated rejection (AMR), although rarely diagnosed, is increasingly understood to overlap with T cell-mediated rejection. Isolated liver allograft recipients are at increased risk of early allograft immunologic injury when preformed DSA are high titer and persist posttransplantation. Persons who undergo simultaneous liver-kidney transplantation are at risk of renal AMR when Class II DSA persist posttransplantation. Other under-appreciated DSA associations include ductopenia and fibrosis, plasma cell hepatitis, biliary strictures and accelerated fibrosis associated with recurrent liver disease. Standardized DSA testing and diagnostic criteria for both acute and chronic AMR are needed to distil existing associations into etiological processes in order to develop responsive therapeutic strategies.


Subject(s)
Graft Rejection/immunology , HLA Antigens/immunology , Isoantibodies/immunology , Liver Diseases/immunology , Liver Transplantation , Practice Guidelines as Topic , Tissue Donors , Humans , Liver Diseases/surgery , Prognosis , Research Report
5.
Am J Transplant ; 14(4): 750-63, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24592900

ABSTRACT

Owing to the adverse effects of immunosuppression and an inability to prevent chronic rejection, there is a pressing need for alternative strategies to control alloimmunity. In three decades, regulatory T cells (Tregs) have evolved from a hypothetical mediator of adoptively transferred tolerance to a well-defined population that can be expanded ex vivo and returned safely to patients in clinical trials. Herein, we review the historical developments that have permitted these advances and the current status of clinical trials examining Tregs as a cellular therapy in transplantation. We conclude by discussing the critical unanswered questions that face this field in the coming years.


Subject(s)
Cell- and Tissue-Based Therapy , Organ Transplantation , T-Lymphocytes, Regulatory/immunology , Transplantation Tolerance/immunology , Humans , Prognosis
6.
Am J Transplant ; 13(7): 1880-90, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23763435

ABSTRACT

Assessment of donor-specific alloreactive memory/effector T cell responses using an IFN-γ Elispot assay has been suggested to be a novel immune-monitoring tool for evaluating the cellular immune risk in renal transplantation. Here, we report the cross-validation data of the IFN-γ Elispot assay performed within different European laboratories taking part of the EU RISET consortium. For this purpose, development of a standard operating procedure (SOP), comparisons of lectures of IFN-γ plates assessing intra- and interlaboratory assay variability of allogeneic or peptide stimuli in both healthy and kidney transplant individuals have been the main objectives. We show that the use of a same SOP and count-settings of the Elispot bioreader allow low coefficient variation between laboratories. Frozen and shipped samples display slightly lower detectable IFN-γ frequencies than fresh samples. Importantly, a close correlation between different laboratories is obtained when measuring high frequencies of antigen-specific primed/memory T cell alloresponses. Interestingly, significant high donor-specific alloreactive T cell responses can be similarly detected among different laboratories in kidney transplant patients displaying histological patterns of acute T cell mediated rejection. In conclusion, assessment of circulating alloreactive memory/effector T cells using an INF-γ Elispot assay can be accurately achieved using the same SOP, Elispot bioreader and experienced technicians in kidney transplantation.


Subject(s)
Enzyme-Linked Immunospot Assay/methods , Graft Rejection/immunology , Immunity, Cellular/immunology , Immunologic Memory , Interferon-gamma/immunology , Kidney Transplantation/immunology , Enzyme-Linked Immunosorbent Assay/methods , Humans , T-Lymphocytes/immunology
7.
Am J Transplant ; 13(7): 1655-64, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23750800

ABSTRACT

The engineered Fc-nonbinding (crystallizable fragment-nonbinding) CD3 antibody has lower mitogenicity and a precise therapeutic window for disease remission in patients with type 1 diabetes. Before anti-CD3 can be considered for use in transplantation, the most effective timing of treatment relative to transplantation needs to be elucidated. In this study anti-CD3F(ab')2 fragments or saline were administered intravenously for 5 consecutive days (early: d1-3 or delayed: d3-7) to mice transplanted with a cardiac allograft (H2(b)-to-H2(k); d0). Survival of allografts was prolonged in mice treated with the early protocol (MST = 48 days), but most were rejected by d100. In contrast, in mice treated with the delayed protocol allografts continued to survive long term. The delayed protocol significantly inhibited donor alloreactivity at d30 as compared to the early protocol. A marked increase in Foxp3(+) T cells (50.3 ± 1.6%) infiltrating the allografts in mice treated with the delayed protocol was observed (p < 0.0001 vs. early (24.9 ± 2.1%)) at d10; a finding that was maintained in the accepted cardiac allografts at d100. We conclude that the timing of treatment with anti-CD3 therapy is critical for inducing long-term graft survival. Delaying administration effectively inhibits the alloreactivity and promotes the dominance of intragraft Foxp3(+) T cells allowing long-term graft acceptance.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , CD3 Complex/drug effects , CD4-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/immunology , Graft Rejection/prevention & control , Graft Survival/drug effects , Heart Transplantation/immunology , Repressor Proteins/immunology , Animals , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/pathology , Cells, Cultured , Disease Models, Animal , Enzyme-Linked Immunospot Assay , Female , Flow Cytometry , Follow-Up Studies , Forkhead Transcription Factors/drug effects , Graft Rejection/immunology , Graft Rejection/pathology , Graft Survival/immunology , Heart Transplantation/pathology , Immunohistochemistry , Immunosuppression Therapy/methods , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Repressor Proteins/drug effects , Time Factors , Transplantation, Homologous
8.
Am J Transplant ; 12(8): 2008-16, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22500984

ABSTRACT

Regulatory T cells (T(reg)) are currently being tested in clinical trials as a potential therapy in cell and solid organ transplantation. The immunosuppressive drug rapamycin has been shown to preferentially promote T(reg) expansion. Here, we hypothesized that adjunctive rapamycin therapy might potentiate the ability of ex vivo expanded human T(reg) to inhibit vascular allograft rejection in a humanized mouse model of arterial transplantation. We studied the influence of combined treatment with low-dose rapamycin and subtherapeutic T(reg) numbers on the development of transplant arteriosclerosis (TA) in human arterial grafts transplanted into immunodeficient BALB/cRag2(-/-) Il2rg(-/-) mice reconstituted with allogeneic human peripheral blood mononuclear cell. In addition, we assessed the effects of the treatment on the proliferation and apoptosis of naïve/effector T cells. The combined therapy efficiently suppressed T-cell proliferation in vivo and in vitro. Neointima formation in the human arterial allografts was potently inhibited compared with each treatment alone. Interestingly, CD4(+) but not CD8(+) T lymphocytes were sensitive to T(reg) and rapamycin-induced apoptosis in vitro. Our data support the concept that rapamycin can be used as an adjunctive therapy to improve efficacy of T(reg)-based immunosuppressive protocols in clinical practice. By inhibiting TA, T(reg) and rapamycin may prevent chronic transplant dysfunction and improve long-term allograft survival.


Subject(s)
Arteriosclerosis/prevention & control , Sirolimus/pharmacology , T-Lymphocytes, Regulatory/drug effects , Animals , Apoptosis , Arteries/transplantation , Arteriosclerosis/etiology , Cell Proliferation , Dose-Response Relationship, Drug , Humans , Mice , Mice, Inbred BALB C , Transplantation/adverse effects
9.
Am J Transplant ; 12(7): 1784-92, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22420490

ABSTRACT

In organ transplantation, the composition of the B-cell compartment is increasingly identified as an important determinant for graft outcome. Whereas naïve and transitional B cells have been associated with long-term allograft survival and operational tolerance, memory B cells have been linked to decreased allograft survival. Alemtuzumab induction therapy effectively depletes B cells, but is followed by rapid repopulation up to levels exceeding base line. The characteristics of the repopulating B cells are currently unknown. We studied the phenotypic and functional characteristics of B cells longitudinally in 19 kidney transplant recipients, before and at 6, 9 and 12 months after alemtuzumab induction therapy. A transient increase in transitional B cells and cells with phenotypic characteristics of regulatory B cells, as well as a long-term dominance in naïve B cells was found in alemtuzumab-treated kidney transplant recipients, which was not influenced by conversion from tacrolimus to sirolimus. At all time-points after treatment, B cells showed unaltered proliferative and IgM-producing capacity as compared to pretransplant samples, whereas the ability to produce IgG was inhibited long-term. In conclusion, induction therapy with alemtuzumab results in a long-term shift toward naïve B cells with altered phenotypic and functional characteristics.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , B-Lymphocytes/cytology , Immunosuppressive Agents/administration & dosage , Kidney Transplantation , Alemtuzumab , B-Lymphocytes/immunology , Cell Division , Flow Cytometry , Humans
10.
Am J Transplant ; 12(3): 590-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22070799

ABSTRACT

It is well established that iNKT cells can be activated by both exogenous and a limited number of endogenous glycolipids. However, although iNKT cells have been implicated in the immune response to transplanted organs, the mechanisms by which iNKT cells are activated in this context remain unknown. Here we demonstrate that iNKT cells are not activated by allogeneic cells per se, but expand, both in vitro and in vivo, in the presence of a concomitant conventional T-cell response to alloantigen. This form of iNKT activation was found to occur independently of TCR-glycolipid/CD1d interactions but rather was a result of sequestration of IL-2 produced by conventional alloreactive T cells. These results show for the first time that IL-2, produced by activated conventional T cells, can activate iNKT cells independently of glycolipid/CD1d recognition. Therefore, we propose that the well-documented involvement of iNKT cells in autoimmunity, the control of cancer as well as following transplantation need not involve recognition of endogenous or exogenous glycolipids but alternatively may be a consequence of specific adaptive immune responses.


Subject(s)
Antigen Presentation/immunology , Bystander Effect , Interleukin-2/immunology , Lymphocyte Activation/immunology , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Skin Transplantation/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD1d/immunology , Apoptosis , Blotting, Western , Cell Proliferation , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Graft Rejection/immunology , Graft Survival/immunology , Interleukin-2/metabolism , Isoantigens/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Natural Killer T-Cells/cytology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transplantation Tolerance/immunology
11.
Transplant Proc ; 43(6): 2135-6, 2011.
Article in English | MEDLINE | ID: mdl-21839214

ABSTRACT

T Regulatory cells (Treg) play an important role in the induction and maintenance of immunological tolerance to self and alloantigens. Recent findings in experimental transplant models have demonstrated that Treg can control acute and delayed allograft rejection. Preclinical attempts to use Treg as a cellular therapy have been successfully undertaken demonstrating the safety and feasibility of such treatment, suggesting that they have therapeutic potential.


Subject(s)
Graft Rejection/prevention & control , Graft Survival , Organ Transplantation/adverse effects , T-Lymphocytes, Regulatory/immunology , Transplantation Immunology , Animals , Graft Rejection/immunology , Humans , Immune Tolerance , Isoantigens/immunology
12.
Placenta ; 32 Suppl 4: S285-90, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21575989

ABSTRACT

The International Placenta Stem Cell Society (IPLASS) was founded in June 2010. Its goal is to serve as a network for advancing research and clinical applications of stem/progenitor cells isolated from human term placental tissues, including the amnio-chorionic fetal membranes and Wharton's jelly. The commitment of the Society to champion placenta as a stem cell source was realized with the inaugural meeting of IPLASS held in Brescia, Italy, in October 2010. Officially designated as an EMBO-endorsed scientific activity, international experts in the field gathered for a 3-day meeting, which commenced with "Meet with the experts" sessions, IPLASS member and board meetings, and welcome remarks by Dr. Ornella Parolini, President of IPLASS. The evening's highlight was a keynote plenary lecture by Dr. Diana Bianchi. The subsequent scientific program consisted of morning and afternoon oral and poster presentations, followed by social events. Both provided many opportunities for intellectual exchange among the 120 multi-national participants. This allowed a methodical and deliberate evaluation of the status of placental cells in research in regenerative and reparative medicine. The meeting concluded with Dr. Parolini summarizing the meeting's highlights. This further prepared the fertile ground on which to build the promising potential of placental cell research. The second IPLASS meeting will take place in September 2012 in Vienna, Austria. This meeting report summarizes the thought-provoking lectures delivered at the first meeting of IPLASS.


Subject(s)
Fetal Stem Cells/cytology , Placenta/cytology , Female , Fetus , Humans , Pregnancy
13.
Am J Transplant ; 10(1): 69-80, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19889125

ABSTRACT

CD4+CD25+Foxp3+ regulatory T cells (Tregs) play a key role in the induction and maintenance of peripheral tolerance. Rapid and transient production of IFN-gamma by Tregs from mice tolerized to alloantigen in vivo has been shown to be critical for their regulatory function. This IFN-gamma has the potential to affect the function of cells present in the same local microenvironment as the Tregs, including the Tregs themselves. Here we investigated the mechanism by which IFN-gamma produced by Tregs triggered signaling pathways in alloantigen reactive Tregs themselves thereby influencing their function in vivo. We show that IFN-gamma production and STAT1 activation was increased, while STAT1-dependent PKB/AKT activation was downregulated in alloantigen reactive Tregs. Further, the activation of STAT1 was blocked in IFN-gamma receptor deficient as well as IFN-gamma-deficient Tregs, suggesting that IFN-gamma produced by the alloantigen reactive Tregs might act in an autocrine manner to induce STAT1 activation. Importantly, STAT1-deficient Tregs failed to control allograft rejection in vivo. Overall, these findings suggest that the IFN-gamma-induced STAT1-PKB/AKT signaling pathway plays a key role in upregulating the ability of alloantigen reactive Tregs to control graft rejection in vivo.


Subject(s)
Interferon-gamma/biosynthesis , Isoantigens/administration & dosage , Proto-Oncogene Proteins c-akt/metabolism , STAT1 Transcription Factor/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Graft Rejection/immunology , Immunosuppression Therapy , Interferon-gamma/deficiency , Interferon-gamma/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Models, Immunological , Receptors, Interferon/metabolism , STAT1 Transcription Factor/deficiency , STAT1 Transcription Factor/genetics , Signal Transduction/immunology , Skin Transplantation/immunology , Transplantation, Homologous , Interferon gamma Receptor
14.
Am J Transplant ; 8(2): 338-47, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18211507

ABSTRACT

Alemtuzumab (CAMPATH-1H) is a depleting agent introduced recently in transplantation and often used with reduced maintenance immunosuppression. In the current study we investigated the immune response of 13 kidney allograft recipients treated with alemtuzumab followed by weaned immunosuppression with reduced dose of mycophenolate mofetil (MMF) and tacrolimus. Tacrolimus was switched to sirolimus at 6 months and MMF withdrawn at 12 months after transplantation. We found that after alemtuzumab induction the recovery of CD8(+) T cells was much faster than that of CD4(+) T cells. It was complete 6 months posttransplant while CD4(+) T cells did not fully recover even 15 months posttransplant. Repopulating CD8(+) T cells were mainly of immunosenescent CD28(-)CD8(+) phenotype. In a series of in vitro experiments we showed that CD28(-)CD8(+) T cells might suppress proliferation of CD4(+) T cells. There were three successfully treated acute rejections during the study (first at +70 day, two others +12 months) that occurred in patients with the lowest level of CD28(-)CD8(+) T cells. We hypothesize that expanded CD28(-)CD8(+) T cells might compete for 'immune space' with CD4(+) T cells suppressing their proliferation and therefore delaying CD4(+) T-cells recovery. This delay might be associated with the clinical outcome as CD4(+) T cells, notably CD4(+) T effector memory cells, were shown to be associated with rejection.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , CD28 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Immunosuppression Therapy/methods , Immunosuppressive Agents/therapeutic use , Kidney Transplantation/immunology , Lymphocyte Depletion , T-Lymphocytes, Regulatory/immunology , Alemtuzumab , Antibodies, Monoclonal, Humanized , Antigens, CD/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Interleukins/blood , Lymphocyte Activation , Polymerase Chain Reaction
15.
Transplant Proc ; 37(1): 37-9, 2005.
Article in English | MEDLINE | ID: mdl-15808539

ABSTRACT

UNLABELLED: Recent evidence suggests that CD4+CD25+ regulatory T cells (Tregs) affect immune responses, including those to alloantigens in organ transplants. We have followed a group of liver allograft recipients with good liver graft function who have been weaned off immunosuppression (IS). The purpose of this study was to determine whether Tregs contributed functionally to the mechanisms of graft acceptance. MATERIAL AND METHODS: The functional assay used peripheral blood obtained from LTx recipients free of immunosuppression. The Whole population of CD4+ T cells and the CD4+ T cells depleted of CD4+CD25 high cells were tested for proliferation against donor versus third party stimulators. Moreover to determine the antigen-specificity of the Tregs, serially diluted numbering of CD4+CD25+ T cells were co-cultured with CD4+CD25- T cells. The proliferation responses were examined toward donor versus third party stimulators. RESULT: CD4+ T cells from all LTx recipients off immunosuppression showed hyporesponsiveness to the donor but not to third party stimulators. However, even after depletion of the CD4+CD25 high population, the cells continued to be hyporesponsive toward the donor. In four out of five cases, the suppression exhibited by CD4+CD25+ T cells was more specific for the donor. DISCUSSION: These findings suggest that donor alloantigen specific regulation by Tregs is one of multiple mechanisms that may contribute to the maintenance of liver graft survival in the absence of immunosuppression.


Subject(s)
CD4 Antigens/immunology , Liver Transplantation/immunology , Living Donors , Receptors, Interleukin-2/immunology , T-Lymphocytes/immunology , Transplantation Tolerance , Antigens, CD/immunology , Child , Humans , Lymphocyte Culture Test, Mixed , Reference Values
16.
Scand J Immunol ; 57(4): 333-41, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12662296

ABSTRACT

Anti-CD4 monoclonal antibodies are potential therapeutic agents for the prevention of autoimmune disease and treatment of rejection after organ transplantation and are capable of both restoring tolerance to self-antigens and inducing tolerance to antigens introduced under the cover of the antibody therapy in vivo. Tolerance to donor alloantigens can be induced in vivo by administering donor alloantigen in combination with either depleting (YTA 3.1) or nondepleting (YTS 177) anti-CD4, 28 days before heart transplantation in the mouse. The effect of anti-CD4 on proximal T-cell receptor (TCR) signalling pathways and proliferation was investigated in vitro and in vivo in the presence and absence of YTA 3.1 or YTS 177. Anti-CD4 was found to perturb proximal signalling events upon TCR/CD3 ligation, resulting in reduced tyrosine phosphorylation of Zap-70 and LAT (linker for activation of T cells) and reduced association of tyrosine-phosphorylated LAT with lck. This ultimately resulted in severely reduced proliferation of the responding CD4+ T cells. The signalling profile of the anti-CD4-treated cells resembled that of anergic T cells. This could be a result of a common mechanism involving perturbation in the formation of the central supramolecular activation cluster of the immunological synapse by impaired recruitment of CD4 and CD28, thereby resulting in severely reduced lck activation.


Subject(s)
Adaptor Proteins, Signal Transducing , Antibodies, Monoclonal/immunology , CD4-Positive T-Lymphocytes/immunology , Carrier Proteins/metabolism , Membrane Proteins , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Receptors, Antigen, T-Cell/immunology , Animals , Antibodies, Monoclonal/pharmacology , Blotting, Western , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Carrier Proteins/immunology , Cell Division/immunology , Female , Ionomycin/immunology , Lymphocyte Activation/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Phosphoproteins/immunology , Phosphorylation , Precipitin Tests , Protein-Tyrosine Kinases/immunology , Receptors, Interleukin-2/immunology , Signal Transduction/immunology , Tetradecanoylphorbol Acetate/pharmacology , ZAP-70 Protein-Tyrosine Kinase
17.
Gene Ther ; 9(3): 220-6, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11859426

ABSTRACT

We investigated the delivery of a donor-specific MHC class I gene, H-2K(b), using a newly constructed replication-defective recombinant adenovirus (AdSV40K(b)) to recipient tissue before transplantation as a means of inducing donor-specific immunological unresponsiveness. AdSV40K(b) was able to transduce both a fibroblast cell line and freshly isolated bone marrow cells (BMCs) resulting in cell surface expression of H2-K(b) protein. Intravenous infusion of AdSV40K(b)-transduced syngeneic CBA/Ca (H-2(k)) BMCs into CBA recipient mice treated with an anti-CD4 monoclonal antibody 27 days before transplantation of a fully MHC-mismatched, C57BL/10 (H-2K(b+)), cardiac allograft resulted in significant long-term graft survival when compared with mice receiving the same dose of syngeneic BMCs transduced with a control adenovirus, AdRSVbetagal. Despite the induction of H-2K(b)-specific hyporesponsiveness following pretreatment with AdSV40K(b)-transduced CBA BMCs, persistence of H-2K(b) mRNA in central or peripheral tissues could not be demonstrated by RT-PCR. This result was in contrast to the observed persistence of K(b) mRNA both in the periphery and thymus following the infusion of transgenic CBK (H-2(k) + K(b)) BMCs. We conclude that ex vivo adenoviral gene transfer of a single donor MHC class I gene to recipient BMCs in combination with transient depletion of CD4(+) cells is sufficient to induce long-term graft survival of a fully allogeneic cardiac graft. In addition, detectable microchimerism is not a prerequisite for graft survival.


Subject(s)
Bone Marrow Cells/immunology , Genes, MHC Class I , Genetic Therapy/methods , Graft vs Host Disease/prevention & control , Heart Transplantation , Adenoviridae/genetics , Animals , Antibodies, Monoclonal/administration & dosage , Bone Marrow Transplantation , CD4 Antigens/immunology , Genetic Vectors/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Transplantation Immunology , Transplantation, Homologous
20.
Transplantation ; 72(3): 369-76, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11502963

ABSTRACT

BACKGROUND: In this study, we have investigated the mechanisms involved in both the induction of suppressive anergy, the stability of the anergy induced, and the possible mechanisms by which the response of immunocompetent CD4+ T cells are suppressed. METHODS: We used immobilized anti-CD3 monoclonal antibody (mAb) to induce anergy in T helper (Th) 1 and Th0 cells reactive with MHC class II molecule H2 I-Ab. RESULTS: We observed that suppressive anergy was induced independently of costimulation in Th0 but not Th1 cells. Although the anergic and suppressive states of Th0 cells were stable in the presence of exogenous interleukin-2, this was not the case for Th1 cells. No evidence for linked epitope suppression was observed for any of the I-Ab reactive cells investigated. Neither anergy nor suppression was observed in Th0 cells upon restimulation with anti-CD3 in the presence of syngeneic antigen-presenting cells (APCs). However, anergy but not suppression was observed in co-cultures restimulated with anti-T-cell antigen receptor (TCR) mAbs/syngeneic APCs and suppression could be restored by the addition of I-Ab+ APCs. CONCLUSIONS: Overall, these data suggested that the MHC-peptide complex recognized by the Th0 cells was required for suppression of the response of immunocompetent cells. We propose that suppression is mediated either by down-modulation of the MHC-peptide complex recognized by the anergic T cells or that a molecule specific to the MHC-peptide/TCR interaction facilitates negative regulation by APC:T or T:T interactions.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Clonal Anergy/physiology , Immune Tolerance/physiology , Major Histocompatibility Complex/physiology , Animals , Antibodies, Monoclonal/pharmacology , CD3 Complex/immunology , Cell Movement/drug effects , Clonal Anergy/drug effects , Histocompatibility Antigens Class II/physiology , Interleukin-2/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Peptides/physiology , Receptors, Antigen, T-Cell/physiology , Th1 Cells/drug effects , Th1 Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...