Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Transplant Direct ; 9(6): e1490, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37250484

ABSTRACT

Presensitized patients awaiting a kidney transplant have a lower graft survival and a longer waiting time because of the limited number of potential donors and the higher risk of antibody-mediated rejection (AMR), particularly in the early posttransplant period, because of preformed donor-specific antibodies binding major histocompatibility complex (MHC) molecules expressed by the graft endothelium followed by the activation of the complement. Advances in kidney preservation techniques allow the development of ex vivo treatment of transplants. We hypothesized that masking MHC ex vivo before transplantation could help to prevent early AMR in presensitized recipients. We evaluated a strategy of MHC I masking by an antibody during ex vivo organ perfusion in a porcine model of kidney transplantation in alloimmunized recipients. Methods: Through the in vitro calcein-release assay and flow cytometry, we evaluated the protective effect of a monoclonal anti-swine leukocyte antigen class I antibody (clone JM1E3) against alloreactive IgG complement-dependent cytotoxicity toward donor endothelial cells. Kidneys perfused ex vivo with JM1E3 during hypothermic machine perfusion were transplanted to alloimmunized recipients. Results: In vitro incubation of endothelial cells with JM1E3 decreased alloreactive IgG cytotoxicity (mean complement-dependent cytotoxicity index [% of control condition] with 1 µg/mL 74.13% ± 35.26 [calcein assay] and 66.88% ± 33.46 [cytometry]), with high interindividual variability. After transplantation, acute AMR occurred in all recipients on day 1, with signs of complement activation (C5b-9 staining) as soon as 1 h after transplantation, despite effective JM1E3 binding on graft endothelium. Conclusions: Despite a partial protective effect of swine leukocyte antigen I masking with JM1E3 in vitro, ex vivo perfusion of the kidney with JM1E3 before transplantation was not sufficient alone at preventing or delaying AMR in highly sensitized recipients.

2.
J Autoimmun ; 129: 102826, 2022 05.
Article in English | MEDLINE | ID: mdl-35378380

ABSTRACT

The critical role of neutrophils in pathological inflammation, notably in various autoimmune disorders, is currently the focus of renewed interest. Here, we demonstrate for the first time that activation of neutrophils with various inflammatory stimuli induces the release of extracellular vesicles (EVs) that are internalized by endothelial cells (ECs), thus leading to the transfer of miR-223, miR-142-3p and miR-451 and subsequent endothelial damage. Indeed, while miR-223 has little effect on EC responses, we show that the induced expression of miR-142-3p and miR-451 in ECs results in profound cell damage, especially in inflammatory conditions, characterized by a dramatic increase in cell apoptosis, impaired angiogenic repair responses, and the induction of IL-6, IL-8, CXCL10 and CXCL11 expression. We show that the strong deleterious effect of miR-142-3p may be due in part to its ability to block the activation of ERK1/2 and eNOS-mediated signals in ECs. miR-142-3p also inhibits the expression of RAC1, ROCK2 and CLIC4, three genes that are critical for EC migration and angiogenic responses. Importantly, miR-223, miR-142-3p and miR-451 are markedly increased in kidney biopsies from patients with active ANCA-associated vasculitis, a severe autoimmune disease that is prototypical of a neutrophil-induced microvascular damage. Taken together, our results suggest that miR-142-3p and miR-451 released in EVs by activated neutrophils can target EC to trigger an inflammatory cascade and induce direct vascular damage, and that therapeutic strategies based on the inhibition of these miRNAs in ECs will have implications for neutrophil-mediated inflammatory diseases.


Subject(s)
Extracellular Vesicles , MicroRNAs , Chloride Channels/metabolism , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , Humans , Inflammation/metabolism , MicroRNAs/genetics , Neutrophils/metabolism
3.
J Clin Invest ; 130(11): 6109-6123, 2020 11 02.
Article in English | MEDLINE | ID: mdl-33074246

ABSTRACT

T cell exclusion causes resistance to cancer immunotherapies via immune checkpoint blockade (ICB). Myeloid cells contribute to resistance by expressing signal regulatory protein-α (SIRPα), an inhibitory membrane receptor that interacts with ubiquitous receptor CD47 to control macrophage phagocytosis in the tumor microenvironment. Although CD47/SIRPα-targeting drugs have been assessed in preclinical models, the therapeutic benefit of selectively blocking SIRPα, and not SIRPγ/CD47, in humans remains unknown. We report a potent synergy between selective SIRPα blockade and ICB in increasing memory T cell responses and reverting exclusion in syngeneic and orthotopic tumor models. Selective SIRPα blockade stimulated tumor nest T cell recruitment by restoring murine and human macrophage chemokine secretion and increased anti-tumor T cell responses by promoting tumor-antigen crosspresentation by dendritic cells. However, nonselective SIRPα/SIRPγ blockade targeting CD47 impaired human T cell activation, proliferation, and endothelial transmigration. Selective SIRPα inhibition opens an attractive avenue to overcoming ICB resistance in patients with elevated myeloid cell infiltration in solid tumors.


Subject(s)
Immunologic Memory , Immunotherapy , Mammary Neoplasms, Experimental/therapy , Neoplasm Proteins/immunology , Receptors, Immunologic/immunology , T-Lymphocytes/immunology , Animals , Female , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Neoplasm Proteins/genetics , Receptors, Immunologic/genetics , T-Lymphocytes/pathology
4.
J Am Soc Nephrol ; 31(4): 876-891, 2020 04.
Article in English | MEDLINE | ID: mdl-32165419

ABSTRACT

BACKGROUND: Identifying biomarkers to predict kidney transplant failure and to define new therapeutic targets requires more comprehensive understanding of the immune response to chronic allogeneic stimulation. METHODS: We investigated the frequency and function of CD8+ T cell subsets-including effector memory (EM) and terminally differentiated EM (TEMRA) CD8+ T cells-in blood samples from 284 kidney transplant recipients recruited 1 year post-transplant and followed for a median of 8.3 years. We also analyzed CD8+ T cell reactivity to donor-specific PBMCs in 24 patients who had received living-donor kidney transplants. RESULTS: Increased frequency of circulating TEMRA CD8+ T cells at 1 year post-transplant associated with increased risk of graft failure during follow-up. This association remained after adjustment for a previously reported composite of eight clinical variables, the Kidney Transplant Failure Score. In contrast, increased frequency of EM CD8+ T cells associated with reduced risk of graft failure. A distinct TEMRA CD8+ T cell subpopulation was identified that was characterized by expression of FcγRIIIA (CD16) and by high levels of proinflammatory cytokine secretion and cytotoxic activity. Although donor-specific stimulation induced a similar rapid, early response in EM and TEMRA CD8+ T cells, CD16 engagement resulted in selective activation of TEMRA CD8+ T cells, which mediated antibody-dependent cytotoxicity. CONCLUSIONS: At 1 year post-transplant, the composition of memory CD8+ T cell subsets in blood improved prediction of 8-year kidney transplant failure compared with a clinical-variables score alone. A subpopulation of TEMRA CD8+ T cells displays a novel dual mechanism of activation mediated by engagement of the T-cell receptor or of CD16. These findings suggest that TEMRA CD8+ T cells play a pivotal role in humoral and cellular rejection and reveal the potential value of memory CD8+ T cell monitoring for predicting risk of kidney transplant failure.


Subject(s)
CD8-Positive T-Lymphocytes , Graft Rejection/etiology , Graft Survival , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/surgery , Kidney Transplantation , Adult , Aged , Biomarkers/blood , Cohort Studies , Female , Graft Rejection/blood , Graft Rejection/diagnosis , Humans , Immunologic Memory , Male , Middle Aged , Predictive Value of Tests , ROC Curve , Risk Assessment , Treatment Outcome
5.
Am J Transplant ; 19(12): 3263-3275, 2019 12.
Article in English | MEDLINE | ID: mdl-31207067

ABSTRACT

Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of immature hematopoietic precursors known to suppress immune responses. Interaction of SIRP alpha (SIRPα), expressed by myeloid cells, with the ubiquitous receptor CD47 is an important immune checkpoint of the innate response regulating macrophages and dendritic cells functions. We previously described that MDSC expressing SIRPα accumulated after transplantation and maintained kidney allograft tolerance. However, the role of the SIRPα/CD47 axis on MDSC function remained unknown. Here, we found that blocking SIRPα or CD47 with monoclonal antibodies (mAbs) induced differentiation of MDSC into myeloid cells overexpressing MHC class II, CD86 costimulatory molecule and increased secretion of macrophage-recruiting chemokines (eg, MCP-1). Using a model of long-term kidney allograft tolerance sustained by MDSC, we observed that administration of blocking anti-SIRPα or CD47 mAbs induced graft dysfunction and rejection. Loss of tolerance came along with significant decrease of MDSC and increase in MCP-1 concentration in the periphery. Graft histological and transcriptomic analyses revealed an inflammatory (M1) macrophagic signature at rejection associated with overexpression of MCP-1 mRNA and protein in the graft. These findings indicate that the SIRPα-CD47 axis regulates the immature phenotype and chemokine secretion of MDSC and contributes to the induction and the active maintenance of peripheral acquired immune tolerance.


Subject(s)
CD47 Antigen/metabolism , Graft Rejection/immunology , Kidney Transplantation/adverse effects , Myeloid Cells/immunology , Myeloid-Derived Suppressor Cells/immunology , Receptors, Immunologic/metabolism , Transplantation Tolerance/immunology , Animals , Antibodies, Monoclonal/administration & dosage , CD47 Antigen/antagonists & inhibitors , CD47 Antigen/immunology , Chemokines , Graft Rejection/pathology , Graft Survival/immunology , Myeloid Cells/cytology , Rats , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/immunology
6.
J Clin Invest ; 129(5): 1910-1925, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30939120

ABSTRACT

It remains unknown what causes inflammatory bowel disease (IBD), including signaling networks perpetuating chronic gastrointestinal inflammation in Crohn's disease (CD) and ulcerative colitis (UC), in humans. According to an analysis of up to 500 patients with IBD and 100 controls, we report that key transcripts of the IL-7 receptor (IL-7R) pathway are accumulated in inflamed colon tissues of severe CD and UC patients not responding to either immunosuppressive/corticosteroid, anti-TNF, or anti-α4ß7 therapies. High expression of both IL7R and IL-7R signaling signature in the colon before treatment is strongly associated with nonresponsiveness to anti-TNF therapy. While in mice IL-7 is known to play a role in systemic inflammation, we found that in humans IL-7 also controlled α4ß7 integrin expression and imprinted gut-homing specificity on T cells. IL-7R blockade reduced human T cell homing to the gut and colonic inflammation in vivo in humanized mouse models, and altered effector T cells in colon explants from UC patients grown ex vivo. Our findings show that failure of current treatments for CD and UC is strongly associated with an overexpressed IL-7R signaling pathway and point to IL-7R as a relevant therapeutic target and potential biomarker to fill an unmet need in clinical IBD detection and treatment.


Subject(s)
Colitis, Ulcerative/metabolism , Colon/metabolism , Crohn Disease/metabolism , Receptors, Interleukin-7/metabolism , T-Lymphocytes/cytology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adolescent , Adult , Aged , Animals , Colon/pathology , Cytokines/metabolism , Endoscopy , Female , Gene Expression Profiling , Gene Expression Regulation , Graft vs Host Disease/metabolism , Humans , Inflammation , Integrins/metabolism , Intestinal Mucosa/metabolism , Leukocytes, Mononuclear/cytology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Signal Transduction , Young Adult
7.
Nat Commun ; 9(1): 4483, 2018 10 26.
Article in English | MEDLINE | ID: mdl-30367166

ABSTRACT

Targeting the expansion of pathogenic memory immune cells is a promising therapeutic strategy to prevent chronic autoimmune attacks. Here we investigate the therapeutic efficacy and mechanism of new anti-human IL-7Rα monoclonal antibodies (mAb) in non-human primates and show that, depending on the target epitope, a single injection of antagonistic anti-IL-7Rα mAbs induces a long-term control of skin inflammation despite repeated antigen challenges in presensitized monkeys. No modification in T cell numbers, phenotype, function or metabolism is observed in the peripheral blood or in response to polyclonal stimulation ex vivo. However, long-term in vivo hyporesponsiveness is associated with a significant decrease in the frequency of antigen-specific T cells producing IFN-γ upon antigen restimulation ex vivo. These findings indicate that chronic antigen-specific memory T cell responses can be controlled by anti-IL-7Rα mAbs, promoting and maintaining remission in T-cell mediated chronic inflammatory diseases.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunologic Memory/drug effects , Inflammation/drug therapy , Receptors, Interleukin-7/antagonists & inhibitors , T-Lymphocytes/immunology , Animals , Chronic Disease , Clonal Deletion/immunology , Disease Models, Animal , Humans , Immunologic Memory/immunology , Inflammation/immunology , Interferon-gamma/immunology , Papio , Receptors, Interleukin-7/agonists , Receptors, Interleukin-7/immunology , Signal Transduction/drug effects , Skin/immunology , Skin/pathology
8.
JCI Insight ; 2(3): e90088, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28194440

ABSTRACT

Rat and human CD4+ and CD8+ Tregs expressing low levels of CD45RC have strong immunoregulatory properties. We describe here that human CD45 isoforms are nonredundant and identify distinct subsets of cells. We show that CD45RC is not expressed by CD4+ and CD8+ Foxp3+ Tregs, while CD45RA/RB/RO are. Transient administration of a monoclonal antibody (mAb) targeting CD45RC in a rat cardiac allotransplantation model induced transplant tolerance associated with inhibition of allogeneic humoral responses but maintained primary and memory responses against cognate antigens. Anti-CD45RC mAb induced rapid death of CD45RChigh T cells through intrinsic cell signaling but preserved and potentiated CD4+ and CD8+ CD45RClow/- Tregs, which are able to adoptively transfer donor-specific tolerance to grafted recipients. Anti-CD45RC treatment results in distinct transcriptional signature of CD4+ and CD8+ CD45RClow/- Tregs. Finally, we demonstrate that anti-human CD45RC treatment inhibited graft-versus-host disease (GVHD) in immune-humanized NSG mice. Thus, short-term anti-CD45RC is a potent therapeutic candidate to induce transplantation tolerance in human.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Graft vs Host Disease/drug therapy , Leukocyte Common Antigens/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Graft vs Host Disease/immunology , Heart Transplantation , Humans , Immunity, Humoral/drug effects , Mice , Rats , Transplantation Tolerance
9.
PLoS One ; 11(6): e0156775, 2016.
Article in English | MEDLINE | ID: mdl-27280712

ABSTRACT

Polyclonal xenogenic IgGs, although having been used in the prevention and cure of severe infectious diseases, are highly immunogenic, which may restrict their usage in new applications such as Ebola hemorrhagic fever. IgG glycans display powerful xenogeneic antigens in humans, for example α1-3 Galactose and the glycolyl form of neuraminic acid Neu5Gc, and IgGs deprived of these key sugar epitopes may represent an advantage for passive immunotherapy. In this paper, we explored whether low immunogenicity IgGs had a protective effect on a guinea pig model of Ebola virus (EBOV) infection. For this purpose, a double knock-out pig lacking α1-3 Galactose and Neu5Gc was immunized against virus-like particles displaying surface EBOV glycoprotein GP. Following purification from serum, hyper-immune polyclonal IgGs were obtained, exhibiting an anti-EBOV GP titer of 1:100,000 and a virus neutralizing titer of 1:100. Guinea pigs were injected intramuscularly with purified IgGs on day 0 and day 3 post-EBOV infection. Compared to control animals treated with IgGs from non-immunized double KO pigs, the anti-EBOV IgGs-treated animals exhibited a significantly prolonged survival and a decreased virus load in blood on day 3. The data obtained indicated that IgGs lacking α1-3 Galactose and Neu5Gc, two highly immunogenic epitopes in humans, have a protective effect upon EBOV infection.


Subject(s)
Antibodies, Viral/blood , Ebola Vaccines/therapeutic use , Galactose/deficiency , Hemorrhagic Fever, Ebola/prevention & control , Immunoglobulin G/immunology , Neuraminic Acids/metabolism , Viral Envelope Proteins/immunology , Animals , Antibodies, Anti-Idiotypic/immunology , Ebola Vaccines/immunology , Ebolavirus/immunology , Guinea Pigs , Hemorrhagic Fever, Ebola/blood , Hemorrhagic Fever, Ebola/immunology , Male , Swine , Vaccination , Viral Load
10.
J Immunol ; 195(12): 5805-15, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26553075

ABSTRACT

In transplantation tolerance, numerous regulatory populations have the capacity to inhibit allograft rejection; however, their compensatory capacities have never been clearly evidenced. We have previously demonstrated that the tolerogenic effect mediated by CD8(+)CD45RC(low) regulatory T cells (Tregs) in a model of organ transplantation with CD40Ig could be abrogated by permanent depletion of CD8(+) cells that resulted in allograft rejection in half of the recipients. This result demonstrated that CD8(+) Tregs were essential, but also that half of the recipients still survived indefinitely. We also demonstrated that no other regulatory populations, besides CD8(+) Tregs, could induce and maintain allograft tolerance in CD40Ig-treated tolerant animals. In the current study, we analyzed the mechanisms that arose following CD8(+) Treg depletion and allowed establishment of networks of new regulatory cells to maintain allograft survival. We identified regulatory B cells (Bregs) and regulatory myeloid cells (RegMCs) as being responsible of the maintenance of the long-term allograft survival. We demonstrated that both regulatory cell subsets efficiently inhibited antidonor immune responses in adoptively transferred recipients. Although Bregs were induced, they were not essential for the maintenance of the graft as demonstrated in IgM-deficient recipients. In addition, we showed that RegMCs were the most suppressive and acted alone, whereas Bregs activity was associated with increased suppressive activity of other subsets in adoptively transferred recipients. Altogether, to our knowledge, we demonstrated in this study for the first time the emergence of both Bregs and RegMCs following Tregs depletion and highlighted the importance of regulatory cell networks and their synergistic potential in transplantation.


Subject(s)
B-Lymphocytes, Regulatory/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Graft Rejection/immunology , Heart Transplantation , Myeloid Cells/immunology , Adoptive Transfer , Animals , B-Lymphocytes, Regulatory/transplantation , Cell Communication , Cells, Cultured , Immunomodulation , Myeloid Cells/transplantation , Rats , Rats, Inbred Lew , Transplantation Tolerance
11.
J Clin Invest ; 124(4): 1723-33, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24569454

ABSTRACT

T cell depletion is commonly used in organ transplantation for immunosuppression; however, a restoration of T cell homeostasis following depletion leads to increased memory T cells, which may promote transplant rejection. The cytokine IL-7 is important for controlling lymphopoiesis under both normal and lymphopenic conditions. Here, we investigated whether blocking IL-7 signaling with a mAb that targets IL-7 receptor α (IL-7Rα) alone or following T cell depletion confers an advantage for allograft survival in murine transplant models. We found that IL-7R blockade alone induced indefinite pancreatic islet allograft survival if anti-IL-7R treatment was started 3 weeks before graft. IL-7R blockade following anti-CD4- and anti-CD8-mediated T cell depletion markedly prolonged skin allograft survival. Furthermore, IL-7 inhibition in combination with T cell depletion synergized with either CTLA-4Ig administration or suboptimal doses of tacrolimus to induce long-term skin graft acceptance in this stringent transplant model. Together, these therapies inhibited T cell reconstitution, decreased memory T cell numbers, increased the relative frequency of Tregs, and abrogated both cellular and humoral alloimmune responses. Our data suggest that IL-7R blockade following T cell depletion has potential as a robust, immunosuppressive therapy in transplantation.


Subject(s)
Graft Survival/immunology , Immunosuppression Therapy/methods , Lymphocyte Depletion , Receptors, Interleukin-7/antagonists & inhibitors , T-Lymphocytes/immunology , Abatacept , Allografts , Animals , Antibodies, Monoclonal/administration & dosage , Immunity, Cellular , Immunity, Humoral , Immunoconjugates/administration & dosage , Immunosuppressive Agents/administration & dosage , Islets of Langerhans Transplantation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Nude , Skin Transplantation , Tacrolimus/administration & dosage , Transplantation Immunology , Transplantation Tolerance/immunology
12.
MAbs ; 5(1): 47-55, 2013.
Article in English | MEDLINE | ID: mdl-23221503

ABSTRACT

Antagonist antibodies targeting CD28 have been proposed as an alternative to the use of CD80/86 antagonists to modulate T cell responses in autoimmunity and transplantation. Advantages would be the blockade of CD28-mediated co-stimulatory signals without impeding the co-inhibitory signals dependent on CD80 interactions with CTLA-4 and PD-L1 that are important for the control of immune responses and for the function of regulatory T cells. Anti-CD28 antibodies are candidate antagonists only if they prevent access to the CD80/86 ligands without simultaneously stimulating CD28 itself, a process that is believed to depend on receptor multimerization. In this study, we evaluated the impact of different formats of a potentially antagonist anti-human CD28 antibody on T cell activation. In particular, we examined the role of valency and of the presence of an Fc domain, two components that might affect receptor multimerization either directly or in the presence of accessory cells expressing Fc receptors. Among monovalent (Fab', scFv), divalent (Fab'2), monovalent-Fc (Fv-Fc) and divalent-Fc (IgG) formats, only the monovalent formats showed consistent absence of induced CD28 multimerization and absence of associated activation of phosphoinositol-3-kinase, and clear antagonist properties in T cell stimulation assays. In contrast, divalent antibodies showed agonist properties that resulted in cell proliferation and cytokine release in an Fc-independent manner. Conjugation of monovalent antibodies with polyethylene glycol, α-1-antitrypsin or an Fc domain significantly extended their in vivo half-life without modifying their antagonist properties. In conclusion, these data indicate that monovalency is mandatory for maintaining the antagonistic activity of anti-CD28 monoclonal antibodies.


Subject(s)
Antibodies, Monoclonal/immunology , CD28 Antigens/antagonists & inhibitors , Single-Domain Antibodies/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , CD28 Antigens/immunology , Female , Half-Life , Humans , Immunoconjugates/immunology , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/immunology , Jurkat Cells , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Mice , Polyethylene Glycols/chemistry , Single-Domain Antibodies/chemistry
13.
J Immunol ; 188(9): 4209-16, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22450806

ABSTRACT

Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of immature cells that are believed to inhibit immune responses in the contexts of cancer and organ transplantation, in association with regulatory T cells (Treg). However, the way in which MDSC cooperate with Treg remains elusive. In this study, we used DNA microarrays to analyze gene expression in blood-derived MDSC from rat recipients of kidney allografts. We found CCL5 (Rantes), a chemotactic C-C motif 5 chemokine, to be strongly downregulated after treatment with a tolerizing regimen. The amount of CCL5 protein was also lower in the plasma of tolerant recipients, whereas intragraft CCL5 was unchanged. Because CCL5 is chemotactic for Treg, we hypothesized that a gradient of CCL5 between the graft and peripheral blood might contribute to the intragraft localization of Treg in tolerant animals. To test this hypothesis, we treated tolerant rat recipients of kidney allografts with recombinant rat CCL5 to restore normal plasma concentrations. This led to a strong reduction in intragraft Treg monitored by immunohistofluorescence and by quantitative real-time PCR measurement of Foxp3 mRNA. Ultimately, this treatment led to an increase in serum creatinine concentrations and to kidney graft rejection after about a month. The kidney function of syngeneic grafts was not affected by a similar administration of CCL5. These data highlight the contribution of MDSC to the establishment of a graft-to-periphery CCL5 gradient in tolerant kidney allograft recipients, which controls recruitment of Treg to the graft where they likely contribute to maintaining tolerance.


Subject(s)
Chemokine CCL5/immunology , Kidney Transplantation , Kidney/immunology , Myeloid Cells/immunology , T-Lymphocytes, Regulatory/immunology , Transplantation Tolerance , Transplants , Animals , Cell Line , Chemokine CCL5/metabolism , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/immunology , Gene Expression Profiling , Gene Expression Regulation/immunology , Male , Mice , Myeloid Cells/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/biosynthesis , RNA, Messenger/immunology , Rats , Rats, Inbred Lew , T-Lymphocytes, Regulatory/metabolism , Transplantation, Homologous
14.
Sci Transl Med ; 2(17): 17ra10, 2010 Feb 03.
Article in English | MEDLINE | ID: mdl-20371478

ABSTRACT

Transplantation is the treatment of choice for patients with end-stage organ failure. Its success is limited by side effects of immunosuppressive drugs, such as inhibitors of the calcineurin pathway that prevent rejection by reducing synthesis of interleukin-2 by T cells. Moreover, none of the existing drugs efficiently prevent the eventual rejection of the organ. Blocking the CD28-mediated T cell costimulation pathway is a nontoxic alternative immunosuppression strategy that is now achieved by blockade of CD80/86, the receptor for CD28 on antigen-presenting cells. However, interaction of CD80/86 with cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) is required for immune regulation. Therefore, CD28 blockade, instead of CD80/86 blockade, might preserve regulatory signals mediated by CTLA-4 and preserve immune regulation. By using monovalent antibodies, we identified true CD28 antagonists that induced CTLA-4-dependent decreased T cell function compatible with regulatory T (Treg) cell suppression. In transplantation experiments in primates, blocking CD28 augmented intragraft and peripheral blood Treg cells, induced molecular signatures of immune regulation, and prevented graft rejection and vasculopathy in synergy with calcineurin inhibition. These findings suggest that targeting costimulation blockade at CD28 preserves CTLA-4-dependent immune regulation and promotes allograft survival.


Subject(s)
Antigens, CD/metabolism , CD28 Antigens/metabolism , Organ Transplantation/methods , Animals , B7-1 Antigen/biosynthesis , B7-2 Antigen/biosynthesis , CTLA-4 Antigen , Calcineurin/metabolism , Graft Survival , Humans , Immune System , Lymphocytes/cytology , Macaca fascicularis , Papio , T-Lymphocytes, Regulatory/cytology
15.
J Immunol ; 180(12): 7898-906, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18523253

ABSTRACT

The immune tolerance to rat kidney allografts induced by a perioperative treatment with anti-CD28 Abs is associated with a severe unresponsiveness of peripheral blood cells to donor Ags. In this model, we identified an accumulation in the blood of CD3(-)class II(-)CD11b(+)CD80/86(+) plastic-adherent cells that additionally expressed CD172a as well as other myeloid markers. These cells were able to inhibit proliferation, but not activation, of effector T cells and to induce apoptosis in a contact-dependent manner. Their suppressive action was found to be under the control of inducible NO synthase, an enzyme also up-regulated in tolerated allografts. Based on these features, these cells can be defined as myeloid-derived suppressor cells (MDSC). Interestingly, CD4(+)CD25(high)FoxP3(+) regulatory T cells were insensitive in vitro to MDSC-mediated suppression. Although the adoptive transfer of MDSC failed to induce kidney allograft tolerance in recently transplanted recipients, the maintenance of tolerance after administration of anti-CD28 Abs was found to be dependent on the action of inducible NO synthase. These results suggest that increased numbers of MDSC can inhibit alloreactive T cell proliferation in vivo and that these cells may participate in the NO-dependent maintenance phase of tolerance.


Subject(s)
Cell Differentiation/immunology , Cell Movement/immunology , Immune Tolerance , Kidney Transplantation/immunology , Myeloid Cells/cytology , Myeloid Cells/immunology , Animals , B7-1 Antigen/biosynthesis , B7-2 Antigen/biosynthesis , CD11b Antigen/biosynthesis , Cell Adhesion , Cells, Cultured , Cytotoxicity Tests, Immunologic , Immunophenotyping , Male , Models, Immunological , Myeloid Cells/metabolism , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/physiology , Rats , Rats, Inbred Lew , Spleen/cytology , Spleen/immunology , Spleen/pathology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology
16.
Xenotransplantation ; 14(1): 25-33, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17214702

ABSTRACT

BACKGROUND: In heart allograft in the rat, a sustained costimulation blockade with CTLA4Ig prevents alloreactive T-cell activation and promotes a long-term graft survival through the action of tolerogeneic dendritic cells. It is unclear whether similar mechanisms might occur after xenotransplantation. To test that hypothesis, we have analyzed the action of CTLA4Ig in a model of CD4(+)T cell-mediated xenograft rejection. METHODS: Hamster hearts were transplanted into LEW.1A rats receiving an accommodation-inducing treatment consisting of a short course administration of LF15-0195 and a daily administration of cyclosporine A (CSA). To achieve long-term delivery of CTLA4Ig, an intravenous administration of an adenovirus vector coding for mouse CTLA4Ig (Ad-CTLA4Ig) was added to the accommodation induction protocol. On day 40 post-transplantation, rejection was induced by CSA withdrawal. In other xenograft recipients, CD28/B7 costimulation was inhibited at that time only by injections of CTLA4Ig or anti-CD28 antibodies. Graft survival, immunohistology, as well as development of antibodies and regulatory cells were examined. RESULTS: Xenografts survived 6 days after CSA withdrawal in controls and were rejected, as previously described, through the action of CD4(+) xenoreactive T cells. Interfering with CD28/B7 costimulation inhibited this xenoreactive T cell response and delayed rejection to day 10. In recipients that had received Ad-CTLA4Ig, survival was prolonged to day 19 and this was accompanied by the appearance of regulatory cells exhibiting non-donor-specific suppressive activity dependent on IL-2, NO, and IDO. These regulatory cells were different from those previously identified after Ad-CTLA4Ig administration in heart allograft in the rat. In these recipients, rejection occurred as a consequence of an evoked anti-donor IgM response and complement activation and not of a cellular rejection as complement inhibition with cobra venom factor further prolonged xenograft survival. CONCLUSION: CD28/B7 blockade delays CD4(+) T cell-mediated rejection after CSA withdrawal in accommodated recipients of hamster heart xenografts. In addition, a sustained expression of CTLA4Ig has the potential of inducing cellular regulatory mechanisms. However, such treatment does not prevent the development of xenoreactive IgM antibodies that participate in vascular rejection processes in a complement-dependent manner.


Subject(s)
Cardiovascular Diseases/immunology , Cricetinae , Graft Rejection/immunology , Heart Transplantation/immunology , Rats , T-Lymphocytes, Regulatory/immunology , Transplantation, Heterologous/immunology , Abatacept , Adenoviridae/genetics , Animals , B7-1 Antigen/immunology , CD28 Antigens/immunology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cardiovascular Diseases/surgery , Cells, Cultured , Cyclosporine/pharmacology , Graft Rejection/prevention & control , Graft Survival/immunology , Heart Transplantation/adverse effects , Immunoconjugates/genetics , Immunoconjugates/immunology , Immunoconjugates/metabolism , Male , Spleen/cytology , T-Lymphocytes, Regulatory/metabolism
17.
Am J Transplant ; 5(2): 255-67, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15643985

ABSTRACT

Recent studies suggest that particular dendritic cells (DC) subpopulations may be tolerogenic. To test the capacity of different DC subpopulations to modulate allograft rejection, we generated two distinct populations of rat bone marrow-derived DCs (BMDC) with low doses of GM-CSF and IL-4. The non-adherent population (nBMDC), which are the 'classical' DCs was able to stimulate naive allogeneic T cells and could be induced to completely mature using various stimuli. In contrast, the adherent population (aBMDC), which displayed an immature phenotype, was unable to stimulate T cells and was more resistant to maturation. We found that syngeneic aBMDCs, injected one day before transplantation, induced significant prolongation of heart allograft survival and decreased anti-donor humoral and cellular responses. Similarly, syngeneic aBMDCs inhibited T-cell responses to KLH in the spleen but not in lymph node in a KLH immunization model without graft. This effect was not antigen specific and could be reversed using an inhibitor of inducible nitric oxide synthase. This compartmentalized inhibition could be in part explained by the fact that the majority of syngeneic adherent cells administered intravenously were found in the spleen with some of them reaching the T-cell areas. These data suggest that syngeneic aBMDCs can modulate immune responses.


Subject(s)
Bone Marrow Cells/physiology , Dendritic Cells/physiology , Graft Survival/physiology , Heart Transplantation , Stem Cells/physiology , Animals , Bone Marrow Cells/immunology , Cell Adhesion/physiology , Cell Movement/physiology , Graft Rejection/immunology , Hemocyanins/immunology , Mice , Rats , Spleen/physiology , Stem Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...