Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
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.
Front Immunol ; 12: 732530, 2021.
Article in English | MEDLINE | ID: mdl-34925315

ABSTRACT

A numerous number of positive and negative signals via various molecules modulate T-cell activation. Within the various transmembrane proteins, SIRPγ is of interest since it is not expressed in rodents. SIRPγ interaction with CD47 is reevaluated in this study. Indeed, we show that the anti-SIRPγ mAb clone LSB2.20 previously used by others has not been appropriately characterized. We reveal that the anti-SIRPα clone KWAR23 is a Pan anti-SIRP mAb which efficiently blocks SIRPα and SIRPγ interactions with CD47. We show that SIRPγ expression on T cells varies with their differentiation and while being expressed on Tregs, is not implicated in their suppressive functions. SIRPγ spatial reorganization at the immune synapse is independent of its interaction with CD47. In vitro SIRPα-γ/CD47 blockade with KWAR23 impairs IFN-γ secretion by chronically activated T cells. In vivo in a xeno-GvHD model in NSG mice, the SIRPγ/CD47 blockade with the KWAR23 significantly delays the onset of the xeno-GvHD and deeply impairs human chimerism. In conclusion, we have shown that T-cell interaction with CD47 is of importance notably in chronic stimulation.


Subject(s)
Antigens, Differentiation/metabolism , CD47 Antigen/metabolism , Graft vs Host Disease/immunology , Lymphocyte Activation/drug effects , Muromonab-CD3/administration & dosage , Receptors, Immunologic/metabolism , Signal Transduction/drug effects , T-Lymphocytes/immunology , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/immunology , Blood Donors , CD47 Antigen/genetics , Disease Models, Animal , Female , Gene Knock-In Techniques , Gene Knockout Techniques , Healthy Volunteers , Heterografts , Humans , Jurkat Cells , Lymphocyte Activation/genetics , Male , Mice , Muromonab-CD3/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Signal Transduction/genetics
3.
Cell Metab ; 30(6): 1075-1090.e8, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31801055

ABSTRACT

Cell therapy is a promising strategy for treating patients suffering from autoimmune or inflammatory diseases or receiving a transplant. Based on our preclinical studies, we have generated human autologous tolerogenic dendritic cells (ATDCs), which are being tested in a first-in-man clinical trial in kidney transplant recipients. Here, we report that ATDCs represent a unique subset of monocyte-derived cells based on phenotypic, transcriptomic, and metabolic analyses. ATDCs are characterized by their suppression of T cell proliferation and their expansion of Tregs through secreted factors. ATDCs produce high levels of lactate that shape T cell responses toward tolerance. Indeed, T cells take up ATDC-secreted lactate, leading to a decrease of their glycolysis. In vivo, ATDCs promote elevated levels of circulating lactate and delay graft-versus-host disease by reducing T cell proliferative capacity. The suppression of T cell immunity through lactate production by ATDCs is a novel mechanism that distinguishes ATDCs from other cell-based immunotherapies.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immune Tolerance , Immunosuppression Therapy , Lactic Acid/biosynthesis , Animals , Autoimmune Diseases/therapy , CD4-Positive T-Lymphocytes/cytology , Cells, Cultured , Dendritic Cells/metabolism , Female , Humans , Lymphocyte Activation , Male , Mice , Mice, Inbred NOD , Mice, SCID , Monocytes/immunology
4.
Blood Adv ; 3(22): 3522-3538, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31730699

ABSTRACT

Polyclonal CD8+CD45RClow/- Tregs are potent regulatory cells able to control solid organ transplantation rejection and even induce tolerance. However, donor major histocompatibility complex (MHC)-specific Tregs are more potent than polyclonal Tregs in suppressing T-cell responses and preventing acute as well as chronic rejection in rodent models. The difficulty of identifying disease-relevant antigens able to stimulate Tregs has reduced the possibility of obtaining antigen-specific Tregs. To bypass this requirement and gain the advantage of antigen specificity, and thus improve the therapeutic potential of CD8+ Tregs, we stably introduced a chimeric antigen receptor (CAR) derived from a HLA-A*02 antigen-specific antibody (A2-CAR) in human CD8+ Tregs and developed a clinically compatible protocol of transduction and expansion. We demonstrated that A2-CAR CD8+ Tregs were not phenotypically altered by the process, were specifically activated, and did not exhibit cytotoxic activity toward HLA-A*02+ kidney endothelial cells (ECs). We showed that A2-CAR CD8+ Tregs were more potent suppressors of immune responses induced by HLA-A*02 mismatch than control-CAR CD8+ Tregs, both in vitro and in vivo, in models of human skin graft rejection and graft-versus-host disease (GVHD) in NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ (NSG) mice. We showed that integrity of human skin graft was preserved with A2-CAR CD8+ Tregs at least 100 days in vivo after administration, and that interaction between the A2-CAR CD8+ Tregs and HLA-A*02+ kidney ECs resulted in a fine-tuned and protolerogenic activation of the ECs without cytotoxicity. Together, our results demonstrated the relevance of the CAR engineering approach to develop antigen-specific CAR-CD8+ Tregs for clinical trials in transplantation, and potentially in other diseases.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Graft vs Host Disease/therapy , HLA Antigens/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Biomarkers , Cell Communication , Disease Models, Animal , Gene Expression , Genetic Engineering , Graft Rejection/genetics , Graft Rejection/immunology , Graft vs Host Disease/etiology , HLA Antigens/immunology , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Humans , Immune Tolerance , Immunophenotyping , Mice , Mice, Knockout , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , Transduction, Genetic
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.
Transplantation ; 102(8): 1271-1278, 2018 08.
Article in English | MEDLINE | ID: mdl-29688994

ABSTRACT

BACKGROUND: Immunodeficient mice are invaluable tools to analyze the long-term effects of potentially immunogenic molecules in the absence of adaptive immune responses. Nevertheless, there are models and experimental situations that would beneficiate of larger immunodeficient recipients. Rats are ideally suited to perform experiments in which larger size is needed and are still a small animal model suitable for rodent facilities. Additionally, rats reproduce certain human diseases better than mice, such as ankylosing spondylitis and Duchenne disease, and these disease models would greatly benefit from immunodeficient rats to test different immunogenic treatments. METHODS: We describe the generation of Il2rg-deficient rats and their crossing with previously described Rag1-deficient rats to generate double-mutant RRG animals. RESULTS: As compared with Rag1-deficient rats, Il2rg-deficient rats were more immunodeficient because they partially lacked not only T and B cells but also NK cells. RRG animals showed a more profound immunossuppressed phenotype because they displayed undetectable levels of T, B, and NK cells. Similarly, all immunoglobulin isotypes in sera were decreased in Rag1- or Il2rg-deficient rats and undetectable in Rats Rag1 and Il2rg (RRG) animals. Rag1- or Il2rg-deficient rats rejected allogeneic skin transplants and human tumors, whereas animals not only accepted allogeneic rat skin but also xenogeneic human tumors, skin, and hepatocytes. Immune humanization of RRG animals was unsuccessful. CONCLUSIONS: Thus, immunodeficient RRG animals are useful recipients for long-term studies in which immune responses could be an obstacle, including tissue humanization of different tissues.


Subject(s)
Gene Deletion , Homeodomain Proteins/genetics , Interleukin Receptor Common gamma Subunit/genetics , Animals , Animals, Genetically Modified , Crosses, Genetic , Disease Models, Animal , Exons , Female , Genotype , Hepatocytes/cytology , Humans , Immune System , Liver/immunology , Male , Mutation , Rats , Rats, Sprague-Dawley , Skin Transplantation , Transplantation, Heterologous , Transplants
9.
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
10.
Front Immunol ; 8: 2014, 2017.
Article in English | MEDLINE | ID: mdl-29445370

ABSTRACT

Both CD4+ and CD8+ Tregs play a critical role in the control of immune responses and immune tolerance; however, our understanding of CD8+ Tregs is limited while they are particularly promising for therapeutic application. We report here existence of highly suppressive human CD8+CD45RClow/- Tregs expressing Foxp3 and producing IFNγ, IL-10, IL-34, and TGFß to mediate their suppressive activity. We demonstrate that total CD8+CD45RClow/- Tregs can be efficiently expanded in the presence of anti-CD3/28 mAbs, high-dose IL-2 and IL-15 and that such expanded Tregs efficiently delay GVHD and human skin transplantation rejection in immune humanized mice. Robustly expanded CD8+ Tregs displayed a specific gene signature, upregulated cytokines and expansion in the presence of rapamycin greatly improved proliferation and suppression. We show that CD8+CD45RClow/- Tregs are equivalent to canonical CD4+CD25highCD127low/- Tregs for suppression of allogeneic immune responses in vitro. Altogether, our results open new perspectives to tolerogenic strategies in human solid organ transplantation and GVHD.

11.
J Am Soc Nephrol ; 27(12): 3577-3588, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27160407

ABSTRACT

Belatacept is a biologic that targets CD80/86 and prevents its interaction with CD28 and its alternative ligand, cytotoxic T lymphocyte antigen 4 (CTLA-4). Clinical experience in kidney transplantation has revealed a high incidence of rejection with belatacept, especially with intensive regimens, suggesting that blocking CTLA-4 is deleterious. We performed a head to head assessment of FR104 (n=5), a selective pegylated Fab' antibody fragment antagonist of CD28 that does not block the CTLA-4 pathway, and belatacept (n=5) in kidney allotransplantation in baboons. The biologics were supplemented with an initial 1-month treatment with low-dose tacrolimus. In cases of acute rejection, animals also received steroids. In the belatacept group, four of five recipients developed severe, steroid-resistant acute cellular rejection, whereas FR104-treated animals did not. Assessment of regulatory T cell-specific demethylated region methylation status in 1-month biopsy samples revealed a nonsignificant trend for higher regulatory T cell frequencies in FR104-treated animals. Transcriptional analysis did not reveal significant differences in Th17 cytokines but did reveal higher levels of IL-21, the main cytokine secreted by CD4 T follicular helper (Tfh) cells, in belatacept-treated animals. In vitro, FR104 controlled the proliferative response of human preexisting Tfh cells more efficiently than belatacept. In mice, selective CD28 blockade also controlled Tfh memory cell responses to KLH stimulation more efficiently than CD80/86 blockade. Our data reveal that selective CD28 blockade and belatacept exert different effects on mechanisms of renal allograft rejection, particularly at the level of Tfh cell stimulation.


Subject(s)
Abatacept/pharmacology , Antibodies/drug effects , Antibodies/immunology , CD28 Antigens/immunology , Graft Rejection/immunology , Immunosuppressive Agents/pharmacology , Animals , Mice , Papio
13.
Glia ; 63(12): 2298-312, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26212105

ABSTRACT

Among the costimulatory factors widely studied in the immune system is the CD28/cytotoxic T-lymphocyte antigen-4 (CTLA4)-CD80/CD86 pathway, which critically controls the nature and duration of the T-cell response. In the brain, up-regulated expression of CD80/CD86 during inflammation has consistently been reported in microglia. However, the role of CD80/CD86 molecules has mainly been studied in a context of microglia-T cell interactions in pathological conditions, while the function of CD80/CD86 in the regulation of intrinsic brain cells remains largely unknown. In this study, we used a transgenic pig line in which neurons express releasable CTLA4-Ig, a synthetic molecule mimicking CTLA4 and binding to CD80/CD86. The effects of CTLA4-Ig on brain cells were analyzed after intracerebral transplantation of CTLA4-Ig-expressing neurons or wild-type neurons as control. This model provided in vivo evidence that CTLA4-Ig stimulated axonal outgrowth, in correlation with a shift of the nearby microglia from a compact to a ramified morphology. In a culture system, we found that the CTLA4-Ig-induced morphological change of microglia was mediated through CD86, but not CD80. This was accompanied by microglial up-regulated expression of the anti-inflammatory molecule Arginase 1 and the neurotrophic factor BDNF, in an astrocyte-dependent manner through the purinergic P2Y1 receptor pathway. Our study identifies for the first time CD86 as a key player in the modulation of microglia phenotype and suggests that CTLA4-Ig-derived compounds might represent new tools to manipulate CNS microglia.


Subject(s)
Abatacept/metabolism , Axons/physiology , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , Microglia/physiology , Abatacept/genetics , Animals , Animals, Genetically Modified , Astrocytes/cytology , Astrocytes/physiology , Brain Tissue Transplantation , Brain-Derived Neurotrophic Factor/metabolism , Cell Enlargement , Cells, Cultured , Coculture Techniques , Corpus Striatum/cytology , Corpus Striatum/physiology , Corpus Striatum/surgery , Humans , Male , Microglia/cytology , RNA, Messenger/metabolism , Rats, Inbred Lew , Rats, Sprague-Dawley , Swine
14.
J Cell Mol Med ; 19(1): 124-34, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25310920

ABSTRACT

Foetal pig neuroblasts are interesting candidates as a cell source for transplantation, but xenotransplantation in the brain requires the development of adapted immunosuppressive treatments. As systemic administration of high doses of cyclosporine A has side effects and does not protect xenotransplants forever, we focused our work on local control of the host immune responses. We studied the advantage of cotransplanting syngenic mesenchymal stem cells (MSC) with porcine neuroblasts (pNb) in immunocompetent rat striata. Two groups of animals were transplanted, either with pNb alone or with both MSC and pNb. At day 63, no porcine neurons were detected in the striata that received only pNb, while four of six rats transplanted with both pNb and MSC exhibited healthy porcine neurons. Interestingly, 50% of the cotransplanted rats displayed healthy grafts with pNF70+ and TH+ neurons at 120 days post-transplantation. qPCR analyses revealed a general dwindling of pro- and anti-inflammatory cytokines in the striata that received the cotransplants. Motor recovery was also observed following the transplantation of pNb and MSC in a rat model of Parkinson's disease. Taken together, the present data indicate that the immunosuppressive properties of MSC are of great interest for the long-term survival of xenogeneic neurons in the brain.


Subject(s)
Brain/immunology , Immunity , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Transplantation, Heterologous , Animals , CD11b Antigen/metabolism , Cell Survival , Chemokines/genetics , Chemokines/metabolism , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Graft Survival/immunology , Immunity, Cellular , Immunocompetence , Male , Mesencephalon/cytology , Molecular Sequence Data , Motor Activity , Neurons/cytology , Neurons/metabolism , Neurons/transplantation , Oxidopamine , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Inbred Lew , Recovery of Function , Sus scrofa
15.
Xenotransplantation ; 21(5): 431-43, 2014.
Article in English | MEDLINE | ID: mdl-25040113

ABSTRACT

BACKGROUND: Human corneal allografting is an established procedure to cure corneal blindness. However, a shortage of human donor corneas as well as compounding economic, cultural, and organizational reasons in many countries limit its widespread use. Artificial corneas as well as porcine corneal xenografts have been considered as possible alternatives. To date, all preclinical studies using de-cellularized pig corneas have shown encouraging graft survival results; however, relatively few studies have been conducted in pig to non-human primate (NHP) models, and particularly using genetically engineered donors. METHODS: In this study, we assessed the potential benefit of using either hCTLA4-Ig transgenic or α1,3-Galactosyl Transferase (GT) Knock-Out (KO) plus transgenic hCD39/hCD55/hCD59/fucosyl-transferase pig lines in an anterior lamellar keratoplasty pig to NHP model. RESULTS: Corneas from transgenic animals expressing hCTLA4-Ig under the transcriptional control of a neuron-specific enolase promoter showed transgene expression in corneal keratocytes of the stroma and expression was maintained after transplantation. Although a first acute rejection episode occurred in all animals during the second week post-keratoplasty, the median final rejection time was 70 days in the hCTLA4-Ig group vs. 21 days in the wild-type (WT) control group. In contrast, no benefit for corneal xenograft survival from the GTKO/transgenic pig line was found. At rejection, cell infiltration in hCTLA4Ig transgenic grafts was mainly composed of macrophages with fewer CD3+ CD4+ and CD79+ cells than in other types of grafts. Anti-donor xenoantibodies increased dramatically between days 9 and 14 post-surgery in all animals. CONCLUSIONS: Local expression of the hCTLA4-Ig transgene dampens rejection of xenogeneic corneal grafts in this pig-to-NHP lamellar keratoplasty model. The hCTLA4-Ig transgene seems to target T-cell responses without impacting humoral responses, the control of which would presumably require additional peripheral immunosuppression.


Subject(s)
Corneal Keratocytes/metabolism , Corneal Transplantation/methods , Graft Rejection/prevention & control , Immunoconjugates/metabolism , Transgenes , Transplantation, Heterologous/methods , Abatacept , Animals , Animals, Genetically Modified , Biomarkers/metabolism , Corneal Keratocytes/immunology , Graft Rejection/genetics , Graft Rejection/immunology , Graft Survival/genetics , Graft Survival/immunology , Immunoconjugates/genetics , Macaca fascicularis , Male , Models, Animal , Sus scrofa/genetics
16.
MAbs ; 6(3): 697-707, 2014.
Article in English | MEDLINE | ID: mdl-24598534

ABSTRACT

Antagonist anti-CD28 antibodies prevent T-cell costimulation and are functionally different from CTLA4Ig since they cannot block CTLA-4 and PDL-1 co-inhibitory signals. They demonstrated preclinical efficacy in suppressing effector T cells while enhancing immunoregulatory mechanisms. Because a severe cytokine release syndrome was observed during the Phase 1 study with the superagonist anti-CD28 TGN1412, development of other anti-CD28 antibodies requires careful preclinical evaluation to exclude any potential immunotoxicity side-effects. The failure to identify immunological toxicity of TGN1412 using macaques led us to investigate more relevant preclinical models. We report here that contrary to macaques, and like in man, all baboon CD4-positive T lymphocytes express CD28 in their effector memory cells compartment, a lymphocyte subtype that is the most prone to releasing cytokines after reactivation. Baboon lymphocytes are able to release pro-inflammatory cytokines in vitro in response to agonist or superagonist anti-CD28 antibodies. Furthermore, we compared the reactivity of human and baboon lymphocytes after transfer into non obese diabetic/severe combined immunodeficiency (NOD/SCID) interleukin-2rγ knockout mice and confirmed that both cell types could release inflammatory cytokines in situ after injection of agonistic anti-CD28 antibodies. In contrast, FR104, a monovalent antagonistic anti-CD28 antibody, did not elicit T cell activation in these assays, even in the presence of anti-drug antibodies. Infusion to baboons also resulted in an absence of cytokine release. In conclusion, the baboon represents a suitable species for preclinical immunotoxicity evaluation of anti-CD28 antibodies because their effector memory T cells do express CD28 and because cytokine release can be assessed in vitro and trans vivo.


Subject(s)
Antibodies, Blocking/toxicity , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/toxicity , CD28 Antigens/antagonists & inhibitors , Papio anubis/immunology , Animals , Antibodies, Blocking/immunology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/toxicity , CD28 Antigens/immunology , Cytokines/biosynthesis , Drug Evaluation, Preclinical , Humans , Immunologic Memory , Lymphocyte Activation , Macaca fascicularis , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Models, Animal , Species Specificity , T-Lymphocytes/immunology
17.
Cell Transplant ; 23(11): 1407-23, 2014.
Article in English | MEDLINE | ID: mdl-23879897

ABSTRACT

Induced pluripotent stem cells (iPSCs) offer certain advantages over embryonic stem cells in cell replacement therapy for a variety of neurological disorders. However, reliable procedures, whereby transplanted iPSCs can survive and differentiate into functional neurons, without forming tumors, have yet to be devised. Currently, retroviral or lentiviral reprogramming methods are often used to reprogram somatic cells. Although the use of these viruses has proven to be effective, formation of tumors often results following in vivo transplantation, possibly due to the integration of the reprogramming genes. The goal of the current study was to develop a new approach, using an adenovirus for reprogramming cells, characterize the iPSCs in vitro, and test their safety, survivability, and ability to differentiate into region-appropriate neurons following transplantation into the rat brain. To this end, iPSCs were derived from bone marrow-derived mesenchymal stem cells and tail-tip fibroblasts using a single cassette lentivirus or a combination of adenoviruses. The reprogramming efficiency and levels of pluripotency were compared using immunocytochemistry, flow cytometry, and real-time polymerase chain reaction. Our data indicate that adenovirus-generated iPSCs from tail-tip fibroblasts are as efficient as the method we used for lentiviral reprogramming. All generated iPSCs were also capable of differentiating into neuronal-like cells in vitro. To test the in vivo survivability and the ability to differentiate into region-specific neurons in the absence of tumor formation, 400,000 of the iPSCs derived from tail-tip fibroblasts that were transfected with the adenovirus pair were transplanted into the striatum of adult, immune-competent rats. We observed that these iPSCs produced region-specific neuronal phenotypes, in the absence of tumor formation, at 90 days posttransplantation. These results suggest that adenovirus-generated iPSCs may provide a safe and viable means for neuronal replacement therapies.


Subject(s)
Adenoviridae/physiology , Cellular Reprogramming/physiology , Corpus Striatum/surgery , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/virology , Stem Cell Transplantation/methods , Animals , Cell Differentiation/physiology , Cell Survival/physiology , Corpus Striatum/cytology , Male , Rats , Rats, Sprague-Dawley , Transfection
18.
Stem Cells ; 30(10): 2342-53, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22888011

ABSTRACT

Besides their therapeutic benefit as cell source, neural stem/progenitor cells (NSPCs) exhibit immunosuppressive properties of great interest for modulating immune response in the central nervous system. To decipher the mechanisms of NSPC-mediated immunosuppression, activated T cells were exposed to NSPCs isolated from fetal rat brains. Analyses revealed that NSPCs inhibited T-cell proliferation and interferon-gamma production in a dose-dependent manner. A higher proportion of helper T cells (CD4+ T cells) was found in the presence of NSPCs, but analyses of FoxP3 population indicated that T-cell suppression was not secondary to an induction of suppressive regulatory T cells (FoxP3+ CD4+ CD25+). Conversely, induction of the high affinity interleukin-2 (IL-2) receptor (CD25) and the inability of IL-2 to rescue T-cell proliferation suggest that NSPCs display immunosuppressive activity without affecting T-cell activation. Cultures in Transwell chambers or addition of NSPC-conditioned medium to activated T cells indicated that part of the suppressive activity was not contact dependent. We therefore searched for soluble factors that mediate NSPC immunosuppression. We found that NSPCs express several immunosuppressive molecules, but the ability of these cells to inhibit T-cell proliferation was only counteracted by heme oxygenase (HO) inhibitors in association or not with nitric oxide synthase inhibitors. Taken together, our findings highlight a dynamic crosstalk between NSPCs and T lymphocytes and provide the first evidence of an implication of HO-1 in mediating the immunosuppressive effects of the NSPCs.


Subject(s)
Brain/metabolism , Heme Oxygenase-1/metabolism , Immunity, Innate , Neural Stem Cells/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Brain/cytology , Brain/immunology , Cell Communication/immunology , Cell Proliferation , Coculture Techniques , Embryo, Mammalian , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression/immunology , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/genetics , Interferon-gamma/immunology , Lymphocyte Activation/drug effects , Neural Stem Cells/cytology , Neural Stem Cells/immunology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology
19.
Methods Mol Biol ; 885: 233-43, 2012.
Article in English | MEDLINE | ID: mdl-22565999

ABSTRACT

Intracerebral cell transplantation offers the possibility of replacing lost neurons in case of neurodegenerative disorders. To date, the best functional recovery for Parkinson's patients has been obtained using neuroblasts derived from human foetal mesencephalon, but the ethical and practical problems relative to the use of human foetal tissue lead to consideration of alternative sources of cells. In this regard, porcine neuroblasts appear as a valuable source as these cells are available in large quantity and programmed to extend long neurites as human neurons. However, the potential use of pig neural cells in the clinical setting depends on efficient and safe immunosuppression. So, most experimental work in this domain aims at developing immunosuppressive treatments specifically adapted to the central nervous system. In such perspective, transplantation of porcine mesencephalic neuroblasts into the striatum of the adult rat brain is of great interest. Indeed, rejection of intracerebral xenografts has been quite well described in rats, and graft survival can be easily monitored in a rat model of Parkinson's disease. In the present chapter, we describe the methods for isolating neuroblasts from foetal porcine mesencephalon as well as the technique of intracerebral transplantation in adult immunocompetent rats.


Subject(s)
Mesencephalon/cytology , Neurons/transplantation , Transplantation, Heterologous/methods , Animals , Corpus Striatum/surgery , Disease Models, Animal , Fetus , Graft Survival , Humans , Immunohistochemistry , Parkinson Disease/therapy , Perfusion , Rats , Swine
20.
J Mol Neurosci ; 46(2): 431-41, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21809042

ABSTRACT

Immune signaling and neuroinflammatory mediators have recently emerged as influential variables that regulate neural precursor/stem cell (NPC) behavior and function. In this study, we investigated whether the signaling adaptor protein CD3ζ, a transmembrane protein involved in T cell differentiation and function and recently shown to regulate neuronal development in the central nervous system (CNS), may have a role in NPC differentiation. We analyzed the expression profile of CD3ζ in embryonic rat brain during neurogenic periods and in neurosphere-derived neural cells, and we investigated the action of CD3ζ on cell differentiation. We found that CD3ζ expression coincided with neuronal commitment, but its forced expression in NPCs prevented the production of neurons and oligodendrocytes, but not astroglial cells. This blockade of neuronal differentiation was operated through an ITAM-independent mechanism, but required the Asp36 of the CD3ζ transmembrane domain involved in membrane receptor interaction. Together, our findings show that ectopic CD3ζ expression in NPCs impaired their normal cell-fate specification and suggest that variations of CD3ζ expression in the developing CNS might result in neurodevelopmental anomalies.


Subject(s)
CD3 Complex/physiology , Cell Lineage , Gene Expression Regulation, Developmental , Nerve Tissue Proteins/physiology , Neural Stem Cells/enzymology , Neurogenesis/physiology , Amino Acid Substitution , Animals , CD3 Complex/biosynthesis , CD3 Complex/chemistry , CD3 Complex/genetics , Cell Survival , Cells, Cultured/enzymology , Enzyme Induction , Female , Gestational Age , Hippocampus/cytology , Hippocampus/embryology , Microscopy, Fluorescence , Mutagenesis, Site-Directed , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neural Stem Cells/pathology , Neurons/cytology , Neurons/enzymology , Olfactory Bulb/embryology , Olfactory Bulb/enzymology , Pregnancy , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...