Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
BMC Biol ; 21(1): 8, 2023 01 12.
Article in English | MEDLINE | ID: mdl-36635667

ABSTRACT

BACKGROUND: Regulatory T cells (Treg) in diverse species include CD4+ and CD8+ T cells. In all species, CD8+ Treg have been only partially characterized and there is no rat model in which CD4+ and CD8+ FOXP3+ Treg are genetically tagged. RESULTS: We generated a Foxp3-EGFP rat transgenic line in which FOXP3 gene was expressed and controlled EGFP. CD4+ and CD8+ T cells were the only cells that expressed EGFP, in similar proportion as observed with anti-FOXP3 antibodies and co-labeled in the same cells. CD4+EGFP+ Treg were 5-10 times more frequent than CD8+EGFP+ Treg. The suppressive activity of CD4+ and CD8+ Treg was largely confined to EGFP+ cells. RNAseq analyses showed similarities but also differences among CD4+ and CD8+ EGFP+ cells and provided the first description of the natural FOXP3+CD8+ Treg transcriptome. In vitro culture of CD4+ and CD8+ EGFP- cells with TGFbeta and IL-2 generated induced EGFP+ Treg. CD4+ and CD8+ EGFP+ Treg were expanded upon in vivo administration of a low dose of IL-2. CONCLUSIONS: This new and unique rat line constitutes a useful model to identify and isolate viable CD4+ and CD8+ FOXP3+ Treg. Additionally, it allows to identify molecules expressed in CD8+ Treg that may allow to better define their phenotype and function not only in rats but also in other species.


Subject(s)
CD8-Positive T-Lymphocytes , T-Lymphocytes, Regulatory , Rats , Animals , T-Lymphocytes, Regulatory/metabolism , CD8-Positive T-Lymphocytes/metabolism , Interleukin-2/genetics , Interleukin-2/metabolism , Transforming Growth Factor beta/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism
2.
J Adv Res ; 43: 163-174, 2023 01.
Article in English | MEDLINE | ID: mdl-36585106

ABSTRACT

INTRODUCTION: Although the physiological role of the C-terminal hydrolase domain of the soluble epoxide hydrolase (sEH-H) is well investigated, the function of its N-terminal phosphatase activity (sEH-P) remains unknown. OBJECTIVES: This study aimed to assess in vivo the physiological role of sEH-P. METHODS: CRISPR/Cas9 was used to generate a novel knock-in (KI) rat line lacking the sEH-P activity. RESULTS: The sEH-P KI rats has a decreased metabolism of lysophosphatidic acids to monoacyglycerols. KI rats grew almost normally but with less weight and fat mass gain while insulin sensitivity was increased compared to wild-type rats. This lean phenotype was more marked in males than in female KI rats and mainly due to decreased food consumption and enhanced energy expenditure. In fact, sEH-P KI rats had an increased lipolysis allowing to supply fatty acids as fuel to potentiate brown adipose thermogenesis under resting condition and upon cold exposure. The potentiation of thermogenesis was abolished when blocking PPARγ, a nuclear receptor activated by intracellular lysophosphatidic acids, but also when inhibiting simultaneously sEH-H, showing a functional interaction between the two domains. Furthermore, sEH-P KI rats fed a high-fat diet did not gain as much weight as the wild-type rats, did not have increased fat mass and did not develop insulin resistance or hepatic steatosis. In addition, sEH-P KI rats exhibited enhanced basal cardiac mitochondrial activity associated with an enhanced left ventricular contractility and were protected against cardiac ischemia-reperfusion injury. CONCLUSION: Our study reveals that sEH-P is a key player in energy and fat metabolism and contributes together with sEH-H to the regulation of cardiometabolic homeostasis. The development of pharmacological inhibitors of sEH-P appears of crucial importance to evaluate the interest of this promising therapeutic strategy in the management of obesity and cardiac ischemic complications.


Subject(s)
Epoxide Hydrolases , Heart Injuries , Obesity , Animals , Female , Male , Rats , CRISPR-Cas Systems , Epoxide Hydrolases/genetics , Epoxide Hydrolases/metabolism , Heart Diseases/genetics , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Injuries/genetics , Heart Injuries/metabolism , Heart Injuries/pathology , Insulin Resistance/genetics , Lysophospholipids , Obesity/genetics , Obesity/metabolism , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Reperfusion Injury/genetics
3.
Clin Transl Med ; 12(8): e988, 2022 08.
Article in English | MEDLINE | ID: mdl-36030499

ABSTRACT

BACKGROUND: Immune homeostasis requires fully functional Tregs with a stable phenotype to control autoimmunity. Although IL-34 is a cytokine first described as mainly involved in monocyte cell survival and differentiation, we recently described its expression by CD8+ Tregs in a rat model of transplantation tolerance and by activated FOXP3+ CD4+ and CD8+ Tregs in human healthy individuals. However, its role in autoimmunity and potential in human diseases remains to be determined. METHODS: We generated Il34-/- rats and using both Il34-/- rats and mice, we investigated their phenotype under inflammatory conditions. Using Il34-/- rats, we further analyzed the impact of the absence of expression of IL-34 for CD4+ Tregs suppressive function. We investigated the potential of IL-34 in human disease to prevent xenogeneic GVHD and human skin allograft rejection in immune humanized immunodeficient NSG mice. Finally, taking advantage of a biocollection, we investigated the correlation between presence of IL-34 in the serum and kidney transplant rejection. RESULTS: Here we report that the absence of expression of IL-34 in Il34-/- rats and mice leads to an unstable immune phenotype, with production of multiple auto-antibodies, exacerbated under inflammatory conditions with increased susceptibility to DSS- and TNBS-colitis in Il34-/- animals. Moreover, we revealed the striking inability of Il34-/- CD4+ Tregs to protect Il2rg-/- rats from a wasting disease induced by transfer of pathogenic cells, in contrast to Il34+/+ CD4+ Tregs. We also showed that IL-34 treatment delayed EAE in mice as well as GVHD and human skin allograft rejection in immune humanized immunodeficient NSG mice. Finally, we show that presence of IL-34 in the serum is associated with a longer rejection-free period in kidney transplanted patients. CONCLUSION: Altogether, our data emphasize on the crucial necessity of IL-34 for immune homeostasis and for CD4+ Tregs suppressive function. Our data also shows the therapeutic potential of IL-34 in human transplantation and auto-immunity. HIGHLIGHTS: -Absence of expression of IL-34 in Il34-/- rats and mice leads to an unstable immune phenotype, with a production of multiple auto-antibodies and exacerbated immune pathology under inflammatory conditions. -Il34-/- CD4+ Tregs are unable to protect Il2rg-/- rats from colitis induced by transfer of pathogenic cells. -IL-34 treatment delayed EAE in mice, as well as acute GVHD and human skin allograft rejection in immune-humanized immunodeficient NSG mice.


Subject(s)
Colitis , Graft vs Host Disease , Interleukins , T-Lymphocytes, Regulatory , Animals , Colitis/immunology , Forkhead Transcription Factors , Graft vs Host Disease/immunology , Homeostasis , Humans , Immune Tolerance , Interleukins/deficiency , Interleukins/genetics , Mice , Rats , T-Lymphocytes, Regulatory/immunology
4.
Methods Mol Biol ; 2495: 233-244, 2022.
Article in English | MEDLINE | ID: mdl-35696036

ABSTRACT

CRISPR/Cas9 system is a promising method for the generation of human disease models by genome editing in non-conventional experimental animals. Medium/large-sized animals like sheep have several advantages to study human diseases and medicine. Here, we present a protocol that describes the generation of an otoferlin edited sheep model via CRISPR-assisted single-stranded oligodinucleotide-mediated Homology-Directed Repair (HDR), through direct cytoplasmic microinjection in in vitro produced zygotes.Otoferlin is a protein expressed in the cochlear inner hair cells, with different mutations at the OTOF gene being the major cause of nonsyndromic recessive auditory neuropathy spectrum disorder in humans. By using this protocol, we reported for the first time an OTOF KI model in sheep with 17.8% edited lambs showing indel mutations, and 61.5% of them bearing knock-in mutations by HDR . The reported method establishes the bases to produce a deafness model to test novel therapies in human disorders related to OTOF mutations.


Subject(s)
CRISPR-Cas Systems , Deafness , Animals , Deafness/genetics , Gene Editing/methods , Humans , Mutation , Recombinational DNA Repair , Sheep
5.
Hepatology ; 75(4): 866-880, 2022 04.
Article in English | MEDLINE | ID: mdl-34687060

ABSTRACT

BACKGROUND AND AIMS: Pluripotent stem cell-derived hepatocytes differentiated in monolayer culture are known to have more fetal than adult hepatocyte characteristics. If numerous studies tend to show that this immature phenotype might not necessarily be an obstacle to their use in transplantation, other applications such as drug screening, toxicological studies, or bioartificial livers are reliant on hepatocyte functionality and require full differentiation of hepatocytes. New technologies have been used to improve the differentiation process in recent years, usually evaluated by measuring the albumin production and CYP450 activity. Here we used the complex production and most importantly the activity of the coagulation factor IX (FIX) produced by mature hepatocytes to assess the differentiation of hemophilia B (HB) patient's induced pluripotent stem cells (iPSCs) in both monolayer culture and organoids. APPROACH AND RESULTS: Indeed, HB is an X-linked monogenic disease due to an impaired activity of FIX synthesized by hepatocytes in the liver. We have developed an in vitro model of HB hepatocytes using iPSCs generated from fibroblasts of a severe HB patient. We used CRISPR/Cas9 technology to target the genomic insertion of a coagulation factor 9 minigene bearing the Padua mutation to enhance FIX activity. Noncorrected and corrected iPSCs were differentiated into hepatocytes under both two-dimensional and three-dimensional differentiation protocols and deciphered the production of active FIX in vitro. Finally, we assessed the therapeutic efficacy of this approach in vivo using a mouse model of HB. CONCLUSIONS: Functional FIX, whose post-translational modifications only occur in fully mature hepatocytes, was only produced in corrected iPSCs differentiated in organoids. Immunohistochemistry analyses of mouse livers indicated a good cell engraftment, and the FIX activity detected in the plasma of transplanted animals confirmed rescue of the bleeding phenotype.


Subject(s)
Hemophilia B , Induced Pluripotent Stem Cells , Liver, Artificial , Animals , Biomarkers , Cell Differentiation , Factor IX/genetics , Hemophilia B/genetics , Hemophilia B/therapy , Hepatocytes , Humans
6.
Transl Res ; 239: 103-123, 2022 01.
Article in English | MEDLINE | ID: mdl-34461306

ABSTRACT

CD160 is a member of the immunoglobulin superfamily with a pattern of expression mainly restricted to cytotoxic cells. To assess the functional relevance of the HVEM/CD160 signaling pathway in allogeneic cytotoxic responses, exon 2 of the CD160 gene was targeted by CRISPR/Cas9 to generate CD160 deficient mice. Next, we evaluated the impact of CD160 deficiency in the course of an alloreactive response. To that aim, parental donor WT (wild-type) or CD160 KO (knock-out) T cells were adoptively transferred into non-irradiated semiallogeneic F1 recipients, in which donor alloreactive CD160 KO CD4 T cells and CD8 T cells clonally expanded less vigorously than in WT T cell counterparts. This differential proliferative response rate at the early phase of T cell expansion influenced the course of CD8 T cell differentiation and the composition of the effector T cell pool that led to a significant decreased of the memory precursor effector cells (MPECs) / short-lived effector cells (SLECs) ratio in CD160 KO CD8 T cells compared to WT CD8 T cells. Despite these differences in T cell proliferation and differentiation, allogeneic MHC class I mismatched (bm1) skin allograft survival in CD160 KO recipients was comparable to that of WT recipients. However, the administration of CTLA-4.Ig showed an enhanced survival trend of bm1 skin allografts in CD160 KO with respect to WT recipients. Finally, CD160 deficient NK cells were as proficient as CD160 WT NK cells in rejecting allogeneic cellular allografts or MHC class I deficient tumor cells. CD160 may represent a CD28 alternative costimulatory molecule for the modulation of allogeneic CD8 T cell responses either in combination with costimulation blockade or by direct targeting of alloreactive CD8 T cells that upregulate CD160 expression in response to alloantigen stimulation.


Subject(s)
Antigens, CD/immunology , CD8-Positive T-Lymphocytes/immunology , Graft Rejection/etiology , Receptors, Immunologic/immunology , 4-1BB Ligand/metabolism , Allografts , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , CRISPR-Cas Systems , Cell Differentiation , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , Gene Expression Regulation , Genes, MHC Class I , Graft Rejection/immunology , Killer Cells, Natural/immunology , Lectins, C-Type/metabolism , Mice, Inbred Strains , Mice, Knockout , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Skin Transplantation , Thymocytes/immunology
7.
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
8.
PLoS One ; 15(12): e0243769, 2020.
Article in English | MEDLINE | ID: mdl-33320872

ABSTRACT

The success of inducing human pluripotent stem cells (hIPSC) offers new opportunities for cell-based therapy. Since B cells exert roles as effector and as regulator of immune responses in different clinical settings, we were interested in generating B cells from hIPSC. We differentiated human embryonic stem cells (hESC) and hIPSC into B cells onto OP9 and MS-5 stromal cells successively. We overcame issues in generating CD34+CD43+ hematopoietic progenitors with appropriate cytokine conditions and emphasized the difficulties to generate proper hematopoietic progenitors. We highlight CD31intCD45int phenotype as a possible marker of hematopoietic progenitors suitable for B cell differentiation. Defining precisely proper lymphoid progenitors will improve the study of their lineage commitment and the signals needed during the in vitro process.


Subject(s)
B-Lymphocytes/cytology , Cell Differentiation , Hematopoietic Stem Cells/cytology , Antigens, CD/metabolism , Embryonic Stem Cells/cytology , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Humans , Phenotype
9.
J Immunother Cancer ; 8(1)2020 01.
Article in English | MEDLINE | ID: mdl-32001504

ABSTRACT

BACKGROUND: Genome editing offers unique perspectives for optimizing the functional properties of T cells for adoptive cell transfer purposes. So far, PDCD1 editing has been successfully tested mainly in chimeric antigen receptor T (CAR-T) cells and human primary T cells. Nonetheless, for patients with solid tumors, the adoptive transfer of effector memory T cells specific for tumor antigens remains a relevant option, and the use of high avidity T cells deficient for programmed cell death-1 (PD-1) expression is susceptible to improve the therapeutic benefit of these treatments. METHODS: Here we used the transfection of CAS9/sgRNA ribonucleoproteic complexes to edit PDCD1 gene in human effector memory CD8+ T cells specific for the melanoma antigen Melan-A. We cloned edited T cell populations and validated PDCD1 editing through sequencing and cytometry in each T cell clone, together with T-cell receptor (TCR) chain's sequencing. We also performed whole transcriptomic analyses on wild-type (WT) and edited T cell clones. Finally, we documented in vitro and in vivo through adoptive transfer in NOD scid gamma (NSG) mice, the antitumor properties of WT and PD-1KO T cell clones, expressing the same TCR. RESULTS: Here we demonstrated the feasibility to edit PDCD1 gene in human effector memory melanoma-specific T lymphocytes. We showed that PD-1 expression was dramatically reduced or totally absent on PDCD1-edited T cell clones. Extensive characterization of a panel of T cell clones expressing the same TCR and exhibiting similar functional avidity demonstrated superior antitumor reactivity against a PD-L1 expressing melanoma cell line. Transcriptomic analysis revealed a downregulation of genes involved in proliferation and DNA replication in PD-1-deficient T cell clones, whereas genes involved in metabolism and cell signaling were upregulated. Finally, we documented the superior ability of PD-1-deficient T cells to significantly delay the growth of a PD-L1 expressing human melanoma tumor in an NSG mouse model. CONCLUSION: The use of such lymphocytes for adoptive cell transfer purposes, associated with other approaches modulating the tumor microenvironment, would be a promising alternative to improve immunotherapy efficacy in solid tumors.


Subject(s)
Immunotherapy, Adoptive/methods , Melanoma/immunology , Melanoma/therapy , Programmed Cell Death 1 Receptor/deficiency , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Line, Tumor , Female , Gene Editing , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Random Allocation , Transfection , Xenograft Model Antitumor Assays
10.
Stem Cell Res Ther ; 10(1): 221, 2019 07 29.
Article in English | MEDLINE | ID: mdl-31358055

ABSTRACT

BACKGROUND: Familial hypercholesterolemia type IIA (FH) is due to mutations in the low-density lipoprotein receptor (LDLR) resulting in elevated levels of low-density lipoprotein cholesterol (LDL-c) in plasma and in premature cardiovascular diseases. As hepatocytes are the only cells capable of metabolizing cholesterol, they are therefore the target cells for cell/gene therapy approaches in the treatment of lipid metabolism disorders. Furthermore, the LDLR has been reported to be involved in hepatitis C virus (HCV) entry into hepatocytes; however, its role in the virus infection cycle is still disputed. METHODS: We generated induced pluripotent stem cells (iPSCs) from a homozygous LDLR-null FH-patient (FH-iPSCs). We constructed a correction cassette bearing LDLR cDNA under the control of human hepatic apolipoprotein A2 promoter that targets the adeno-associated virus integration site AAVS1. We differentiated both FH-iPSCs and corrected FH-iPSCs (corr-FH-iPSCs) into hepatocytes to study statin-mediated regulation of genes involved in cholesterol metabolism. Upon HCV particle inoculation, viral replication and production were quantified in these cells. RESULTS: We showed that FH-iPSCs displayed the disease phenotype. Using homologous recombination mediated by the CRISPR/Cas9 system, FH-iPSCs were genetically corrected by the targeted integration of a correction cassette at the AAVS1 locus. Both FH-iPSCs and corr-FH-iPSCs were then differentiated into functional polarized hepatocytes using a stepwise differentiation approach (FH-iHeps and corr-FH-iHeps). The correct insertion and expression of the correction cassette resulted in restoration of LDLR expression and function (LDL-c uptake) in corr-FH-iHeps. We next demonstrated that pravastatin treatment increased the expression of genes involved in cholesterol metabolism in both cell models. Moreover, LDLR expression and function were also enhanced in corr-FH-iHeps after pravastatin treatment. Finally, we demonstrated that both FH-iHeps and corr-FH-iHeps were as permissive to viral infection as primary human hepatocytes but that virus production in FH-iHeps was significantly decreased compared to corr-FH-iHeps, suggesting a role of the LDLR in HCV morphogenesis. CONCLUSIONS: Our work provides the first LDLR-null FH cell model and its corrected counterpart to study the regulation of cholesterol metabolism and host determinants of HCV life cycle, and a platform to screen drugs for treating dyslipidemia and HCV infection.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing , Hepatitis C/pathology , Hyperlipoproteinemia Type II/pathology , Receptors, LDL/genetics , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Apolipoprotein A-II/genetics , Cell Differentiation , Cholesterol/metabolism , Hepacivirus/drug effects , Hepacivirus/physiology , Hepatitis C/drug therapy , Hepatitis C/virology , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Hyperlipoproteinemia Type II/metabolism , Induced Pluripotent Stem Cells/cytology , Phenotype , Plasmids/genetics , Plasmids/metabolism , Promoter Regions, Genetic , Proprotein Convertase 9/genetics , Proprotein Convertase 9/metabolism , Receptors, LDL/metabolism , Sofosbuvir/pharmacology , Sofosbuvir/therapeutic use , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism
11.
Sci Rep ; 7(1): 16554, 2017 11 29.
Article in English | MEDLINE | ID: mdl-29185448

ABSTRACT

The generation of gene-edited animals using the CRISPRs/Cas9 system is based on microinjection into zygotes which is inefficient, time consuming and demands high technical skills. We report the optimization of an electroporation method for intact rat zygotes using sgRNAs and Cas9 protein in combination or not with ssODNs (~100 nt). This resulted in high frequency of knockouts, between 15 and 50% of analyzed animals. Importantly, using ssODNs as donor template resulted in precise knock-in mutations in 25-100% of analyzed animals, comparable to microinjection. Electroporation of long ssDNA or dsDNA donors successfully used in microinjection in the past did not allow generation of genome-edited animals despite dsDNA visualization within zygotes. Thus, simultaneous electroporation of a large number of intact rat zygotes is a rapid, simple, and efficient method for the generation of a variety of genome-edited rats.


Subject(s)
CRISPR-Associated Protein 9/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats/physiology , Zygote/metabolism , Animals , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Electroporation/methods , Female , Genotype , Microscopy, Confocal , Mutation , Rats
12.
J Genet Genomics ; 43(5): 341-8, 2016 05 20.
Article in English | MEDLINE | ID: mdl-27209567

ABSTRACT

The recent emergence and application of engineered endonucleases have led to the development of genome editing tools capable of rapidly implementing various targeted genome editions in a wide range of species. Moreover, these novel tools have become easier to use and have resulted in a great increase of applications. Whilst gene knockout (KO) or knockin (KI) animal models are relatively easy to achieve, there is a bottleneck in the detection and analysis of these mutations. Although several methods exist to detect these targeted mutations, we developed a heteroduplex mobility assay on an automated microfluidic capillary electrophoresis system named HMA-CE in order to accelerate the genotyping process. The HMA-CE method uses a simple PCR amplification of genomic DNA (gDNA) followed by an automated capillary electrophoresis step which reveals a heteroduplexes (HD) signature for each mutation. This allows efficient discrimination of wild-type and genome-edited animals down to the single base pair level.


Subject(s)
Cost-Benefit Analysis , Electrophoresis, Capillary/instrumentation , Gene Editing , Genotyping Techniques/economics , Lab-On-A-Chip Devices , Animals , Electrophoresis, Capillary/economics , Genotyping Techniques/instrumentation , Mutation , Rats , Time Factors
13.
J Immunol ; 180(3): 1317-25, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18209025

ABSTRACT

In rats, tolerance to MHC-incompatible renal allografts can be induced by the administration of anti-donor class II Abs on the day of transplantation. In this study we explored the mechanisms involved in the maintenance phase of this tolerance by analyzing intragraft gene expression profiles by microarray in long-term accepted kidneys. Comparison of the gene expression patterns of tolerated to syngeneic kidneys revealed 5,954 differentially expressed genes (p < 0.05). Further analysis of this gene set revealed a key role for the wingless-type (WNT) signaling pathway, one of the pivotal pathways involved in cell regulation that has not yet been implicated in transplantation. Several genes within this pathway were significantly up-regulated in the tolerated grafts, particularly matrix metalloproteinase 7 (MMP7; fold change > 40). Analysis of several other pathway-related molecules indicated that MMP7 overexpression was the result of the noncanonical WNT signaling pathway. MMP7 expression was restricted to vascular smooth muscle cells and was specific to anti-class II Ab-induced tolerance, as it was undetectable in other models of renal and heart transplant tolerance and chronic rejection induced across the same strain combination. These results suggest a novel role for noncanonical WNT signaling in maintaining kidney transplant tolerance in this model, with MMP7 being a key target. Determining the mechanisms whereby MMP7 contributes to transplant tolerance may help in the development of new strategies to improve long-term graft outcome.


Subject(s)
Graft Survival/genetics , Histocompatibility Antigens Class II/drug effects , Immune Tolerance , Kidney Transplantation/immunology , Matrix Metalloproteinase 7/physiology , Wnt Proteins/physiology , Animals , Antibodies/administration & dosage , Gene Expression , Graft Survival/drug effects , Histocompatibility Antigens Class II/immunology , Kidney/drug effects , Kidney/immunology , Male , Matrix Metalloproteinase 7/genetics , Models, Animal , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Rats , Rats, Inbred Lew , Signal Transduction , Tissue Donors , Wnt Proteins/genetics
14.
Int Immunol ; 19(2): 193-201, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17208927

ABSTRACT

Toll-like receptors (TLRs) play a crucial role in the initiation of innate responses following microbial infection and also in adaptive immune responses by orchestrating the activation of different cell populations. TLRs are expressed at high levels in antigen-presenting cells and recent studies have demonstrated the expression and biological role of TLRs in mouse and human CD4(+) T cells. In this study, we analyzed TLR mRNA expression in rat CD4(+) T cells using stringent quantitative reverse transcription-PCR conditions enabling a direct comparison of the levels of each TLR. We show that TLR3, 5, 6 and 9 mRNAs are the highest TLRs expressed in rat CD4(+) T cells and that TLR5 mRNA is highly expressed in regulatory CD4(+) CD25(+) T cells. In addition, we show that the TLR9 ligand (TLR9L), CpG oligodeoxynucleotide, synergizes with anti-CD3 to induce proliferation of both CD4(+) CD25(-) and regulatory CD4(+) CD25(+) T cells and that TLR9L partially abrogates the suppressive activity mediated by regulatory CD4(+) CD25(+) T cells. This loss of suppression is in part due to the direct effect of TLR9L on effector T cells that are rendered more resistant to the regulation exerted by regulatory T cells. To our knowledge, this is the first study describing the expression of TLR mRNA in rat CD4(+) T cells and the capacity of TLR9L to directly regulate rat T cell responses. Thus, TLR9L may rapidly increase the host's adaptive immunity by expanding effector cells and also by attenuating the suppressive activity mediated by regulatory T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , T-Lymphocyte Subsets/immunology , Toll-Like Receptor 9/biosynthesis , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , CpG Islands , Interleukin-2 Receptor alpha Subunit/metabolism , Ligands , Oligodeoxyribonucleotides , RNA, Messenger/analysis , Rats , Rats, Inbred Lew , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocyte Subsets/metabolism
15.
Transplantation ; 82(9): 1185-93, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17102770

ABSTRACT

BACKGROUND: Progressively better therapies have largely prevented or at least effectively treated acute allograft rejection. Consequently, the long-term survival of solid organ transplants has increasingly become limited primarily by the development of chronic allograft rejection. The mechanisms of chronic rejection remain largely unknown and the induction of specific tolerance would be the ultimate achievement in transplant immunology. We previously demonstrated, in a fully major histocompatibility complex (MHC)-mismatched rat cardiac allograft combination, that a 20-day treatment with a deoxyspergualin (DSG) analogue, LF15-0195, induces allograft tolerance with the development of potent CD4CD25 regulatory T cells. In order to better characterize the mechanisms involved in allograft tolerance, we compared long-term tolerated allografts with allografts exhibiting signs of chronic rejection induced by donor-specific blood transfusion. METHODS: We analyzed both types of allografts for infiltration, alloantibody production and gene expression by histology, exhaustive microarray and quantitative reverse-transcriptase polymerase chain reaction. RESULTS: Interestingly, we observed in tolerated allografts an infiltrate as dense as the one observed in chronically rejected allografts and alloantibody deposits on graft endothelial cells. Prominent gene expression of many putative proinflammatory cytokines and genes related to cell activation or cytotoxicity were observed in tolerated allografts. However, we observed a specific upregulation of cytoprotective genes such as nitric oxide synthase, BclXL, and indoleamine 2,3 dioxygenase, and a poor in situ expression of immunoglobulin chain gene. CONCLUSIONS: This study demonstrates a state of accommodation of tolerated allografts and suggests the importance of early control of humoral immunity for the prevention of chronic rejection and the maintenance of long-term tolerance.


Subject(s)
Graft Rejection/immunology , Graft Survival/immunology , Guanidines/administration & dosage , Heart Transplantation/immunology , Immunosuppressive Agents/administration & dosage , Transplantation Tolerance/drug effects , Animals , Cytotoxicity, Immunologic/genetics , Gene Expression , Gene Expression Profiling , Genes, Immunoglobulin/genetics , Graft Rejection/genetics , Graft Rejection/prevention & control , Graft Survival/drug effects , Graft Survival/genetics , Inflammation Mediators/metabolism , Isoantibodies/blood , Male , Models, Biological , Rats , T-Lymphocytes, Regulatory/immunology , Transplantation Tolerance/genetics
16.
J Immunol ; 177(2): 1007-16, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16818757

ABSTRACT

Dendritic cells (DC) are a heterogeneous population of APC endowed with specific functions. The nature of the DC subset involved in the course of an immune response to a specific pathogen might be important for inducing the appropriate effectors. In addition, each DC subset might also exhibit intrinsic functional plasticity. In the rat, spleen DC can be separated into three morphological and phenotypical distinct subsets, namely CD4+, CD4-, and plasmacytoid DC (pDC), whose frequencies are strain dependent. We correlated the expression of TLR and nucleotide-binding oligomerization domain 2 (NOD2) in these DC subsets to their in vitro responsiveness to specific ligands. CD4- DC expressed high levels of TLR1, 2, 3, and 10 mRNA, low TLR4, 5, 6, 7, and 9, and very low, if any, TLR8. pDC had a restricted repertoire characterized by high TLR7 and 9. CD4+ DC expressed all TLR and 10-fold higher levels of NOD2 mRNA than CD4- and pDC. Upon stimulation by TLR and NOD2 ligands, each DC subset responded in quite a stereotyped fashion. TLR2/6, 3, 4, 5, 9, and NOD2 triggering induced CD4- DC to mature and produce high IL-12p40, low IL-10, and TNF-alpha. TLR7/8 and 9 triggering induced pDC to mature and produce copious amounts of IL-6, IL-12p40, and TNF-alpha and low IFN-alpha. CD4+ DC were very poor producers of inflammatory cytokines. This study suggests that the nature of spleen DC responses to pathogens is dependent on subset specific-stimulation rather than intrinsic plasticity.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Spleen/immunology , Spleen/metabolism , Toll-Like Receptors/biosynthesis , Animals , Cell Differentiation/immunology , Cell Separation , Cell Survival/immunology , Cells, Cultured , Cytokines/biosynthesis , Dendritic Cells/classification , Dendritic Cells/cytology , Interleukin-10/biosynthesis , Interleukin-12/biosynthesis , Interleukin-12 Subunit p40 , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Leukocyte Count , Ligands , Lymphocyte Culture Test, Mixed , Nod2 Signaling Adaptor Protein , Protein Subunits/biosynthesis , RNA, Messenger/biosynthesis , Rats , Rats, Inbred BN , Rats, Inbred Lew , Rats, Sprague-Dawley , Spleen/cytology , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
17.
J Immunol ; 176(2): 839-47, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16393967

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) is an instructive model for the human demyelinating disease multiple sclerosis. Lewis (LEW) rats immunized with myelin-basic protein (MBP) develop EAE characterized by a single episode of paralysis, from which they recover spontaneously and become refractory to a second induction of disease. LF 15-0195 is a novel molecule that has potent immunosuppressive effects in several immune-mediated pathological manifestations, including EAE. In the present study, we show that a 30-day course of LF 15-0195 treatment not only prevents MBP-immunized LEW rats from developing EAE but also preserves their refractory phase to reinduction of disease. This effect is Ag driven since it requires priming by the autoantigen during the drug administration. In contrast to other immunosuppressive drugs, short-term treatment with this drug induces a persistent tolerance with no rebound of EAE up to 4 mo after treatment withdrawal. This beneficial effect of LF 15-0195 on EAE does not result from the deletion of MBP-specific Vbeta8.2 encephalitogenic T cells. In contrast, this drug favors the differentiation of MBP-specific CD4 T cells into Foxp3-expressing regulatory T cells that, upon adoptive transfer in syngeneic recipients, prevent the development of actively induced EAE. Finally, we demonstrate that the tolerance induced by LF 15-0195 treatment is not dependent on the presence of TGF-beta. Together, these data demonstrate that short-term treatment with LF 15-0195 prevents MBP-immunized LEW rats from EAE by favoring the development of Foxp-3-expressing regulatory CD4 T cells.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Forkhead Transcription Factors/genetics , Guanidines/pharmacology , Immunosuppressive Agents/pharmacology , Adoptive Transfer , Animals , Base Sequence , CD4-Positive T-Lymphocytes/metabolism , DNA/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Gene Expression/drug effects , Humans , Male , Multiple Sclerosis/immunology , Multiple Sclerosis/prevention & control , Myelin Basic Protein/immunology , Neutralization Tests , Rats , Rats, Inbred Lew , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Chemokine/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism
18.
Transplantation ; 80(10): 1476-84, 2005 Nov 27.
Article in English | MEDLINE | ID: mdl-16340794

ABSTRACT

BACKGROUND: We have previously demonstrated that a short-course treatment with LF15-0195, a 15-deoxyspergualin analogue, induces donor-specific tolerance of cardiac allografts in rats and expansion of splenic CD4CD25 regulatory T cells. METHODS: To further characterize long-term tolerance in this model, we have analyzed the phenotype, regulatory properties and TCR-Vbeta usage of the T cells infiltrating the tolerated allografts. RESULTS: We demonstrate that the tolerated allografts express high levels of FoxP3 transcripts and contain a large number of CD4 T cells, half of which express CD25. Moreover, T cells from these tolerated allografts are very powerful at transferring tolerance to a subsequent allograft recipient, demonstrating the presence of potent regulatory T cells at the site of the graft. Interestingly, the T cells infiltrating the tolerated allografts systematically display restricted Vbeta7 TCR rearrangements. CONCLUSION: These results demonstrate in this model of tolerance, a specific accumulation of T cells with potent regulatory properties and exhibiting restricted Vbeta7-TCR rearrangements at the graft site.


Subject(s)
Gene Rearrangement, beta-Chain T-Cell Antigen Receptor/immunology , Guanidines/pharmacology , Heart Transplantation/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes, Regulatory/physiology , Transplantation Tolerance/immunology , Animals , Heart Transplantation/pathology , Male , Rats , Rats, Inbred Lew , Transplantation Tolerance/drug effects
19.
Am J Transplant ; 5(9): 2143-53, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16095493

ABSTRACT

We identified a novel rat gene specifically overexpressed in tolerated heart allografts in a model of tolerance induced by donor-specific blood transfusion (DST). We named this gene TORID, for tolerance-related and induced transcript. We show that TORID expression can be attributed to non-T cells infiltrating tolerated grafts. Interestingly, TORID overexpression was also observed in long-term grafts from a different model of tolerance in which chronic rejection does not occur. Comparison of the predicted amino acid sequence of TORID and of its human counterpart LR8 showed an homology with the four-transmembrane CD20/FcepsilonRIbeta family proteins. We investigated TORID expression in naive rat immune cells and lymphoid tissues. TORID was found to be preferentially expressed in cells of the myeloid lineage such as macrophages and dendritic cells (DCs). Its expression dramatically decreased following activation/maturation. Similar results were obtained in human monocyte-derived DCs. Interestingly, TORID overexpression in bone marrow-derived DCs alters expression of MHC II and CD86 and production of IL12p40 following activation. These results suggest that TORID may be involved in the control of DC maturation and may, therefore, play a role in the induction or maintenance of allograft tolerance.


Subject(s)
Antigens, CD20/metabolism , Receptors, IgE/metabolism , Transplantation, Homologous/methods , Adenoviridae/genetics , Amino Acid Sequence , Animals , Antigens, CD20/chemistry , Blood Transfusion , Bone Marrow Cells/cytology , Cell Lineage , Chromosome Mapping , Cloning, Molecular , DNA, Complementary/metabolism , Dendritic Cells/cytology , Down-Regulation , Gene Transfer Techniques , Graft Rejection , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Interleukin-12/metabolism , Interleukin-12 Subunit p40 , Lymphocyte Activation , Macrophages/cytology , Macrophages/metabolism , Mice , Molecular Sequence Data , Monocytes/cytology , Multigene Family , Nucleic Acid Hybridization , Oligonucleotides/chemistry , Phylogeny , Polymerase Chain Reaction , Protein Subunits/metabolism , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Receptors, IgE/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Spleen/metabolism , Time Factors , Transplantation Tolerance
20.
Transplantation ; 78(9): 1259-66, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15548961

ABSTRACT

BACKGROUND: Donor-specific tolerance to heart allografts in the rat can be achieved by donor-specific blood transfusions (DST) before transplantation. This tolerance induction requires the presence of host CD8 T cells and is characterized by the infiltration of numerous leukocytes. METHODS: To identify new mediators involved in tolerance induction, gene searching was performed and resulted in the identification of the Fractalkine receptor, CX3CR1, as being highly expressed in tolerated allografts. RESULTS: We showed that the high CX3CR1 mRNA accumulation found in tolerated allografts was related to the active recruitment of monocytes/macrophages. CX3CR1 transcript accumulation was preceded by an early expression of its ligand, Fractalkine, by graft endothelial cells. Interestingly, depletion of recipient CD8 cells led to a dramatic decrease in both CX3CR1 and Fractalkine mRNA levels. Moreover, in vitro, CD8 T cells from DST-primed animals were found to strongly induce Fractalkine expression in an allogeneic endothelial cell line. CONCLUSION: This is the first report describing Fractalkine, a chemokine usually described in inflammatory processes, as being expressed in a model of allograft tolerance.


Subject(s)
Blood Transfusion , CD8-Positive T-Lymphocytes/immunology , Chemokines, CX3C/biosynthesis , Immune Tolerance , Membrane Proteins/biosynthesis , Receptors, Cytokine/biosynthesis , Receptors, HIV/biosynthesis , Animals , CX3C Chemokine Receptor 1 , Chemokine CX3CL1 , Male , RNA, Messenger/analysis , Rats , Rats, Inbred Lew , Receptors, Cytokine/genetics , Receptors, HIV/genetics , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...