Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
Front Immunol ; 15: 1285215, 2024.
Article En | MEDLINE | ID: mdl-38629063

The analytical capability of flow cytometry is crucial for differentiating the growing number of cell subsets found in human blood. This is important for accurate immunophenotyping of patients with few cells and a large number of parameters to monitor. Here, we present a 43-parameter panel to analyze peripheral blood mononuclear cells from healthy individuals using 41 fluorescence-labelled monoclonal antibodies, an autofluorescent channel, and a viability dye. We demonstrate minimal population distortions that lead to optimized population identification and reproducible results. We have applied an advanced approach in panel design, in selection of sample acquisition parameters and in data analysis. Appropriate autofluorescence identification and integration in the unmixing matrix, allowed for resolution of unspecific signals and increased dimensionality. Addition of one laser without assigned fluorochrome resulted in decreased fluorescence spill over and improved discrimination of cell subsets. It also increased the staining index when autofluorescence was integrated in the matrix. We conclude that spectral flow cytometry is a highly valuable tool for high-end immunophenotyping, and that fine-tuning of major experimental steps is key for taking advantage of its full capacity.


Fluorescent Dyes , Leukocytes, Mononuclear , Humans , Antibodies, Monoclonal , Leukocyte Count , Light
2.
J Crohns Colitis ; 14(5): 669-679, 2020 Jun 19.
Article En | MEDLINE | ID: mdl-31784737

BACKGROUND AND AIMS: Nucleotide oligomerization domain 2 [NOD2] mutations are key risk factors for Crohn's disease [CD]. NOD2 contributes to intestinal homeostasis by regulating innate and adaptive immunity together with intestinal epithelial function. However, the exact roles of NOD2 in CD and other NOD2-associated disorders remain poorly known. METHODS: We initially observed that NOD2 expression was increased in epithelial cells away from inflamed areas in CD patients. To explore this finding, Nod2 mRNA expression, inflammation, and cytokines expression were examined in the small bowel of wild-type [WT], Nod2 knockout and Nod2 mutant mice after rectal instillation of 2,4,6-trinitrobenzene sulphonic acid [TNBS]. RESULTS: In WT mice, Nod2 upregulation upstream to rectal injury was associated with pro-inflammatory cytokine expression but no overt histological inflammatory lesions. Conversely, in Nod2-deficient mice the inflammation spread from colitis to ileum and duodenum. CONCLUSIONS: Nod2 protects the gut from colitis spreading to small intestine.


Colitis/genetics , Duodenitis/genetics , Ileitis/genetics , Intestinal Mucosa/metabolism , Nod2 Signaling Adaptor Protein/genetics , RNA, Messenger/metabolism , Animals , Cecum/metabolism , Cecum/pathology , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Crohn Disease/metabolism , Crohn Disease/pathology , Duodenitis/chemically induced , Duodenitis/metabolism , Duodenitis/pathology , Duodenum/metabolism , Duodenum/pathology , Gene Expression , Humans , Ileitis/chemically induced , Ileitis/metabolism , Ileitis/pathology , Ileum/metabolism , Ileum/pathology , Interferon-gamma/metabolism , Interleukin-12/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Nod2 Signaling Adaptor Protein/metabolism , Trinitrobenzenesulfonic Acid , Tumor Necrosis Factor-alpha/metabolism
3.
Inflamm Bowel Dis ; 23(7): 1109-1119, 2017 07.
Article En | MEDLINE | ID: mdl-28520587

BACKGROUND: Crohn's disease (CD) pathogenesis is multifactorial involving genetic and environmental factors. Loss of function mutations in the nucleotide oligomerization domain 2 (NOD2) gene are the main genetic risk factor for CD. Like patients with CD, Nod2 mice are characterized by an enhanced Th1 immune response and a defective mucosal barrier function evidenced by increased intestinal permeability. We previously showed that the latter is related to hematopoietic Nod2 deficiency. Our aim was to explore the mechanisms by which Nod2 expressed in the hematopoietic and in the nonhematopoietic compartments interplay to control epithelial paracellular permeability. METHODS: Depletion of CD4 T cells in Nod2 mice and treatments with inhibitors were conducted in chimeric mice transplanted with bone marrow cells from Nod2-deficient donors into Nod2-sufficient recipients or vice versa. Caco-2 cells overexpressing a NOD2 gene which did or did not include a CD-associated polymorphism were treated with inhibitors or siRNAs and cocultured with hematopoietic cells from Peyer's patches. RESULTS: In vivo and in vitro Nod2 in hematopoietic cells regulates epithelial paracellular permeability through cytokine production influencing myosin light chain kinase (MLCK) activity. Indeed, tumor necrosis factor-α and interferon-γ secretion by CD4 T cells upregulated expression and activity of epithelial MLCK leading to increased epithelial tight junction opening. When stimulated by muramyl dipeptide, Nod2 in the nonhematopoietic compartment normalized the permeability and T-cell cytokine secretion and regulated MLCK activity. This MLCK regulation is mediated by TAK1 and RICK-dependent mechanisms. CONCLUSIONS: Our study demonstrates how hematopoietic and nonhematopoietic Nod2 regulate intestinal barrier function, improving our knowledge on the mechanisms involved in CD pathogenesis.


Hematopoietic Stem Cells/metabolism , Intestinal Mucosa/metabolism , Myosin-Light-Chain Kinase/metabolism , Nod2 Signaling Adaptor Protein/physiology , Peyer's Patches/metabolism , Animals , Caco-2 Cells , Cell Membrane Permeability , Humans , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Tight Junctions
4.
Immunity ; 45(3): 610-625, 2016 09 20.
Article En | MEDLINE | ID: mdl-27612641

The nature of gut intraepithelial lymphocytes (IELs) lacking antigen receptors remains controversial. Herein we showed that, in humans and in mice, innate intestinal IELs expressing intracellular CD3 (iCD3(+)) differentiate along an Id2 transcription factor (TF)-independent pathway in response to TF NOTCH1, interleukin-15 (IL-15), and Granzyme B signals. In NOTCH1-activated human hematopoietic precursors, IL-15 induced Granzyme B, which cleaved NOTCH1 into a peptide lacking transcriptional activity. As a result, NOTCH1 target genes indispensable for T cell differentiation were silenced and precursors were reprogrammed into innate cells with T cell marks including intracellular CD3 and T cell rearrangements. In the intraepithelial lymphoma complicating celiac disease, iCD3(+) innate IELs acquired gain-of-function mutations in Janus kinase 1 or Signal transducer and activator of transcription 3, which enhanced their response to IL-15. Overall we characterized gut T cell-like innate IELs, deciphered their pathway of differentiation and showed their malignant transformation in celiac disease.


Celiac Disease/immunology , Interleukin-15/immunology , Intestines/immunology , Lymphoma/immunology , T-Lymphocyte Subsets/immunology , Animals , CD3 Complex/immunology , Cell Differentiation/immunology , Cells, Cultured , Granzymes/immunology , Humans , Inhibitor of Differentiation Protein 2/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Receptor, Notch1/immunology , STAT3 Transcription Factor/immunology , Signal Transduction/immunology , Transcription, Genetic/immunology
5.
Inflamm Bowel Dis ; 22(10): 2369-81, 2016 10.
Article En | MEDLINE | ID: mdl-27598740

BACKGROUND: Aside from cases of backwash ileitis, the ileal mucosa of patients with ulcerative colitis (UC), an idiotypic inflammatory bowel disease, has received little attention despite the fact that colitis is known to trigger alterations in morphology and/or functions of the small intestine remotely. METHODS: The ileal mucosa was studied in patients with UC and in a spontaneous model of colitis (Il10/Nox1 mice) mimicking the histological and clinical features of UC and was also studied in acute and chronic murine models of chemically induced colitis. Proliferation and apoptosis were assessed using morphological and immunohistological methods and Western blot analysis. Peyer's patch immune cell subsets were analyzed. Cytokines levels were quantified using quantitative PCR and Luminex xMAP technology. Total RNA from isolated ileal crypts was used for whole genome transcriptome analysis. RESULTS: The most striking features were an increased ileal crypt length associated with an enhanced cell proliferation of the transit-amplifying cells along with activation of the Wnt/ß-catenin and MAPkinase pathways. These changes did not result from intestinal inflammation as assessed by histology and/or pro-inflammatory cytokine expression levels. The increased proliferation rate was dependent on the duration but not on the severity of colitis and was observed in different mouse models of colitis, including the Il10/Nox1 model and 2,4,6-trinitrobenzenesulfonic acid-treated mice. Interestingly, the ileal mucosa of patients with UC also displayed longer crypts and enhanced cell proliferation compared with control patients. CONCLUSIONS: These data show that despite the absence of inflammation in the small intestine, alterations in the ileal mucosa homeostasis are present in UC.


Cell Proliferation/physiology , Colitis, Ulcerative/physiopathology , Ileum/physiopathology , Intestinal Mucosa/physiopathology , Animals , Case-Control Studies , Colitis, Ulcerative/etiology , Colitis, Ulcerative/pathology , Humans , Intestinal Mucosa/pathology , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C57BL , Trinitrobenzenesulfonic Acid , Wnt Signaling Pathway/physiology , beta Catenin/physiology
6.
J Crohns Colitis ; 10(12): 1428-1436, 2016 Dec.
Article En | MEDLINE | ID: mdl-27147452

BACKGROUND AND AIMS: Crohn's disease [CD] is a complex disorder characterised by an inappropriate immune response, impaired barrier function and microbial dysbiosis. Mutations in nucleotide oligomeriation domain 2 [NOD2] are CD risk factors. Increase of intestinal permeability, CD4+ T cell infiltration, and bacterial dysbiosis are also seen in Nod2-knockout [Nod2 KO] mice. However, the specificity and relationship between these Nod2-associated abnormalities remain largely unexplored. METHODS: Wild-type [WT], Nod1-knockout [Nod1 KO] and Nod2 KO mice were analysed in parallel. Microbial composition was defined by 454-pyrosequencing of bacterial 16S rRNA genes. Mucin and antimicrobial peptide expression was assessed by RT-PCR. Cell populations from Peyer's patches were determined by flow cytometry. Ussing chambers were used to measure intestinal permeability and bacterial translocation. Finally, to explore the impact of colonisation with mother's microbiota at birth, analyses were also performed in Nod2 KO and WT mice born from WT surrogate mothers after embryo transfer. RESULTS: Nod2 KO mice exhibited colonic bacterial dysbiosis different from WT and Nod1 KO mice. Altered expression of antimicrobial peptides and mucins in ileum and colon was associated with the microbial composition. Bacterial composition of Nod2 KO and WT mice obtained by embryo transfer was similar to that observed in Nod2 KO mice, arguing for a dominant effect of Nod2 KO-associated dysbiosis. In contrast, increased levels of CD4+ T cells and gut barrier defects across Peyer's patches were specific to Nod2 deficiency and independent of Microbial dysbiosis. CONCLUSIONS: Nod2 deficiency is associated with a specific dominant dysbiosis which does not drive mucosal tissue and immune alterations.


Dysbiosis/physiopathology , Gastrointestinal Microbiome/physiology , Intestinal Mucosa/physiopathology , Nod2 Signaling Adaptor Protein/deficiency , Animals , Antimicrobial Cationic Peptides/metabolism , Female , Gastrointestinal Microbiome/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucins/metabolism , Nod2 Signaling Adaptor Protein/physiology , Peyer's Patches/physiopathology , RNA, Ribosomal, 16S/genetics , Real-Time Polymerase Chain Reaction
7.
Inflamm Bowel Dis ; 22(4): 763-73, 2016 Apr.
Article En | MEDLINE | ID: mdl-26963567

BACKGROUND: NOD2 mutations are associated with Crohn's disease (CD). Both CD (in human) and Nod2 deficiency (in mice) are characterized by increased mucosal CD4 T-cells, an altered permeability and a microbial dysbiosis. However, the respective roles of the gut epithelial and immune compartments on the phenotype are not known. METHODS: Microbial composition, epithelial peptide secretion, intestinal permeability, and immune cell composition of Peyer patches were studied in Nod2 knock-out mice transplanted with wild-type bone marrow cells and vice versa. RESULTS: The nonhematopoietic cells control the microbiota composition and epithelial secretion of mucins and antimicrobial peptides. These parameters are correlated with recurrent associations between bacterial species and luminal products. In contrast, Nod2 in the hematopoietic compartment regulates the epithelial permeability and the gut-associated lymphoid tissue independently of the bacterial composition. CONCLUSIONS: The immune system and the gut permeability in one hand and the microbial and epithelial peptide compositions in the other hand are separate couples of interdependent parameters, both controlled by Nod2 in either the hematopoietic or nonhematopoietic lineages.


Dysbiosis/microbiology , Gastrointestinal Microbiome , Hematopoietic Stem Cells/microbiology , Homeostasis/physiology , Intestinal Mucosa/microbiology , Nod2 Signaling Adaptor Protein/physiology , Animals , Dysbiosis/metabolism , Dysbiosis/pathology , Hematopoietic Stem Cells/metabolism , Intestinal Mucosa/metabolism , Mice , Mice, Knockout
8.
Inflamm Bowel Dis ; 21(3): 543-55, 2015 Mar.
Article En | MEDLINE | ID: mdl-25659087

BACKGROUND: Ileal Crohn's disease is related to NOD2 mutations and to a gut barrier dysfunction. Pseudomonas fluorescens has also been associated with ileal Crohn's disease. The aim of this study was to determine the impact of P. fluorescens on the paracellular permeability in ileum and Peyer's patches. METHODS: To explore this question, in vivo and ex vivo experiments were performed in wild-type, Nod2, Nod2, and IL-1R mice together with in vitro analyses using the Caco-2 (epithelial) and the THP-1 (monocyte) human cell lines. RESULTS: Pseudomonas fluorescens increased the paracellular permeability of the intestinal mucosa through the secretion of IL-1ß by the immune cell populations and the activation of myosin light chain kinase in the epithelial cells. Induction of the IL-1ß pathway required the expression of Nod2 in the hematopoietic compartment, and muramyl dipeptide (a Nod2 ligand) had an inhibitory effect. CONCLUSIONS: Pseudomonas fluorescens thus alters the homeostasis of the epithelial barrier function by a mechanism similar to that previously observed for Yersinia pseudotuberculosis. This work further documents a putative role of psychrotrophic bacteria in Crohn's disease.


Hematopoietic Stem Cells/metabolism , Intestinal Mucosa/metabolism , Macrophages/metabolism , Nod2 Signaling Adaptor Protein/physiology , Pseudomonas fluorescens/physiology , Receptors, Interleukin-1/physiology , Animals , Blotting, Western , Caco-2 Cells , Cell Membrane Permeability , Cells, Cultured , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/microbiology , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/microbiology , Macrophages/cytology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
9.
PLoS One ; 9(7): e101669, 2014.
Article En | MEDLINE | ID: mdl-25014110

Ulcerative colitis (UC) is a chronic inflammatory bowel disease affecting the rectum which progressively extents. Its etiology remains unknown and the number of treatments available is limited. Studies of UC patients have identified an unbalanced endoplasmic reticulum (ER) stress in the non-inflamed colonic mucosa. Animal models with impaired ER stress are sensitive to intestinal inflammation, suggesting that an unbalanced ER stress could cause inflammation. However, there are no ER stress-regulating strategies proposed in the management of UC partly because of the lack of relevant preclinical model mimicking the disease. Here we generated the IL10/Nox1dKO mouse model which combines immune dysfunction (IL-10 deficiency) and abnormal epithelium (NADPH oxidase 1 (Nox1) deficiency) and spontaneously develops a UC-like phenotype with similar complications (colorectal cancer) than UC. Our data identified an unanticipated combined role of IL10 and Nox1 in the fine-tuning of ER stress responses in goblet cells. As in humans, the ER stress was unbalanced in mice with decreased eIF2α phosphorylation preceding inflammation. In IL10/Nox1dKO mice, salubrinal preserved eIF2α phosphorylation through inhibition of the regulatory subunit of the protein phosphatase 1 PP1R15A/GADD34 and prevented colitis. Thus, this new experimental model highlighted the central role of epithelial ER stress abnormalities in the development of colitis and defined the defective eIF2α pathway as a key pathophysiological target for UC. Therefore, specific regulators able to restore the defective eIF2α pathway could lead to the molecular remission needed to treat UC.


Colitis, Ulcerative/etiology , Disease Models, Animal , Endoplasmic Reticulum Stress , Inflammation/etiology , Interleukin-10/physiology , NADH, NADPH Oxidoreductases/physiology , Animals , Blotting, Western , Case-Control Studies , Cell Proliferation , Cells, Cultured , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Colon/immunology , Colon/metabolism , Colon/pathology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Inflammation/metabolism , Inflammation/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 1 , Phosphorylation , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Unfolded Protein Response
10.
Gastroenterology ; 146(4): 1017-27, 2014 Apr.
Article En | MEDLINE | ID: mdl-24361466

BACKGROUND & AIMS: CD4(+) T cells specific for dietary gluten and interleukin 15 (IL15) contribute to the pathogenesis of celiac disease. We investigated whether and how they interact to damage the intestine using mice that overexpress human IL15 in the intestinal epithelium and have CD4(+) T cells specific for ovalbumin, a dietary antigen. METHODS: We crossed mice with CD4(+) T cells specific for ovalbumin (OTII) with mice that overexpress human IL15 under an intestine-specific promoter (B6 × IL15Tge). The offspring (OTII × IL15Tge mice) received control or ovalbumin-containing diets until 3 months of age. Enteropathy was monitored by weight, ratio of villous:crypt length, and the number of intestinal lymphocytes. Phenotype, cytokine production, and degranulation of mucosal and spleen lymphocytes were analyzed by multicolor flow cytometry or enzyme-linked immunosorbent assay. Regulatory T-cell function and CD8(+) T-cell activation were analyzed in co-culture assays. RESULTS: Exposure to ovalbumin reduced growth and led to enteropathy in OTII × IL15Tge mice but not in control OTII × B6 littermates. Enteropathy was associated with expansion of mucosal granzyme B(+) CD8(+) T cells, and developed despite increased frequency of functional ovalbumin-specific regulatory T cells. Ovalbumin-activated CD4(+) T cells secreted IL2, which along with IL15 stimulated expansion of noncognate intestinal cytotoxic CD8(+) T cells, which did not respond to regulatory T cells and induced epithelial damage. CONCLUSIONS: We observed that in mice given food antigen, cooperation between IL15 and CD4(+) T cells is necessary and sufficient to activate CD8(+) T cells and damage the small intestine. We propose that this process is involved in the development of celiac disease.


Antigens , CD4-Positive T-Lymphocytes/immunology , Celiac Disease/immunology , Diet , Interleukin-15/metabolism , Intestine, Small/immunology , Ovalbumin/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Celiac Disease/metabolism , Celiac Disease/pathology , Cell Degranulation , Cell Proliferation , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Disease Models, Animal , Granzymes/metabolism , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Humans , Immunity, Mucosal , Interleukin-15/genetics , Interleukin-2/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Intestine, Small/pathology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/administration & dosage , Phenotype , Signal Transduction , Spleen/immunology , Spleen/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , beta 2-Microglobulin/deficiency , beta 2-Microglobulin/genetics
11.
Gastroenterology ; 143(6): 1470-1472.e2, 2012 Dec.
Article En | MEDLINE | ID: mdl-22922421

Large granular lymphocyte leukemia (LGL) is characterized by clonal expansion of CD3+ T cells or CD3(-) natural killer cells and frequently is associated with autoimmune diseases. We describe 2 patients with celiac disease who no longer responded to gluten-free diets after they developed T-cell LGL, with intestinal localization of malignant lymphocytes. Flow cytometry phenotyping of isolated intestinal intraepithelial and lamina propria cells eliminated type II refractory celiac disease, identifying large-sized CD8(+)CD57(+) T cells. Treatment with a combination of cyclosporine and methotrexate restored the patients' sensitivity to gluten-free diets. LGL therefore might be a cause of refractory celiac disease that is sensitive to immunosuppressive therapy.


Celiac Disease/diet therapy , Diet, Gluten-Free , Immunosuppressive Agents/therapeutic use , Leukemia, Large Granular Lymphocytic/drug therapy , Aged , Celiac Disease/pathology , Cyclosporine/therapeutic use , Drug Therapy, Combination , Female , Humans , Leukemia, Large Granular Lymphocytic/pathology , Methotrexate/therapeutic use , Middle Aged , Treatment Failure , Treatment Outcome
12.
Immunity ; 37(1): 108-21, 2012 Jul 27.
Article En | MEDLINE | ID: mdl-22705105

With the goal in mind to define how interleukin-15 (IL-15) contributes to acute intestinal inflammation, we have used a mouse model of ileitis induced by oral infection with Toxoplasma gondii. We observed that a crosstalk between IL-15 and interleukin-18 (IL-18) promoted intestinal recruitment of inflammatory monocytes, where these cells participated in parasite control but also in tissue damage. A stromal source of IL-15 controlled the development of lamina propria NKp46(+)NK1.1(+) cells, whereas IL-18 produced during T. gondii infection stimulated their production of the chemokine CCL3. In turn, CCL3 attracted inflammatory monocytes via their chemokine receptor CCR1, which was indispensable for their recruitment into the inflamed gut. Collectively, these results identify the IL-15-dependent subset of intestinal NKp46(+) cells as an important source of CCL3, which can amplify intestinal inflammation via the recruitment of CCR1(+) inflammatory monocytes. Preliminary evidence suggests that this pathway might operate in Crohn's disease.


Antigens, Ly/metabolism , Enteritis/immunology , Interleukin-15/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Monocytes/immunology , Natural Cytotoxicity Triggering Receptor 1/metabolism , Adolescent , Animals , Chemokine CCL3/metabolism , Child , Crohn Disease/immunology , Crohn Disease/metabolism , Enteritis/metabolism , Enteritis/parasitology , Humans , Interleukin-15/genetics , Interleukin-18/immunology , Interleukin-18/metabolism , Interleukin-7 Receptor alpha Subunit/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/parasitology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Receptors, CCR1/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Toxoplasma/immunology , Toxoplasma/metabolism
13.
J Clin Invest ; 122(6): 2239-51, 2012 Jun.
Article En | MEDLINE | ID: mdl-22565313

Intestinal barrier function requires intricate cooperation between intestinal epithelial cells and immune cells. Enteropathogens are able to invade the intestinal lymphoid tissue known as Peyer's patches (PPs) and disrupt the integrity of the intestinal barrier. However, the underlying molecular mechanisms of this process are poorly understood. In mice infected with Yersinia pseudotuberculosis, we found that PP barrier dysfunction is dependent on the Yersinia virulence plasmid and the expression of TLR-2 by hematopoietic cells, but not by intestinal epithelial cells. Upon TLR-2 stimulation, Y. pseudotuberculosis-infected monocytes activated caspase-1 and produced IL-1ß. In turn, IL-1ß increased NF-κB and myosin light chain kinase activation in intestinal epithelial cells, thus disrupting the intestinal barrier by opening the tight junctions. Therefore, Y. pseudotuberculosis subverts intestinal barrier function by altering the interplay between immune and epithelial cells during infection.


Hematopoietic Stem Cells/immunology , Intestinal Mucosa/immunology , Peyer's Patches/immunology , Signal Transduction/immunology , Toll-Like Receptor 2/immunology , Yersinia pseudotuberculosis Infections/immunology , Yersinia pseudotuberculosis/immunology , Animals , Caco-2 Cells , Caspase 1/genetics , Caspase 1/immunology , Enzyme Activation/genetics , Enzyme Activation/immunology , Hematopoietic Stem Cells/microbiology , Hematopoietic Stem Cells/pathology , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Monocytes/immunology , Monocytes/microbiology , Monocytes/pathology , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Peyer's Patches/microbiology , Peyer's Patches/pathology , Signal Transduction/genetics , Toll-Like Receptor 2/genetics , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis Infections/genetics , Yersinia pseudotuberculosis Infections/pathology
15.
J Clin Invest ; 120(6): 2131-43, 2010 Jun.
Article En | MEDLINE | ID: mdl-20440074

Enteropathy-associated T cell lymphoma is a severe complication of celiac disease (CD). One mechanism suggested to underlie its development is chronic exposure of intraepithelial lymphocytes (IELs) to potent antiapoptotic signals initiated by IL-15, a cytokine overexpressed in the enterocytes of individuals with CD. However, the signaling pathway by which IL-15 transmits these antiapoptotic signals has not been firmly established. Here we show that the survival signals delivered by IL-15 to freshly isolated human IELs and to human IEL cell lines derived from CD patients with type II refractory CD (RCDII) - a clinicopathological entity considered an intermediary step between CD and enteropathy-associated T cell lymphoma - depend on the antiapoptotic factors Bcl-2 and/or Bcl-xL. The signals also required IL-15Rbeta, Jak3, and STAT5, but were independent of PI3K, ERK, and STAT3. Consistent with these data, IELs from patients with active CD and RCDII contained increased amounts of Bcl-xL, phospho-Jak3, and phospho-STAT5. Furthermore, incubation of patient duodenal biopsies with a fully humanized human IL-15-specific Ab effectively blocked Jak3 and STAT5 phosphorylation. In addition, treatment with this Ab induced IEL apoptosis and wiped out the massive IEL accumulation in mice overexpressing human IL-15 in their gut epithelium. Together, our results delineate the IL-15-driven survival pathway in human IELs and demonstrate that IL-15 and its downstream effectors are meaningful therapeutic targets in RCDII.


Apoptosis/immunology , Celiac Disease/immunology , Inflammation/immunology , Interleukin-15/immunology , Lymphocytes/immunology , Adult , Apoptosis/drug effects , Celiac Disease/complications , Celiac Disease/metabolism , Cytokines/immunology , Cytokines/metabolism , Cytokines/pharmacology , Enterocytes/immunology , Enterocytes/metabolism , Humans , Inflammation/complications , Inflammation/metabolism , Interleukin-15/metabolism , Interleukin-15/pharmacology , Intestinal Mucosa/metabolism , Intestine, Small/immunology , Intestine, Small/metabolism , Intestines/immunology , Janus Kinase 3/immunology , Janus Kinase 3/metabolism , Leukemia/complications , Leukemia/immunology , Leukemia/metabolism , Lymphocytes/metabolism , Phosphorylation , Protein Binding/immunology , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology , Signal Transduction/drug effects , Signal Transduction/immunology
16.
Immunology ; 125(3): 320-30, 2008 Nov.
Article En | MEDLINE | ID: mdl-18445006

CD3- and CD28-activated T cells expanded for 12 days ex vivo to produce suicide gene-modified T cells are hyporesponsive to alloantigens. To investigate whether this impaired alloreactivity is a result of preferential expansion of regulatory T (Treg) cells, we compared peripheral blood mononuclear cells (PBMC) activated with CD3 and CD28 antibodies co-immobilized on beads and expanded for 12 days with interleukin (IL)-2 (Co(CD3/CD28) cells) to the respective unactivated PBMC in terms of proliferation, cytokine production, and expression of Treg markers [cytotoxic T-lymphocyte antigen 4 (CTLA4), glucocorticoid-induced tumour necrosis factor receptor (GITR) and forkhead box P3 (FoxP3)] after allostimulation. Alloreactive cells were identified by carboxyfluoresceine succinimidyl ester staining dilution. Alloreactive cells in Co(CD3/CD28) cells had a lower proliferative response and a lower potential for IL-2 and interferon-gamma secretion than did those in PBMC, demonstrating a functional impairment of alloreactive cells during ex vivo expansion. Expression of Treg markers transiently increased during ex vivo expansion and was unaffected by depletion of CD25(+) cells (containing Treg cells) before ex vivo PBMC expansion. Such prior CD25(+) depletion did not restore the alloreactivity of Co(CD3/CD28) cells. After allostimulation, expression of Treg markers was restricted to proliferative (alloreactive) cells among PBMC or Co(CD3/CD28) cells. Lastly, CD4(+) CD25(+) cells purified from Co(CD3/CD28) cells lacked suppressive activity when used as a third party, in contrast to CD4(+) CD25(+) cells purified from PBMC. In conclusion, the impaired alloreactivity of T cells expanded ex vivo is not a result of preferential Treg cell expansion and/or enhanced suppressive Treg activity.


T-Lymphocytes, Regulatory/immunology , Antigens, CD/metabolism , CTLA-4 Antigen , Cell Proliferation , Cells, Cultured , Forkhead Transcription Factors/metabolism , Glucocorticoid-Induced TNFR-Related Protein , Humans , Immune Tolerance , Immunophenotyping , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Lymphocyte Culture Test, Mixed , Receptors, Nerve Growth Factor/metabolism , Receptors, Tumor Necrosis Factor/metabolism
17.
Mol Immunol ; 45(4): 1112-25, 2008 Feb.
Article En | MEDLINE | ID: mdl-17825913

A suicide gene introduced by retroviral means can allow in vivo control of alloreactivity mediated by donor gene-modified T cells (GMTC) after allogeneic hematopoietic stem cell transplantation. The present study establishes the transcriptomic profile of GMTC prepared according to the GMTC production process used in our clinical trial (activation/selection methods, CD3/NeoR), which was previously demonstrated to induce phenotypical and functional alterations. This transcriptomic profile was compared with that of GMTC prepared by a novel process (CD3-CD28/DeltaNGFR-MACS) that limits alterations. Using a human pan-genomic microarray and GeneSpring software, we determined the gene expression profiles of CD8+ T cells from four healthy donors before and after the different steps required for gene modification. This analysis revealed that the gene expression pattern of GMTC is affected mainly by the activation step. Specific analysis of GMTC production processes showed that DeltaNGFR-MACS selection combined with CD3-CD28 activation limits the aberrant expression of genes involved in immunological functions and apoptotic pathways. Furthermore, our results indicate a limited risk of oncogenesis associated with retroviral-mediated gene transfer in CD8+ cells, a lower perturbation of the cell cycle regulation pathway after CD3-CD28 activation than after CD3 activation, and no significant involvement of the DeltaNGFR transduction signaling pathway when DeltaNGFR is used for selection. Moreover, genes that might be targeted to limit T cell functional alterations after ex vivo manipulation and culture were identified. These findings should be relevant to further adoptive T cell immunotherapy trials using ex vivo-expanded, gene-modified or unmodified T cells.


CD8-Positive T-Lymphocytes/metabolism , Gene Expression Profiling , Gene Transfer Techniques , Retroviridae/genetics , Adult , Female , Humans , Lymphocyte Activation , Male , Middle Aged , Oligonucleotide Array Sequence Analysis/methods , Polymorphism, Genetic , Transgenes
...