Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
2.
Gut ; 71(3): 497-508, 2022 03.
Article in English | MEDLINE | ID: mdl-33579790

ABSTRACT

OBJECTIVE: Enteropathy-associated T-cell lymphoma (EATL) is a rare but severe complication of coeliac disease (CeD), often preceded by low-grade clonal intraepithelial lymphoproliferation, referred to as type II refractory CeD (RCDII). Knowledge on underlying oncogenic mechanisms remains scarce. Here, we analysed and compared the mutational landscape of RCDII and EATL in order to identify genetic drivers of CeD-associated lymphomagenesis. DESIGN: Pure populations of RCDII-cells derived from intestinal biopsies (n=9) or sorted from blood (n=2) were analysed by whole exome sequencing, comparative genomic hybridisation and RNA sequencing. Biopsies from RCDII (n=50), EATL (n=19), type I refractory CeD (n=7) and uncomplicated CeD (n=18) were analysed by targeted next-generation sequencing. Moreover, functional in vitro studies and drug testing were performed in RCDII-derived cell lines. RESULTS: 80% of RCDII and 90% of EATL displayed somatic gain-of-functions mutations in the JAK1-STAT3 pathway, including a remarkable p.G1097 hotspot mutation in the JAK1 kinase domain in approximately 50% of cases. Other recurrent somatic events were deleterious mutations in nuclear factor kappa-light-chain-enhancer of activated B-cells (NF-κB) regulators TNFAIP3 and TNIP3 and potentially oncogenic mutations in TET2, KMT2D and DDX3X. JAK1 inhibitors, and the proteasome inhibitor bortezomib could block survival and proliferation of malignant RCDII-cell lines. CONCLUSION: Mutations activating the JAK1-STAT3 pathway appear to be the main drivers of CeD-associated lymphomagenesis. In concert with mutations in negative regulators of NF-κB, they may favour the clonal emergence of malignant lymphocytes in the cytokine-rich coeliac intestine. The identified mutations are attractive therapeutic targets to treat RCDII and block progression towards EATL.


Subject(s)
Celiac Disease/complications , Celiac Disease/genetics , Enteropathy-Associated T-Cell Lymphoma/etiology , Gain of Function Mutation/genetics , Lymphocytes/physiology , Adult , Aged , Aged, 80 and over , Celiac Disease/pathology , Cohort Studies , Enteropathy-Associated T-Cell Lymphoma/pathology , Female , France , Humans , Janus Kinase 1/genetics , Male , Middle Aged , STAT3 Transcription Factor/genetics , Young Adult
3.
Gastroenterology ; 160(1): 287-301.e20, 2021 01.
Article in English | MEDLINE | ID: mdl-32980345

ABSTRACT

BACKGROUND AND AIMS: The presence of gastrointestinal symptoms and high levels of viral RNA in the stool suggest active severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication within enterocytes. METHODS: Here, in multiple, large cohorts of patients with inflammatory bowel disease (IBD), we have studied the intersections between Coronavirus Disease 2019 (COVID-19), intestinal inflammation, and IBD treatment. RESULTS: A striking expression of ACE2 on the small bowel enterocyte brush border supports intestinal infectivity by SARS-CoV-2. Commonly used IBD medications, both biologic and nonbiologic, do not significantly impact ACE2 and TMPRSS2 receptor expression in the uninflamed intestines. In addition, we have defined molecular responses to COVID-19 infection that are also enriched in IBD, pointing to shared molecular networks between COVID-19 and IBD. CONCLUSIONS: These data generate a novel appreciation of the confluence of COVID-19- and IBD-associated inflammation and provide mechanistic insights supporting further investigation of specific IBD drugs in the treatment of COVID-19. Preprint doi: https://doi.org/10.1101/2020.05.21.109124.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19/enzymology , Inflammatory Bowel Diseases/enzymology , Intestinal Mucosa/enzymology , SARS-CoV-2/pathogenicity , Serine Endopeptidases/metabolism , Angiotensin-Converting Enzyme 2/genetics , Animals , Anti-Inflammatory Agents/therapeutic use , Antiviral Agents/therapeutic use , COVID-19/genetics , COVID-19/virology , Case-Control Studies , Clinical Trials as Topic , Cross-Sectional Studies , Disease Models, Animal , Female , Gene Regulatory Networks , Host-Pathogen Interactions , Humans , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/genetics , Intestinal Mucosa/drug effects , Intestinal Mucosa/virology , Longitudinal Studies , Male , Mice , SARS-CoV-2/drug effects , Serine Endopeptidases/genetics , Signal Transduction , COVID-19 Drug Treatment
4.
F1000Res ; 82019.
Article in English | MEDLINE | ID: mdl-31297187

ABSTRACT

Celiac disease (CeD), defined as gluten-induced enteropathy, is a frequent and largely underdiagnosed disease. Diagnosis relies on the detection of highly specific serum IgA anti-transglutaminase auto-antibodies and on the demonstration of duodenal villous atrophy. Treatment necessitates a strict gluten-free diet, which resolves symptoms and enables histological recovery. However, regular follow-up is necessary to assess mucosal healing, which emerges as an important prognostic factor. Recent work on CeD pathogenesis has highlighted how the cross-talk between gluten-specific CD4 + T cells and interleukin-15 can activate cytotoxic intraepithelial lymphocytes and trigger epithelial lesions. Moreover, acquisition by a subset of intraepithelial lymphocytes of somatic gain-of-function mutations in the JAK-STAT pathway was shown to be a decisive step in the progression toward lymphomas complicating CeD, thus opening new therapeutic perspectives for these rare but life-threatening complications.


Subject(s)
Celiac Disease , Celiac Disease/immunology , Celiac Disease/metabolism , Celiac Disease/therapy , Diet, Gluten-Free , Disease Progression , Glutens , Humans , Transglutaminases
5.
Gut ; 68(8): 1396-1405, 2019 08.
Article in English | MEDLINE | ID: mdl-30448772

ABSTRACT

OBJECTIVES: Primary GI T-cell lymphoproliferative diseases (T-LPD) are heterogeneous entities, which raise difficult diagnosis and therapeutic challenges. We have recently provided evidences that lymphomas complicating coeliac disease (CD) arise from innate-like lymphocytes, which may carry NK receptors (NKRs). DESIGN: NKRs expression was compared by flow cytometry in intraepithelial lymphocytes (IEL) from CD, type I or type II refractory CD (RCD). NKp46 was next assessed by immunohistochemistry in paraffin-embedded biopsies from 204 patients with CD, RCDI, RCDII or GI T-cell lymphomas and from a validation cohort of 61 patients. The cytotoxic properties of an anti-NKp46 monoclonal antibody conjugated to pyrrolobenzodiazepine (PBD) was tested ex vivo in human primary tumour cells isolated from fresh duodenal biopsies. RESULTS: NKp46 (but not CD94, NKG2A, NKG2C, NKG2D) was significantly more expressed by malignant RCDII IEL than by normal IEL in CD and RCDI. In paraffin biopsies, detection of >25 NKp46+ IEL per 100 epithelial cells discriminated RCDII from CD and RCDI. NKp46 was also detected in enteropathy-associated T-cell lymphomas (EATL, 24/29) and in monomorphic epitheliotropic intestinal T-cell lymphomas (MEITL, 4/4) but not in indolent T-LPD (0/15). Treatment with anti-NKp46-PBD could efficiently and selectively kill human NKp46+ primary IEL ex vivo. CONCLUSION: NKp46 is a novel biomarker useful for diagnosis and therapeutic stratification of GI T-LPD. Strong preclinical rationale identifies anti-NKp46-PBD as a promising therapy for RCDII, EATL and MEITL.


Subject(s)
Celiac Disease , Enteropathy-Associated T-Cell Lymphoma , Intestinal Mucosa , Killer Cells, Natural/immunology , Natural Cytotoxicity Triggering Receptor 1/immunology , Antibodies, Monoclonal/immunology , Biomarkers/blood , Biopsy/methods , Celiac Disease/complications , Celiac Disease/diagnosis , Celiac Disease/immunology , Celiac Disease/pathology , Cells, Cultured , Enteropathy-Associated T-Cell Lymphoma/diagnosis , Enteropathy-Associated T-Cell Lymphoma/etiology , Enteropathy-Associated T-Cell Lymphoma/immunology , Enteropathy-Associated T-Cell Lymphoma/pathology , Female , France , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestine, Small/pathology , Male , Middle Aged , Prognosis
6.
Eur J Immunol ; 48(8): 1271-1280, 2018 08.
Article in English | MEDLINE | ID: mdl-29974461

ABSTRACT

Natural killer (NK) cells and lymphoid tissue inducer (LTi) cells were discovered more than 40 and 20 years ago, respectively. These two cell types were initially studied for their unique functions in the elimination of infected or transformed cells, and in the development of lymphoid tissues. It took an additional 10 years to realize that NK cells and LTi cells were members of a larger family of innate lymphoid cells (ILCs), whose phenotypes and functions mirror those of T cells. Many mouse models have since been developed to identify and isolate ILCs, map their developmental pathways and characterize their functions. Because of the similarity between ILCs and T cells, this exploration remains a challenge. In spite of this, a broad range of mouse models available to researchers has lead to significant progress in untangling the unique roles of ILCs early in defense, regulation of adaptive immunity and homeostasis. Here, we review these mouse models, and discuss their strengths and limitations.


Subject(s)
Killer Cells, Natural/immunology , Lymphocytes/immunology , Models, Animal , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cell Lineage , Immunity, Innate/immunology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Mice , Mice, Knockout
7.
Microbes Infect ; 20(6): 317-322, 2018.
Article in English | MEDLINE | ID: mdl-29852240

ABSTRACT

Innate lymphoid cells (ILCs) are the innate counterpart of T cells. Upon infection or injury, ILCs react promptly to direct the developing immune response to the one most adapted to the threat facing the organism. Therefore, ILCs play an important role early in resistance to infection, but also to maintain homeostasis with the symbiotic microbiota following perturbations induced by diet and pathogens. Such roles of ILCs have been best characterized in the intestine and lung, mucosal sites that are exposed to the environment and are therefore colonized with diverse but specific types of microbes. Understanding the dialogue between pathogens, microbiota and ILCs may lead to new strategies to re-inforce immunity for prevention, vaccination and therapy.


Subject(s)
Host-Pathogen Interactions/immunology , Immunity, Innate , Lymphocytes/immunology , Microbiota/immunology , Animals , Cytokines/immunology , Homeostasis/immunology , Humans , Intestinal Mucosa/immunology , Lymphocytes/cytology , Respiratory Mucosa/immunology , Symbiosis/immunology
9.
Science ; 350(6264): 1079-84, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26541610

ABSTRACT

Antibodies targeting CTLA-4 have been successfully used as cancer immunotherapy. We find that the antitumor effects of CTLA-4 blockade depend on distinct Bacteroides species. In mice and patients, T cell responses specific for B. thetaiotaomicron or B. fragilis were associated with the efficacy of CTLA-4 blockade. Tumors in antibiotic-treated or germ-free mice did not respond to CTLA blockade. This defect was overcome by gavage with B. fragilis, by immunization with B. fragilis polysaccharides, or by adoptive transfer of B. fragilis-specific T cells. Fecal microbial transplantation from humans to mice confirmed that treatment of melanoma patients with antibodies against CTLA-4 favored the outgrowth of B. fragilis with anticancer properties. This study reveals a key role for Bacteroidales in the immunostimulatory effects of CTLA-4 blockade.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Bacteroides/immunology , CTLA-4 Antigen/antagonists & inhibitors , Gastrointestinal Microbiome/immunology , Melanoma/therapy , Skin Neoplasms/therapy , Adult , Aged , Aged, 80 and over , Animals , Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/adverse effects , CTLA-4 Antigen/immunology , Dysbiosis/immunology , Fecal Microbiota Transplantation , Female , Gastrointestinal Microbiome/drug effects , Germ-Free Life/immunology , Humans , Immunologic Memory , Immunotherapy , Intestines/immunology , Intestines/microbiology , Ipilimumab , Male , Mice , Mice, Inbred C57BL , Middle Aged , T-Lymphocytes/immunology
10.
J Invest Dermatol ; 135(12): 2982-2991, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26203637

ABSTRACT

Murine contact hypersensitivity (CHS) is a dendritic cell (DC)-dependent T-cell-mediated inflammation with CD8+ T cells as effectors and CD4+ T cells as regulators (Treg cells) that models human allergic contact dermatitis. The integrin αE(CD103) is expressed by some T-cell and DC subsets and has been implicated in epithelial lymphocyte localization, but its role in immune regulation remains enigmatic. We have identified a function for CD103 in the development of cutaneous allergic immune responses. CHS responses, but not irritant contact dermatitis, were significantly augmented in CD103-deficient mice in hapten-challenged skin. Phenotype and function of skin DCs during sensitization were normal, whereas adoptive transfer experiments revealed that the elevated CHS response in CD103-deficient mice is transferred by primed T cells and is independent of resident cells in recipient mice. While T-cell counts were elevated in challenged skin of CD103-deficient mice, the FoxP3 expression level of CD4+CD25+ Treg cells was significantly reduced, indicating impaired functionality. Indeed, Treg cells from CD103-deficient mice were not able to suppress CHS reactions during the elicitation phase. Further, CD103 on FoxP3+ Treg cells was involved in Treg retention to inflamed skin. These findings indicate an unexpected dichotomous functional role for CD103 on Treg cells by modulating FoxP3 expression.


Subject(s)
Antigens, CD/physiology , Dermatitis, Allergic Contact/immunology , Integrin alpha Chains/physiology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Dendritic Cells/immunology , Forkhead Transcription Factors/analysis , Forkhead Transcription Factors/physiology , Homeodomain Proteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Skin/immunology
11.
Science ; 349(6251): 989-93, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26160380

ABSTRACT

Changes to the symbiotic microbiota early in life, or the absence of it, can lead to exacerbated type 2 immunity and allergic inflammations. Although it is unclear how the microbiota regulates type 2 immunity, it is a strong inducer of proinflammatory T helper 17 (T(H)17) cells and regulatory T cells (T(regs)) in the intestine. Here, we report that microbiota-induced T(regs) express the nuclear hormone receptor RORγt and differentiate along a pathway that also leads to T(H)17 cells. In the absence of RORγt(+) T(regs), T(H)2-driven defense against helminths is more efficient, whereas T(H)2-associated pathology is exacerbated. Thus, the microbiota regulates type 2 responses through the induction of type 3 RORγt(+) T(regs) and T(H)17 cells and acts as a key factor in balancing immune responses at mucosal surfaces.


Subject(s)
Immunity, Mucosal , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestines/microbiology , Microbiota/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Colitis, Ulcerative/immunology , Colon/immunology , Colon/microbiology , Germ-Free Life , Homeostasis , Intestine, Small/immunology , Intestine, Small/microbiology , Intestines/immunology , Mice , Models, Immunological , Nematospiroides dubius , Specific Pathogen-Free Organisms , Strongylida Infections/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/immunology , Th2 Cells/immunology , Vitamin A/metabolism
12.
FEBS Lett ; 588(22): 4176-81, 2014 Nov 17.
Article in English | MEDLINE | ID: mdl-24681095

ABSTRACT

RORγt(+) innate lymphoid cells (ILCs), or ILC3, play a fundamental role in the development of lymphoid tissues, as well as in homeostasis and defence of mucosal tissues. These cells produce IL-22, IL-17A and LTα1ß2, key cytokines for the activation of epithelial defences and the recruitment of polymorphonuclear phagocytes. In the absence of ILC3, the early defence to infection and resistance to injury are compromised. Given the importance of ILC3 in mucosal immunity, significant efforts are made to discover their multiple functions and decipher their mode of action and regulation.


Subject(s)
Immunity, Innate , Lymphocytes/immunology , Lymphocytes/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Animals , Humans , Lymphocytes/cytology
13.
Eur J Microbiol Immunol (Bp) ; 3(1): 1-10, 2013 Mar.
Article in English | MEDLINE | ID: mdl-24265914

ABSTRACT

Compelling evidence demonstrates that intestinal commensal microbiota modulate conventional and regulatory T cell (Treg) responses that are required for effective host defence against pathogens and avoidance of autoimmunity and other immunopathologic conditions. Here, we investigated the contribution of the commensal microbiota and Toll-like receptor (TLR) signaling to homeostasis of Foxp3(-) conventional CD4(+) T cells and Foxp3(+) Tregs. Upon long-term antibiotics treatment, we observed a significant reduction of conventional CD4(+) T cell proliferation in a systemic manner, whereas Foxp3(+) Treg proliferation was locally impaired in gut-draining mesenteric lymph nodes and Peyer's patches. The proliferative response to microbial components was not mediated by TLRs as MyD88- and various TLR-deficient mice displayed normal or even increased conventional T cell and Foxp3(+) Treg proliferation. Thus, commensal microbiota-derived stimuli support cycling of both conventional CD4(+) T cells and Foxp3(+) Tregs with TLR-mediated recognition of bacterial components not being the major mechanism controlling microbiota-driven T cell homeostasis.

14.
J Mol Med (Berl) ; 88(10): 1029-40, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20574810

ABSTRACT

The forkhead-box protein P3 (Foxp3) is a key transcription factor for the development and suppressive activity of regulatory T cells (Tregs), a T cell subset critically involved in the maintenance of self-tolerance and prevention of over-shooting immune responses. However, the transcriptional regulation of Foxp3 expression remains incompletely understood. We have previously shown that epigenetic modifications in the CpG-rich Treg-specific demethylated region (TSDR) in the Foxp3 locus are associated with stable Foxp3 expression. We now demonstrate that the methylation state of the CpG motifs within the TSDR controls its transcriptional activity rather than a Treg-specific transcription factor network. By systematically mutating every CpG motif within the TSDR, we could identify four CpG motifs, which are critically determining the transcriptional activity of the TSDR and which serve as binding sites for essential transcription factors, such as CREB/ATF and NF-κB, which have previously been shown to bind to this element. The transcription factor Ets-1 was here identified as an additional molecular player that specifically binds to the TSDR in a demethylation-dependent manner in vitro. Disruption of the Ets-1 binding sites within the TSDR drastically reduced its transcriptional enhancer activity. In addition, we found Ets-1 bound to the demethylated TSDR in ex vivo isolated Tregs, but not to the methylated TSDR in conventional CD4(+) T cells. We therefore propose that Ets-1 is part of a larger protein complex, which binds to the TSDR only in its demethylated state, thereby restricting stable Foxp3 expression to the Treg lineage.


Subject(s)
Forkhead Transcription Factors/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Base Sequence , Cells, Cultured , CpG Islands , Enhancer Elements, Genetic , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Humans , Methylation , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Proto-Oncogene Protein c-ets-1/genetics , T-Lymphocytes, Regulatory/cytology
15.
J Immunol ; 180(1): 146-55, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18097014

ABSTRACT

Naturally occurring Foxp3+CD25+CD4+ regulatory T cells (Treg) have initially been described as anergic cells; however, more recent in vivo studies suggest that Tregs vigorously proliferate under both homeostatic as well as inflammatory conditions. We have previously identified a subset of murine CD4+ Tregs, which is characterized by expression of the integrin alphaEbeta7 and which displays an effector/memory-like phenotype indicative of Ag-specific expansion and differentiation. In the present study, the alphaE+ Treg subset was found to contain a large fraction of cycling cells under homeostatic conditions in healthy mice. Using an adoptive transfer system of Ag-specific T cells, we could demonstrate that the vast majority of transferred natural, naive-like CD25+CD4+ Tregs acquired expression of the integrin alphaEbeta7 upon tolerogenic application of Ag via the oral route. In addition, using the same system, Foxp3+ Tregs could be de novo induced from conventional naive CD25-CD4+ T cells, and this conversion was associated with concomitant expression of alphaE. These findings suggest that Tregs expressing the integrin alphaE are effector/memory Tregs with a high turnover rate that can develop in the periphery upon Ag contact under tolerogenic conditions, both from thymic-derived CD25+CD4+ Tregs with a naive-like phenotype as well as from conventional naive T cells.


Subject(s)
Forkhead Transcription Factors/analysis , Immunologic Memory , Integrins/analysis , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Proliferation , Homeostasis , Interleukin-2 Receptor alpha Subunit/analysis , Mice , Mice, Inbred Strains
SELECTION OF CITATIONS
SEARCH DETAIL