Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 242
Filtrer
1.
PLoS Pathog ; 20(5): e1011820, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38718306

RÉSUMÉ

The production of IFN-γ is crucial for control of multiple enteric infections, but its impact on intestinal epithelial cells (IEC) is not well understood. Cryptosporidium parasites exclusively infect epithelial cells and the ability of interferons to activate the transcription factor STAT1 in IEC is required for parasite clearance. Here, the use of single cell RNA sequencing to profile IEC during infection revealed an increased proportion of mid-villus enterocytes during infection and induction of IFN-γ-dependent gene signatures that was comparable between uninfected and infected cells. These analyses were complemented by in vivo studies, which demonstrated that IEC expression of the IFN-γ receptor was required for parasite control. Unexpectedly, treatment of Ifng-/- mice with IFN-γ showed the IEC response to this cytokine correlates with a delayed reduction in parasite burden but did not affect parasite development. These data sets provide insight into the impact of IFN-γ on IEC and suggest a model in which IFN-γ signalling to uninfected enterocytes is important for control of Cryptosporidium.


Sujet(s)
Cryptosporidiose , Interféron gamma , Muqueuse intestinale , Souris knockout , Animaux , Interféron gamma/métabolisme , Interféron gamma/immunologie , Cryptosporidiose/immunologie , Cryptosporidiose/parasitologie , Souris , Muqueuse intestinale/parasitologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/immunologie , Cryptosporidium , Cellules épithéliales/parasitologie , Cellules épithéliales/métabolisme , Cellules épithéliales/immunologie , Entérocytes/parasitologie , Entérocytes/métabolisme , Entérocytes/immunologie , Souris de lignée C57BL , , Facteur de transcription STAT-1/métabolisme , Récepteur interféron/métabolisme , Récepteur interféron/génétique , Transduction du signal
2.
Nat Commun ; 13(1): 17, 2022 01 10.
Article de Anglais | MEDLINE | ID: mdl-35013162

RÉSUMÉ

Defense against intracellular infection has been extensively studied in vertebrate hosts, but less is known about invertebrate hosts; specifically, the transcription factors that induce defense against intracellular intestinal infection in the model nematode Caenorhabditis elegans remain understudied. Two different types of intracellular pathogens that naturally infect the C. elegans intestine are the Orsay virus, which is an RNA virus, and microsporidia, which comprise a phylum of fungal pathogens. Despite their molecular differences, these pathogens induce a common host transcriptional response called the intracellular pathogen response (IPR). Here we show that zip-1 is an IPR regulator that functions downstream of all known IPR-activating and regulatory pathways. zip-1 encodes a putative bZIP transcription factor, and we show that zip-1 controls induction of a subset of genes upon IPR activation. ZIP-1 protein is expressed in the nuclei of intestinal cells, and is at least partially required in the intestine to upregulate IPR gene expression. Importantly, zip-1 promotes resistance to infection by the Orsay virus and by microsporidia in intestinal cells. Altogether, our results indicate that zip-1 represents a central hub for triggers of the IPR, and that this transcription factor has a protective function against intracellular pathogen infection in C. elegans.


Sujet(s)
Facteurs de transcription à motif basique et à glissière à leucines , Caenorhabditis elegans , Entérocytes , Interactions hôte-pathogène/physiologie , Animaux , Facteurs de transcription à motif basique et à glissière à leucines/immunologie , Facteurs de transcription à motif basique et à glissière à leucines/métabolisme , Caenorhabditis elegans/immunologie , Caenorhabditis elegans/microbiologie , Caenorhabditis elegans/virologie , Protéines de Caenorhabditis elegans/immunologie , Protéines de Caenorhabditis elegans/métabolisme , Entérocytes/immunologie , Entérocytes/microbiologie , Entérocytes/virologie , Immunité innée/physiologie , Intestins/microbiologie , Intestins/virologie , Invertébrés/immunologie , Microsporidia/pathogénicité , Virus à ARN/pathogénicité
3.
Cell Rep ; 37(13): 110150, 2021 12 28.
Article de Anglais | MEDLINE | ID: mdl-34965418

RÉSUMÉ

Enteric pathogens overcome barrier immunity within the intestinal environment that includes the endogenous flora. The microbiota produces diverse ligands, and the full spectrum of microbial products that are sensed by the epithelium and prime protective immunity is unknown. Using Drosophila, we find that the gut presents a high barrier to infection, which is partially due to signals from the microbiota, as loss of the microbiota enhances oral viral infection. We report cyclic dinucleotide (CDN) feeding is sufficient to protect microbiota-deficient flies from enhanced oral infection, suggesting that bacterial-derived CDNs induce immunity. Mechanistically, we find CDN protection is dSTING- and dTBK1-dependent, leading to NF-kB-dependent gene expression. Furthermore, we identify the apical nucleoside transporter, CNT2, as required for oral CDN protection. Altogether, our studies define a role for bacterial products in priming immune defenses in the gut.


Sujet(s)
Infections à alphavirus/immunologie , Antiviraux/pharmacologie , Drosophila melanogaster/immunologie , Entérocytes/immunologie , Protéines membranaires/métabolisme , Protéines de transport membranaire/métabolisme , Nucléotides cycliques/administration et posologie , Infections à alphavirus/traitement médicamenteux , Infections à alphavirus/virologie , Animaux , Drosophila melanogaster/effets des médicaments et des substances chimiques , Drosophila melanogaster/génétique , Drosophila melanogaster/métabolisme , Entérocytes/effets des médicaments et des substances chimiques , Entérocytes/virologie , Femelle , Immunité innée , Protéines membranaires/génétique , Protéines de transport membranaire/génétique , Facteur de transcription NF-kappa B/génétique , Facteur de transcription NF-kappa B/métabolisme , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme , Virus Sindbis/immunologie
4.
Cells ; 10(9)2021 08 30.
Article de Anglais | MEDLINE | ID: mdl-34571900

RÉSUMÉ

Iron is crucial to the regulation of the host innate immune system and the outcome of many infections. Hepatitis C virus (HCV), one of the major viral human pathogens that depends on iron to complete its life cycle, is highly skilled in evading the immune system. This study presents the construction and validation of a physiologically relevant triple-cell co-culture model that was used to investigate the input of iron in HCV infection and the interplay between HCV, iron, and determinants of host innate immunity. We recorded the expression patterns of key proteins of iron homeostasis involved in iron import, export and storage and examined their relation to the iron regulatory hormone hepcidin in hepatocytes, enterocytes and macrophages in the presence and absence of HCV. We then assessed the transcriptional profiles of pro-inflammatory cytokines Interleukin-6 (IL-6) and interleukin-15 (IL-15) and anti-inflammatory interleukin-10 (IL-10) under normal or iron-depleted conditions and determined how these were affected by infection. Our data suggest the presence of a link between iron homeostasis and innate immunity unfolding among liver, intestine, and macrophages, which could participate in the deregulation of innate immune responses observed in early HCV infection. Coupled with iron-assisted enhanced viral propagation, such a mechanism may be important for the establishment of viral persistence and the ensuing chronic liver disease.


Sujet(s)
Entérocytes/anatomopathologie , Hépatite C/anatomopathologie , Hépatocytes/anatomopathologie , Homéostasie , Immunité innée , Fer/métabolisme , Macrophages/anatomopathologie , Techniques de coculture , Cytokines/métabolisme , Entérocytes/immunologie , Entérocytes/métabolisme , Entérocytes/virologie , Hepacivirus/immunologie , Hepacivirus/métabolisme , Hépatite C/immunologie , Hépatite C/métabolisme , Hépatite C/virologie , Hépatocytes/immunologie , Hépatocytes/métabolisme , Hépatocytes/virologie , Humains , Macrophages/immunologie , Macrophages/métabolisme , Macrophages/virologie
5.
Clin Transl Gastroenterol ; 12(8): e00387, 2021 08 01.
Article de Anglais | MEDLINE | ID: mdl-34333499

RÉSUMÉ

INTRODUCTION: Adult-onset autoimmune enteropathy (AIE) is a rare cause of severe chronic diarrhea because of small intestinal villous atrophy. We report on patients with adult-onset AIE in an European referral center. METHODS: Retrospective study including patients diagnosed with AIE in the Amsterdam UMC, location VUmc, between January 2003 and December 2019. Clinical, serological, and histological features and response to treatment were reported. The specificity of antienterocyte antibodies (AEA) was evaluated by examining the prevalence of AEA in (i) controls (n = 30) and in patients with (ii) AIE (n = 13), (iii) celiac disease (CD, n = 52), (iv) refractory celiac disease type 2 (n = 18), and (v) enteropathy-associated T-cell lymphoma (EATL, n = 10). RESULTS: Thirteen AIE patients were included, 8 women (62%), median age of 52 years (range 23-73), and 6 (46%) with an autoimmune disease. AEA were observed in 11 cases (85%), but were also found in CD (7.7%), refractory celiac disease type 2 (16.7%), and EATL (20%). Ten patients (77%) were human leukocyte antigen DQ2.5 heterozygous. Total parenteral nutrition was required in 8 cases (62%). Steroids induced clinical remission in 8 cases (62%). Step-up therapy with rituximab, cyclosporine, infliximab, and cladribine in steroid-refractory patients was only moderately effective. Four patients died (31%), but 4 (31%) others are in long-term drug-free remission after receiving immunosuppressive treatment, including 1 patient who underwent autologous stem cell transplantation. DISCUSSION: Adult-onset AIE is a rare but severe enteropathy that occurs in patients susceptible for autoimmune disease. Four patients (31%) died secondary to therapy-refractory malabsorption, while immunosuppressive therapy leads to a long-lasting drug-free remission in one-third of patients.


Sujet(s)
Autoanticorps/analyse , Entérocytes/immunologie , Polyendocrinopathies auto-immunes/diagnostic , Polyendocrinopathies auto-immunes/immunologie , Adulte , Sujet âgé , Atrophie , Maladie chronique , Diarrhée/étiologie , Duodénum/anatomopathologie , Protéines de liaison aux acides gras/sang , Femelle , Antigènes HLA-DQ/sang , Humains , Immunosuppresseurs/usage thérapeutique , Mâle , Adulte d'âge moyen , Pays-Bas , Nutrition parentérale , Polyendocrinopathies auto-immunes/traitement médicamenteux , Polyendocrinopathies auto-immunes/anatomopathologie , Études rétrospectives , Sous-populations de lymphocytes T/métabolisme , Centres de soins tertiaires , Jeune adulte
6.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Article de Anglais | MEDLINE | ID: mdl-34290141

RÉSUMÉ

"Taste-like" tuft cells in the intestine trigger type 2 immunity in response to worm infection. The secretion of interleukin-13 (IL-13) from type 2 innate lymphoid cells (ILC2) represents a key step in the tuft cell-ILC2 cell-intestinal epithelial cell circuit that drives the clearance of worms from the gut via type 2 immune responses. Hallmark features of type 2 responses include tissue remodeling, such as tuft and goblet cell expansion, and villus atrophy, yet it remains unclear if additional molecular changes in the gut epithelium facilitate the clearance of worms from the gut. Using gut organoids, we demonstrated that IL-4 and IL-13, two type 2 cytokines with similar functions, not only induced the classical type 2 responses (e.g., tuft cell expansion) but also drastically up-regulated the expression of gasdermin C genes (Gsdmcs). Using an in vivo worm-induced type 2 immunity model, we confirmed the up-regulation of Gsdmcs in Nippostrongylus brasiliensis-infected wild-type C57BL/6 mice. Consistent with gasdermin family members being principal effectors of pyroptosis, overexpression of Gsdmc2 in human embryonic kidney 293 (HEK293) cells triggered pyroptosis and lytic cell death. Moreover, in intestinal organoids treated with IL-4 or IL-13, or in wild-type mice infected with N. brasiliensis, lytic cell death increased, which may account for villus atrophy observed in worm-infected mice. Thus, we propose that the up-regulated Gsdmc family may be major effectors for type 2 responses in the gut and that Gsdmc-mediated pyroptosis may provide a conduit for the release of antiparasitic factors from enterocytes to facilitate the clearance of worms.


Sujet(s)
Mort cellulaire , Protéines de liaison à l'ADN/métabolisme , Entérocytes/anatomopathologie , Immunité innée/immunologie , Intestin grêle/anatomopathologie , Infections à Strongylida/complications , Lymphocytes auxiliaires Th2/immunologie , Animaux , Prolifération cellulaire , Protéines de liaison à l'ADN/génétique , Entérocytes/immunologie , Entérocytes/métabolisme , Entérocytes/parasitologie , Femelle , Interleukine-13/métabolisme , Interleukine-4/métabolisme , Intestin grêle/immunologie , Intestin grêle/métabolisme , Intestin grêle/parasitologie , Mâle , Souris , Souris de lignée C57BL , Nippostrongylus/physiologie , Transduction du signal , Infections à Strongylida/immunologie , Infections à Strongylida/métabolisme , Infections à Strongylida/parasitologie
7.
Monoclon Antib Immunodiagn Immunother ; 40(3): 101-106, 2021 Jun.
Article de Anglais | MEDLINE | ID: mdl-34161159

RÉSUMÉ

CC chemokine receptor 9 (CCR9) belongs to the beta chemokine receptor family and is mainly distributed on the surface of immature T lymphocytes and enterocytes. This receptor is highly expressed in rheumatoid arthritis, colitis, type 2 diabetes, and various tumors. Therefore, more sensitive monoclonal antibodies (mAbs) need to be developed to predict the prognosis of many high CCR9 expression diseases. Because CCR9 is a structurally unstable G protein-coupled receptor, it has been difficult to develop anti-CCR9 mAbs using the traditional method. This study developed anti-human CCR9 (hCCR9) mAbs for flow cytometry using a Cell-Based Immunization and Screening (CBIS) method. Two mice were immunized with hCCR9-overexpressed Chinese hamster ovary (CHO)-K1 cells (CHO/hCCR9), and hybridomas showing strong signals from CHO/hCCR9 and no signals from CHO-K1 cells were selected by flow cytometry. We established an anti-hCCR9 mAb, C9Mab-1 (IgG1, kappa), which detected hCCR9 in MOLT-4 leukemia T lymphoblast cells and CHO/hCCR9 cells by flow cytometry. Our study showed that an anti-hCCR9 mAb was developed more rapidly by the CBIS method than the previous method.


Sujet(s)
Anticorps anti-idiotypiques/immunologie , Anticorps monoclonaux/immunologie , Récepteurs CCR/immunologie , Lymphocytes T/immunologie , Animaux , Anticorps anti-idiotypiques/pharmacologie , Anticorps monoclonaux/pharmacologie , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/thérapie , Cellules CHO , Colite/immunologie , Colite/thérapie , Cricetinae , Cricetulus , Diabète de type 2/immunologie , Diabète de type 2/thérapie , Entérocytes/immunologie , Épitopes/immunologie , Cytométrie en flux , Humains , Immunoglobuline G/immunologie , Souris , Récepteurs CCR/antagonistes et inhibiteurs , Lymphocytes T/effets des médicaments et des substances chimiques
8.
Commun Biol ; 4(1): 631, 2021 05 27.
Article de Anglais | MEDLINE | ID: mdl-34045640

RÉSUMÉ

IL22 is an important cytokine involved in the intestinal defense mechanisms against microbiome. By using ileum-derived organoids, we show that the expression of anti-microbial peptides (AMPs) and anti-viral peptides (AVPs) can be induced by IL22. In addition, we identified a bacterial and a viral route, both leading to IL22 production by T cells, but via different pathways. Bacterial products, such as LPS, induce enterocyte-secreted SAA1, which triggers the secretion of IL6 in fibroblasts, and subsequently IL22 in T cells. This IL22 induction can then be enhanced by macrophage-derived TNFα in two ways: by enhancing the responsiveness of T cells to IL6 and by increasing the expression of IL6 by fibroblasts. Viral infections of intestinal cells induce IFNß1 and subsequently IL7. IFNß1 can induce the expression of IL6 in fibroblasts and the combined activity of IL6 and IL7 can then induce IL22 expression in T cells. We also show that IL22 reduces the expression of viral entry receptors (e.g. ACE2, TMPRSS2, DPP4, CD46 and TNFRSF14), increases the expression of anti-viral proteins (e.g. RSAD2, AOS, ISG20 and Mx1) and, consequently, reduces the viral infection of neighboring cells. Overall, our data indicates that IL22 contributes to the innate responses against both bacteria and viruses.


Sujet(s)
Interleukines/biosynthèse , Interleukines/métabolisme , Animaux , Antibactériens/métabolisme , Antiviraux/métabolisme , Techniques de culture cellulaire , Cytokines/immunologie , Cytokines/métabolisme , Modèles animaux de maladie humaine , Entérocytes/immunologie , Entérocytes/métabolisme , Femelle , Fibroblastes/immunologie , Fibroblastes/métabolisme , Interleukines/immunologie , Muqueuse intestinale/métabolisme , Intestins/physiologie , Souris , Souris de lignée C57BL , Cellules myéloïdes/immunologie , Cellules myéloïdes/métabolisme , Organoïdes/métabolisme , Perforines/génétique , Perforines/métabolisme ,
9.
Mucosal Immunol ; 14(5): 1017-1037, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-33859369

RÉSUMÉ

The mechanisms that maintain intestinal homeostasis despite constant exposure of the gut surface to multiple environmental antigens and to billions of microbes have been scrutinized over the past 20 years with the goals to gain basic knowledge, but also to elucidate the pathogenesis of inflammatory bowel diseases (IBD) and to identify therapeutic targets for these severe diseases. Considerable insight has been obtained from studies based on gene inactivation in mice as well as from genome wide screens for genetic variants predisposing to human IBD. These studies are, however, not sufficient to delineate which pathways play key nonredundant role in the human intestinal barrier and to hierarchize their respective contribution. Here, we intend to illustrate how such insight can be derived from the study of human Mendelian diseases, in which severe intestinal pathology results from single gene defects that impair epithelial and or hematopoietic immune cell functions. We suggest that these diseases offer the unique opportunity to study in depth the pathogenic mechanisms leading to perturbation of intestinal homeostasis in humans. Furthermore, molecular dissection of monogenic intestinal diseases highlights key pathways that might be druggable and therapeutically targeted in common forms of IBD.


Sujet(s)
Interactions hôte-microbes/immunologie , Immunité muqueuse , Immunomodulation , Muqueuse intestinale/physiologie , Immunité acquise , Animaux , Transport biologique , Marqueurs biologiques , Différenciation cellulaire/génétique , Différenciation cellulaire/immunologie , Prédisposition aux maladies , Entérocytes/cytologie , Entérocytes/immunologie , Entérocytes/métabolisme , Régulation de l'expression des gènes , Maladies génétiques congénitales/génétique , Maladies génétiques congénitales/immunologie , Prédisposition génétique à une maladie , Hématopoïèse/génétique , Hématopoïèse/immunologie , Homéostasie , Interactions hôte-microbes/génétique , Humains , Immunité innée , Mutation , Transduction du signal
10.
Mucosal Immunol ; 14(3): 751-761, 2021 05.
Article de Anglais | MEDLINE | ID: mdl-33674763

RÉSUMÉ

Although they globally cause viral gastroenteritis in children, astroviruses are understudied due to the lack of well-defined animal models. While murine astroviruses (muAstVs) chronically infect immunodeficient mice, a culture system and understanding of their pathogenesis is lacking. Here, we describe a platform to cultivate muAstV using air-liquid interface (ALI) cultures derived from mouse enteroids, which support apical infection and release. Chronic muAstV infection occurs predominantly in the small intestine and correlates with higher interferon-lambda (IFN-λ) expression. MuAstV stimulates IFN-λ production in ALI, recapitulating our in vivo findings. We demonstrate that goblet cells and enterocytes are targets for chronic muAstV infection in vivo, and that infection is enhanced by parasite co-infection or type 2 cytokine signaling. Depletion of goblet cells from ALI limits muAstV infection in vitro. During chronic infection, muAstV stimulates IFN-λ production in infected cells and induces ISGs throughout the intestinal epithelium in an IFN-λ-receptor-dependent manner. Collectively, our study provides insights into the cellular tropism and innate immune responses to muAstV and establishes an enteroid-based culture system to propagate muAstV in vitro.


Sujet(s)
Infections à Astroviridae/immunologie , Astroviridae/physiologie , Cytokines/métabolisme , Entérocytes/virologie , Gastroentérite/immunologie , Cellules caliciformes/virologie , Lymphocytes auxiliaires Th2/immunologie , Animaux , Cellules cultivées , Co-infection , Entérocytes/immunologie , Cellules caliciformes/immunologie , Humains , Immunité innée , Souris , Souris de lignée C57BL , Souris knockout , Techniques de culture d'organes , Tropisme viral
11.
Mucosal Immunol ; 14(3): 615-629, 2021 05.
Article de Anglais | MEDLINE | ID: mdl-33731826

RÉSUMÉ

The gut epithelium is a critical protective barrier. Its NAIP/NLRC4 inflammasome senses infection by Gram-negative bacteria, including Salmonella Typhimurium (S.Tm) and promotes expulsion of infected enterocytes. During the first ~12-24 h, this reduces mucosal S.Tm loads at the price of moderate enteropathy. It remained unknown how this NAIP/NLRC4-dependent tradeoff would develop during subsequent infection stages. In NAIP/NLRC4-deficient mice, S.Tm elicited severe enteropathy within 72 h, characterized by elevated mucosal TNF (>20 pg/mg) production from bone marrow-derived cells, reduced regeneration, excessive enterocyte loss, and a collapse of the epithelial barrier. TNF-depleting antibodies prevented this destructive pathology. In hosts proficient for epithelial NAIP/NLRC4, a heterogeneous enterocyte death response with both apoptotic and pyroptotic features kept S.Tm loads persistently in check, thereby preventing this dire outcome altogether. Our results demonstrate that immediate and selective removal of infected enterocytes, by locally acting epithelium-autonomous NAIP/NLRC4, is required to avoid a TNF-driven inflammatory hyper-reaction that otherwise destroys the epithelial barrier.


Sujet(s)
Protéines régulatrices de l'apoptose/métabolisme , Protéines de liaison au calcium/métabolisme , Entérocytes/immunologie , Inflammation/immunologie , Muqueuse intestinale/anatomopathologie , Protéine inhibitrice de l'apoptose neuronale/métabolisme , Salmonelloses/immunologie , Salmonella typhimurium/physiologie , Animaux , Protéines régulatrices de l'apoptose/génétique , Protéines de liaison au calcium/génétique , Cellules cultivées , Cytotoxicité immunologique , Souris , Souris de lignée C57BL , Souris knockout , Protéine inhibitrice de l'apoptose neuronale/génétique , Jonctions serrées/métabolisme , Facteur de nécrose tumorale alpha/métabolisme
12.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Article de Anglais | MEDLINE | ID: mdl-33372132

RÉSUMÉ

The apicomplexan parasite Cryptosporidium infects the intestinal epithelium. While infection is widespread around the world, children in resource-poor settings suffer a disproportionate disease burden. Cryptosporidiosis is a leading cause of diarrheal disease, responsible for mortality and stunted growth in children. CD4 T cells are required to resolve this infection, but powerful innate mechanisms control the parasite prior to the onset of adaptive immunity. Here, we use the natural mouse pathogen Cryptosporidium tyzzeri to demonstrate that the inflammasome plays a critical role in initiating this early response. Mice lacking core inflammasome components, including caspase-1 and apoptosis-associated speck-like protein, show increased parasite burden and caspase 1 deletion solely in enterocytes phenocopies whole-body knockout (KO). This response was fully functional in germfree mice and sufficient to control Cryptosporidium infection. Inflammasome activation leads to the release of IL-18, and mice that lack IL-18 are more susceptible to infection. Treatment of infected caspase 1 KO mice with recombinant IL-18 is remarkably efficient in rescuing parasite control. Notably, NOD-like receptor family pyrin domain containing 6 (NLRP6) was the only NLR required for innate parasite control. Taken together, these data support a model of innate recognition of Cryptosporidium infection through an NLRP6-dependent and enterocyte-intrinsic inflammasome that leads to the release of IL-18 required for parasite control.


Sujet(s)
Cryptosporidiose/immunologie , Entérocytes/métabolisme , Inflammasomes/métabolisme , Interleukine-18/métabolisme , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines de liaison aux phosphates/métabolisme , Récepteurs de surface cellulaire/métabolisme , Animaux , Caspase-1/métabolisme , Cryptosporidium/physiologie , Entérocytes/immunologie , Interactions hôte-pathogène , Souris
13.
Food Funct ; 11(10): 8516-8526, 2020 Oct 21.
Article de Anglais | MEDLINE | ID: mdl-32975271

RÉSUMÉ

Lactoferrin (LF) plays critical roles in various physiological processes. However, its protective effects on small intestinal epithelial cells remain poorly understood. This study aimed to investigate its protective effects and underlying mechanisms in vitro on lipopolysaccharide (LPS)-challenged intestinal porcine epithelial cells (IPEC-J2 cells). The IPEC-J2 cells were treated with or without LPS and LF for 24 h and analyzed using various assays. The results indicated that the LPS treatment induced the secretion of pro-inflammatory cytokines [interleukin (IL)-1ß, IL-8, and TNF-α], increased cell permeability, and enhanced reactive oxygen species (ROS) production. The LF treatment decreased the secretion and gene expression of IL-1ß and downregulated the phosphorylation levels of NF-κB, IκB, P38, and ERK1/2 in LPS-challenged cells. Moreover, the LF treatment decreased cell permeability, enhanced the expression of claudin-1 protein, and inhibited the expression of the myosin light-chain kinase (MLCK) protein in LPS-challenged cells. It also reduced the ROS and MDA production as well as upregulated the GSH-Px activity and the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) protein. Taken together, these results suggested that LF alleviated the LPS-induced cellular inflammation through the attenuation of nuclear factor kappa B (NF-κB)/mitogen-activated protein kinase (MAPK) pathways, maintaining cellular barrier integrity and mitigating oxidative stress.


Sujet(s)
Entérocytes/immunologie , Inflammation , Lactoferrine/pharmacologie , Lipopolysaccharides/immunologie , Système de signalisation des MAP kinases , Facteur-2 apparenté à NF-E2/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Animaux , Lignée cellulaire , Cytokines/métabolisme , Entérocytes/métabolisme , Protéines I-kappa B/métabolisme , Interleukine-1 bêta/génétique , Interleukine-1 bêta/métabolisme , Mitogen-Activated Protein Kinase 1/métabolisme , Mitogen-Activated Protein Kinase 3/métabolisme , NADPH dehydrogenase (quinone)/métabolisme , Stress oxydatif , Perméabilité , Phosphorylation , Espèces réactives de l'oxygène/métabolisme , Transduction du signal , Suidae , p38 Mitogen-Activated Protein Kinases/métabolisme
14.
Acta Trop ; 211: 105592, 2020 Nov.
Article de Anglais | MEDLINE | ID: mdl-32565198

RÉSUMÉ

Elastase-1 is one member of serine protease family, distributes in organisms widely and plays a crucial role in the invasion and development of Trichinella spiralis. In order to identify the binding of T. spiralis elastase-1 (TsEla) with host's intestinal epithelial cells (IECs) and its role in Trichinella larval intrusion, TsEla gene was cloned and expressed in our previous study. The recombinant TsEla (rTsEla) has the enzymatic activity to degrade specific peptide substrate. A specific binding between rTsEla and IECs was detected by Far Western blot and ELISA. In an in vitro invasion assay, rTsEla promoted the larval intrusion, whereas anti-rTsEla serum inhibited the larval penetration. The larval intrusion was also suppressed after the silencing of TsEla by siRNA. Silencing of TsEla gene by siRNA-291 meditated RNA interference suppressed TsEla protein expression, reduced the worm infectivity, development and reproductive capacity. These results indicated that TsEla plays an important role in the T. spiralis intrusion of host's intestinal epithelia, and it could be a prospective vaccine molecular target against T. spiralis infection.


Sujet(s)
Entérocytes/physiologie , Protéines d'helminthes/métabolisme , Muqueuse intestinale/physiologie , Pancreatic elastase/métabolisme , Trichinella spiralis/enzymologie , Trichinellose/parasitologie , Animaux , Entérocytes/immunologie , Cellules épithéliales , Régulation de l'expression des gènes codant pour des enzymes , Protéines d'helminthes/immunologie , Intestins , Larve/physiologie , Souris , Souris de lignée BALB C , Pancreatic elastase/composition chimique , Pancreatic elastase/génétique , Études prospectives , Trichinella spiralis/génétique , Trichinellose/immunologie
15.
Cell ; 181(5): 1016-1035.e19, 2020 05 28.
Article de Anglais | MEDLINE | ID: mdl-32413319

RÉSUMÉ

There is pressing urgency to understand the pathogenesis of the severe acute respiratory syndrome coronavirus clade 2 (SARS-CoV-2), which causes the disease COVID-19. SARS-CoV-2 spike (S) protein binds angiotensin-converting enzyme 2 (ACE2), and in concert with host proteases, principally transmembrane serine protease 2 (TMPRSS2), promotes cellular entry. The cell subsets targeted by SARS-CoV-2 in host tissues and the factors that regulate ACE2 expression remain unknown. Here, we leverage human, non-human primate, and mouse single-cell RNA-sequencing (scRNA-seq) datasets across health and disease to uncover putative targets of SARS-CoV-2 among tissue-resident cell subsets. We identify ACE2 and TMPRSS2 co-expressing cells within lung type II pneumocytes, ileal absorptive enterocytes, and nasal goblet secretory cells. Strikingly, we discovered that ACE2 is a human interferon-stimulated gene (ISG) in vitro using airway epithelial cells and extend our findings to in vivo viral infections. Our data suggest that SARS-CoV-2 could exploit species-specific interferon-driven upregulation of ACE2, a tissue-protective mediator during lung injury, to enhance infection.


Sujet(s)
Pneumocytes/métabolisme , Entérocytes/métabolisme , Cellules caliciformes/métabolisme , Interféron de type I/métabolisme , Muqueuse nasale/cytologie , Peptidyl-Dipeptidase A/génétique , Adolescent , Pneumocytes/immunologie , Angiotensin-converting enzyme 2 , Animaux , Betacoronavirus/physiologie , COVID-19 , Lignée cellulaire , Cellules cultivées , Enfant , Infections à coronavirus/virologie , Entérocytes/immunologie , Cellules caliciformes/immunologie , Infections à VIH/immunologie , Humains , Grippe humaine/immunologie , Interféron de type I/immunologie , Poumon/cytologie , Poumon/anatomopathologie , Macaca mulatta , Souris , Mycobacterium tuberculosis , Muqueuse nasale/immunologie , Pandémies , Peptidyl-Dipeptidase A/métabolisme , Pneumopathie virale/virologie , Récepteurs viraux/génétique , SARS-CoV-2 , Serine endopeptidases/métabolisme , Analyse sur cellule unique , Tuberculose/immunologie , Régulation positive
16.
Vet Microbiol ; 242: 108596, 2020 Mar.
Article de Anglais | MEDLINE | ID: mdl-32122600

RÉSUMÉ

Gut is often subject to infection by different pathogens like Y. enterocolitica. To date, biotypes (BTs) 1A have been considered as non-pathogenic, because they do not express plasmid of virulence pYV; however, BTs 1A strains present other chromosomic virulence genes and recent studies suggest an implication of this microorganism in reactive arthritis. Although many studies highlighted the molecular basis of pathogenesis of Ye infection, scanty data are available about several environmental BTs 1A strains, often isolated in cases of foodborne disease but not included in pathogenicity studies. The aim of our work was to verify the ability of different Ye 1A strains to adhere and penetrate IPEC-J2 cells and to modulate intestinal innate immunity. Our results showed that all strains under study were able to adhere and penetrate enterocytes, causing inflammatory responses. Indeed, adhesion and invasion of enterocytes is an essential step in Ye pathogenesis (Fàbrega and Vila, 2012). Moreover, our data suggest the possible involvement of strains Ye2/O:9 in reactive arthritis, due to their ability (i) to penetrate enterocytes as pathogenic Ye1/O:8 strains do, and (ii) to increase IL-6, IL-8, IL-12 and IL-18 release. Lastly, our results confirm that IPEC-J2 cells are a very good model to evaluate host-pathogen interaction, and indicate IL-8, TNF-α, TLRs1 and 4 as possible markers of the ability of Ye strains to penetrate enterocytes. Moreover, we showed that Ye strains differently affect the host's innate immune responses.


Sujet(s)
Entérocytes/immunologie , Entérocytes/microbiologie , Interactions hôte-microbes/immunologie , Immunité innée , Jéjunum/cytologie , Yersinioses/immunologie , Animaux , Adhérence bactérienne , Lignée cellulaire , Cytokines/immunologie , Jéjunum/immunologie , Jéjunum/microbiologie , Suidae , Virulence , Yersinia enterocolitica/classification
17.
Int J Biol Macromol ; 153: 600-607, 2020 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-32165203

RÉSUMÉ

Glucans are complex polysaccharides consisting of repeated units of d-glucose linked by glycosidic bonds. The nutritional contribution in α-glucans is mainly given by starch and glycogen while in ß-glucans by mushrooms, yeasts and whole grains, such as barley and spelt well represented in the Mediterranean Diet. Numerous and extensive studies performed on glucans highlighted their marked anti-tumor, antioxidant and immunomodulatory activity. It has recently been shown that rather than merely being a passive barrier, the intestinal epithelium is an essential modulator of immunity. Indeed, epithelial absorptive enterocytes and mucin secreting goblet cells can produce specific immune modulating factors, driving innate immunity to pathogens as well as preventing autoimmunity. Despite the clear evidence of the effects of glucans on immune system cells, there are only limited data about their effects on immune activity of mucosal intestinal cells strictly related to intestinal barrier integrity. The aim of the study was to evaluate the effects of α and ß glucans, alone or in combination with other substances with antioxidant properties, on reactive oxygen species (ROS) levels, on the expression of ROS-generating enzyme DUOX-2 and of the immune modulating factors Tumor Necrosis Factor (TNF-α), Interleukin 1 ß (IL-1ß) and cyclooxygenase-2 (COX-2) in two intestinal epithelial cells, the enterocyte-like Caco-2 cells and goblet cell-like LS174T. In our research, the experiments were carried out incubating the cells with glucans for 18 h in culture medium containing 0.2% FBS and measuring ROS levels fluorimetrically as dihydrodichlorofluoresce diacetate (DCF-DA) fluorescence, protein levels of DUOX-2 by Western blotting and mRNA levels of, TNF-α, IL-1ß and COX-2 by qRT-PCR. α and ß glucans decreased ROS levels in Caco-2 and LS 174T cells. The expression levels of COX-2, TNF-α, and IL-1ß were also reduced by α- and ß-glucans. Additive effects on the expression of these immune modulating factors were exerted by vitamin C. In Caco-2 cells, the dual oxidase DUOX-2 expression is positively modulated by ROS. Accordingly, in Caco-2 or LS174T cells treated with α and ß-glucans alone or in combination with Vitamin C, the decrease of ROS levels was associated with a reduced expression of DUOX-2. The treatment of cells with the NADPH oxidase (NOX) inhibitor apocynin decrease ROS, DUOX-2, COX-2, TNF-α and IL-1ß levels indicating that NOX dependent ROS regulate the expression of immune modulating factors of intestinal cells. However, the combination of vitamin C, α and ß-glucans with apocynin did not exert an additive effect on COX-2, TNF-α and IL-1ß levels when compared with α-, ß-glucans and Vitamin C alone. The present study showing a modulatory effect of α and ß-glucans on ROS and on the expression of immune modulating factors in intestinal epithelial cells suggests that the assumption of food containing high levels of these substances or dietary supplementation can contribute to normal immunomodulatory function of intestinal barrier.


Sujet(s)
Entérocytes/immunologie , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Glucanes/pharmacologie , Cellules caliciformes/immunologie , Cellules Caco-2 , Cyclooxygenase 2/immunologie , Dual oxydases/immunologie , Entérocytes/cytologie , Régulation de l'expression des gènes/immunologie , Cellules caliciformes/cytologie , Humains , Interleukine-1 bêta/immunologie , Facteur de nécrose tumorale alpha/immunologie
18.
mBio ; 11(1)2020 01 14.
Article de Anglais | MEDLINE | ID: mdl-31937650

RÉSUMÉ

Hepatitis E virus (HEV), a single-strand positive-sense RNA virus, is an understudied but important human pathogen. The virus can establish infection at a number of host tissues, including the small intestine and liver, causing acute and chronic hepatitis E as well as certain neurological disorders. The retinoic acid-inducible gene I (RIG-I) pathway is essential to induce the interferon (IFN) response during HEV infection. However, the pathogen-associated motif patterns (PAMPs) in the HEV genome that are recognized by RIG-I remain unknown. In this study, we first identified that HEV RNA PAMPs derived from the 3' untranslated region (UTR) of the HEV genome induced higher levels of IFN mRNA, interferon regulatory factor-3 (IRF3) phosphorylation, and nuclear translocation than the 5' UTR of HEV. We revealed that the U-rich region in the 3' UTR of the HEV genome acts as a potent RIG-I PAMP, while the presence of poly(A) tail in the 3' UTR further increases the potency. We further demonstrated that HEV UTR PAMPs induce differential type I and type III IFN responses in a cell type-dependent fashion. Predominant type III IFN response was observed in the liver tissues of pigs experimentally infected with HEV as well as in HEV RNA PAMP-induced human hepatocytes in vitro In contrast, HEV RNA PAMPs induced a predominant type I IFN response in swine enterocytes. Taken together, the results from this study indicated that the IFN response during HEV infection depends both on viral RNA motifs and host target cell types. The results have important implications in understanding the mechanism of HEV pathogenesis.IMPORTANCE Hepatitis E virus (HEV) is an important human pathogen causing both acute and chronic viral hepatitis E infection. Currently, the mechanisms of HEV replication and pathogenesis remain poorly understood. The innate immune response acts as the first line of defense during viral infection. The retinoic acid-inducible gene I (RIG-I)-mediated interferon (IFN) response has been implicated in establishing antiviral response during HEV infection, although the HEV RNA motifs that are recognized by RIG-I are unknown. This study identified that the U-rich region in the 3' untranslated region (UTR) of the HEV genome acts as a potent RIG-I agonist compared to the HEV 5' UTR. We further revealed that the HEV RNA pathogen-associated motif patterns (PAMPs) induced a differential IFN response in a cell type-dependent manner: a predominantly type III IFN response in hepatocytes, and a predominantly type I IFN response in enterocytes. These data demonstrate the complexity by which both host and viral factors influence the IFN response during HEV infection.


Sujet(s)
Virus de l'hépatite E/immunologie , Interactions hôte-microbes/immunologie , Immunité innée , Interféron de type I/immunologie , Interférons/immunologie , Régions non traduites/immunologie , Régions 3' non traduites/immunologie , Régions 5' non traduites/immunologie , Animaux , Entérocytes/immunologie , Entérocytes/virologie , Génome viral , Virus de l'hépatite E/pathogénicité , Hépatocytes/immunologie , Hépatocytes/virologie , Humains , Facteur-3 de régulation d'interféron/métabolisme , Molécules contenant des motifs associés aux pathogènes/immunologie , Phosphorylation , Suidae , Interféron lambda
19.
Front Immunol ; 11: 604075, 2020.
Article de Anglais | MEDLINE | ID: mdl-33679694

RÉSUMÉ

Several formulas are available for the dietary treatment of cow's milk allergy (CMA). Clinical data suggest potentially different effect on immune tolerance elicited by these formulas. We aimed to comparatively evaluate the tolerogenic effect elicited by the protein fraction of different formulas available for the dietary treatment of CMA. Five formulas were compared: extensively hydrolyzed whey formula (EHWF), extensively hydrolyzed casein formula (EHCF), hydrolyzed rice formula (HRF), soy formula (SF), and amino acid-based formula (AAF). The formulas were reconstituted in water according to the manufacturer's instructions and subjected to an in vitro infant gut simulated digestion using a sequential gastric and duodenal static model. Protein fraction was then purified and used for the experiments on non-immune and immune components of tolerance network in human enterocytes and in peripheral mononuclear blood cells (PBMCs). We assessed epithelial layer permeability and tight junction proteins (occludin and zonula occludens-1, ZO-1), mucin 5AC, IL-33, and thymic stromal lymphopoietin (TSLP) in human enterocytes. In addition, Th1/Th2 cytokine response and Tregs activation were investigated in PBMCs from IgE-mediated CMA infants. EHCF-derived protein fraction positively modulated the expression of gut barrier components (mucin 5AC, occludin and ZO-1) in human enterocytes, while SF was able to stimulate the expression of occludin only. EHWF and HRF protein fractions elicited a significant increase in TSLP production, while IL-33 release was significantly increased by HRF and SF protein fractions in human enterocytes. Only EHCF-derived protein fraction elicited an increase of the tolerogenic cytokines production (IL-10, IFN-γ) and of activated CD4+FoxP3+ Treg number, through NFAT, AP1, and Nf-Kb1 pathway. The effect paralleled with an up-regulation of FoxP3 demethylation rate. Protein fraction from all the study formulas was unable to induce Th2 cytokines production. The results suggest a different regulatory action on tolerogenic mechanisms elicited by protein fraction from different formulas commonly used for CMA management. EHCF-derived protein fraction was able to elicit tolerogenic effect through at least in part an epigenetic modulation of FoxP3 gene. These results could explain the different clinical effects observed on immune tolerance acquisition in CMA patients and on allergy prevention in children at risk for atopy observed using EHCF.


Sujet(s)
Entérocytes/métabolisme , Tolérance immunitaire , Préparation pour nourrissons , Muqueuse intestinale/métabolisme , Hypersensibilité au lait/diétothérapie , Hydrolysats de protéines/métabolisme , Lymphocytes T/métabolisme , Acides aminés/immunologie , Acides aminés/métabolisme , Animaux , Cellules Caco-2 , Caséines/immunologie , Caséines/métabolisme , Cytokines/métabolisme , Impédance électrique , Entérocytes/immunologie , Épigenèse génétique , Facteurs de transcription Forkhead/génétique , Facteurs de transcription Forkhead/métabolisme , Humains , Nourrisson , Muqueuse intestinale/immunologie , Hypersensibilité au lait/immunologie , Hypersensibilité au lait/métabolisme , Oryza , Perméabilité , Hydrolysats de protéines/immunologie , Protéines de soja/immunologie , Protéines de soja/métabolisme , Lymphocytes T/immunologie , Protéines de lactosérum/immunologie , Protéines de lactosérum/métabolisme
20.
Cell Mol Gastroenterol Hepatol ; 9(3): 403-423, 2020.
Article de Anglais | MEDLINE | ID: mdl-31756560

RÉSUMÉ

BACKGROUND & AIMS: Necrotizing enterocolitis (NEC) is a devastating disease of premature infants characterized by Toll-like receptor 4 (TLR4)-dependent intestinal inflammation and enterocyte death. Given that necroptosis is a proinflammatory cell death process that is linked to bacterial signaling, we investigated its potential role in NEC, and the mechanisms involved. METHODS: Human and mouse NEC intestine were analyzed for necroptosis gene expression (ie, RIPK1, RIPK3, and MLKL), and protein activation (phosphorylated RIPK3). To evaluate a potential role for necroptosis in NEC, the effects of genetic (ie, Ripk3 knockout or Mlkl knockout) or pharmacologic (ie, Nec1s) inhibition of intestinal inflammation were assessed in a mouse NEC model, and a possible upstream role of TLR4 was assessed in Tlr4-deficient mice. The NEC-protective effects of human breast milk and its constituent milk oligosaccharides on necroptosis were assessed in a NEC-in-a-dish model, in which mouse intestinal organoids were cultured as either undifferentiated or differentiated epithelium in the presence of NEC bacteria and hypoxia. RESULTS: Necroptosis was activated in the intestines of human and mouse NEC in a TLR4-dependent manner, and was up-regulated specifically in differentiated epithelium of the immature ileum. Inhibition of necroptosis genetically and pharmacologically reduced intestinal-epithelial cell death and mucosal inflammation in experimental NEC, and ex vivo in the NEC-in-a-dish system. Strikingly, the addition of human breast milk, or the human milk oligosaccharide 2 fucosyllactose in the ex vivo system, reduced necroptosis and inflammation. CONCLUSIONS: Necroptosis is activated in the intestinal epithelium upon TLR4 signaling and is required for NEC development, and explains in part the protective effects of breast milk.


Sujet(s)
Entérocolite nécrosante/anatomopathologie , Entérocytes/anatomopathologie , Muqueuse intestinale/anatomopathologie , Lait humain/composition chimique , Nécroptose/immunologie , Animaux , Modèles animaux de maladie humaine , Entérocolite nécrosante/traitement médicamenteux , Entérocolite nécrosante/génétique , Entérocolite nécrosante/immunologie , Entérocytes/effets des médicaments et des substances chimiques , Entérocytes/immunologie , Femelle , Humains , Nouveau-né , Muqueuse intestinale/effets des médicaments et des substances chimiques , Souris , Souris knockout , Nécroptose/effets des médicaments et des substances chimiques , Protein kinases/génétique , Receptor-Interacting Protein Serine-Threonine Kinases/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/génétique , Transduction du signal/immunologie , Récepteur de type Toll-4/génétique , Récepteur de type Toll-4/métabolisme , Triholosides/pharmacologie , Triholosides/usage thérapeutique , Régulation positive
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...