Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 434
Filter
1.
Wiad Lek ; 75(5 pt 2): 1309-1312, 2022.
Article in English | MEDLINE | ID: mdl-35758449

ABSTRACT

OBJECTIVE: The aim: Based on the above cytological signs of M-cells, we set the goal of more detailed clarification of some of their topological relationships with other enterocytes in the follicle-associated epithelium of Peyer's patches of albino rat small intestine. PATIENTS AND METHODS: Materials and methods: 10 mature albino male rats weighted 200,0±20,0 g were involved into the study. Anatomical dissection with the sampling of the sections of the small intestine containing Peyer's patches was carried out with subsequent embedment of the latter into paraffin blocks and making of serial histological sections of 4 µm thick in the cross-section of the small intestine, followed with hematoxylin-eosin staining. The specimens were studied and documented on the "Konus" light microscope equipped. Morphometric characteristics of the specimen tissue structures were studied using the Sigeta X 1 mm/100 Div.x0.01mm stage micrometer. RESULTS: Results: The findings of the study revealed enterocytes with phagocytic properties found in the lymphoid-associated epithelium of Peyer's patches of the small intestine of albino rats. Moreover, if they are clearly visualized at the light-optical level, then M-cells are poorly recognizable, which is consistent with a similar assessment made by other authors. CONCLUSION: Conclusions: Given this, the issue on the topology and functional purpose of M-cells remains uncertain to date and, thereby, the prospect of further research is being outlined, which, in our opinion, can be successful using the method of stereomorphological analysis. For this purpose, multilayer plastic reconstruction methods can be used for serial semi-thin sections of Peyer's patches embedded in epoxy resin, according to the requirements of transmission electron microscopy.


Subject(s)
Intestine, Small , Peyer's Patches , Epithelium , Humans , Peyer's Patches/chemistry , Peyer's Patches/pathology , Rats , Staining and Labeling
2.
Front Immunol ; 12: 697725, 2021.
Article in English | MEDLINE | ID: mdl-34804004

ABSTRACT

The intestinal mucosal immune environment requires multiple immune cells to maintain homeostasis. Although intestinal B cells are among the most important immune cells, little is known about the mechanism that they employ to regulate immune homeostasis. In this study, we found that CD11b+ B cells significantly accumulated in the gut lamina propria and Peyer's patches in dextran sulfate sodium-induced colitis mouse models and patients with ulcerative colitis. Adoptive transfer of CD11b+ B cells, but not CD11b-/- B cells, effectively ameliorated colitis and exhibited therapeutic effects. Furthermore, CD11b+ B cells were found to produce higher levels of IgA than CD11b- B cells. CD11b deficiency in B cells dampened IgA production, resulting in the loss of their ability to ameliorate colitis. Mechanistically, CD11b+ B cells expressed abundant TGF-ß and TGF-ß receptor II, as well as highly activate phosphorylated Smad2/3 signaling pathway, consequently promoting the class switch to IgA. Collectively, our findings demonstrate that CD11b+ B cells are essential intestinal suppressive immune cells and the primary source of intestinal IgA, which plays an indispensable role in maintaining intestinal homeostasis.


Subject(s)
B-Lymphocytes/immunology , CD11b Antigen/immunology , Colitis, Ulcerative/immunology , Colitis/immunology , Immunoglobulin A, Secretory/immunology , Peyer's Patches/immunology , Adoptive Transfer , Animals , B-Lymphocytes/pathology , CD11b Antigen/genetics , Colitis/chemically induced , Colitis/pathology , Colitis, Ulcerative/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Humans , Immunoglobulin Class Switching , Intestinal Mucosa/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Peyer's Patches/pathology , Signal Transduction , Smad2 Protein/metabolism
3.
Virology ; 552: 43-51, 2021 01 02.
Article in English | MEDLINE | ID: mdl-33059319

ABSTRACT

This study focused on intestinal restitution including phenotype switching of absorptive enterocytes and the abundance of different enterocyte subtypes in weaned pigs after porcine epidemic diarrhea virus (PEDV) infection. At 10 days post-PEDV-inoculation, the ratio of villus height to crypt depth in both jejunum and ileum had restored, and the PEDV antigen was not detectable. However, enterocytes at the villus tips revealed epithelial-mesenchymal transition (EMT) in the jejunum in which E-cadherin expression decreased while expression of N-cadherin, vimentin, and Snail increased. Additionally, there was reduced expression of actin in microvilli and Zonula occludens-1 (ZO-1) in tight junctions. Moreover, the protein concentration of transforming growth factor ß1 (TGFß1), which mediates EMT and cytoskeleton alteration, was increased. We also found a decreased number of Peyer's patch M cells in the ileum. These results reveal incomplete restitution of enterocytes in the jejunum and potentially impaired immune surveillance in the ileum after PEDV infection.


Subject(s)
Coronavirus Infections/veterinary , Enterocytes/pathology , Epithelial-Mesenchymal Transition , Gastroenteritis, Transmissible, of Swine/pathology , Peyer's Patches/pathology , Porcine epidemic diarrhea virus/pathogenicity , Animals , Cadherins/metabolism , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Coronavirus Infections/virology , Gastroenteritis, Transmissible, of Swine/immunology , Gastroenteritis, Transmissible, of Swine/virology , Ileum/immunology , Ileum/pathology , Intestinal Mucosa/pathology , Jejunum/immunology , Jejunum/pathology , Microvilli/pathology , Swine , Tight Junctions/pathology , Transforming Growth Factor beta1/metabolism , Weaning
4.
Nutrients ; 12(11)2020 Nov 04.
Article in English | MEDLINE | ID: mdl-33158132

ABSTRACT

There is no effective therapy for milk allergy. The role of lactic acid bacteria (LAB) and probiotics in protection against allergy-related outcomes is still under investigation. The aim of the study was to evaluate the immunomodulative and therapeutic potential of yogurt drinks in cow's milk allergy (CMA) management. We compared immunoreactivity of α-casein (α-CN), ß-casein (ß-CN), κ-casein (κ-CN), α-lactalbumin (α-LA), and ß-lactoglobulin (ß-LG) in 27 yogurt drinks fermented with different basic yogurt cultures, or yogurt cultures enriched with Lactobacillus plantarum and/or Bifidobacterium lactis strains, by competitive ELISA assay. Drinks with the lowest antigenic potential were used as allergoids for CMA therapy. BALB/c mice were sensitized via intraperitoneal injection of α-CN + ß-LG mixture with aluminum adjuvant, and gavaged with increasing doses of selected low-immunogenic drinks (YM-basic, or YM-LB-enriched with L. plantarum and B. lactis) to induce tolerance. Milk- or phosphate-buffered saline (PBS)-dosed mice served as controls. Compared to milk, the immunoreactivity of proteins in drinks increased or decreased, depending on the bacterial sets applied for fermentation. Only a few sets acted synergistically in reducing immunoreactivity. The selected low-immunogenic drinks stimulated allergic mice for profiling Th2 to Th1 response and acquire tolerance, and the effect was greater with YM-LB drink, which during long-lasting interventional feeding strongly increased the secretion of regulatory cytokines, i.e., IL-10 and TGF-ß, and IgA and decreased IL-4, IgE, and anti-(α-CN + ß-LG) IgG1. The studies revealed variations in the potency of yogurt bacteria to change allergenicity of milk proteins and the need for their strict selection to obtain a safe product for allergy sufferers. The YM-LB drink with reduced antigenic potential may be a source of allergoids used in the immunotherapy of IgE mediated CMA, but further clinical or volunteer studies are required.


Subject(s)
Bacteria/immunology , Immune Tolerance , Milk Proteins/immunology , Milk/microbiology , Probiotics/pharmacology , Yogurt/microbiology , Animals , Body Weight , Caseins/immunology , Cecum/microbiology , Cytokines/metabolism , Feeding Behavior , Female , Fermentation , Gastrointestinal Microbiome , Immunity, Humoral , Mice, Inbred BALB C , Peyer's Patches/pathology , Spleen/pathology , Whey Proteins/immunology
6.
J Immunol ; 204(10): 2697-2711, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32238457

ABSTRACT

The adaptive immune system of all jawed vertebrates relies on the presence of B and T cell lymphocytes that aggregate in specific body sites to form primary and secondary lymphoid structures. Secondary lymphoid organs include organized MALT (O-MALT) such as the tonsils and Peyer patches. O-MALT became progressively organized during vertebrate evolution, and the TNF superfamily of genes has been identified as essential for the formation and maintenance of O-MALT and other secondary and tertiary lymphoid structures in mammals. Yet, the molecular drivers of O-MALT structures found in ectotherms and birds remain essentially unknown. In this study, we provide evidence that TNFSFs, such as lymphotoxins, are likely not a universal mechanism to maintain O-MALT structures in adulthood of teleost fish, sarcopterygian fish, or birds. Although a role for TNFSF2 (TNF-α) cannot be ruled out, transcriptomics suggest that maintenance of O-MALT in nonmammalian vertebrates relies on expression of diverse genes with shared biological functions in neuronal signaling. Importantly, we identify that expression of many genes with olfactory function is a unique feature of mammalian Peyer patches but not the O-MALT of birds or ectotherms. These results provide a new view of O-MALT evolution in vertebrates and indicate that different genes with shared biological functions may have driven the formation of these lymphoid structures by a process of convergent evolution.


Subject(s)
B-Lymphocytes/immunology , Lymphoid Tissue/immunology , Mucous Membrane/pathology , Palatine Tonsil/pathology , Peyer's Patches/pathology , T-Lymphocytes/immunology , Tumor Necrosis Factors/metabolism , Adaptive Immunity , Animals , Biological Evolution , Immunity, Mucosal , Neural Conduction , Palatine Tonsil/immunology , Peyer's Patches/immunology , Transcriptome , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factors/genetics , Vertebrates
7.
Nutrients ; 12(3)2020 Mar 14.
Article in English | MEDLINE | ID: mdl-32183266

ABSTRACT

Some lactic acid bacteria (LAB) are known to improve atopic dermatitis (AD) through the regulation and stimulation of the host immune system. In this study, we found that ingestion of yogurt containing Lactococcus lactis 11/19-B1 strain (L. lactis 11/19-B1) daily for 8 weeks significantly improved the severity scoring of atopic dermatitis (SCORAD) system score from 38.8 ± 14.4 to 24.2 ± 12.0 in children suffering from AD. We tried to identify which LAB species among the five species contained in the test yogurt contributed to the improvement in AD pathology using an AD mouse model induced by repeated application of 1-fluoro-2, 4-dinitrobenzene (DNFB). AD-like skin lesions on the dorsal skin and ear were most improved by L. lactis 11/19-B1 intake among the five LAB species. In addition, analysis of CD4+ T cell subsets in Peyer's patches (PPs) and cervical lymph nodes (CLNs) indicated that the intake of L. lactis 11/19-B1 generally suppressed all subsets related to inflammation, i.e., Th1, Th2 and Th17, instead of activating the suppressive system, Treg, in the AD mouse model. Histological observations showed ingestion of L. lactis 11/19-B1 significantly suppressed severe inflammatory findings, such as inflammatory cell filtration, epidermal erosion and eosinophil infiltration. These results suggest that the immunomodulatory effects of L. lactis 11/19-B1 contribute to improvements in AD pathology.


Subject(s)
Dermatitis, Atopic , Lactococcus lactis/immunology , Skin , Yogurt , Adolescent , Animals , Child , Child, Preschool , Dermatitis, Atopic/diet therapy , Dermatitis, Atopic/immunology , Dermatitis, Atopic/pathology , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred BALB C , Peyer's Patches/immunology , Peyer's Patches/pathology , Skin/immunology , Skin/pathology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology
8.
J Immunol ; 204(5): 1075-1083, 2020 03 01.
Article in English | MEDLINE | ID: mdl-32071090

ABSTRACT

The mammalian intestine is a complex environment that is constantly exposed to Ags derived from food, microbiota, and metabolites. Intestinal dendritic cells (DC) have the responsibility of establishing oral tolerance against these Ags while initiating immune responses against mucosal pathogens. We now know that DC are a heterogeneous population of innate immune cells composed of classical and monocyte-derived DC, Langerhans cells, and plasmacytoid DC. In the intestine, DC are found in organized lymphoid tissues, such as the mesenteric lymph nodes and Peyer's patches, as well as in the lamina propria. In this Brief Review, we review recent work that describes a division of labor between and collaboration among gut DC subsets in the context of intestinal homeostasis and inflammation. Understanding relationships between DC subtypes and their biological functions will rationalize oral vaccine design and will provide insights into treatments that quiet pathological intestinal inflammation.


Subject(s)
Immunity, Mucosal , Intestinal Mucosa/immunology , Langerhans Cells/immunology , Peyer's Patches/immunology , Animals , Humans , Inflammation/immunology , Inflammation/pathology , Intestinal Mucosa/pathology , Langerhans Cells/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Mesentery/immunology , Mesentery/pathology , Peyer's Patches/pathology
9.
J Allergy Clin Immunol ; 145(3): 982-992, 2020 03.
Article in English | MEDLINE | ID: mdl-31816409

ABSTRACT

BACKGROUND: Kabuki syndrome (KS) is commonly caused by mutations in the histone-modifying enzyme lysine methyltransferase 2D (KMT2D). Immune dysfunction is frequently observed in individuals with KS, but the role of KMT2D in immune system function has not been identified. OBJECTIVE: We sought to understand the mechanisms driving KS-associated immune deficiency (hypogammaglobulinemia [low IgA], splenomegaly, and diminished immunization responses). METHODS: We performed a comprehensive evaluation of humoral immunity and secondary lymphoid tissues in an established KS (Kmt2d+/ßGeo) mouse model and validated select findings in a patient with KS. RESULTS: Compared with wild-type littermates, Kmt2d+/ßGeo mice demonstrated deficiencies in multiple B-cell lineages and reduced serum IgA and elevated IgM levels across multiple ages. The bone marrow, spleen, and intestine of Kmt2d+/ßGeo mice contained diminished numbers of IgA-secreting cells, while elevated germinal center B cells were found in the mesenteric lymph node and Peyer patches. Kmt2d+/ßGeo mice have decreased size and numbers of Peyer patches, a finding confirmed in human samples. We identified deficiency of Itgb7 RNA and protein expression, a gene encoding an adhesion protein that mediates intestinal homing, and we demonstrated KMT2D-dependent control of ITGB7 expression in a human cell line. CONCLUSIONS: Kmt2d haploinsufficiency has broad deleterious effects on B-cell differentiation, specifically hampering gut lymphocyte homing and IgA+ plasma cell differentiation. Intestinal lymphoid defects caused by ITGB7 deficiency have not previously been recognized in KS, and these results provide new mechanistic insights into the pathogenesis of KS-associated immune deficiency.


Subject(s)
Abnormalities, Multiple/immunology , Abnormalities, Multiple/pathology , B-Lymphocytes/pathology , Face/abnormalities , Hematologic Diseases/immunology , Hematologic Diseases/pathology , Peyer's Patches/pathology , Vestibular Diseases/immunology , Vestibular Diseases/pathology , Animals , B-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Movement/immunology , DNA-Binding Proteins/genetics , Face/pathology , Histone-Lysine N-Methyltransferase/genetics , Humans , IgA Deficiency/genetics , IgA Deficiency/immunology , Integrin beta Chains/metabolism , Intestines/immunology , Mice , Mutation , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasm Proteins/genetics , Peyer's Patches/immunology
10.
J Gastroenterol Hepatol ; 35(7): 1143-1149, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31734952

ABSTRACT

BACKGROUND AND AIM: Peyer's patches (PPs) play a major role in intestinal mucosal immunity; however, their role in ulcerative colitis (UC) is not well investigated. We evaluated endoscopic features of PPs on narrow-band imaging with magnifying endoscopy (NBI-ME) and investigated their association with clinical factors. METHODS: We prospectively recruited 105 patients with UC, 18 with Crohn's disease, 16 with disease control, and 33 healthy control subjects at three institutions from 2014 to 2017. NBI-ME images of the villi of PPs were evaluated according to the Villi Index, and patients were divided into the Villi Index low (L) and high (H) types. The 1-year sustained clinical remission rate was evaluated between L-type and H-type PPs in patients with UC. RESULTS: The proportions of patients with H-type PPs were significantly higher among UC, Crohn's disease, and disease control patients than among healthy control patients (P = 0.0125, 0.018, 0.0007). In UC, age, gender, endoscopic score, and extent of disease involvement were not significantly different between L-type and H-type PPs, whereas the sustained clinical remission rate was significantly higher in L-type PPs than in H-type PPs (88% [57/65] vs 65% [17/26], P = 0.019). Multivariate analysis revealed that the L type of PPs was a significant factor for sustained clinical remission (odds ratio 3.8, 95% confidence interval 1.1-12.9, P = 0.033). CONCLUSIONS: Patients with UC showed endoscopic alterations in PPs on NBI-ME, and highly altered appearance of PPs can be associated with a high risk of clinical relapse in patients with UC.


Subject(s)
Colitis, Ulcerative/diagnostic imaging , Colitis, Ulcerative/pathology , Endoscopy, Gastrointestinal/methods , Peyer's Patches/diagnostic imaging , Peyer's Patches/pathology , Adolescent , Adult , Child , Female , Humans , Male , Narrow Band Imaging/methods , Prospective Studies , Recurrence , Remission Induction , Risk , Young Adult
11.
Front Immunol ; 10: 2345, 2019.
Article in English | MEDLINE | ID: mdl-31649668

ABSTRACT

Microfold (M) cells are located in the epithelium covering mucosa-associated lymphoid tissues, such as the Peyer's patches (PPs) of the small intestine. M cells actively transport luminal antigens to the underlying lymphoid follicles to initiate an immune response. The molecular machinery of M-cell differentiation and function has been vigorously investigated over the last decade. Studies have shed light on the role of M cells in the mucosal immune system and have revealed that antigen uptake by M cells contributes to not only mucosal but also systemic immune responses. However, M-cell studies usually focus on infectious diseases; the contribution of M cells to autoimmune diseases has remained largely unexplored. Accumulating evidence suggests that dysbiosis of the intestinal microbiota is implicated in multiple systemic diseases, including autoimmune diseases. This implies that the uptake of microorganisms by M cells in PPs may play a role in the pathogenesis of autoimmune diseases. We provide an outline of the current understanding of M-cell biology and subsequently discuss the potential contribution of M cells and PPs to the induction of systemic autoimmunity, beyond the mucosal immune response.


Subject(s)
Autoimmune Diseases , Gastrointestinal Microbiome/immunology , Immunity, Mucosal , Peyer's Patches , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/microbiology , Autoimmune Diseases/pathology , Dysbiosis/immunology , Dysbiosis/microbiology , Dysbiosis/pathology , Humans , Peyer's Patches/immunology , Peyer's Patches/microbiology , Peyer's Patches/pathology
12.
Cell ; 178(5): 1072-1087.e14, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31442401

ABSTRACT

Nutritional status potentially influences immune responses; however, how nutritional signals regulate cellular dynamics and functionality remains obscure. Herein, we report that temporary fasting drastically reduces the number of lymphocytes by ∼50% in Peyer's patches (PPs), the inductive site of the gut immune response. Subsequent refeeding seemingly restored the number of lymphocytes, but whose cellular composition was conspicuously altered. A large portion of germinal center and IgA+ B cells were lost via apoptosis during fasting. Meanwhile, naive B cells migrated from PPs to the bone marrow during fasting and then back to PPs during refeeding when stromal cells sensed nutritional signals and upregulated CXCL13 expression to recruit naive B cells. Furthermore, temporal fasting before oral immunization with ovalbumin abolished the induction of antigen-specific IgA, failed to induce oral tolerance, and eventually exacerbated food antigen-induced diarrhea. Thus, nutritional signals are critical in maintaining gut immune homeostasis.


Subject(s)
B-Lymphocytes/physiology , Immunity, Mucosal , Animals , Antigens/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bone Marrow/immunology , Bone Marrow/metabolism , Chemokine CXCL13/genetics , Chemokine CXCL13/metabolism , Fasting , Gene Expression Regulation , Glycolysis , Immunoglobulin A/metabolism , Male , Mice , Mice, Inbred BALB C , Nutritional Status , Ovalbumin/immunology , Peyer's Patches/immunology , Peyer's Patches/metabolism , Peyer's Patches/pathology , Receptors, CXCR5/genetics , Receptors, CXCR5/metabolism , Signal Transduction , Stromal Cells/cytology , Stromal Cells/metabolism , TOR Serine-Threonine Kinases/metabolism
13.
Mycopathologia ; 184(4): 461-478, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31230200

ABSTRACT

The details of how gut-associated lymphoid tissues such as Peyer's patches (PPs) in the small intestine play a role in immune surveillance, microbial differentiation and the mucosal barrier protection in response to fungal organisms such as Candida albicans are still unclear. We particularly focus on PPs as they are the immune sensors and inductive sites of the gut that influence inflammation and tolerance. We have previously demonstrated that CD11c+ phagocytes that include dendritic cells and macrophages are located in the sub-epithelial dome within PPs sample C. albicans. To gain insight on how specific cells within PPs sense and respond to the sampling of fungi, we gavaged naïve mice with C. albicans strains ATCC 18804 and SC5314 as well as Saccharomyces cerevisiae. We measured the differential gene expression of sorted CD45+ B220+ B-cells, CD3+ T-cells and CD11c+ DCs within the first 24 h post-gavage using nanostring nCounter® technology. The results reveal that at 24 h, PP phagocytes were the cell type that displayed differential gene expression. These phagocytes were able to sample C. albicans and discriminate between strains. In particular, strain ATCC 18804 upregulated fungal-specific pro-inflammatory genes pertaining to innate and adaptive immune responses. Interestingly, PP CD11c+ phagocytes also differentially expressed genes in response to C. albicans that were important in the protection of the mucosal barrier. These results highlight that the mucosal barrier not only responds to C. albicans, but also aids in the protection of the host.


Subject(s)
Candida albicans/immunology , Gene Expression Profiling , Host-Pathogen Interactions , Inflammation/pathology , Peyer's Patches/immunology , Peyer's Patches/pathology , Administration, Oral , Animals , Antigens, CD/analysis , B-Lymphocytes/chemistry , B-Lymphocytes/immunology , Dendritic Cells/chemistry , Dendritic Cells/immunology , Female , Mice , Saccharomyces cerevisiae/immunology , T-Lymphocytes/chemistry , T-Lymphocytes/immunology
14.
JCI Insight ; 52019 06 04.
Article in English | MEDLINE | ID: mdl-31162138

ABSTRACT

Excess dietary salt contributes to inflammation and hypertension via poorly understood mechanisms. Antigen presenting cells including dendritic cells (DCs) play a key role in regulating intestinal immune homeostasis in part by surveying the gut epithelial surface for pathogens. Previously, we found that highly reactive γ-ketoaldehydes or isolevuglandins (IsoLGs) accumulate in DCs and act as neoantigens, promoting an autoimmune-like state and hypertension. We hypothesized that excess dietary salt alters the gut microbiome leading to hypertension and this is associated with increased immunogenic IsoLG-adduct formation in myeloid antigen presenting cells. To test this hypothesis, we performed fecal microbiome analysis and measured blood pressure of healthy human volunteers with salt intake above or below the American Heart Association recommendations. We also performed 16S rRNA analysis on cecal samples of mice fed normal or high salt diets. In humans and mice, high salt intake was associated with changes in the gut microbiome reflecting an increase in Firmicutes, Proteobacteria and genus Prevotella bacteria. These alterations were associated with higher blood pressure in humans and predisposed mice to vascular inflammation and hypertension in response to a sub-pressor dose of angiotensin II. Mice fed a high salt diet exhibited increased intestinal inflammation including the mesenteric arterial arcade and aorta, with a marked increase in the B7 ligand CD86 and formation of IsoLG-protein adducts in CD11c+ myeloid cells. Adoptive transfer of fecal material from conventionally housed high salt-fed mice to germ-free mice predisposed them to increased intestinal inflammation and hypertension. These findings provide novel insight into the mechanisms underlying inflammation and hypertension associated with excess dietary salt and may lead to interventions targeting the microbiome to prevent and treat this important disease.


Subject(s)
Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dysbiosis , Hypertension/metabolism , Sodium Chloride, Dietary/adverse effects , Sodium Chloride/adverse effects , Adolescent , Adoptive Transfer , Adult , Angiotensin II , Animals , Aorta/metabolism , Bacteria/classification , Bacteria/genetics , Blood Pressure , CD11c Antigen/immunology , Colon/microbiology , Colon/pathology , Cytokines/metabolism , Dendritic Cells/pathology , Disease Models, Animal , Female , Gastrointestinal Microbiome , Humans , Inflammation/metabolism , Lipids , Lymph Nodes , Male , Mice , Mice, Inbred C57BL , Middle Aged , Myeloid Cells/metabolism , Peyer's Patches/microbiology , Peyer's Patches/pathology , RNA, Ribosomal, 16S/genetics , Sodium Chloride/administration & dosage , Sodium Chloride, Dietary/administration & dosage , Young Adult
15.
Sci Rep ; 9(1): 6674, 2019 04 30.
Article in English | MEDLINE | ID: mdl-31040320

ABSTRACT

The early replication of some orally-acquired prion strains upon stromal-derived follicular dendritic cells (FDC) within the small intestinal Peyer's patches is essential to establish host infection, and for the disease to efficiently spread to the brain. Factors that influence the early accumulation of prions in Peyer's patches can directly influence disease pathogenesis. The host's immune response to a gastrointestinal helminth infection can alter susceptibility to co-infection with certain pathogenic bacteria and viruses. Here we used the natural mouse small intestine-restricted helminth pathogen Heligmosomoides polygyrus to test the hypothesis that pathology specifically within the small intestine caused by a helminth co-infection would influence oral prion disease pathogenesis. When mice were co-infected with prions on d 8 after H. polygyrus infection the early accumulation of prions within Peyer's patches was reduced and survival times significantly extended. Natural prion susceptible hosts such as sheep, deer and cattle are regularly exposed to gastrointestinal helminth parasites. Our data suggest that co-infections with small intestine-restricted helminth pathogens may be important factors that influence oral prion disease pathogenesis.


Subject(s)
Coinfection , Disease Susceptibility , Helminthiasis, Animal/complications , Helminthiasis, Animal/parasitology , Helminths , Intestinal Diseases, Parasitic/complications , Intestinal Diseases, Parasitic/parasitology , Prion Diseases/etiology , Animals , Disease Models, Animal , Disease Susceptibility/immunology , Helminthiasis, Animal/pathology , Immunity, Mucosal , Immunohistochemistry , Intestinal Diseases, Parasitic/pathology , Mice , Peyer's Patches/immunology , Peyer's Patches/pathology
16.
Front Immunol ; 10: 361, 2019.
Article in English | MEDLINE | ID: mdl-30891036

ABSTRACT

The intestinal mucosa in inflammatory bowel disease (IBD) contains increased frequencies of lymphocytes and a disproportionate increase in plasma cells secreting immunoglobulin (Ig)G relative to other isotypes compared to healthy controls. Despite consistent evidence of B lineage cells in the mucosa in IBD, little is known of B cell recruitment to the gut in IBD. Here we analyzed B cells in blood of patients with Crohn's disease (CD) and ulcerative colitis (UC) with a range of disease activities. We analyzed the frequencies of known B cell subsets in blood and observed a consistent reduction in the proportion of CD27-IgD- B cells expressing all Ig isotypes in the blood in IBD (independent of severity of disease and treatment) compared to healthy controls. Successful treatment of patients with biologic therapies did not change the profile of B cell subsets in blood. By mass cytometry we demonstrated that CD27-IgD- B cells were proportionately enriched in the gut-associated lymphoid tissue (GALT) in IBD. Since production of TNFα is a feature of IBD relevant to therapies, we sought to determine whether B cells in GALT or the CD27-IgD- subset in particular could contribute to pathology by secretion of TNFα or IL-10. We found that donor matched GALT and blood B cells are capable of producing TNFα as well as IL-10, but we saw no evidence that CD27-IgD- B cells from blood expressed more TNFα compared to other subsets. The reduced proportion of CD27-IgD- B cells in blood and the increased proportion in the gut implies that CD27-IgD- B cells are recruited from the blood to the gut in IBD. CD27-IgD- B cells have been implicated in immune responses to intestinal bacteria and recruitment to GALT, and may contribute to the intestinal inflammatory milieu in IBD.


Subject(s)
B-Lymphocyte Subsets/immunology , Colitis, Ulcerative/blood , Colitis, Ulcerative/immunology , Crohn Disease/blood , Crohn Disease/immunology , Immunoglobulin D/metabolism , Intestinal Mucosa/immunology , Peyer's Patches/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Adult , Antibodies, Monoclonal, Humanized/therapeutic use , Biopsy , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/pathology , Crohn Disease/drug therapy , Crohn Disease/pathology , Female , Gastrointestinal Agents/therapeutic use , Humans , Infliximab/therapeutic use , Interleukin-10/metabolism , Intestinal Mucosa/pathology , Male , Peyer's Patches/pathology , Severity of Illness Index , Tumor Necrosis Factor-alpha/metabolism , Ustekinumab/therapeutic use
17.
Radiother Oncol ; 132: 8-15, 2019 03.
Article in English | MEDLINE | ID: mdl-30825974

ABSTRACT

OBJECTIVE: Exposure to lethal doses of radiation has severe effects on normal tissues. Exposed individuals experience a plethora of symptoms in different organ systems including the gastrointestinal (GI) tract, summarized as Acute Radiation Syndrome (ARS). There are currently no approved drugs for mitigating GI-ARS. A recent high-throughput screen performed at the UCLA Center for Medical Countermeasures against Radiation identified compounds containing sulfonylpiperazine groups with radiation mitigation properties to the hematopoietic system and the gut. Among these 1-[(4-Nitrophenyl)sulfonyl]-4-phenylpiperazine (Compound #5) efficiently mitigated gastrointestinal ARS. However, the mechanism of action and target cells of this drug is still unknown. In this study we examined if Compound #5 affects gut-associated lymphoid tissue (GALT) with its subepithelial domes called Peyer's patches. METHODS: C3H mice were irradiated with 0 or 12 Gy total body irradiation (TBI). A single dose of Compound #5 or solvent was administered subcutaneously 24 h later. 48 h after irradiation the mice were sacrificed, and the guts examined for changes in the number of visible Peyer's patches. In some experiments the mice received 4 daily injections of treatment and were sacrificed 96 h after TBI. For immune histochemistry gut tissues were fixed in formalin and embedded in paraffin blocks. Sections were stained with H&E, anti-Ki67 or a TUNEL assay to assess the number of regenerating crypts, mitotic and apoptotic indices. Cells isolated from Peyer's patches were subjected to immune profiling using flow cytometry. RESULTS: Compound #5 significantly increased the number of visible Peyer's patches when compared to its control in non-irradiated and irradiated mice. Additionally, assessment of total cells per Peyer's patch isolated from these mice demonstrated an overall increase in the total number of Peyer's patch cells per mouse in Compound #5-treated mice. In non-irradiated animals the number of CD11bhigh in Peyer's patches increased significantly. These Compound #5-driven increases did not coincide with a decrease in apoptosis or an increase in proliferation in the germinal centers inside Peyer's patches 24 h after drug treatment. A single dose of Compound #5 significantly increased the number of CD45+ cells after 12 Gy TBI. Importantly, 96 h after 12 Gy TBI Compound #5 induced a significant rise in the number of visible Peyer's patches and the number of Peyer's patch-associated regenerating crypts. CONCLUSION: In summary, our study provides evidence that Compound #5 leads to an influx of immune cells into GALT, thereby supporting crypt regeneration preferentially in the proximity of Peyer's patches.


Subject(s)
Intestine, Small/drug effects , Nitrobenzenes/pharmacology , Peyer's Patches/drug effects , Piperazines/pharmacology , Radiation Injuries/drug therapy , Radiation-Protective Agents/pharmacology , Regeneration/drug effects , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Female , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestinal Mucosa/radiation effects , Intestine, Small/pathology , Intestine, Small/radiation effects , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Peyer's Patches/immunology , Peyer's Patches/pathology , Peyer's Patches/radiation effects , Radiation Injuries/pathology , Random Allocation , Regeneration/radiation effects , Whole-Body Irradiation
18.
Nat Commun ; 9(1): 4870, 2018 11 19.
Article in English | MEDLINE | ID: mdl-30451821

ABSTRACT

RORγt controls the differentiation of TH17 cells, which are mediators of autoimmune conditions such as experimental autoimmune encephalomyelitis (EAE). RORγt also regulates thymocyte development and lymph node genesis. Here we show that the function of RORγt is regulated by its sumoylation. Loss of Sumo3, but not Sumo1, dampens TH17 differentiation and delays the progression of thymic CD8+ immature single-positive cells (ISPs). RORγt is SUMO3-modified by E3 ligase PIAS4 at lysine 31 (K31), and the mutation of K31 to arginine in mice prevents RORγt sumoylation, leading to impaired TH17 differentiation, resistance to TH17-mediated EAE, accumulation of thymic ISPs, and a lack of Peyer's patches. Mechanistically, sumoylation of RORγt-K31 recruits histone acetyltransferase KAT2A, which stabilizes the binding of SRC1 to enhance RORγt transcription factor activity. This study thus demonstrates that sumoylation is a critical mechanism for regulating RORγt function, and reveals new drug targets for preventing TH17-mediated autoimmunity.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Protein Processing, Post-Translational , Th17 Cells/immunology , Thymocytes/microbiology , Thymus Gland/immunology , Ubiquitins/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Hematopoiesis/genetics , Hematopoiesis/immunology , Lymph Nodes/immunology , Lymph Nodes/pathology , Male , Mice , Mice, Transgenic , Nuclear Receptor Coactivator 1/genetics , Nuclear Receptor Coactivator 1/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Peyer's Patches/immunology , Peyer's Patches/pathology , SUMO-1 Protein/deficiency , SUMO-1 Protein/genetics , SUMO-1 Protein/immunology , Sumoylation , Th17 Cells/pathology , Thymocytes/immunology , Thymocytes/pathology , Thymus Gland/pathology , Ubiquitins/deficiency , Ubiquitins/immunology , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/immunology
19.
J Exp Med ; 215(11): 2936-2954, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30355616

ABSTRACT

The foodborne pathogen Listeria monocytogenes (Lm) crosses the intestinal villus epithelium via goblet cells (GCs) upon the interaction of Lm surface protein InlA with its receptor E-cadherin. Here, we show that Lm infection accelerates intestinal villus epithelium renewal while decreasing the number of GCs expressing luminally accessible E-cadherin, thereby locking Lm portal of entry. This novel innate immune response to an enteropathogen is triggered by the infection of Peyer's patch CX3CR1+ cells and the ensuing production of IL-23. It requires STAT3 phosphorylation in epithelial cells in response to IL-22 and IL-11 expressed by lamina propria gp38+ stromal cells. Lm-induced IFN-γ signaling and STAT1 phosphorylation in epithelial cells is also critical for Lm-associated intestinal epithelium response. GC depletion also leads to a decrease in colon mucus barrier thickness, thereby increasing host susceptibility to colitis. This study unveils a novel innate immune response to an enteropathogen, which implicates gp38+ stromal cells and locks intestinal villus invasion, but favors colitis.


Subject(s)
Colitis/immunology , Intestinal Mucosa/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Membrane Glycoproteins/immunology , Myeloid Cells/immunology , Peyer's Patches/immunology , Animals , Colitis/genetics , Colitis/microbiology , Colitis/pathology , Cytokines/genetics , Cytokines/immunology , Immunity, Innate/genetics , Immunity, Mucosal/genetics , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Listeriosis/genetics , Listeriosis/pathology , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Myeloid Cells/microbiology , Myeloid Cells/pathology , Peyer's Patches/microbiology , Peyer's Patches/pathology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , Stromal Cells/immunology , Stromal Cells/microbiology , Stromal Cells/pathology
20.
Immunology ; 155(3): 356-366, 2018 11.
Article in English | MEDLINE | ID: mdl-29969845

ABSTRACT

Regulatory T (Treg) cells play an essential role in the maintenance of intestinal homeostasis. In Peyer's patches (PPs), which comprise the most important IgA induction site in the gut-associated lymphoid tissue, Treg cells promote IgA isotype switching. However, the mechanisms underlying their entry into PPs and isotype switching facilitation in activated B cells remain unknown. This study, based on the dextran sulphate sodium (DSS)-induced colitis model, revealed that Treg cells are significantly increased in PPs, along with CD11b+ B-cell induction. Immunofluorescence staining showed that infiltrated Treg cells were located around CD11b+ B cells and produced transforming growth factor-ß, thereby inducing IgA+ B cells. Furthermore, in vivo and in vitro studies revealed that CD11b+ B cells in PPs had the capacity to recruit Treg cells into PPs rather than promoting their proliferation. Finally, we found that Treg cell recruitment was mediated by the chemokine CXCL9 derived from CD11b+ B cells in PPs. These findings demonstrate that CD11b+ B cells induced in PPs during colitis actively recruit Treg cells to accomplish IgA isotype switch in a CXCL9-dependent manner.


Subject(s)
B-Lymphocytes/immunology , CD11b Antigen/immunology , Chemokine CXCL9/immunology , Colitis/immunology , Peyer's Patches/immunology , T-Lymphocytes, Regulatory/immunology , Animals , B-Lymphocytes/pathology , CD11b Antigen/genetics , Chemokine CXCL9/genetics , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Mice , Mice, Knockout , Peyer's Patches/pathology , T-Lymphocytes, Regulatory/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...