Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Poult Sci ; 100(2): 615-622, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33518114

ABSTRACT

The rapid renewal and repair of the intestinal mucosa are based on intestinal stem cells (ISC), which are located at the crypt bottom. Paneth cells are an essential component in the crypt, which served as the niche for ISC development. However, in the chicken, how the function of Paneth cells changes during intestinal inflammation is unclear and is the key to understand the mechanism of mucosal repair. In the present study, 36 HyLine White chickens (7 d of age, n = 6) were randomly divided into 1 control and 5 lipopolysaccharide (LPS) injection groups. The chickens were injected (i.p.) with PBS in the control group, however, were injected (i.p.) with LPS (10 mg/kg BW) in the LPS injection groups, which would be sampled at 5 time points (1 h postinjection [hpi], 2 hpi, 4 hpi, 6 hpi, and 8 hpi). Results showed that tumor necrosis factor-α mRNA transcription in duodenal tissue increased gradually since 1 hpi, peaked at 4 hpi, and then reduced remarkably, indicating that 4 hpi of LPS was the early stage of intestinal inflammation. Meanwhile, the MUC2 expression in duodenal tissue was dramatically reduced since 1 hpi of LPS. The ISC marker, Lgr5 and Bmi1, in the duodenal crypt were reduced from 1 hpi to 4 hpi and elevated later. Accordingly, the hydroethidine staining showed that the reactive oxygen species level, which drives the differentiation of ISC, in the duodenal crypt reduced obviously at 1 hpi and recovered gradually since 4 hpi. The analysis of Paneth cells showed that many swollen mitochondria appeared in Paneth cells at 4 hpi of LPS. Meanwhile, the Lysozyme transcription in the duodenal crypt was substantially decreased since 1 hpi of LPS. However, the Wnt3a and Dll1 in duodenal crypt decreased at 1 hpi of LPS, then increased gradually. In conclusion, Paneth cells were impaired at the early stage of intestinal inflammation, then recovered rapidly. Thus, the ISC activity was reduced at first and recovery soon.


Subject(s)
Chickens , Gastroenteritis/veterinary , Paneth Cells/pathology , Poultry Diseases/pathology , Animals , Duodenum/cytology , Duodenum/pathology , Duodenum/ultrastructure , Gastroenteritis/pathology , Intestinal Mucosa/pathology , Microscopy, Electron, Transmission/veterinary , Paneth Cells/ultrastructure , Random Allocation , Stem Cells/pathology
2.
Front Immunol ; 11: 560110, 2020.
Article in English | MEDLINE | ID: mdl-33224134

ABSTRACT

HPS1, a BLOC-3 subunit that acts as a guanine nucleotide exchange factor of Rab32/38, may play a role in the removal of VAMP7 during the maturation of large dense core vesicles of Paneth cells. Loss of HPS1 impairs lysozyme secretion and alters the composition of intestinal microbiota, which may explain the susceptibility of HPS-associated inflammatory bowel disease. Hermansky-Pudlak syndrome (HPS) is characterized by oculocutaneous albinism, bleeding tendency, and other chronic organ lesions due to defects in tissue-specific lysosome-related organelles (LROs). For some HPS subtypes, such as HPS-1, it is common to have symptoms of HPS-associated inflammatory bowel disease (IBD). However, its underlying mechanism is largely unknown. HPS1 is a subunit of the BLOC-3 complex which functions in the biogenesis of LROs. Large dense core vesicles (LDCVs) in Paneth cells of the intestine are a type of LROs. We here first report the abnormal LDCV morphology (increased number and enlarged size) in HPS1-deficient pale ear (ep) mice. Similar to its role in melanosome maturation, HPS1 plays an important function in the removal of VAMP7 from LDCVs to promote the maturation of LDCVs. The immature LDCVs in ep mice are defective in regulated secretion of lysozyme, a key anti-microbial peptide in the intestine. We observed changes in the composition of intestinal microbiota in both HPS-1 patients and ep mice. These findings provide insights into the underlying mechanism of HPS-associated IBD development, which may be implicated in possible therapeutic intervention of this devastating condition.


Subject(s)
Lysosomes/metabolism , Membrane Proteins/metabolism , Paneth Cells/metabolism , Secretory Vesicles/metabolism , Animals , Child , Disease Models, Animal , Feces/microbiology , Female , Fluorescent Antibody Technique , Gastrointestinal Microbiome , Hermanski-Pudlak Syndrome/etiology , Hermanski-Pudlak Syndrome/metabolism , Humans , Immunohistochemistry , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Membrane Proteins/genetics , Metagenomics/methods , Mice , Mice, Knockout , Paneth Cells/ultrastructure , Protein Transport , R-SNARE Proteins/genetics , R-SNARE Proteins/metabolism
3.
Anat Rec (Hoboken) ; 301(6): 1074-1085, 2018 06.
Article in English | MEDLINE | ID: mdl-29316348

ABSTRACT

Paneth cells secrete bactericidal substances in response to bacterial proliferation on the mucosal surface without directly contacting bacteria. However, the induction mechanism of this transient secretion has not been clarified, although nervous system and/or immunocompetent cells in the lamina propria (LP) might be involved. In this study, we ultrastructurally and immunohistochemically investigated which LP cells are localized beneath Paneth cells and examined the relationship between the Paneth cell-derived cellular processes which extended into the LP and the LP cells. The results showed that various cells-including blood capillary, subepithelial stromal cell, and nerve fiber-were present in the LP beneath Paneth cells. Endothelial cells of blood capillary were the cells most frequently found in this location; they were situated within 1 µm of the Paneth cells and possessed fenestration on the surfaces adjacent to Paneth cells. The Paneth cells rarely extended the cellular processes toward the LP across the basal lamina. Most of the cellular processes of Paneth cells contacted the subepithelial stromal cells. Immunohistochemistry revealed that the CD34+ CD31- αSMA- stromal cells preferentially localized in the LP beneath the intestinal crypt base, while PDGFRαhi αSMA+ stromal cells mainly localized around the lateral portions of the intestinal crypt and PDGFRαhi αSMA- stromal cells localized in the intestinal villus. From these findings, the existence of blood capillaries beneath Paneth cells might reflect the active exocrine function of Paneth cells. Furthermore, subepithelial stromal cells, probably with a CD34+ CD31- αSMA- PDGFRα-/lo phenotype, beneath the crypt base might affect Paneth cell activity by interacting with their cellular processes. Anat Rec, 301:1074-1085, 2018. © 2018 Wiley Periodicals, Inc.


Subject(s)
Ileum/ultrastructure , Mucous Membrane/ultrastructure , Paneth Cells/ultrastructure , Animals , Ileum/metabolism , Immunohistochemistry , Male , Microscopy, Electron , Mucous Membrane/metabolism , Paneth Cells/metabolism , Rats , Rats, Wistar
4.
Environ Sci Pollut Res Int ; 24(7): 6657-6666, 2017 03.
Article in English | MEDLINE | ID: mdl-28083743

ABSTRACT

Environmental and occupational exposure to aluminum along with ionizing radiation results in serious health problems. This study was planned to investigate the impact of oxidative stress provoked by exposure to ionizing radiation with aluminum administration upon cellular ultra structure and apoptotic changes in Paneth cells of rat small intestine . Animals received daily aluminum chloride by gastric gavage at a dose 0.5 mg/Kg BW for 4 weeks. Whole body gamma irradiation was applied at a dose 2 Gy/week up to 8 Gy. Ileum malondialdehyde, advanced oxidative protein products, protein carbonyl and tumor necrosis factor-alpha were assessed as biomarkers of lipid peroxidation, protein oxidation and inflammation respectively along with superoxide dismutase, catalase, and glutathione peroxidase activities as enzymatic antioxidants. Moreover, analyses of cell cycle division and apoptotic changes were evaluated by flow cytometry. Intestinal cellular ultra structure was investigated using transmission electron microscope.Oxidative and inflammatory stresses assessment in the ileum of rats revealed that aluminum and ionizing radiation exposures exhibited a significant effect upon the increase in oxidative stress biomarkers along with the inflammatory marker tumor necrosis factor-α accompanied by a significant decreases in the antioxidant enzyme activities. Flow cytometric analyses showed significant alterations in the percentage of cells during cell cycle division phases along with significant increase in apoptotic cells. Ultra structurally, intestinal cellular alterations with marked injury in Paneth cells at the sites of bacterial translocation in the crypt of lumens were recorded. The results of this study have clearly showed that aluminum and ionizing radiation exposures induced apoptosis with oxidative and inflammatory disturbance in the Paneth cells of rat intestine, which appeared to play a major role in the pathogenesis of cellular damage. Furthermore, the interaction of these two intestinal toxic routes was found to be synergistic.


Subject(s)
Aluminum/toxicity , Apoptosis , Gamma Rays/adverse effects , Oxidative Stress , Paneth Cells/drug effects , Paneth Cells/radiation effects , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Apoptosis/radiation effects , Catalase/metabolism , Ileum/drug effects , Ileum/metabolism , Ileum/radiation effects , Ileum/ultrastructure , Lipid Peroxidation/drug effects , Male , Malondialdehyde/metabolism , Oxidation-Reduction , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Paneth Cells/metabolism , Paneth Cells/ultrastructure , Rats , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/metabolism , Whole-Body Irradiation
5.
Tissue Cell ; 48(4): 383-8, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27233914

ABSTRACT

Paneth cells are secretory epithelial cells of the innate immune system of the intestine of several mammals, including alpacas. Little is known about the latter; thus, in the present study we described the morphology and histochemical characteristics of Paneth cells in healthy fetuses, and young and adult alpacas. For this purpose, samples of duodenum, jejunum and ileum were taken from 6 fetuses at different days of pregnancy (between days 221-330), 66 offsprings (between 0 and 45-days-old) and 5 adult alpacas (>2-years-old). Samples were fixed in 10% buffered formalin and processed for histological and morphometrical analysis using HE and Masson Trichomics technique. Immunohistochemistry was used to identify Paneth cells using anti-lysozyme antibody. In addition, the lectinhistochemichal binding-pattern of Paneth cells granules was evaluated. Lyzozyme was immunohistochemically detected in the granules of Paneth cells from day 283 of pregnancy in all the small intestinal sections of the studied fetuses. In newborn alpacas Paneth cells were initially found in the duodenum, but the following days (days 18-21 after birth) they were also found in the ileum. Their size gradually increased after birth, but then no significant differences were found. In adult alpacas the number was lower than offsprings. We suggest that Paneth cells early differentiate in the small intestine of alpacas, and the increase in their number during the first two weeks of life strongly support their possible involvement in the intestinal defensive functions against the enteric diseases that occur during the lactancy stage.


Subject(s)
Camelids, New World , Fetus/ultrastructure , Paneth Cells/ultrastructure , Animals , Cytoplasmic Granules/ultrastructure , Duodenum/ultrastructure , Female , Ileum/ultrastructure , Jejunum/ultrastructure , Paneth Cells/metabolism , Pregnancy
6.
Poult Sci ; 95(7): 1631-1635, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27030696

ABSTRACT

The Paneth cells are highly specialized cells in the epithelium of the small intestine of many vertebrate species. These cells reside at the base of crypts of the Lieberkühn and contain abundant secretory granules. Previous studies suggesting the existence of Paneth cells in the chicken (Gallus gallus) remained controversial. Here we seek to identify the Paneth cells in the chicken small intestine through morphological examination and specific gene expression. Histological staining and transmission electron microscope confirmed the presence of granulated secretory cells at the base of the crypts in the chicken small intestine. Western blotting experiment also manifested the expression of lysozyme protein, which is specifically secreted by the Paneth cells in the small intestine. Moreover, lysozyme c and lysozyme g mRNAs were expressed in the small intestine of chickens at different ages. Lysozyme c mRNA, in particular, was located at the base of the small intestinal crypts as displayed by in situ hybridization. Collectively, we provide evidences that the Paneth cells indeed exist in the small intestine of the chicken.


Subject(s)
Chickens/anatomy & histology , Chickens/genetics , Paneth Cells/ultrastructure , Animals , Avian Proteins/genetics , Avian Proteins/metabolism , Chickens/metabolism , Intestine, Small/enzymology , Intestine, Small/ultrastructure , Microscopy, Electron, Transmission/veterinary , Muramidase/genetics , Muramidase/metabolism , Paneth Cells/enzymology , RNA, Messenger/metabolism
7.
Mucosal Immunol ; 8(4): 828-40, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25425265

ABSTRACT

Tumor necrosis factor (TNF) is a powerful activator of the immune system and a well-validated target for treatment of autoimmune diseases. Injection of TNF induces systemic lethal inflammation characterized by hypothermia, induction of multiple cytokines, and extensive damage to multiple organs. Previously, we reported that TNF-induced lethal inflammation is strictly TNFR1(P55)-dependent. We also uncovered a crucial role for P55 expression levels in intestinal epithelial cells (IECs), in which P55+/+ expression is sufficient to sensitize to TNF lethality in an otherwise fully protected P55+/- background. Here, we investigated the molecular mechanism that drives TNF toxicity in IECs. Unexpectedly, we found that the degree of TNF-induced enterocyte damage and apoptosis in IECs is equally strong in TNF-sensitive P55+/+ mice and TNF-resistant P55+/- mice. Our results suggest that P55+/+-induced signaling causes goblet and Paneth cell dysfunction, leading to severe epithelial barrier dysfunction. As a result, intestinal permeability and systemic bacterial spread are induced, causing lethal systemic inflammation. In conclusion, we identified P55-induced goblet and Paneth cell dysfunction as a crucial mechanism for TNF-induced systemic and lethal inflammation.


Subject(s)
Goblet Cells/metabolism , Inflammation/metabolism , Paneth Cells/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Animals , Apoptosis/drug effects , Dexamethasone/pharmacology , Goblet Cells/drug effects , Goblet Cells/ultrastructure , Inflammation/mortality , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/ultrastructure , Mice , Mice, Knockout , Models, Biological , Paneth Cells/drug effects , Paneth Cells/ultrastructure , Permeability/drug effects , Receptors, Tumor Necrosis Factor, Type I/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/toxicity
8.
Anat Rec (Hoboken) ; 297(8): 1462-71, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24788798

ABSTRACT

Paneth cells (PCs) contribute to the host defense against indigenous bacteria in the small intestine. We found Paneth cell-like cells (PLCs) in the rat ascending colon, but the nature of PLCs is never clarified. Therefore, the present study aimed to clarify the cytological characteristics of PLCs and discuss their cellular differentiation. PLCs were localized in the bases of intestinal crypts, especially follicle-associated intestinal crypts in proximal colonic lymphoid tissue, but were very seldom found in the ordinary intestinal crypts of the ascending colon. PLCs possessed specific granules with highly electron-dense cores and haloes, as well as PCs in the small intestine. The secretory granules of PLCs were positive for PAS reaction, lysozyme and soluble phospholipase A2, but negative for Alcian blue staining, ß-defensin-1 and -2, as well as the ones of PCs. Furthermore, intermediate cells possessing both the PLC-specific granules and the mucus granules similar to those of goblet cells (GCs) were occasionally found in the vicinity of PLCs. Intermediate cells ranged from goblet cell-like cells rich in mucus granules to PLC-like cells with few mucus granules. The cellular condensation and fragmentation were exclusively found in PLCs but never seen in intermediate cells or GCs. The PLCs, which were identified as PC, were suggested to be transformed from GCs through intermediate cells and finally to die by apoptosis in intestinal crypts of proximal colonic lymphoid tissue in the rat ascending colon.


Subject(s)
Colon, Ascending/ultrastructure , Goblet Cells/ultrastructure , Intestine, Small/ultrastructure , Lymphoid Tissue/ultrastructure , Paneth Cells/ultrastructure , Secretory Vesicles/ultrastructure , Animals , Biomarkers/metabolism , Cells, Cultured , Colon, Ascending/cytology , Colon, Ascending/metabolism , Goblet Cells/cytology , Goblet Cells/metabolism , Immunoenzyme Techniques , Intestine, Small/cytology , Intestine, Small/metabolism , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Male , Microscopy, Electron, Transmission , Paneth Cells/cytology , Paneth Cells/metabolism , Rats , Rats, Wistar , Secretory Vesicles/metabolism
9.
Inflamm Bowel Dis ; 20(4): 712-22, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24577114

ABSTRACT

BACKGROUND: Recent studies link endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) to inflammatory bowel disease. Altered eIF2α phosphorylation (eIF2α-P), a regulatory hub of the UPR, was observed in mucosal tissue of patients with inflammatory bowel disease. In this study, we examined the mechanistic role of eIF2α-P in intestinal epithelial cell (IEC) function and intestinal homeostasis in mice. METHODS: We generated mice with villin-Cre-mediated conditional expression of nonphosphorylatable Ser51Ala mutant eIF2α in IECs (AA mice). We analyzed AA mice under normal conditions and on challenge with oral infection of Salmonella Typhimurium or dextran sulfate sodium-induced colitis. RESULTS: Loss of eIF2α-P did not affect the normal proliferation or differentiation of IECs. However, AA mice expressed decreased secretory proteins including lysozyme, suggesting eIF2α-P is required for Paneth cell function. The ultrastructure of AA Paneth cells exhibited a reduced number of secretory granules, a fragmented ER, and distended mitochondria under normal conditions. UPR gene expression was defective in AA IECs. Translation of Paneth cell specific messenger RNAs encoding lysozyme and cryptidins was significantly defective leading to the observed granule-deficient phenotype, which was associated with reduced ribosomal recruitment of these messenger RNAs to the ER membrane. Consequently, AA mice were more susceptible to oral Salmonella infection and dextran sulfate sodium-induced colitis. CONCLUSIONS: We conclude eIF2α phosphorylation is required for the normal function of intestinal Paneth cells and mucosal homeostasis by activating UPR signaling and promoting messenger RNA recruitment to the ER membrane for translation.


Subject(s)
Endoplasmic Reticulum/metabolism , Eukaryotic Initiation Factor-2/metabolism , Homeostasis , Muramidase/biosynthesis , Paneth Cells/metabolism , Paneth Cells/ultrastructure , Animals , Apoptosis , Cell Differentiation , Cell Proliferation , Colitis/chemically induced , Colitis/metabolism , Dextran Sulfate , Disease Susceptibility , Endoplasmic Reticulum/ultrastructure , Eukaryotic Initiation Factor-2/genetics , Interferon-gamma/metabolism , Mice , Mice, Inbred C3H , Mice, Transgenic , Molecular Chaperones , Muramidase/genetics , Paneth Cells/immunology , Phosphorylation , Protein Biosynthesis , RNA, Messenger/metabolism , Ribosomes/physiology , Salmonella Infections, Animal/immunology , Secretory Vesicles/ultrastructure , Signal Transduction , Stress, Physiological , Unfolded Protein Response/genetics
10.
Autophagy ; 9(8): 1188-200, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23782979

ABSTRACT

The identification of inflammatory bowel disease (IBD) susceptibility genes by genome-wide association has linked this pathology to autophagy, a lysosomal degradation pathway that is crucial for cell and tissue homeostasis. Here, we describe autophagy-related 4B, cysteine peptidase/autophagin-1 (ATG4B) as an essential protein in the control of inflammatory response during experimental colitis. In this pathological condition, ATG4B protein levels increase in parallel with the induction of autophagy. Moreover, ATG4B expression is significantly reduced in affected areas of the colon from IBD patients. Consistently, atg4b (-/-) mice present Paneth cell abnormalities, as well as an increased susceptibility to DSS-induced colitis. atg4b-deficient mice exhibit significant alterations in proinflammatory cytokines and mediators of the immune response to bacterial infections, which are reminiscent of those found in patients with Crohn disease or ulcerative colitis. Additionally, antibiotic treatments and bone marrow transplantation from wild-type mice reduced colitis in atg4b (-/-) mice. Taken together, these results provided additional evidence for the importance of autophagy in intestinal pathologies and describe ATG4B as a novel protective protein in inflammatory colitis. Finally, we propose that atg4b-null mice are a suitable model for in vivo studies aimed at testing new therapeutic strategies for intestinal diseases associated with autophagy deficiency.


Subject(s)
Autophagy , Colitis/pathology , Colitis/prevention & control , Cysteine Endopeptidases/metabolism , Homeostasis , Intestinal Mucosa/metabolism , Intestines/pathology , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Autophagy/drug effects , Autophagy-Related Proteins , Colitis/drug therapy , Cysteine Endopeptidases/deficiency , Cytokines/metabolism , Dextran Sulfate , Disease Susceptibility , Hematopoiesis/drug effects , Homeostasis/drug effects , Ileum/drug effects , Ileum/pathology , Inflammation Mediators/metabolism , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/pathology , Intestines/drug effects , Mice , Mice, Inbred C57BL , Paneth Cells/drug effects , Paneth Cells/pathology , Paneth Cells/ultrastructure
11.
Proc Natl Acad Sci U S A ; 109(31): 12650-5, 2012 Jul 31.
Article in English | MEDLINE | ID: mdl-22802641

ABSTRACT

Using an environmentally sensitized genetic screen we identified mutations that cause inflammatory colitis in mice. The X-linked Klein-Zschocher (KLZ) mutation created a null allele of Yipf6, a member of a gene family believed to regulate vesicular transport in yeast, but without known functions in mammals. Yipf6 is a five transmembrane-spanning protein associated with Golgi compartments. Klein-Zschocher mutants were extremely sensitive to colitis induced by dextran sodium sulfate (DSS) and developed spontaneous ileitis and colitis after 16 mo of age in specific pathogen-free housing conditions. Electron microscopy, gene expression, and immunocytochemistry analyses provided evidence that impaired intestinal homeostasis stemmed from defective formation and secretion of large secretory granules from Paneth and goblet cells. These studies support a tissue- and organ-specific function for Yipf6 in the maintenance of intestinal homeostasis and implicate the orthologous human gene as a disease susceptibility locus.


Subject(s)
Colitis/metabolism , Goblet Cells/metabolism , Membrane Proteins/metabolism , Mutation , Paneth Cells/metabolism , Animals , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Dextran Sulfate/toxicity , Female , Gene Expression Regulation , Genetic Loci , Genetic Predisposition to Disease , Goblet Cells/ultrastructure , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Humans , Ileitis/chemically induced , Ileitis/genetics , Ileitis/metabolism , Ileitis/pathology , Male , Membrane Proteins/genetics , Mice , Mice, Transgenic , Paneth Cells/ultrastructure
12.
Gastroenterology ; 142(5): 1097-1099.e4, 2012 May.
Article in English | MEDLINE | ID: mdl-22285936

ABSTRACT

Autophagy-related 16 like-1 (ATG16L-1), immunity-related GTPase-M (IRGM), and nucleotide-binding oligomerization domain-containing 2 (NOD2) regulate autophagy, and variants in these genes have been associated with predisposition to Crohn's disease (CD). However, little is known about the role of autophagy in CD. Intestinal biopsies from untreated pediatric patients with CD, celiac disease, or ulcerative colitis were analyzed by immunohistochemistry and electron microscopy. We observed that autophagy was specifically activated in Paneth cells from patients with CD, independently of mucosal inflammation or disease-associated variants of ATG16L1 or IRGM. In these cells, activation of autophagy was associated with a significant decrease in number of secretory granules and features of crinophagy. These observations might account for the disorganization of secretory granules previously reported in Paneth cells from patients with CD.


Subject(s)
Autophagy/physiology , Crohn Disease/pathology , Paneth Cells/ultrastructure , Secretory Vesicles/physiology , Autophagy-Related Proteins , Carrier Proteins/genetics , Child , Female , GTP-Binding Proteins/genetics , Humans , Infant , Male , Microtubule-Associated Proteins/analysis , Nod2 Signaling Adaptor Protein/genetics , Polymorphism, Single Nucleotide
13.
Am J Pathol ; 179(6): 2885-93, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21986443

ABSTRACT

Lack of enteral feeding, with or without parenteral nutritional support, is associated with increased intestinal permeability and translocation of bacteria. Such translocation is thought to be important in the high morbidity and mortality rates of patients who receive nothing by mouth. Recently, Paneth cells, important constituents of innate intestinal immunity, were found to be crucial in host protection against invasion of both commensal and pathogenic bacteria. This study investigates the influence of food deprivation on Paneth cell function in a mouse starvation model. Quantitative PCR showed significant decreases in mRNA expression of typical Paneth cell antimicrobials, lysozyme, cryptdin, and RegIIIγ, in ileal tissue after 48 hours of food deprivation. Protein expression levels of lysozyme and RegIIIγ precursor were also significantly diminished, as shown by Western blot analysis and IHC. Late degenerative autophagolysosomes and aberrant Paneth cell granules in starved mice were evident by electron microscopy, Western blot analysis, and quantitative PCR. Furthermore, increased bacterial translocation to mesenteric lymph nodes coincided with Paneth cell abnormalities. The current study demonstrates the occurrence of Paneth cell abnormalities during enteral starvation. Such changes may contribute to loss of epithelial barrier function, causing the apparent bacterial translocation in enteral starvation.


Subject(s)
Bacterial Translocation/immunology , Paneth Cells/physiology , Starvation/physiopathology , Animals , Autophagy/immunology , Ileum/immunology , Ileum/metabolism , Immunity, Innate , Immunologic Techniques , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron , Muramidase/metabolism , Pancreatitis-Associated Proteins , Paneth Cells/immunology , Paneth Cells/ultrastructure , Permeability , Protein Precursors/metabolism , Proteins/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Starvation/immunology , Starvation/pathology
14.
Cell Mol Immunol ; 8(4): 325-32, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21499290

ABSTRACT

Intestinal stem cells may have important roles in the maintenance of epithelial integrity during tissue repair. Alemtuzumab is a humanized anti-CD52 lymphocytic antibody that is increasingly being used to induce immunosuppression; intestinal barrier function is impaired during treatment with alemtuzumab. We investigated the response of intestinal stem cells to epithelial damage resulting from alemtuzumab treatment. Intestinal epithelial cell loss and abnormal Paneth cell morphology were found following a single dose of alemtuzumab. The animals receiving alemtuzumab exhibited increased apoptosis in the villi 3 days after alemtuzumab treatment and in the crypt on day 9, but apoptosis was scarce on day 35. We assessed expression of Musashi-1- and Lgr5-positive stem cells following alemtuzumab treatment. Increased numbers of cells staining positive for both Musashi-1 and Lgr5 were found in the stem cell zone after alemtuzumab treatment for 3 and 9 days. These data indicated that the epithelial cells were injured following alemtuzumab treatment, with the associated expansion of intestinal stem cells. After alemtuzumab treatment for 35 days, the numbers of intestinal epithelial cells and intestinal stem cells returned to normal. This study suggests that alemtuzumab treatment induced the increase in stem cells, resulting in the availability of more enterocytes for repair.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Neoplasm/pharmacology , Epithelium/drug effects , Paneth Cells/drug effects , Stem Cells/drug effects , Alemtuzumab , Animals , Apoptosis/drug effects , Cell Count , Epithelium/metabolism , Epithelium/ultrastructure , Macaca , Male , Paneth Cells/cytology , Paneth Cells/metabolism , Paneth Cells/ultrastructure , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Stem Cells/cytology , Stem Cells/metabolism
15.
Dev Biol ; 345(1): 49-63, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20599897

ABSTRACT

Elucidating the mechanisms determining multipotent progenitor cell fate remains a fundamental project of contemporary biology. Various tissues of mice and men with defects in the zinc-finger transcriptional repressor Gfi1 have dramatic perturbations in the proportions of their differentiated cell types. In Gfi1-deficient intestinal epithelium there is a shift from mucous and Paneth towards enteroendocrine cells, leading to the proposal that Gfi1 functions in the allocation of the progeny derived from a hypothetical common granulocytic progenitor. However, studies of clones have yielded no evidence of such a common progenitor prompting us to investigate alternate mechanisms explaining the Gfi1-deficient phenotype. We report that mucous and Paneth but not enteroendocrine lineage cells normally express Gfi1. Sporadic mucous and Paneth lineage cells in the crypts of Gfi1-deficient mice aberrantly express the pro-enteroendocrine transcription factor Neurog3, indicating that stable repression of Neurog3 in these lineages requires Gfi1. Importantly, we also find mucous and Paneth lineage cells in various stages of cellular reprogramming into the enteroendocrine lineage in Gfi1-deficient mice. We propose that mucous and Paneth cell lineage metastability, rather than reallocation at the level of a hypothetical common granulocytic progenitor, is responsible for the shifts in cell type proportions observed in Gfi1-deficient intestinal epithelium.


Subject(s)
Cell Lineage , DNA-Binding Proteins/metabolism , Intestinal Mucosa/metabolism , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , DNA-Binding Proteins/genetics , Enteroendocrine Cells/cytology , Enteroendocrine Cells/metabolism , Enteroendocrine Cells/ultrastructure , Flow Cytometry , Gene Expression , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/ultrastructure , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Paneth Cells/cytology , Paneth Cells/metabolism , Paneth Cells/ultrastructure , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Zinc Fingers
16.
Clin Immunol ; 136(3): 375-86, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20605528

ABSTRACT

Alemtuzumab has been recently introduced for induction therapy in organ transplantation. However, the pathogenesis and molecular mechanism of the impact of such induction therapy on bacterial infections remain to be clarified. We found the alterations of Paneth cells including abnormal Paneth cell granules and expression of lysozyme and defensin 5 in response to lymphocyte depletion by alemtuzumab. Lymphocyte depletion resulted in decreased expression of TNF-alpha, IFN-gamma, IL-10 and TGF-beta in the intestine. The diversity of gut bacteria varied significantly between different times of alemtuzumab treatment. Abnormal expression of granule peptides might result in impairment of host gut microflora. The alterations in bacterial microflora had almost reversed 56days after alemtuzumab treatment, which was consistent with our results that Paneth cells were recovered to secrete antimicrobial peptides to govern gut microflora. These findings indicated the associations between changes of Paneth cell function and gut microflora and supported the important role of Paneth cells to barrier impairment with the use of alemtuzumab in organ transplantation.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/pharmacology , Intestines/immunology , Intestines/microbiology , Paneth Cells/immunology , Paneth Cells/physiology , Alemtuzumab , Animals , Antibodies, Monoclonal, Humanized , Bacteria, Aerobic/genetics , Bacteria, Aerobic/isolation & purification , Bacteria, Anaerobic/genetics , Bacteria, Anaerobic/isolation & purification , Base Sequence , Cytokines/metabolism , Cytoplasmic Granules/physiology , Cytoplasmic Granules/ultrastructure , DNA, Bacterial/genetics , DNA, Bacterial/isolation & purification , Defensins/metabolism , Lymphocyte Depletion , Macaca , Mice , Microscopy, Electron, Transmission , Models, Animal , Muramidase/metabolism , Paneth Cells/ultrastructure
17.
J Pathol ; 219(2): 242-52, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19681094

ABSTRACT

Macroautophagy (mAG) is a lysosomal mechanism of degradation of cell self-constituents damaged due to variety of stress factors, including ionizing irradiation. Activation of mAG requires expression of mAG protein Atg8 (LC3) and conversion of its form I (LC3-I) to form II (LC3-II), mediated by redox-sensitive Atg4 protease. We have demonstrated upregulation of this pathway in the innate host defense Paneth cells of the small intestine (SI) due to ionizing irradiation and correlation of this effect with induction of pro-oxidant inducible nitric oxide synthase (iNOS). CD2F1 mice were exposed to 9.25 Gy gamma-ionizing irradiation. Small intestinal specimens were collected during 7 days after ionizing irradiation. Assessment of ionizing irradiation-associated alterations in small intestinal crypt and villus cells and activation of the mAG pathway was conducted using microscopical and biochemical techniques. Analysis of iNOS protein and the associated formation of nitrites and lipid peroxidation products was performed using immunoblotting and biochemical analysis, and revealed increases in iNOS protein, nitrate levels and oxidative stress at day 1 following ionizing irradiation. Increase in immunoreactivity of LC3 protein in the crypt cells was observed at day 7 following ionizing irradiation. This effect predominantly occurred in the CD15-positive Paneth cells and was associated with accumulation of LC3-II isoform. The formation of autophagosomes in Paneth cells was confirmed by transmission electron microscopy (TEM). Up-regulation of LC3 pathway in the irradiated SI was accompanied by a decreased protein-protein interaction between LC3 and chaperone heat shock protein 70. A high-level of LC3-immunoreactivity in vacuole-shaped structures was spatially co-localized with immunoreactivity of 3-nitro-tyrosine. The observed effects were diminished in iNOS knockout B6.129P2-NOS2(tm1Lau)/J mice subjected to the same treatments. We postulate that the observed up-regulation of mAG in the irradiated small intestine is at least in part mediated by the iNOS signalling mechanism.


Subject(s)
Autophagy/radiation effects , Gamma Rays , Intestine, Small/radiation effects , Paneth Cells/radiation effects , Up-Regulation/radiation effects , Animals , In Situ Nick-End Labeling , Intestine, Small/ultrastructure , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron, Scanning , Nitric Oxide Synthase Type II/physiology , Oxidative Stress/radiation effects , Paneth Cells/ultrastructure , Whole-Body Irradiation
18.
Gastroenterology ; 137(4): 1333-45.e1-3, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19549527

ABSTRACT

BACKGROUND & AIMS: Stem cells within the intestinal epithelium generate daughter cells that undergo lineage commitment and maturation through the combined action of the Wnt and Notch signaling cascades. Both pathways, in turn, regulate transcription factor networks that further define differentiation toward either enterocytes or 1 of 3 secretory cell lineages (Paneth, goblet, or enteroendocrine cells). In this study, we investigated the role of the Wnt-responsive, Ets-domain transcription factor Spdef in the differentiation of goblet and Paneth cells. METHODS: The in vivo function of Spdef was examined by disrupting the Spdef gene in mice (Spdef(-/-) mice) and analyzing the intestinal phenotype using a range of histologic techniques and DNA microarray profiling. RESULTS: In accordance with expression data, we found that loss of Spdef severely impaired the maturation of goblet and Paneth cells and, conversely, led to an accumulation of immature secretory progenitors. Spdef appears to positively and negatively regulate a specific subset of goblet and Paneth cell genes, including Cryptdins, Mmp7, Ang4, Kallikreins, and Muc2. CONCLUSIONS: Spdef acts downstream of Math1 to promote terminal differentiation of a secretory progenitor pool into Paneth and goblet cells.


Subject(s)
Cell Differentiation , Colon/metabolism , Goblet Cells/metabolism , Intestine, Small/metabolism , Paneth Cells/metabolism , Proto-Oncogene Proteins c-ets/metabolism , Stem Cells/metabolism , Animals , Biomarkers/metabolism , Cell Differentiation/genetics , Cell Lineage , Colon/ultrastructure , Gene Expression Profiling/methods , Gene Expression Regulation , Genotype , Goblet Cells/ultrastructure , Intestine, Small/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Paneth Cells/ultrastructure , Phenotype , Proto-Oncogene Proteins c-ets/deficiency , Proto-Oncogene Proteins c-ets/genetics , Stem Cells/ultrastructure , Transcription, Genetic
19.
J Clin Invest ; 119(5): 1241-50, 2009 May.
Article in English | MEDLINE | ID: mdl-19349688

ABSTRACT

The accumulation of certain species of bacteria in the intestine is involved in both tissue homeostasis and immune-mediated pathologies. The host mechanisms involved in controlling intestinal colonization with commensal bacteria are poorly understood. We observed that under specific pathogen-free or germ-free conditions, intragastric administration of Pseudomonas aeruginosa, E. coli, Staphylococcus aureus, or Lactobacillus gasseri resulted in increased colonization of the small intestine and bacterial translocation in mice lacking Cd1d, an MHC class I-like molecule, compared with WT mice. In contrast, activation of Cd1d-restricted T cells (NKT cells) with alpha-galactosylceramide caused diminished intestinal colonization with the same bacterial strains. We also found prominent differences in the composition of intestinal microbiota, including increased adherent bacteria, in Cd1d-/- mice in comparison to WT mice under specific pathogen-free conditions. Germ-free Cd1d-/- mice exhibited a defect in Paneth cell granule ultrastructure and ability to degranulate after bacterial colonization. In vitro, NKT cells were shown to induce the release of lysozyme from intestinal crypts. Together, these data support a role for Cd1d in regulating intestinal colonization through mechanisms that include the control of Paneth cell function.


Subject(s)
Antigens, CD1d/physiology , Bacteria/immunology , Intestines/immunology , Intestines/microbiology , Animals , Cell Degranulation/physiology , Escherichia coli/immunology , Feces/microbiology , Galactosylceramides/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestine, Small/immunology , Intestine, Small/microbiology , Lactobacillus/immunology , Lymph Nodes/microbiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/immunology , Paneth Cells/physiology , Paneth Cells/ultrastructure , Pseudomonas aeruginosa/immunology , RNA, Ribosomal, 16S/analysis , Secretory Vesicles/chemistry , Secretory Vesicles/ultrastructure , Specific Pathogen-Free Organisms/immunology , Staphylococcus aureus/immunology
20.
Semin Immunol ; 21(3): 156-63, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19237300

ABSTRACT

The unfolded protein response as a consequence of endoplasmic reticulum (ER) stress has recently been implicated as a novel mechanism that may lead to inflammatory bowel disease (IBD). Impairment of proper ER stress resolution in highly secretory Paneth and, to a lesser extent, goblet cells within the epithelium can primarily lead to intestinal inflammation. An inability to manage ER stress may not only be a primary originator of intestinal inflammation as exemplified by genetic polymorphisms in XBP1 that are associated with IBD but also a perpetuator of inflammation when ER stress is induced secondarily to inflammatory mediators or microbial factors. Furthermore, ER stress pathways may interact with other processes that lead to IBD, notably autophagy.


Subject(s)
DNA-Binding Proteins/metabolism , Endoplasmic Reticulum/immunology , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology , Transcription Factors/metabolism , Animals , Autophagy/immunology , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Endoplasmic Reticulum/ultrastructure , Gene Expression Regulation/immunology , Genetic Predisposition to Disease , Goblet Cells/immunology , Goblet Cells/ultrastructure , Humans , Inflammation , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/pathology , Paneth Cells/immunology , Paneth Cells/ultrastructure , Polymorphism, Genetic , Protein Folding , Regulatory Factor X Transcription Factors , Stress, Physiological/immunology , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/immunology , X-Box Binding Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...