Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Host Microbe ; 24(6): 833-846.e6, 2018 12 12.
Article in English | MEDLINE | ID: mdl-30543778

ABSTRACT

Symbionts play an indispensable role in gut homeostasis, but underlying mechanisms remain elusive. To clarify the role of lactic-acid-producing bacteria (LAB) on intestinal stem-cell (ISC)-mediated epithelial development, we fed mice with LAB-type symbionts such as Bifidobacterium and Lactobacillus spp. Here we show that administration of LAB-type symbionts significantly increased expansion of ISCs, Paneth cells, and goblet cells. Lactate stimulated ISC proliferation through Wnt/ß-catenin signals of Paneth cells and intestinal stromal cells. Moreover, Lactobacillus plantarum strains lacking lactate dehydrogenase activity, which are deficient in lactate production, elicited less ISC proliferation. Pre-treatment with LAB-type symbionts or lactate protected mice in response to gut injury provoked by combined treatments with radiation and a chemotherapy drug. Impaired ISC-mediated epithelial development was found in mice deficient of the lactate G-protein-coupled receptor, Gpr81. Our results demonstrate that LAB-type symbiont-derived lactate plays a pivotal role in promoting ISC-mediated epithelial development in a Gpr81-dependent manner.


Subject(s)
Goblet Cells/cytology , Lactic Acid/metabolism , Lactobacillus plantarum/metabolism , Paneth Cells/cytology , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Goblet Cells/drug effects , Goblet Cells/radiation effects , HEK293 Cells , Humans , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Lactobacillus plantarum/genetics , Methotrexate/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Paneth Cells/drug effects , Paneth Cells/radiation effects , Receptors, G-Protein-Coupled/genetics
2.
Stem Cell Reports ; 10(6): 1947-1958, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29805107

ABSTRACT

Two principal stem cell pools orchestrate the rapid cell turnover in the intestinal epithelium. Rapidly cycling Lgr5+ stem cells are intercalated between the Paneth cells at the crypt base (CBCs) and injury-resistant reserve stem cells reside above the crypt base. The intermediate filament Keratin 15 (Krt15) marks either stem cells or long-lived progenitor cells that contribute to tissue repair in the hair follicle or the esophageal epithelium. Herein, we demonstrate that Krt15 labels long-lived and multipotent cells in the small intestinal crypt by lineage tracing. Krt15+ crypt cells display self-renewal potential in vivo and in 3D organoid cultures. Krt15+ crypt cells are resistant to high-dose radiation and contribute to epithelial regeneration following injury. Notably, loss of the tumor suppressor Apc in Krt15+ cells leads to adenoma and adenocarcinoma formation. These results indicate that Krt15 marks long-lived, multipotent, and injury-resistant crypt cells that may function as a cell of origin in intestinal cancer.


Subject(s)
Cell Transformation, Neoplastic , Intestinal Mucosa/cytology , Keratin-15/metabolism , Radiation Tolerance , Stem Cells/metabolism , Stem Cells/radiation effects , Animals , Biomarkers , Cell Differentiation , Cell Proliferation , Cell Self Renewal , Cell Transformation, Neoplastic/metabolism , Dose-Response Relationship, Radiation , Fluorescent Antibody Technique , Immunohistochemistry , Mice , Paneth Cells/cytology , Paneth Cells/metabolism , Paneth Cells/radiation effects , Stem Cells/cytology
3.
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
4.
Nature ; 526(7575): 715-8, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26503053

ABSTRACT

The gut epithelium has remarkable self-renewal capacity that under homeostatic conditions is driven by Wnt signalling in Lgr5(+) intestinal stem cells (ISCs). However, the mechanisms underlying ISC regeneration after injury remain poorly understood. The Hippo signalling pathway mediates tissue growth and is important for regeneration. Here we demonstrate in mice that Yap, a downstream transcriptional effector of Hippo, is critical for recovery of intestinal epithelium after exposure to ionizing radiation. Yap transiently reprograms Lgr5(+) ISCs by suppressing Wnt signalling and excessive Paneth cell differentiation, while promoting cell survival and inducing a regenerative program that includes Egf pathway activation. Accordingly, growth of Yap-deficient organoids is rescued by the Egfr ligand epiregulin, and we find that non-cell-autonomous production of stromal epiregulin may compensate for Yap loss in vivo. Consistent with key roles for regenerative signalling in tumorigenesis, we further demonstrate that Yap inactivation abolishes adenomas in the Apc(Min) mouse model of colon cancer, and that Yap-driven expansion of Apc(-/-) organoids requires the Egfr module of the Yap regenerative program. Finally, we show that in vivo Yap is required for progression of early Apc mutant tumour-initiating cells, suppresses their differentiation into Paneth cells, and induces a regenerative program and Egfr signalling. Our studies reveal that upon tissue injury, Yap reprograms Lgr5(+) ISCs by inhibiting the Wnt homeostatic program, while inducing a regenerative program that includes activation of Egfr signalling. Moreover, our findings reveal a key role for the Yap regenerative pathway in driving cancer initiation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Differentiation , Colonic Neoplasms/pathology , Intestines/cytology , Phosphoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Regeneration , Stem Cells/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adenoma/metabolism , Adenoma/pathology , Animals , Cell Cycle Proteins , Cell Differentiation/radiation effects , Cell Survival/radiation effects , Cell Transformation, Neoplastic , Colonic Neoplasms/metabolism , Disease Models, Animal , Epiregulin/metabolism , ErbB Receptors/metabolism , Female , Hippo Signaling Pathway , Homeostasis/radiation effects , Intestinal Mucosa/metabolism , Intestines/radiation effects , Male , Mice , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Organoids/metabolism , Paneth Cells/cytology , Paneth Cells/radiation effects , Phosphoproteins/deficiency , Protein Serine-Threonine Kinases/metabolism , Radiation, Ionizing , Regeneration/radiation effects , Stem Cells/cytology , Stem Cells/radiation effects , Wnt Signaling Pathway , YAP-Signaling Proteins
5.
Cell Stem Cell ; 14(2): 149-59, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24332836

ABSTRACT

The intestinal epithelium continually self-renews and can rapidly regenerate after damage. Lgr5 marks mitotically active intestinal stem cells (ISCs). Importantly, intestinal homeostasis can be maintained after depletion of Lgr5(+) cells due to the activation of Lgr5(-) reserve ISCs. The Lgr5(-) ISC populations are thought to play a similar role during intestinal regeneration following radiation-induced damage. We tested this regeneration hypothesis by combining depletion of Lgr5(+) ISCs with radiation exposure. In contrast to the negligible effect of Lgr5(+) ISC loss during homeostasis, depletion of Lgr5(+) cells during radiation-induced damage and subsequent repair caused catastrophic crypt loss and deterioration of crypt-villus architecture. Interestingly though, we found that crypts deficient for Lgr5(+) cells are competent to undergo hyperplasia upon loss of Apc. These data argue that Lgr5(-) reserve stem cells are radiosensitive and that Lgr5(+) cells are crucial for robust intestinal regeneration following radiation exposure but are dispensable for premalignant hyperproliferation.


Subject(s)
Intestines/physiology , Intestines/radiation effects , Radiation, Ionizing , Receptors, G-Protein-Coupled/metabolism , Regeneration , Stem Cells/cytology , Adenomatous Polyposis Coli Protein/metabolism , Animals , Colitis/chemically induced , Colitis/pathology , Dextran Sulfate , Diphtheria Toxin/pharmacology , Dose-Response Relationship, Radiation , Hyperplasia , Intestines/drug effects , Mice , Models, Animal , Paneth Cells/cytology , Paneth Cells/drug effects , Paneth Cells/radiation effects , Regeneration/drug effects , Regeneration/radiation effects , Stem Cells/drug effects , Stem Cells/radiation effects
6.
Int J Immunopathol Pharmacol ; 23(4): 1111-23, 2010.
Article in English | MEDLINE | ID: mdl-21244760

ABSTRACT

Ionizing irradiation causes damage and functional failure of irradiation-sensitive systems and tissues such as small intestine. The molecular mechanisms underlying inflammatory and adaptive responses to acute irradiation damage are poorly understood. Using a mouse model of total-body γ-irradiation, we assessed the irradiation response of crypt host-defense Paneth cells by measuring alpha-defensin 4 (AD4) expression and correlated the gathered data with activation of the caspase-1/IL-1ß inflammatory signaling cascade. The irradiation injury was produced in CD2F1 mice exposed to 9.25 Gy γ-radiation. This dose resulted in 85-100 percent mortality at the 15(th) day post-irradiation. Small intestine tissue samples were collected at the 7th day post-irradiation. Assessment of irradiation-associated pro-inflammatory alterations in small intestine tissue and expression of AD4 in Paneth cells was conducted using confocal immunofluorescence imaging, transmission electron microscopy (TEM), light microscopy, and immunoblotting techniques. The small intestine analysis revealed an increase in the precursor form of IL-1ß, the activated form of IL-1ß, and the activated form of caspase-1 (p10 CASP-1) at the 7(th) day post-irradiation. Immunoprecipitation analysis showed increased interaction between IL-1ß and p10 CASP-1 after irradiation. This effect was observed in the irradiated small intestine and CD15-positive Paneth cells using confocal imaging techniques. The pro-inflammatory alterations in Paneth cells were accompanied by increases in AD4 mRNA and its 8 kD peptide product. Paneth cell secretory activity was observed at the sites of bacterial translocation in the crypt lumens. These data suggest that Paneth cells can contribute to small intestine inflammatory remodeling during the post-irradiation period.


Subject(s)
Intestine, Small/radiation effects , Paneth Cells/radiation effects , Whole-Body Irradiation , Animals , Caspase 1/analysis , Gamma Rays , Interleukin-1beta/analysis , Interleukin-1beta/physiology , Intestine, Small/chemistry , Intestine, Small/pathology , Male , Mice , Paneth Cells/chemistry , Paneth Cells/pathology , alpha-Defensins/analysis , alpha-Defensins/genetics , alpha-Defensins/physiology
7.
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
8.
J Cell Sci ; 115(Pt 11): 2381-8, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12006622

ABSTRACT

The stem cells in the crypts of the small intestinal mucosa divide about a thousand times during the lifespan of a laboratory mouse, and yet they show little evidence of any decline in proliferative potential and rarely develop carcinogenic mutations, suggesting that their genome is extremely well protected. Protection against DNA-replication-induced errors can be achieved by the selective sorting of old (template) and new DNA strands with all template strands retained in the stem cell line. The template strands in the stem cells can be labelled during development or during tissue regeneration using tritiated thymidine ((3)HTdR). Labelling newly synthesised strands with a different marker (bromodeoxyuridine, BrdUrd) allows segregation of the two markers to be studied. Template strand label is retained ((3)HTdR), whereas label in the newly synthesised strands (BrdUrd) is lost following the second division of the stem cell. Random errors may occur in the template strands owing to environmental elements. These are protected against by the altruistic cell suicide (apoptosis) of the cells incurring such errors. A final level of protection for the tissue compensates for excessive deletion of stem cells via the apoptosis pathway. This is achieved by a hierarchical age structure in the stem cell compartment, with some cells being able to efficiently repair DNA damage and hence being more radioresistant. The presence of these protective mechanisms ensures that the small intestine rarely develops cancer and that stem cells can sustain the extensive cell proliferation needed during life.


Subject(s)
Cell Division/genetics , Chromosome Segregation/genetics , DNA Repair/genetics , DNA Replication/genetics , Intestinal Mucosa/metabolism , Stem Cells/metabolism , Templates, Genetic , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Bromodeoxyuridine , Cell Division/radiation effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/radiation effects , Chromosome Segregation/radiation effects , DNA Repair/radiation effects , DNA Replication/radiation effects , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/radiation effects , Gamma Rays , Genome , Intestinal Mucosa/cytology , Intestinal Mucosa/radiation effects , Intestinal Neoplasms/genetics , Intestine, Small/cytology , Intestine, Small/metabolism , Intestine, Small/radiation effects , Mice , Mice, Inbred Strains , Mutation/genetics , Mutation/radiation effects , Paneth Cells/cytology , Paneth Cells/metabolism , Paneth Cells/radiation effects , Stem Cells/cytology , Stem Cells/radiation effects
9.
Pathol Res Pract ; 170(1-3): 104-14, 1980 Dec.
Article in English | MEDLINE | ID: mdl-18788156

ABSTRACT

Attempts were made to learn about an optimal condition for the induction of intestinal metaplasia in the gastric mucosa. The gastric region of 5-week-old female Wistar rats was irradiated with 500 rad of X-ray daily for 6 times (Group I) or with 1,000 rad of X-ray every two days for 3 times (Group III). In addition, the effect of immunization by allogeneic stomach antigen on the intestinalization was studied in rats irradiated with 500 rad of X-ray daily for 6 times (Group II). In a group of rats (Group II) injected with allogeneic stomach antigen and X-irradiated the process of intestinalization was more accelerated as compared to that in rats treated with X-ray (Group I). The similar results were obtained in rats irradiated with 1,000 rad of X-ray 3 times (Group II). Intestinal metaplasia developed more later in the fundic gland mucosa which became usually atrophic due to the loss of parietal cell mass. There was an intimate association among the parietal cell loss in the fundic gland, a rise in pH value and the development of intestinal metaplasia. In all groups, no case of gastric adenocarcinoma was detected during observation period up to 52nd week.


Subject(s)
Gastric Mucosa/radiation effects , Paneth Cells/radiation effects , Parietal Cells, Gastric/radiation effects , X-Rays , Animals , Antigens , Atrophy , Dose-Response Relationship, Radiation , Female , Gastric Fundus/pathology , Gastric Fundus/radiation effects , Gastric Mucosa/immunology , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Hydrogen-Ion Concentration , Metaplasia , Paneth Cells/metabolism , Paneth Cells/pathology , Parietal Cells, Gastric/metabolism , Parietal Cells, Gastric/pathology , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...