Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Proc Natl Acad Sci U S A ; 119(3)2022 01 18.
Article in English | MEDLINE | ID: mdl-35027453

ABSTRACT

Paneth cells are intestinal epithelial cells that release antimicrobial peptides, such as α-defensin as part of host defense. Together with mesenchymal cells, Paneth cells provide niche factors for epithelial stem cell homeostasis. Here, we report two subtypes of murine Paneth cells, differentiated by their production and utilization of fucosyltransferase 2 (Fut2), which regulates α(1,2)fucosylation to create cohabitation niches for commensal bacteria and prevent invasion of the intestine by pathogenic bacteria. The majority of Fut2- Paneth cells were localized in the duodenum, whereas the majority of Fut2+ Paneth cells were in the ileum. Fut2+ Paneth cells showed higher granularity and structural complexity than did Fut2- Paneth cells, suggesting that Fut2+ Paneth cells are involved in host defense. Signaling by the commensal bacteria, together with interleukin 22 (IL-22), induced the development of Fut2+ Paneth cells. IL-22 was found to affect the α-defensin secretion system via modulation of Fut2 expression, and IL-17a was found to increase the production of α-defensin in the intestinal tract. Thus, these intestinal cytokines regulate the development and function of Fut2+ Paneth cells as part of gut defense.


Subject(s)
Cytokines/metabolism , Fucosyltransferases/metabolism , Gastrointestinal Microbiome/physiology , Paneth Cells/metabolism , Animals , Fucosyltransferases/genetics , Ileum , Interleukin-17/metabolism , Interleukins/metabolism , Mice , Symbiosis , alpha-Defensins/metabolism , Interleukin-22 , Galactoside 2-alpha-L-fucosyltransferase
2.
Biochem Biophys Res Commun ; 545: 14-19, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33529805

ABSTRACT

Paneth cells and Lgr5+ intestinal stem cells (Lgr5+ ISCs) constitute the stem cell niche and maintain small intestinal epithelial integrity by recognizing various niche factors derived from subepithelial cells and external antigens. Although it has been known that interferon-γ (IFN-γ), a Th1 cytokine, is associated with intestinal epithelial disruption during inflammation as a niche factor, dynamics of Paneth cells and Lgr5+ ISCs in response to IFN-γ remain to be understood. Here we show that CAG-tdTomato;Lgr5-EGFP (CT-LE) mice generated in this study enable to identify Paneth cells and Lgr5+ ISCs separately by fluorescence signals. Lgr5+ ISCs underwent cell death a little earlier than Paneth cells in response to IFN-γ by simultaneous tracking using CT-LE mice. In addition, the timing of cell death in most Paneth cells overlapped with Lgr5+ ISCs, suggesting that Paneth cell depletion is induced directly by IFN-γ. Taken together, we established a novel simultaneous stem cell niche tracking method and clarified the involvement of both Paneth cells and Lgr5+ ISCs in stem cell niche damage induced by IFN-γ, further contribute to understanding the mechanism for maintaining intestinal homeostasis by stem cell niche.


Subject(s)
Interferon-gamma/pharmacology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Paneth Cells/drug effects , Paneth Cells/pathology , Stem Cells/drug effects , Stem Cells/pathology , Animals , Cell Death/drug effects , Cell Death/physiology , Computer Systems , Homeostasis/drug effects , Homeostasis/physiology , Interferon-gamma/physiology , Intestinal Mucosa/physiology , Mice , Mice, Transgenic , Paneth Cells/physiology , Receptors, G-Protein-Coupled/metabolism , Receptors, Interferon/metabolism , Stem Cell Niche/drug effects , Stem Cell Niche/physiology , Stem Cells/physiology , Interferon gamma Receptor
3.
Biochem Biophys Res Commun ; 504(1): 231-237, 2018 09 26.
Article in English | MEDLINE | ID: mdl-30193733

ABSTRACT

Near-haploidy is observed in certain cancer types, but ploidy-dependent alterations in gene regulation in the haploid state remain elusive. Here, by comparative transcriptome analysis between human isogenic haploid and diploid cell lines, we found lowering of cyclin D2 level in haploids. Acute genome duplication in haploids restored cyclin D2 expression to diploid level, indicating that the regulation of cyclin D2 expression is directly linked to ploidy. Downstream pathways of cyclin D2, such as Rb phosphorylation and p27 sequestration remained intact in haploids, suggesting that they adapt to lowered cyclin D level. Interestingly, however, haploid cells were more susceptible to cdk4/6 inhibition compared to diploids. Our finding indicates feasibility of selective growth suppression of haploid cells based on ploidy-linked gene regulation.


Subject(s)
Cyclin D2/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Gene Expression Regulation , Haploidy , Ploidies , Cell Line , Cell Proliferation , Humans , Phosphorylation , RNA Interference
4.
Environ Monit Assess ; 185(7): 5627-37, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23108714

ABSTRACT

Perchlorate contamination was investigated in groundwater and surface water from Sivakasi and Madurai in the Tamil Nadu State of South India. Sensitive determination of perchlorate (LOQ = 0.005 µg/L) was achieved by large-volume (500 µL) injection ion chromatography coupled with tandem mass spectrometry. Concentrations of perchlorate were <0.005-7,690 µg/L in groundwater (n = 60), <0.005-30.2 µg/L in surface water (n = 11), and 0.063-0.393 µg/L in tap water (n = 3). Levels in groundwater were significantly higher in the fireworks factory area than in the other locations, indicating that the fireworks and safety match industries are principal sources of perchlorate pollution. This is the first study that reports the contamination status of perchlorate in this area and reveals firework manufacture to be the pollution source. Since perchlorate levels in 17 out of 57 groundwater samples from Sivakasi, and none from Madurai, exceeded the drinking water guideline level proposed by USEPA (15 µg/L), further investigation on human health is warranted.


Subject(s)
Environmental Monitoring , Groundwater/chemistry , Perchlorates/analysis , Water Pollutants, Chemical/analysis , Explosive Agents/analysis , India
5.
Life Sci Alliance ; 3(6)2020 06.
Article in English | MEDLINE | ID: mdl-32345659

ABSTRACT

Crohn's disease (CD) is an intractable inflammatory bowel disease, and dysbiosis, disruption of the intestinal microbiota, is associated with CD pathophysiology. ER stress, disruption of ER homeostasis in Paneth cells of the small intestine, and α-defensin misfolding have been reported in CD patients. Because α-defensins regulate the composition of the intestinal microbiota, their misfolding may cause dysbiosis. However, whether ER stress, α-defensin misfolding, and dysbiosis contribute to the pathophysiology of CD remains unknown. Here, we show that abnormal Paneth cells with markers of ER stress appear in SAMP1/YitFc, a mouse model of CD, along with disease progression. Those mice secrete reduced-form α-defensins that lack disulfide bonds into the intestinal lumen, a condition not found in normal mice, and reduced-form α-defensins correlate with dysbiosis during disease progression. Moreover, administration of reduced-form α-defensins to wild-type mice induces the dysbiosis. These data provide novel insights into CD pathogenesis induced by dysbiosis resulting from Paneth cell α-defensin misfolding and they suggest further that Paneth cells may be potential therapeutic targets.


Subject(s)
Crohn Disease/metabolism , Dysbiosis/metabolism , Ileitis/metabolism , Paneth Cells/metabolism , Protein Folding , alpha-Defensins/chemistry , alpha-Defensins/metabolism , Animals , Bacteroidaceae/genetics , Bacteroidetes/genetics , Crohn Disease/microbiology , Disease Models, Animal , Disease Progression , Dysbiosis/microbiology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/microbiology , Endoplasmic Reticulum Stress , Feces/microbiology , Gastrointestinal Microbiome/genetics , Ileitis/microbiology , Ileum/metabolism , Ileum/microbiology , Mice , Mice, Inbred ICR , RNA, Ribosomal, 16S
6.
Nutrients ; 11(11)2019 Nov 18.
Article in English | MEDLINE | ID: mdl-31752111

ABSTRACT

The intestine not only plays a role in fundamental processes in digestion and nutrient absorption, but it also has a role in eliminating ingested pathogenic bacteria and viruses. Paneth cells, which reside at the base of small intestinal crypts, secrete α-defensins and contribute to enteric innate immunity through potent microbicidal activities. However, the relationship between food factors and the innate immune functions of Paneth cells remains unknown. Here, we examined whether short-chain fatty acids and amino acids induce α-defensin secretion from Paneth cells in the isolated crypts of small intestine. Butyric acid and leucine elicit α-defensin secretion by Paneth cells, which kills Salmonella typhimurium. We further measured Paneth cell secretion in response to butyric acid and leucine using enteroids, a three-dimensional ex vivo culture system of small intestinal epithelial cells. Paneth cells expressed short-chain fatty acid receptors, Gpr41, Gpr43, and Gpr109a mRNAs for butyric acid, and amino acid transporter Slc7a8 mRNA for leucine. Antagonists of Gpr41 and Slc7a8 inhibited granule secretion by Paneth cells, indicating that these receptor and transporter on Paneth cells induce granule secretion. Our findings suggest that Paneth cells may contribute to intestinal homeostasis by secreting α-defensins in response to certain nutrients or metabolites.


Subject(s)
Butyric Acid/immunology , Intestine, Small/metabolism , Leucine/immunology , Paneth Cells/metabolism , alpha-Defensins/metabolism , Amino Acid Transport System y+/antagonists & inhibitors , Amino Acid Transport System y+/metabolism , Animals , Fusion Regulatory Protein 1, Light Chains/antagonists & inhibitors , Fusion Regulatory Protein 1, Light Chains/metabolism , Gene Expression , Homeostasis , Immunity, Innate , Mice , Mice, Inbred ICR , Microbiota , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism
7.
Sci Rep ; 9(1): 2710, 2019 02 25.
Article in English | MEDLINE | ID: mdl-30804449

ABSTRACT

Paneth cells at the base of small intestinal crypts secrete granules containing α-defensins in response to bacteria and maintain the intestinal environment by clearing enteric pathogens and regulating the composition of the intestinal microbiota. However, Paneth cell secretory responses remain debatable and the mechanisms that regulate the secretion are not well understood. Although enteroids, three-dimensional cultures of small intestinal epithelial cells, have proven useful for analyzing intestinal epithelial cell functions including ion transport, their closed structures have imposed limitations to investigating interactions between Paneth cells and the intestinal microbiota. Here, we report that microinjection of bacteria or lipopolysaccharide (LPS) into the enteroid lumen provides an ex vivo system for studying Paneth cell secretion in real-time. The results show that Paneth cells released granules immediately when the apical surfaces of enteroid epithelial cells were exposed to LPS or live bacteria by microinjection. However, Paneth cells did not respond to LPS delivered in culture media to enteroid exterior basolateral surface, although they responded to basolateral carbamyl choline. In addition, Paneth cells replenished their granules after secretion, enabling responses to second stimulation. These findings provide new insight for apically-induced Paneth cell secretory responses in regulating the intestinal environment.


Subject(s)
Paneth Cells/metabolism , Animals , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fluorescent Antibody Technique , LIM Domain Proteins/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Nuclear Proteins/metabolism , Paneth Cells/drug effects
8.
JCI Insight ; 3(18)2018 09 20.
Article in English | MEDLINE | ID: mdl-30232288

ABSTRACT

Paneth cells contribute to small intestinal homeostasis by secreting antimicrobial peptides and constituting the intestinal stem cell (ISC) niche. Certain T cell-mediated enteropathies are characterized by extensive Paneth cell depletion coincident with mucosal destruction and dysbiosis. In this study, mechanisms of intestinal crypt injury have been investigated by characterizing responses of mouse intestinal organoids (enteroids) in coculture with mouse T lymphocytes. Activated T cells induced enteroid damage, reduced Paneth cell and Lgr5+ ISC mRNA levels, and induced Paneth cell death through a caspase-3/7-dependent mechanism. IFN-γ mediated these effects, because IFN-γ receptor-null enteroids were unaffected by activated T cells. In mice, administration of IFN-γ induced enteropathy with crypt hyperplasia, villus shortening, Paneth cell depletion, and modified ISC marker expression. IFN-γ exacerbated radiation enteritis, which was ameliorated by treatment with a selective JAK1/2 inhibitor. Thus, IFN-γ induced Paneth cell death and impaired regeneration of small intestinal epithelium in vivo, suggesting that IFN-γ may be a useful target for treating defective mucosal regeneration in enteric inflammation.


Subject(s)
Inflammation/immunology , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Intestines/immunology , Paneth Cells/drug effects , Paneth Cells/immunology , T-Lymphocytes/immunology , Animals , Bone Marrow Transplantation , Cell Death/drug effects , Cell Proliferation/drug effects , Female , Homeostasis , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestines/drug effects , Intestines/pathology , Janus Kinase 1 , Janus Kinase 2 , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Organoids/immunology , Organoids/injuries , Paneth Cells/pathology , Receptors, G-Protein-Coupled/metabolism , Receptors, Interferon , Stem Cells
9.
J Exp Med ; 214(12): 3507-3518, 2017 Dec 04.
Article in English | MEDLINE | ID: mdl-29066578

ABSTRACT

The intestinal microbial ecosystem is actively regulated by Paneth cell-derived antimicrobial peptides such as α-defensins. Various disorders, including graft-versus-host disease (GVHD), disrupt Paneth cell functions, resulting in unfavorably altered intestinal microbiota (dysbiosis), which further accelerates the underlying diseases. Current strategies to restore the gut ecosystem are bacteriotherapy such as fecal microbiota transplantation and probiotics, and no physiological approach has been developed so far. In this study, we demonstrate a novel approach to restore gut microbial ecology by Wnt agonist R-Spondin1 (R-Spo1) or recombinant α-defensin in mice. R-Spo1 stimulates intestinal stem cells to differentiate to Paneth cells and enhances luminal secretion of α-defensins. Administration of R-Spo1 or recombinant α-defensin prevents GVHD-mediated dysbiosis, thus representing a novel and physiological approach at modifying the gut ecosystem to restore intestinal homeostasis and host-microbiota cross talk toward therapeutic benefits.


Subject(s)
Dysbiosis/etiology , Dysbiosis/prevention & control , Graft vs Host Disease/complications , Paneth Cells/pathology , Thrombospondins/pharmacology , Thrombospondins/therapeutic use , Administration, Oral , Animals , Bacteria/metabolism , Cell Differentiation/drug effects , Cytoprotection/drug effects , Dysbiosis/pathology , Female , Graft vs Host Disease/pathology , Humans , Intestines/pathology , Mice, Inbred C57BL , Paneth Cells/drug effects , Paneth Cells/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Stem Cell Transplantation , Stem Cells/drug effects , Stem Cells/metabolism , alpha-Defensins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL