Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Nature ; 557(7706): 580-584, 2018 05.
Article in English | MEDLINE | ID: mdl-29769727

ABSTRACT

Somatic mutations in tet methylcytosine dioxygenase 2 (TET2), which encodes an epigenetic modifier enzyme, drive the development of haematopoietic malignancies1-7. In both humans and mice, TET2 deficiency leads to increased self-renewal of haematopoietic stem cells with a net developmental bias towards the myeloid lineage1,4,8,9. However, pre-leukaemic myeloproliferation (PMP) occurs in only a fraction of Tet2-/- mice8,9 and humans with TET2 mutations1,3,5-7, suggesting that extrinsic non-cell-autonomous factors are required for disease onset. Here we show that bacterial translocation and increased interleukin-6 production, resulting from dysfunction of the small-intestinal barrier, are critical for the development of PMP in mice that lack Tet2 expression in haematopoietic cells. Furthermore, in symptom-free Tet2-/- mice, PMP can be induced by disrupting intestinal barrier integrity, or in response to systemic bacterial stimuli such as the toll-like receptor 2 agonist. PMP was reversed by antibiotic treatment and failed to develop in germ-free Tet2-/- mice, which illustrates the importance of microbial signals in the development of this condition. Our findings demonstrate the requirement for microbial-dependent inflammation in the development of PMP and provide a mechanistic basis for the variation in PMP penetrance observed in Tet2-/- mice. This study will prompt new lines of investigation that may profoundly affect the prevention and management of haematopoietic malignancies.


Subject(s)
Asymptomatic Diseases , Bacterial Physiological Phenomena , Cell Proliferation , DNA-Binding Proteins/deficiency , Leukemia/microbiology , Leukemia/pathology , Proto-Oncogene Proteins/deficiency , Animals , Bacterial Infections/immunology , Bacterial Infections/microbiology , Bacterial Physiological Phenomena/immunology , DNA-Binding Proteins/genetics , Dioxygenases , Female , Germ-Free Life , Inflammation/microbiology , Interleukin-6/immunology , Intestinal Mucosa/metabolism , Lactobacillus/chemistry , Lactobacillus/cytology , Lactobacillus/immunology , Male , Mice , Penetrance , Permeability , Proto-Oncogene Proteins/genetics , Toll-Like Receptor 2/agonists
2.
Nature ; 487(7405): 104-8, 2012 Jul 05.
Article in English | MEDLINE | ID: mdl-22722865

ABSTRACT

The composite human microbiome of Western populations has probably changed over the past century, brought on by new environmental triggers that often have a negative impact on human health. Here we show that consumption of a diet high in saturated (milk-derived) fat, but not polyunsaturated (safflower oil) fat, changes the conditions for microbial assemblage and promotes the expansion of a low-abundance, sulphite-reducing pathobiont, Bilophila wadsworthia. This was associated with a pro-inflammatory T helper type 1 (T(H)1) immune response and increased incidence of colitis in genetically susceptible Il10(−/−), but not wild-type mice. These effects are mediated by milk-derived-fat-promoted taurine conjugation of hepatic bile acids, which increases the availability of organic sulphur used by sulphite-reducing microorganisms like B. wadsworthia. When mice were fed a low-fat diet supplemented with taurocholic acid, but not with glycocholic acid, for example, a bloom of B. wadsworthia and development of colitis were observed in Il10(−/−) mice. Together these data show that dietary fats, by promoting changes in host bile acid composition, can markedly alter conditions for gut microbial assemblage, resulting in dysbiosis that can perturb immune homeostasis. The data provide a plausible mechanistic basis by which Western-type diets high in certain saturated fats might increase the prevalence of complex immune-mediated diseases like inflammatory bowel disease in genetically susceptible hosts.


Subject(s)
Bilophila/drug effects , Colitis/chemically induced , Colitis/microbiology , Dietary Fats/pharmacology , Interleukin-10/deficiency , Metagenome/drug effects , Taurocholic Acid/metabolism , Animals , Bile Acids and Salts/metabolism , Bilophila/growth & development , Colitis/immunology , Colitis/pathology , Diet, Fat-Restricted , Inflammation/chemically induced , Inflammation/immunology , Inflammation/microbiology , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Interleukin-10/genetics , Mice , Mice, Inbred C57BL , Milk/chemistry , Molecular Sequence Data , Safflower Oil/pharmacology , Sulfites/metabolism , Taurine/metabolism , Taurocholic Acid/pharmacology , Th1 Cells/drug effects , Th1 Cells/immunology
3.
Pediatr Surg Int ; 34(3): 323-330, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29196880

ABSTRACT

BACKGROUND: Necrotizing enterocolitis (NEC) is a gastrointestinal disease of complex etiology resulting in devastating systemic inflammation and often death in premature newborns. We previously demonstrated that formula feeding inhibits ileal expression of heat shock protein-70 (Hsp70), a critical stress protein within the intestine. Barrier function for the premature intestine is critical. We sought to determine whether reduced Hsp70 protein expression increases neonatal intestinal permeability. METHODS: Young adult mouse colon cells (YAMC) were utilized to evaluate barrier function as well as intestine from Hsp70-/- pups (KO). Sections of intestine were analyzed by Western blot, immunohistochemistry, and real time PCR. YAMC cells were sub-lethally heated or treated with expressed milk (EM) to induce Hsp70. RESULTS: Immunostaining demonstrates co-localized Hsp70 and tight junction protein zona occludens-1 (ZO-1), suggesting physical interaction to protect tight junction function. The permeability of YAMC monolayers increases following oxidant injury and is partially blocked by Hsp70 induction either by prior heat stress or EM. RT-PCR analysis demonstrated that the Hsp70 isoforms, 70.1 and 70.3, predominate in WT pup; however, Hsp70.2 predominates in the KO pups. While Hsp70 is present in WT milk, it is not present in KO EM. Hsp70 associates with ZO-1 to maintain epithelial barrier function. CONCLUSION: Both induction of Hsp70 and exposure to EM prevent stress-induced increased permeability. Hsp70.2 is present in both WT and KO neonatal intestine, suggesting a crucial role in epithelial integrity. Induction of the Hsp70.2 isoform appears to be mediated by mother's milk. These results suggest that mother's milk feeding modulates Hsp70.2 expression and could attenuate injury leading to NEC. LEVEL OF EVIDENCE: Level III.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , Intestinal Mucosa/metabolism , Milk/metabolism , Animals , Animals, Newborn , Cytoprotection , HSP70 Heat-Shock Proteins/genetics , Mice , Permeability , Protein Isoforms , Tight Junctions/metabolism , Zonula Occludens-1 Protein/metabolism
4.
Biomed Chromatogr ; 31(4)2017 Apr.
Article in English | MEDLINE | ID: mdl-27606833

ABSTRACT

After ingestion of ginseng, the bioavailability of its parent compounds is low and enteric microbiota plays an important role in parent compound biotransformation to their metabolites. Diet type can influence the enteric microbiota profile. When human subjects on different diets ingest ginseng, their different gut microbiota profiles may influence the metabolism of ginseng parent compounds. In this study, the effects of different diet type on gut microbiota metabolism of American ginseng saponins were investigated. We recruited six healthy adults who regularly consumed different diet types. These subjects received 7 days' oral American ginseng, and their biological samples were collected for LC-Q-TOF-MS analysis. We observed significant ginsenoside Rb1 (a major parent compound) and compound K (a major active metabolite) level differences in the samples from the subjects consuming different diets. Subjects on an Asian diet had much higher Rb1 levels but much lower compound K levels compared with those on a Western diet. Since compound K possesses much better cancer chemoprevention potential, our data suggested that consumers on a Western diet should obtain better cancer prevention effects with American ginseng intake compared with those on an Asian diet. Ginseng compound levels could be enhanced or reduced via gut microbiota manipulation for clinical utility.


Subject(s)
Diet , Gastrointestinal Microbiome , Panax/metabolism , Saponins/pharmacokinetics , Adult , Chromatography, Liquid/methods , Diet, Western , Feces/chemistry , Gastrointestinal Microbiome/drug effects , Ginsenosides/analysis , Ginsenosides/metabolism , Humans , Inactivation, Metabolic , Male , Middle Aged , Saponins/analysis , Saponins/metabolism
5.
Phytother Res ; 31(1): 90-99, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27730672

ABSTRACT

Chemopreventative properties of traditional medicines and underlying mechanisms of action are incompletely investigated. This study demonstrates that dietary daikenchuto (TU-100), comprised of ginger, ginseng, and Japanese pepper effectively suppresses intestinal tumor development and progression in the azoxymethane (AOM) and APCmin/+ mouse models. For the AOM model, TU-100 was provided after the first of six biweekly AOM injections. Mice were sacrificed at 30 weeks. APCmin/+ mice were fed diet without or with TU-100 starting at 6 weeks, and sacrificed at 24 weeks. In both models, dietary TU-100 decreased tumor size. In APC min/+ mice, the number of small intestinal tumors was significantly decreased. In the AOM model, both TU-100 and Japanese ginseng decreased colon tumor numbers. Decreased Ki-67 and ß-catenin immunostaining and activation of numerous transduction pathways involved in tumor initiation and progression were observed. EGF receptor expression and stimulation/phosphorylation in vitro were investigated in C2BBe1 cells. TU-100, ginger, and 6-gingerol suppressed EGF receptor induced Akt activation. TU-100 and ginseng and to a lesser extent ginger or 6-gingerol inhibited EGF ERK1/2 activation. TU-100 and some of its components and metabolites of these components inhibit tumor progression in two mouse models of colon cancer by blocking downstream pathways of EGF receptor activation. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Azoxymethane/chemistry , Colonic Neoplasms/drug therapy , Plant Extracts/therapeutic use , Animals , Azoxymethane/pharmacology , Colonic Neoplasms/pathology , Disease Models, Animal , Male , Medicine, Traditional , Mice , Panax , Plant Extracts/administration & dosage , Zanthoxylum , Zingiberaceae
6.
Am J Physiol Gastrointest Liver Physiol ; 311(4): G634-G647, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27514476

ABSTRACT

Bifidobacterium breve and other Gram-positive gut commensal microbes protect the gastrointestinal epithelium against inflammation-induced stress. However, the mechanisms whereby these bacteria accomplish this protection are poorly understood. In this study, we examined soluble factors derived from Bifidobacterium breve and their impact on the two major protein degradation systems within intestinal epithelial cells, proteasomes and autophagy. Conditioned media from gastrointestinal Gram-positive, but not Gram-negative, bacteria activated autophagy and increased expression of the autophagy proteins Atg5 and Atg7 along with the stress response protein heat shock protein 27. Specific examination of media conditioned by the Gram-positive bacterium Bifidobacterium breve (Bb-CM) showed that this microbe produces small molecules (<3 kDa) that increase expression of the autophagy proteins Atg5 and Atg7, activate autophagy, and inhibit proteasomal enzyme activity. Upregulation of autophagy by Bb-CM was mediated through MAP kinase signaling. In vitro studies using C2BBe1 cells silenced for Atg7 and in vivo studies using mice conditionally deficient in intestinal epithelial cell Atg7 showed that Bb-CM-induced cytoprotection is dependent on autophagy. Therefore, this work demonstrates that Gram-positive bacteria modify protein degradation programs within intestinal epithelial cells to promote their survival during stress. It also reveals the therapeutic potential of soluble molecules produced by these microbes for prevention and treatment of gastrointestinal disease.


Subject(s)
Autophagy/physiology , Intestinal Mucosa/microbiology , Proteasome Endopeptidase Complex/metabolism , Stress, Physiological/physiology , Animals , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Bifidobacterium breve , Cell Line , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Inflammation/metabolism , Inflammation/microbiology , Intestinal Mucosa/metabolism , Mice , Mice, Knockout , Signal Transduction/physiology
7.
Am J Physiol Gastrointest Liver Physiol ; 310(11): G973-88, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27079612

ABSTRACT

Gut dysbiosis, host genetics, and environmental triggers are implicated as causative factors in inflammatory bowel disease (IBD), yet mechanistic insights are lacking. Longitudinal analysis of ulcerative colitis (UC) patients following total colectomy with ileal anal anastomosis (IPAA) where >50% develop pouchitis offers a unique setting to examine cause vs. effect. To recapitulate human IPAA, we employed a mouse model of surgically created blind self-filling (SFL) and self-emptying (SEL) ileal loops using wild-type (WT), IL-10 knockout (KO) (IL-10), TLR4 KO (T4), and IL-10/T4 double KO mice. After 5 wk, loop histology, host gene/protein expression, and bacterial 16s rRNA profiles were examined. SFL exhibit fecal stasis due to directional motility oriented toward the loop end, whereas SEL remain empty. In WT mice, SFL, but not SEL, develop pouchlike microbial communities without accompanying active inflammation. However, in genetically susceptible IL-10-deficient mice, SFL, but not SEL, exhibit severe inflammation and mucosal transcriptomes resembling human pouchitis. The inflammation associated with IL-10 required TLR4, as animals lacking both pathways displayed little disease. Furthermore, germ-free IL-10 mice conventionalized with SFL, but not SEL, microbiota populations develop severe colitis. These data support essential roles of stasis-induced, colon-like microbiota, TLR4-mediated colonic metaplasia, and genetic susceptibility in the development of pouchitis and possibly UC. However, these factors by themselves are not sufficient. Similarities between this model and human UC/pouchitis provide opportunities for gaining insights into the mechanistic basis of IBD and for identification of targets for novel preventative and therapeutic interventions.


Subject(s)
Colitis, Ulcerative/etiology , Dysbiosis/complications , Gastrointestinal Motility , Interleukin-10/genetics , Toll-Like Receptor 4/genetics , Animals , Female , Humans , Interleukin-10/metabolism , Intestinal Mucosa/metabolism , Intestines/microbiology , Intestines/pathology , Intestines/physiopathology , Mice , Mice, Inbred C57BL , Microbiota , Toll-Like Receptor 4/metabolism
8.
Dig Dis Sci ; 58(3): 668-77, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23329012

ABSTRACT

BACKGROUND: Lubiprostone has been used to treat constipation through its effects to stimulate Cl(-) secretion, resulting in water and electrolyte secretion. AIM: Potential associated changes in intestinal mucus and the colonizing bacteria (microbiome) have not been studied. As mucus obstructions may play a role in cystic fibrosis, the hypothesis that lubiprostone alters intestinal mucus and the microbiome was investigated. METHODS: Ion transport studies were performed ex vivo. For mucus and microbiome studies, mice were gavaged daily with lubiprostone or vehicle. Mucin from intestinal sections was analyzed in Carnoy's fixed tissues stained with Alcian blue. Microbiome composition was analyzed by 16S rRNA gene-based sequencing. RESULTS: Lubiprostone stimulated short circuit current in all mouse intestinal segments after both serosal and mucosal additions, albeit at lower concentrations in the latter. Current was Cl-dependent and blocked by mucosal diphenylcarboxylic acid, serosal bumetanide, and serosal Ba(++). The CFTR inhibitor CFTRinh172 had a marginal effect. Mucus near epithelial cells (inner layer mucus) was not present in the small intestine of any mice. Proximal colon inner mucus layer was thicker in ∆F/∆F compared with +/∆F and +/+ mice. Lubiprostone decreased inner mucus layer thickness in both proximal and distal colon of all mice. Furthermore, lubiprostone altered the intestinal microbiome by increasing abundance of Lactobacillus and Alistipes. CONCLUSIONS: Lubiprostone activates non-CFTR Cl(-) secretion and alters the colonic inner mucus layer, which is associated with changes in the composition of the enteric microbiome.


Subject(s)
Alprostadil/analogs & derivatives , Bacteria/drug effects , Colon/drug effects , Colon/microbiology , Alprostadil/pharmacology , Animals , Anions/metabolism , Bacteria/classification , Bacteria/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Ion Transport/drug effects , Ion Transport/physiology , Lubiprostone , Mice , Mice, Inbred CFTR , Phylogeny , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism
9.
Int J Mol Sci ; 14(2): 2980-95, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23434653

ABSTRACT

Compound K (20-O-beta-D-glucopyranosyl-20(S)-protopanaxadiol, CK), an intestinal bacterial metabolite of ginseng protopanaxadiol saponins, has been shown to inhibit cell growth in a variety of cancers. However, the mechanisms are not completely understood, especially in colorectal cancer (CRC). A xenograft tumor model was used first to examine the anti-CRC effect of CK in vivo. Then, multiple in vitro assays were applied to investigate the anticancer effects of CK including antiproliferation, apoptosis and cell cycle distribution. In addition, a qPCR array and western blot analysis were executed to screen and validate the molecules and pathways involved. We observed that CK significantly inhibited the growth of HCT-116 tumors in an athymic nude mouse xenograft model. CK significantly inhibited the proliferation of human CRC cell lines HCT-116, SW-480, and HT-29 in a dose- and time-dependent manner. We also observed that CK induced cell apoptosis and arrested the cell cycle in the G1 phase in HCT-116 cells. The processes were related to the upregulation of p53/p21, FoxO3a-p27/p15 and Smad3, and downregulation of cdc25A, CDK4/6 and cyclin D1/3. The major regulated targets of CK were cyclin dependent inhibitors, including p21, p27, and p15. These results indicate that CK inhibits transcriptional activation of multiple tumor-promoting pathways in CRC, suggesting that CK could be an active compound in the prevention or treatment of CRC.

10.
Cell Host Microbe ; 30(6): 809-823.e6, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35439436

ABSTRACT

Gut microbial diurnal oscillations are important diet-dependent drivers of host circadian rhythms and metabolism ensuring optimal energy balance. However, the interplay between diet, microbes, and host factors sustaining intestinal oscillations is complex and poorly understood. Here, using a mouse model, we report the host C-type lectin antimicrobial peptide Reg3γ works with key ileal microbes to orchestrate these interactions in a bidirectional manner and does not correlate with the intestinal core circadian clock. High-fat diet is the primary driver of microbial oscillators that impair host metabolic homeostasis, resulting in arrhythmic host Reg3γ expression that secondarily drives abundance and oscillation of key gut microbes. This illustrates transkingdom coordination of biological rhythms primarily influenced by diet and reciprocal sensor-effector signals between host and microbial components, ultimately driving metabolism. Restoring the gut microbiota's capacity to sense dietary signals mediated by specific host factors such as Reg3γ could be harnessed to improve metabolic dysfunction.


Subject(s)
Circadian Clocks , Gastrointestinal Microbiome , Circadian Rhythm , Diet , Diet, High-Fat/adverse effects , Lipid Metabolism
11.
Am J Physiol Gastrointest Liver Physiol ; 301(1): G181-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21512157

ABSTRACT

Heat shock proteins (Hsps) are highly conserved proteins that play a role in cytoprotection and maintaining intestinal homeostasis. Glutamine is essential for the optimal induction of intestinal epithelial Hsp expression, but its mechanisms of action are incompletely understood. Glutamine is a substrate for polyamine synthesis and stimulates the activity of ornithine decarboxylase (ODC), a key enzyme for polyamine synthesis, in intestinal epithelial cells. Thus we investigated whether polyamines (putrescine, spermidine, or spermine) and their precursor ornithine mediate the induction of Hsp expression in IEC-18 rat intestinal epithelial cells. As previously observed, glutamine was required for heat stress induction of Hsp70 and Hsp25, although it had little effect under basal conditions. Under conditions of glutamine depletion, supplementation of ornithine or polyamines restored the heat-induced expression of Hsp70 and Hsp25. When ODC was inhibited by α-difluoromethylornithine (DFMO), an irreversible ODC inhibitor, the heat stress induction of Hsp70 and Hsp25 was decreased significantly, even in the presence of glutamine. Ornithine, polyamines, and DFMO did not modify the nuclear localization of heat shock transcription factor 1 (HSF-1). However, DFMO dramatically reduced glutamine-dependent HSF-1 binding to an oligonucleotide with heat shock elements (HSE), which was increased by glutamine. In addition, exogenous polyamines recovered the DNA-binding activity. These results indicate that polyamines play a critical role in the glutamine-dependent induction of the intestinal epithelial heat shock response through facilitation of HSF-1 binding to HSE.


Subject(s)
DNA-Binding Proteins/metabolism , Glutamine/pharmacology , HSP27 Heat-Shock Proteins/biosynthesis , HSP70 Heat-Shock Proteins/biosynthesis , Heat-Shock Response , Intestinal Mucosa/drug effects , Polyamines/pharmacology , Transcription Factors/metabolism , Animals , Cell Line , Eflornithine/pharmacology , Enzyme Inhibitors/pharmacology , HSP27 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/genetics , Heat Shock Transcription Factors , Intestinal Mucosa/metabolism , Rats , Response Elements
12.
Pediatr Res ; 69(5 Pt 1): 395-400, 2011 May.
Article in English | MEDLINE | ID: mdl-21263375

ABSTRACT

Preterm infants face many challenges in transitioning from the in utero to extrauterine environment while still immature. Failure of the preterm gut to successfully mature to accommodate bacteria and food substrate leads to significant morbidity such as neonatal necrotizing enterocolitis. The intestinal epithelial barrier plays a critical role in gut protection. Heat shock protein 70 (Hsp70) is an inducible cytoprotective molecule shown to protect the intestinal epithelium in adult models. To investigate the hypothesis that Hsp70 may be important for early protection of the immature intestine, Hsp70 expression was evaluated in intestine of immature rat pups. Data demonstrate that Hsp70 is induced by exposure to mother's milk. Hsp70 is found in mother's milk, and increased Hsp70 transcription is induced by mother's milk. This Hsp70 colocalizes with the tight junction protein ZO-1. Mother's milk-induced Hsp70 may contribute to maintenance of barrier function in the face of oxidant stress. Further understanding of the means by which mother's milk increases Hsp70 in the ileum will allow potential means of strengthening the intestinal barrier in at-risk preterm infants.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , Ileum/metabolism , Intestinal Mucosa/metabolism , Milk/metabolism , Animals , Animals, Newborn , Bacterial Infections/metabolism , Bacterial Infections/microbiology , Cytoprotection , Disease Models, Animal , HSP70 Heat-Shock Proteins/genetics , Hypoxia/metabolism , Ileum/microbiology , Ileum/pathology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Membrane Proteins/metabolism , Oxidative Stress , Permeability , Phosphoproteins/metabolism , RNA, Messenger/metabolism , Rats , Tight Junctions/metabolism , Up-Regulation , Zonula Occludens-1 Protein
13.
BMC Complement Altern Med ; 11: 111, 2011 Nov 09.
Article in English | MEDLINE | ID: mdl-22070864

ABSTRACT

BACKGROUND: Western diets increase colon cancer risk. Epidemiological evidence and experimental studies suggest that ginseng can inhibit colon cancer development. In this study we asked if ginseng could inhibit Western diet (20% fat) promoted colonic tumorigenesis and if compound K, a microbial metabolite of ginseng could suppress colon cancer xenograft growth. METHODS: Mice were initiated with azoxymethane (AOM) and, two weeks later fed a Western diet (WD, 20% fat) alone, or WD supplemented with 250-ppm ginseng. After 1 wk, mice received 2.5% dextran sulfate sodium (DSS) for 5 days and were sacrificed 12 wks after AOM. Tumors were harvested and cell proliferation measured by Ki67 staining and apoptosis by TUNEL assay. Levels of EGF-related signaling molecules and apoptosis regulators were determined by Western blotting. Anti-tumor effects of intraperitoneal compound K were examined using a tumor xenograft model and compound K absorption measured following oral ginseng gavage by UPLC-mass spectrometry. Effects of dietary ginseng on microbial diversity were measured by analysis of bacterial 16S rRNA. RESULTS: Ginseng significantly inhibited colonic inflammation and tumorigenesis and concomitantly reduced proliferation and increased apoptosis. The EGFR cascade was up-regulated in colonic tumors and ginseng significantly reduced EGFR and ErbB2 activation and Cox-2 expression. Dietary ginseng altered colonic microbial diversity, and bacterial suppression with metronidazole reduced serum compound K following ginseng gavage. Furthermore, compound K significantly inhibited tumor xenograft growth. CONCLUSIONS: Ginseng inhibited colonic inflammation and tumorigenesis promoted by Western diet. We speculate that the ginseng metabolite compound K contributes to the chemopreventive effects of this agent in colonic tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Dietary Fats/adverse effects , Down-Regulation/drug effects , ErbB Receptors/immunology , Panax/chemistry , Plant Extracts/therapeutic use , Animals , Bacteria/genetics , Bacteria/isolation & purification , Cell Line, Tumor , Colon/drug effects , Colon/immunology , Colon/microbiology , Colonic Neoplasms/microbiology , Colonic Neoplasms/pathology , Disease Models, Animal , ErbB Receptors/genetics , Humans , Male , Mice
14.
Am J Physiol Gastrointest Liver Physiol ; 298(4): G481-92, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20110459

ABSTRACT

Mucosal inflammation, through cytokines such as interferon-gamma (IFN-gamma) and tumor necrosis factor-alpha (TNF-alpha), has many effects on the intestinal epithelium, including selective translational inhibition of the cytoprotective protein heat shock protein 70 (Hsp70). To further elucidate the mechanisms underlying this effect, we examined the role of stress granules in mediating the actions of these proinflammatory cytokines. Using conditionally immortalized young adult mouse colonic epithelial cells, we demonstrate that IFN-gamma and TNF-alpha, which upregulate eukaryotic initiation factor-alpha (eIF-2alpha) phosphorylation and reduce Hsp70 translation, significantly enhance stress granule formation in heat-shocked intestinal epithelial cells. The IFN-gamma and TNF-alpha effects in upregulation of stress granule formation and downregulation of Hsp70 were eIF-2alpha dependent, and the effect could be negated by blocking eIF-2alpha phosphorylation with use of an RNA-dependent protein kinase inhibitor. Correspondingly, IFN-gamma and TNF-alpha increased binding of cytoplasmic proteins to the 3'-untranslated region of Hsp70 mRNA, suggesting specific recruitment of Hsp70 to stress granules as the mechanism of IFN-gamma and TNF-alpha inhibition of Hsp70 translation. We thus report a novel linkage between inflammatory cytokine production, stress granule formation, and Hsp70 translation inhibition, providing additional insights into the response of intestinal epithelial cells to inflammatory stress.


Subject(s)
Cytoplasmic Granules/physiology , Gene Expression Regulation/physiology , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Response/physiology , Interferon-gamma/pharmacology , Protein Biosynthesis/physiology , Tumor Necrosis Factor-alpha/pharmacology , 3' Untranslated Regions/genetics , Animals , Cell Line, Transformed , Colon/cytology , Cytoplasmic Granules/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression/drug effects , Gene Expression/genetics , HSP70 Heat-Shock Proteins/genetics , Mice , Models, Biological , Phosphorylation/drug effects , Protein Binding/genetics , Protein Biosynthesis/drug effects , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Ribonucleoproteins/metabolism , T-Cell Intracellular Antigen-1 , eIF-2 Kinase/antagonists & inhibitors
15.
Am J Physiol Gastrointest Liver Physiol ; 298(2): G203-11, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19926819

ABSTRACT

The apical membrane Na(+)-H(+) exchanger (NHE)3 is regulated by cAMP-dependent phosphorylation, which inhibits its activity through membrane endocytosis. The clathrin complex adaptor protein synaptotagmin 1 (Syt 1) appears to be essential to this process, but little is known about its expression in intestinal epithelial cells or interaction with NHE3. The intestinal epithelial expression and apical location of Syt 1 were determined by Syt 1 mRNA profiling and immunolocalization. Tandem mass spectrometry was used for protein identification. Bis(sulfosuccinimidyl) suberate (BS(3)) cross linking suggested that NHE3 and Syt 1 were in a membrane complex following cAMP stimulation of Caco2BBE (Brush Border Expressions) cells. To investigate the regulation of NHE3 appearance in a Syt 1-containing membrane compartment, doxycycline-inducible hemaglutinin (HA)-tagged NHE3 was expressed in Caco2BBE cells. HA-NHE3 correctly targeted to the apical membrane, where, upon cAMP stimulation, it was internalized with a Syt 1-containing compartment. Site-directed mutagenesis of NHE3 showed that serine 605 (S605) was pivotal to NHE3 and Syt 1 association and internalization. Direct Syt 1 interaction with NHE3 was suggested by fluorescence resonance energy transfer (FRET) analysis. The physiological role of S552 was less clear. By FRET, this serine residue appeared to be involved in cAMP-induced Syt 1 binding of NHE3. However, when HA-tagged NHE3 S552A was expressed in Caco2 cells, the mutated construct was not inserted into the apical membrane. We conclude that intestinal epithelial Syt 1 plays an important role in cAMP-stimulated endocytosis of apical NHE3 through cAMP-dependent phosphorylation of S605 that is required for NHE3 and Syt 1 association.


Subject(s)
Cyclic AMP/metabolism , Endocytosis/physiology , Intestinal Mucosa/cytology , Sodium-Hydrogen Exchangers/metabolism , Synaptotagmin I/metabolism , Amino Acid Sequence , Animals , Caco-2 Cells , Diarrhea/metabolism , Diarrhea/physiopathology , Fluorescence Resonance Energy Transfer , Humans , Intestinal Mucosa/metabolism , Jejunum/physiology , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation/physiology , Protein Binding/physiology , Protein Transport/physiology , RNA, Messenger/metabolism , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/genetics , Synaptotagmin I/genetics
16.
Am J Physiol Gastrointest Liver Physiol ; 299(6): G1266-75, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20864653

ABSTRACT

Cytoprotective heat shock proteins (Hsps) are critical for intestinal homeostasis and are known to be decreased in inflammatory bowel diseases. Signals responsible for maintenance of Hsp expression are incompletely understood. In this study, we find that Hsp25/27 and Hsp70 protein expressions are differentially regulated along the longitudinal length of the large intestine, being highest in the proximal colon and decreasing to the distal colon. This longitudinal gradient was similar in both conventionally colonized mouse colon as well as biopsies of human proximal and distal colon but was abolished in the colon of germ-free mice, suggesting a role of intestinal microbiota in the Hsp regional expression. Correspondingly, analysis of 16S ribosomal RNA genes of bacteria from each colonic segment indicated increased bacterial richness and diversity in the proximal colon. The mechanism of regulation is transcriptional, as Hsp70 mRNA followed a similar pattern to Hsp70 protein expression. Lysates of mucosa-associated bacteria from the proximal colon stimulated greater Hsp25 and Hsp70 mRNA transcription and subsequent protein expression in intestinal epithelial cells than did lysates from distal colon. In addition, transrectal administration of cecal contents stimulated Hsp25 and Hsp70 expression in the distal colon. Thus host-microbial interactions resulting in differential Hsp expression may have significant implications for the maintenance of intestinal homeostasis and possibly for development of inflammatory diseases of the bowel.


Subject(s)
Colon/microbiology , Gene Expression Regulation/physiology , Heat-Shock Proteins/metabolism , Intestinal Mucosa/microbiology , Animals , Biopsy , Cecum/microbiology , Cells, Cultured , Colon/anatomy & histology , Colon/cytology , Colon/metabolism , Germ-Free Life , Heat-Shock Proteins/genetics , Humans , Jejunum/metabolism , Jejunum/microbiology , Mice , Mice, Inbred C57BL , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism
17.
Gastroenterology ; 136(3): 924-32, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19121316

ABSTRACT

BACKGROUND & AIMS: Glutamine plays a protective role in intestinal cells during physiologic stress; however, the protection mechanisms are not fully understood. Autophagy functions in bulk degradation of cellular components, but has been recognized recently as an important mechanism for cell survival under conditions of stress. We therefore sought to see if glutamine's actions involve the induction of autophagy in intestinal cells and, if so, the mechanisms that underlie this action. METHODS: Formation of microtubule-associated protein light chain 3 (LC3)-phospholipid conjugates (LC3-II) in rat intestinal epithelial IEC-18 cells and human colonic epithelial Caco-2(BBE) cells was determined by Western blotting and localized by confocal microscopy. Activation of mammalian target of rapamycin (mTOR) pathway, mitogen-activated protein (MAP) kinases, caspase-3, and poly (ADP-ribose) polymerase were monitored by Western blotting. RESULTS: Glutamine increased LC3-II as well as the number of autophagosomes. Glutamine-induced LC3-II formation was paralleled by inactivation of mTOR and p38 MAP kinase pathways, and inhibition of mTOR and p38 MAP kinase allowed LC3-II induction in glutamine-deprived cells. Under glutamine starvation, LC3-II recovery after heat stress or the increase under oxidative stress was blunted significantly. Glutamine depletion increased caspase-3 and poly (ADP-ribose) polymerase activity after heat stress, which was inhibited by treatment with inhibitors of mTOR and p38 MAP kinase. CONCLUSIONS: Glutamine induces autophagy under basal and stressed conditions, and prevents apoptosis under heat stress through its regulation of the mTOR and p38 MAP kinase pathways. We propose that glutamine contributes to cell survival during physiologic stress by induction of autophagy.


Subject(s)
Autophagy/drug effects , Glutamine/pharmacology , Glutamine/physiology , Intestinal Mucosa/drug effects , Stress, Physiological/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Autophagy/physiology , Caco-2 Cells , Cell Survival/drug effects , Cell Survival/physiology , Colon/cytology , Humans , Intestinal Mucosa/cytology , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Proteins , Rats , TOR Serine-Threonine Kinases , Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Appl Microbiol Biotechnol ; 88(6): 1333-42, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20931185

ABSTRACT

Metagenomic analysis of colonic mucosa-associated microbes has been complicated by technical challenges that disrupt or alter community structure and function. In the present study, we determined the feasibility of laser capture microdissection (LCM) of intact regional human colonic mucosa-associated microbes followed by phi29 multiple displacement amplification (MDA) and massively parallel sequencing for metagenomic analysis. Samples were obtained from the healthy human subject without bowel preparation and frozen sections immediately prepared. Regional mucosa-associated microbes were successfully dissected using LCM with minimal contamination by host cells, their DNA extracted and subjected to phi29 MDA with a high fidelity, prior to shotgun sequencing using the GS-FLX DNA sequencer. Metagenomic analysis of approximately 67 million base pairs of DNA sequences from two samples revealed that the metabolic functional profiles in mucosa-associated microbes were as diverse as those reported in feces, specifically the representation of functional genes associated with carbohydrate, protein, and nucleic acid utilization. In summary, these studies demonstrate the feasibility of the approach to study the structure and metagenomic profiles of human intestinal mucosa-associated microbial communities at small spatial scales.


Subject(s)
Biodiversity , Colon/microbiology , Intestinal Mucosa/microbiology , Metagenome , Microdissection/methods , Cluster Analysis , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Ribosomal/chemistry , DNA, Ribosomal/genetics , High-Throughput Nucleotide Sequencing , Human Experimentation , Humans , Molecular Sequence Data , Nucleic Acid Amplification Techniques , Phylogeny , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA
19.
Proc Natl Acad Sci U S A ; 104(50): 20120-5, 2007 Dec 11.
Article in English | MEDLINE | ID: mdl-18077438

ABSTRACT

The sodium-chloride cotransporter (NCC) is the principal salt-absorptive pathway in the mammalian distal convoluted tubule (DCT) and is the site of action of one of the most effective classes of antihypertensive medications, thiazide diuretics. We developed a cell model system to assess NCC function in a mammalian cell line that natively expresses NCC, the mouse DCT (mDCT) cell line. We used this system to study the complex regulation of NCC by the phorbol ester (PE) 12-O-tetradecanoylphorbol-13-acetate (TPA), a diacylglycerol (DAG) analog. It has generally been thought that PEs mediate their effects on transporters through the activation of PKC. However, there are at least five other DAG/PE targets. Here we describe how one of those alternate targets of DAG/PE effects, Ras guanyl-releasing protein 1 (RasGRP1), mediates the PE-induced suppression of function and the surface expression of NCC. Functional assessment of NCC by using thiazide-sensitive (22)Na(+) uptakes revealed that TPA completely suppresses NCC function. Biotinylation experiments demonstrated that this result was primarily because of decreased surface expression of NCC. Although inhibitors of PKC had no effect on this suppression, MAPK inhibitors completely prevented the TPA effect. RasGRP1 activates the MAPK pathway through activation of the small G protein Ras. Gene silencing of RasGRP1 prevented the PE-mediated suppression of NCC activity, the activation of the H-Ras isoform of Ras, and the activation of ERK1/2 MAPK. This finding confirmed the critical role of RasGRP1 in mediating the PE-induced suppression of NCC activity through the stimulation of the MAPK pathway.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Protein Kinase C/physiology , Signal Transduction/physiology , Sodium Chloride Symporters/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Absorption/drug effects , Absorption/physiology , Animals , Cells, Cultured , Kidney Tubules, Distal/drug effects , Kidney Tubules, Distal/metabolism , Mice , Signal Transduction/drug effects , Sodium Chloride Symporter Inhibitors/pharmacology , Xenopus laevis
20.
J Clin Invest ; 116(10): 2682-94, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17016558

ABSTRACT

Acute T cell-mediated diarrhea is associated with increased mucosal expression of proinflammatory cytokines, including the TNF superfamily members TNF and LIGHT. While we have previously shown that epithelial barrier dysfunction induced by myosin light chain kinase (MLCK) is required for the development of diarrhea, MLCK inhibition does not completely restore water absorption. In contrast, although TNF-neutralizing antibodies completely restore water absorption after systemic T cell activation, barrier function is only partially corrected. This suggests that, while barrier dysfunction is critical, other processes must be involved in T cell-mediated diarrhea. To define these processes in vivo, we asked whether individual cytokines might regulate different events in T cell-mediated diarrhea. Both TNF and LIGHT caused MLCK-dependent barrier dysfunction. However, while TNF caused diarrhea, LIGHT enhanced intestinal water absorption. Moreover, TNF, but not LIGHT, inhibited Na+ absorption due to TNF-induced internalization of the brush border Na+/H+ exchanger NHE3. LIGHT did not cause NHE3 internalization. PKCalpha activation by TNF was responsible for NHE3 internalization, and pharmacological or genetic PKCalpha inhibition prevented NHE3 internalization, Na+ malabsorption, and diarrhea despite continued barrier dysfunction. These data demonstrate the necessity of coordinated Na+ malabsorption and barrier dysfunction in TNF-induced diarrhea and provide insight into mechanisms of intestinal water transport.


Subject(s)
Diarrhea/physiopathology , Intestinal Mucosa/physiopathology , Sodium-Hydrogen Exchangers/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cyclic AMP/metabolism , Diarrhea/chemically induced , Diarrhea/metabolism , Gene Expression/genetics , Intestinal Absorption/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Jejunum/drug effects , Jejunum/metabolism , Jejunum/physiopathology , Lymphocyte Activation/physiology , Lymphotoxin beta Receptor/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Myosin-Light-Chain Kinase/antagonists & inhibitors , Myosin-Light-Chain Kinase/metabolism , Occludin , Permeability/drug effects , Phosphorylation/drug effects , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/metabolism , Signal Transduction/drug effects , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/genetics , T-Lymphocytes/immunology , Tight Junctions/drug effects , Tight Junctions/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/toxicity , Water/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL