Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Mol Cell ; 83(19): 3438-3456.e12, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37738977

ABSTRACT

Transcription factors (TFs) activate enhancers to drive cell-specific gene programs in response to signals, but our understanding of enhancer assembly during signaling events is incomplete. Here, we show that androgen receptor (AR) forms condensates through multivalent interactions mediated by its N-terminal intrinsically disordered region (IDR) to orchestrate enhancer assembly in response to androgen signaling. AR IDR can be substituted by IDRs from selective proteins for AR condensation capacity and its function on enhancers. Expansion of the poly(Q) track within AR IDR results in a higher AR condensation propensity as measured by multiple methods, including live-cell single-molecule microscopy. Either weakening or strengthening AR condensation propensity impairs its heterotypic multivalent interactions with other enhancer components and diminishes its transcriptional activity. Our work reveals the requirement of an optimal level of AR condensation in mediating enhancer assembly and suggests that alteration of the fine-tuned multivalent IDR-IDR interactions might underlie AR-related human pathologies.


Subject(s)
Enhancer Elements, Genetic , Transcription Factors , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Hormones , Signal Transduction
2.
Int J Mol Sci ; 21(22)2020 Nov 13.
Article in English | MEDLINE | ID: mdl-33202845

ABSTRACT

Autophagy is a conserved pathway that plays a key role in cell homeostasis in normal settings, as well as abnormal and stress conditions. Autophagy dysfunction is found in various neurodegenerative diseases, although it remains unclear whether autophagy impairment is a contributor or consequence of neurodegeneration. Axonal injury is an acute neuronal stress that triggers autophagic responses in an age-dependent manner. In this study, we investigate the injury-triggered autophagy response in a C. elegans model of tauopathy. We found that transgenic expression of pro-aggregant Tau, but not the anti-aggregant Tau, abolished axon injury-induced autophagy activation, resulting in a reduced axon regeneration capacity. Furthermore, axonal trafficking of autophagic vesicles were significantly reduced in the animals expressing pro-aggregant F3ΔK280 Tau, indicating that Tau aggregation impairs autophagy regulation. Importantly, the reduced number of total or trafficking autophagic vesicles in the tauopathy model was not restored by the autophagy activator rapamycin. Loss of PTL-1, the sole Tau homologue in C. elegans, also led to impaired injury-induced autophagy activation, but with an increased basal level of autophagic vesicles. Therefore, we have demonstrated that Tau aggregation as well as Tau depletion both lead to disruption of injury-induced autophagy responses, suggesting that aberrant protein aggregation or microtubule dysfunction can modulate autophagy regulation in neurons after injury.


Subject(s)
Animals, Genetically Modified , Autophagy , Axons , Caenorhabditis elegans Proteins , Caenorhabditis elegans , Microtubule-Associated Proteins , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Axons/metabolism , Axons/pathology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Disease Models, Animal , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , tau Proteins/genetics , tau Proteins/metabolism
3.
Infect Immun ; 87(11)2019 11.
Article in English | MEDLINE | ID: mdl-31451622

ABSTRACT

The Bacteroides fragilis enterotoxin (BFT), a virulence factor of enterotoxigenic B. fragilis (ETBF), interacts with intestinal epithelial cells and can provoke signals that induce mucosal inflammation. Although ß-catenin signaling is reported to be associated with inflammatory responses and BFT is known to cleave E-cadherin linked with ß-catenin, little is known about the ß-catenin-mediated regulation of inflammation in ETBF infection. This study was conducted to investigate the role of ß-catenin as a cellular signaling intermediate in the induction of proinflammatory responses to stimulation of intestinal epithelial cells with BFT. Expression of ß-catenin in intestinal epithelial cells was reduced relatively early after stimulation with BFT and then recovered to normal levels relatively late after stimulation. In contrast, phosphorylation of ß-catenin in BFT-exposed cells occurred at high levels early in stimulation and decreased as time passed. Concurrently, late after stimulation the nuclear levels of ß-catenin were relatively higher than those early after stimulation. Suppression of ß-catenin resulted in increased NF-κB activity and interleukin-8 (IL-8) expression in BFT-stimulated cells. However, suppression or enhancement of ß-catenin expression neither altered the phosphorylated IκB kinase α/ß complex nor activated activator protein 1 signals. Furthermore, inhibition of glycogen synthase kinase 3ß was associated with increased ß-catenin expression and attenuated NF-κB activity and IL-8 expression in BFT-exposed cells. These findings suggest the negative regulation of NF-κB-mediated inflammatory responses by ß-catenin in intestinal epithelial cells stimulated with BFT, resulting in attenuation of acute inflammation in ETBF infection.


Subject(s)
Bacterial Toxins/pharmacology , Epithelial Cells/drug effects , Gene Expression Regulation/drug effects , Inflammation/metabolism , Metalloendopeptidases/pharmacology , NF-kappa B/metabolism , beta Catenin/metabolism , HCT116 Cells , Humans , Intestinal Mucosa/cytology , NF-kappa B/genetics , Signal Transduction , beta Catenin/genetics
4.
Infect Immun ; 85(10)2017 10.
Article in English | MEDLINE | ID: mdl-28694294

ABSTRACT

Bacteroides fragilis enterotoxin (BFT), a virulence factor of enterotoxigenic B. fragilis (ETBF), plays an essential role in mucosal inflammation. Although autophagy contributes to the pathogenesis of diverse infectious diseases, little is known about autophagy in ETBF infection. This study was conducted to investigate the role of BFT in the autophagic process in endothelial cells (ECs). Stimulation of human umbilical vein ECs (HUVECs) with BFT increased light chain 3 protein II (LC3-II) conversion from LC3-I and protein expression of p62, Atg5, and Atg12. In addition, BFT-exposed ECs showed increased indices of autophagosomal fusion with lysosomes such as LC3-lysosome-associated protein 2 (LAMP2) colocalization and the percentage of red vesicles monitored by the expression of dual-tagged LC3B. BFT also upregulated expression of C/EBP homologous protein (CHOP), and inhibition of CHOP significantly increased indices of autophagosomal fusion with lysosomes. BFT activated an AP-1 transcription factor, in which suppression of AP-1 activity significantly downregulated CHOP and augmented autophagosomal fusion with lysosomes. Furthermore, suppression of Jun N-terminal protein kinase (JNK) mitogen-activated protein kinase (MAPK) significantly inhibited the AP-1 and CHOP signals, leading to an increase in autophagosomal fusion with lysosomes in BFT-stimulated ECs. These results suggest that BFT induced accumulation of autophagosomes in ECs, but activation of a signaling pathway involving JNK, AP-1, and CHOP may interfere with complete autophagy.


Subject(s)
Autophagosomes/physiology , Autophagy , Bacteroides fragilis/metabolism , Lysosomes/physiology , Metalloendopeptidases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Transcription Factor AP-1/metabolism , Transcription Factor CHOP/metabolism , Autophagosomes/microbiology , Bacteroides fragilis/pathogenicity , Cells, Cultured , Down-Regulation , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Endothelial Cells/ultrastructure , Humans , Lysosomes/microbiology , NF-kappa B/metabolism , Signal Transduction , Umbilical Veins/cytology , Up-Regulation
5.
Infect Immun ; 84(8): 2162-2174, 2016 08.
Article in English | MEDLINE | ID: mdl-27185786

ABSTRACT

Helicobacter pylori sheds outer membrane vesicles (OMVs) that contain many surface elements of bacteria. Dendritic cells (DCs) play a major role in directing the nature of adaptive immune responses against H. pylori, and heme oxygenase-1 (HO-1) has been implicated in regulating function of DCs. In addition, HO-1 is important for adaptive immunity and the stress response. Although H. pylori-derived OMVs may contribute to the pathogenesis of H. pylori infection, responses of DCs to OMVs have not been elucidated. In the present study, we investigated the role of H. pylori-derived crude OMVs in modulating the expression of HO-1 in DCs. Exposure of DCs to crude H. pylori OMVs upregulated HO-1 expression. Crude OMVs obtained from a cagA-negative isogenic mutant strain induced less HO-1 expression than OMVs obtained from a wild-type strain. Crude H. pylori OMVs activated signals of transcription factors such as NF-κB, AP-1, and Nrf2. Suppression of NF-κB or Nrf2 resulted in significant attenuation of crude OMV-induced HO-1 expression. Crude OMVs increased the phosphorylation of Akt and downstream target molecules of mammalian target of rapamycin (mTOR), such as S6 kinase 1 (S6K1). Suppression of Akt resulted in inhibition of crude OMV-induced Nrf2-dependent HO-1 expression. Furthermore, suppression of mTOR was associated with inhibition of IκB kinase (IKK), NF-κB, and HO-1 expression in crude OMV-exposed DCs. These results suggest that H. pylori-derived OMVs regulate HO-1 expression through two different pathways in DCs, Akt-Nrf2 and mTOR-IKK-NF-κB signaling. Following this induction, increased HO-1 expression in DCs may modulate inflammatory responses in H. pylori infection.


Subject(s)
Dendritic Cells/metabolism , Extracellular Vesicles/metabolism , Helicobacter pylori/metabolism , Heme Oxygenase-1/metabolism , Signal Transduction , Animals , Gene Expression , Heme Oxygenase-1/genetics , Humans , I-kappa B Kinase/metabolism , I-kappa B Proteins/metabolism , Mice , Mice, Knockout , NF-E2-Related Factor 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism
6.
Infect Immun ; 84(9): 2541-54, 2016 09.
Article in English | MEDLINE | ID: mdl-27324483

ABSTRACT

The Bacteroides fragilis enterotoxin (BFT), a virulence factor of enterotoxigenic B. fragilis (ETBF), interacts with intestinal epithelial cells and can provoke signals that induce mucosal inflammation. Although expression of heme oxygenase-1 (HO-1) is associated with regulation of inflammatory responses, little is known about HO-1 induction in ETBF infection. This study was conducted to investigate the effect of BFT on HO-1 expression in intestinal epithelial cells. Stimulation of intestinal epithelial cells with BFT resulted in upregulated expression of HO-1. BFT activated transcription factors such as NF-κB, AP-1, and Nrf2 in intestinal epithelial cells. Upregulation of HO-1 in intestinal epithelial cells was dependent on activated IκB kinase (IKK)-NF-κB signals. However, suppression of Nrf2 or AP-1 signals in intestinal epithelial cells did not result in significant attenuation of BFT-induced HO-1 expression. HO-1 induction via IKK-NF-κB in intestinal epithelial cells was regulated by p38 mitogen-activated protein kinases (MAPKs). Furthermore, suppression of HO-1 activity led to increased apoptosis in BFT-stimulated epithelial cells. These results suggest that a signaling pathway involving p38 MAPK-IKK-NF-κB in intestinal epithelial cells is required for HO-1 induction during exposure to BFT. Following this induction, increased HO-1 expression may regulate the apoptotic process in responses to BFT stimulation.


Subject(s)
Apoptosis/immunology , Bacteroides fragilis/immunology , Enterotoxins/immunology , Epithelial Cells/microbiology , Heme Oxygenase-1/metabolism , Intestinal Mucosa/microbiology , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Animals , Epithelial Cells/immunology , Epithelial Cells/metabolism , I-kappa B Proteins/metabolism , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/metabolism , Signal Transduction/immunology , Transcription Factor AP-1/metabolism , Transcriptional Activation/immunology , Up-Regulation/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
7.
J Gastroenterol Hepatol ; 30(1): 99-108, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25041690

ABSTRACT

BACKGROUND AND AIM: Dendritic cells (DCs) are observed on the Helicobacter pylori-infected gastric mucosa. DCs generally play an important role in the regulation of inflammation. Although stimulation of gastric epithelial cells with H. pylori vacuolating cytotoxin (VacA) has been reported to induce apoptosis and endoplasmic reticulum (ER) stress, the effects of VacA on the DC apoptotic response have not been well elucidated. This study was conducted to investigate the role of H. pylori VacA on the apoptotic process and ER stress in DCs. METHODS: Murine and human DCs were generated from specific pathogen-free C57BL/6 mice and human peripheral blood mononuclear cells, respectively. DCs were incubated with purified VacA, after which Bax activation, cytochrome c release, and DNA fragmentation for apoptosis were measured by fluorescent microscopy, immunoblot, and ELISA. ER stress-related molecules such as GRP78 and CHOP were analyzed by immunoblot. RESULTS: Treatment of DCs with purified H. pylori VacA resulted in the induction of apoptosis. DC stimulation with VacA led to the translocation of cytoplasmic Bax to mitochondria and cytochrome c release from mitochondria. H. pylori VacA induced signals for ER stress early during the stimulation process in DCs. Furthermore, suppression of ER stress resulted in a significant inhibition of the VacA-induced apoptosis in DCs. CONCLUSION: These results suggest that ER stress is critical for regulation of DC apoptotic process in response to VacA stimulation.


Subject(s)
Apoptosis/genetics , Bacterial Proteins/physiology , Dendritic Cells/pathology , Endoplasmic Reticulum Stress/genetics , Helicobacter pylori , Animals , Dendritic Cells/physiology , Endoplasmic Reticulum Chaperone BiP , Gastric Mucosa/cytology , Heat-Shock Proteins/metabolism , Humans , Mice, Inbred C57BL , Transcription Factor CHOP/metabolism
8.
Mediators Inflamm ; 2015: 301716, 2015.
Article in English | MEDLINE | ID: mdl-25821353

ABSTRACT

Eosinophil cationic protein (ECP), a cytotoxic protein contained in eosinophils granules, can contribute to various inflammatory responses. Although Helicobacter pylori infection increases infiltration of eosinophils, the mechanisms of eosinophil degranulation by H. pylori infection are largely unknown. The goal of this study was to investigate the role of H. pylori outer membrane vesicles (OMVs) in modulating eosinophil degranulation. We found that eosinophils treated with H. pylori OMVs released significantly more ECP compared with untreated controls. In addition, eosinophils cocultured with OMV-preexposed primary gastric epithelial cells exhibited significantly increased ECP release. Similarly, eosinophils cocultured with culture supernatant (CM) from primary gastric epithelial cells exposed to OMVs (OMV-CM) released significantly higher amounts of ECP compared with eosinophils cocultured with CM from unexposed control cells. Furthermore, OMVs and OMV-CM both induced the upregulation of ICAM-1 on gastric epithelial cells and ß2 integrin CD11b on eosinophils. In addition, both transduction of ICAM-1 shRNA into gastric epithelial cells and treatment with neutralizing mAbs to CD18 significantly decreased OMV-mediated or OMV-CM-mediated release of ECP. These results suggest that the eosinophil degranulation response to H. pylori OMVs occurs via a mechanism that is dependent on both ß2 integrin CD11/CD18 and ICAM-1.


Subject(s)
Bacterial Outer Membrane Proteins/physiology , CD18 Antigens/physiology , Cell Degranulation , Eosinophils/physiology , Helicobacter pylori/physiology , Intercellular Adhesion Molecule-1/physiology , CD11b Antigen/analysis , Eosinophil Cationic Protein/physiology , Gastric Mucosa/metabolism , Helicobacter Infections/etiology , Humans
9.
J Gastroenterol Hepatol ; 29(3): 502-10, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24716225

ABSTRACT

BACKGROUND AND AIM: The aim of this study is to evaluate the effect of metformin on intestinal inflammation. METHODS: COLO205 cells were pretreated with metformin and stimulated with tumor necrosis factor (TNF)-α. Expression of interleukin (IL)-8 was determined by luciferase assay and real-time PCR. Inhibitor of kappaB (IκB) phosphorylation/degradation and adenosine monohosphate-activated protein kinase (AMPK) activity were evaluated by Western blotting. DNA-binding activity of transcription factor nuclear factor-kappaB (NF-κB) was assessed by electrophoretic mobility shift assay. In an acute colitis model, mice were given 4% dextran sulfate sodium (DSS) for 5 days. IL-10−/− mice were used to evaluate the effect of metformin on chronic colitis. In an inflamation-associated tumor model, mice were given a single intraperitoneal injection of azoxymethane followed by three cycles of 2% DSS for 5 days and 2 weeks of free water consumption. RESULTS: Metformin significantly inhibited IL-8 induction in COLO 205 cells stimulated with TNF-α. Metformin attenuated IκBα phosphorylation and NF-κB DNA-binding activity. Administration of metformin significantly reduced the severity of DSS-induced colitis. In addition, DSS-induced IκB kinase (IKK) activation was significantly reduced in mice treated with metformin. Metformin significantly attenuated the severity of colitis in IL-10−/− mice, induced AMPK activity in intestinal epithelial cells, and inhibited the development of colitic cancer in mice. CONCLUSIONS: These results indicate that metformin suppresses NF-κB activation in intestinal epithelial cells and ameliorates murine colitis and colitis-associated tumorigenesis in mice, suggesting that metformin could be a potential therapeutic agent for the treatment of inflammatory bowel disease.


Subject(s)
Colitis/drug therapy , Colitis/genetics , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Metformin/pharmacology , Metformin/therapeutic use , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line, Tumor , Colitis/chemically induced , Colitis/complications , Colonic Neoplasms/etiology , Colonic Neoplasms/metabolism , Dextran Sulfate , Disease Models, Animal , Humans , I-kappa B Kinase/metabolism , Inflammation Mediators/metabolism , Interleukin-8/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy , NF-kappa B/genetics , NF-kappa B/metabolism , Severity of Illness Index , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/adverse effects
10.
Lab Invest ; 93(4): 384-96, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23381626

ABSTRACT

Enterotoxigenic Bacteroides fragilis (ETBF) produces an ≈ 20 kDa B. fragilis enterotoxin (BFT), which plays an essential role in mucosal inflammation. Lipocalin (Lcn)-2, a siderophore-binding antimicrobial protein, is critical for control of bacterial infection; however, expression of Lcn-2 in BFT-exposed intestinal epithelial cells has not been elucidated. In the present study, stimulation of human intestinal epithelial cells with BFT resulted in the upregulation of Lcn-2 expression that was a relatively late response of intestinal epithelial cells compared with human ß-defensin (hBD)-2 expression. The upregulation of Lcn-2 was dependent on AP-1 but not on NF-κB signaling. Lcn-2 induction via AP-1 was regulated by mitogen-activated protein kinases (MAPKs) including ERK and p38. Lcn-2 was secreted from the apical and basolateral surfaces in BFT-treated cells. These results suggest that a signaling pathway involving MAPKs and AP-1 is required for Lcn-2 induction in intestinal epithelial cells exposed to BFT, after which the secreted Lcn-2 may facilitate antimicrobial activity within ETBF-infected mucosa.


Subject(s)
Acute-Phase Proteins/metabolism , Bacterial Toxins/toxicity , Intestinal Mucosa/drug effects , Lipocalins/metabolism , Metalloendopeptidases/toxicity , Proto-Oncogene Proteins/metabolism , Acute-Phase Proteins/genetics , Epithelial Cells/metabolism , HT29 Cells , Humans , Intestinal Mucosa/metabolism , Lipocalin-2 , Lipocalins/genetics , MAP Kinase Signaling System , NF-kappa B/metabolism , Proto-Oncogene Proteins/genetics , Transcription Factor AP-1/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL