Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 13(1): 874, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35169117

ABSTRACT

IL-18 is emerging as an IL-22-induced and epithelium-derived cytokine which contributes to host defence against intestinal infection and inflammation. In contrast to its known role in Goblet cells, regulation of barrier function at the molecular level by IL-18 is much less explored. Here we show that IL-18 is a bona fide IL-22-regulated gate keeper for intestinal epithelial barrier. IL-22 promotes crypt immunity both via induction of phospho-Stat3 binding to the Il-18 gene promoter and via Il-18 independent mechanisms. In organoid culture, while IL-22 primarily increases organoid size and inhibits expression of stem cell genes, IL-18 preferentially promotes organoid budding and induces signature genes of Lgr5+ stem cells via Akt-Tcf4 signalling. During adherent-invasive E. coli (AIEC) infection, systemic administration of IL-18 corrects compromised T-cell IFNγ production and restores Lysozyme+ Paneth cells in Il-22-/- mice, but IL-22 administration fails to restore these parameters in Il-18-/- mice, thereby placing IL-22-Stat3 signalling upstream of the IL-18-mediated barrier defence function. IL-18 in return regulates Stat3-mediated anti-microbial response in Paneth cells, Akt-Tcf4-triggered expansion of Lgr5+ stem cells to facilitate tissue repair, and AIEC clearance by promoting IFNγ+ T cells.


Subject(s)
Escherichia coli Infections/immunology , Immunity, Mucosal/immunology , Interleukin-18/immunology , Interleukins/immunology , Intestinal Mucosa/immunology , Animals , Crohn Disease/microbiology , Crohn Disease/pathology , Dysbiosis/microbiology , Escherichia coli/immunology , Interferon-gamma/immunology , Interleukin-18/genetics , Intestinal Mucosa/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Muramidase/metabolism , Organoids , Paneth Cells/immunology , Promoter Regions, Genetic/genetics , STAT3 Transcription Factor/metabolism , Tight Junctions/immunology , Interleukin-22
2.
Nat Commun ; 11(1): 4286, 2020 08 27.
Article in English | MEDLINE | ID: mdl-32855403

ABSTRACT

Intracellular galectins are carbohydrate-binding proteins capable of sensing and repairing damaged lysosomes. As in the physiological conditions glycosylated moieties are mostly in the lysosomal lumen but not cytosol, it is unclear whether galectins reside in lysosomes, bind to glycosylated proteins, and regulate lysosome functions. Here, we show in gut epithelial cells, galectin-9 is enriched in lysosomes and predominantly binds to lysosome-associated membrane protein 2 (Lamp2) in a Asn(N)-glycan dependent manner. At the steady state, galectin-9 binding to glycosylated Asn175 of Lamp2 is essential for functionality of lysosomes and autophagy. Loss of N-glycan-binding capability of galectin-9 causes its complete depletion from lysosomes and defective autophagy, leading to increased endoplasmic reticulum (ER) stress preferentially in autophagy-active Paneth cells and acinar cells. Unresolved ER stress consequently causes cell degeneration or apoptosis that associates with colitis and pancreatic disorders in mice. Therefore, lysosomal galectins maintain homeostatic function of lysosomes to prevent organ pathogenesis.


Subject(s)
Galectins/metabolism , Lysosomal-Associated Membrane Protein 2/metabolism , Lysosomes/metabolism , Pancreas/pathology , Paneth Cells/pathology , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Autophagy/physiology , Colitis/metabolism , Colitis/pathology , Endoplasmic Reticulum Stress , Galectins/genetics , HT29 Cells , Humans , Lysosomal-Associated Membrane Protein 2/genetics , Lysosomes/genetics , Lysosomes/pathology , Mice, Inbred C57BL , Mice, Knockout , Pancreas/metabolism , Pancreatitis/metabolism , Pancreatitis/pathology , Paneth Cells/metabolism
3.
Front Immunol ; 9: 2522, 2018.
Article in English | MEDLINE | ID: mdl-30455690

ABSTRACT

In gut epithelium, IL-22 transmits signals through STAT3 phosphorylation (pSTAT3) which provides intestinal immunity. Many components in the IL-22-pSTAT3 pathway have been identified as risk factors for inflammatory bowel disease (IBD) and some of them are considered as promising therapeutic targets. However, new perspectives are still needed to understand IL-22-pSTAT3 signaling for effective clinical interventions in IBD patients. Here, we revealed activating transcription factor 3 (ATF3), recently identified to be upregulated in patients with active IBD, as a crucial player in the epithelial IL-22-pSTAT3 signaling cascade. We found ATF3 is central to intestinal homeostasis and provides protection during colitis. Loss of ATF3 led to decreased crypt numbers, more shortened colon length, impaired ileal fucosylation at the steady state, and lethal disease activity during DSS-induced colitis which can be effectively ameliorated by rectal transplantation of wild-type colonic organoids. Epithelial stem cells and Paneth cells form a niche to orchestrate epithelial regeneration and host-microbe interactions, and IL-22-pSTAT3 signaling is a key guardian for this niche. We found ATF3 is critical for niche maintenance as ATF3 deficiency caused compromised stem cell growth and regeneration, as well as Paneth cell degeneration and loss of anti-microbial peptide (AMP)-producing granules, indicative of malfunction of Paneth/stem cell network. Mechanistically, we found IL-22 upregulates ATF3, which is required to relay IL-22 signaling leading to STAT3 phosphorylation and subsequent AMP induction. Intriguingly, ATF3 itself does not act on STAT3 directly, instead ATF3 regulates pSTAT3 by negatively targeting protein tyrosine phosphatases (PTPs) including SHP2 and PTP-Meg2. Furthermore, we identified ATF3 is also involved in IL-6-mediated STAT3 activation in T cells and loss of ATF3 leads to reduced capacity of Th17 cells to produce their signature cytokine IL-22 and IL-17A. Collectively, our results suggest that via IL-22-pSTAT3 signaling in the epithelium and IL-6-pSTAT3 signaling in Th17 cells, ATF3 mediates a cross-regulation in the barrier to maintain mucosal homeostasis and immunity.


Subject(s)
Activating Transcription Factor 3/metabolism , Colitis/pathology , Interleukin-6/metabolism , Interleukins/metabolism , STAT3 Transcription Factor/metabolism , Activating Transcription Factor 3/genetics , Animals , Cell Line, Tumor , Colitis/chemically induced , Colitis/immunology , Colon/pathology , Epithelial Cells/metabolism , Homeostasis/immunology , Ileum/pathology , Immunity, Mucosal/immunology , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Paneth Cells/immunology , Paneth Cells/metabolism , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction/immunology , Stem Cells/metabolism , Th17 Cells/immunology , Interleukin-22
4.
Biol Cell ; 106(10): 359-76, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25055739

ABSTRACT

BACKGROUND INFORMATION: During the initiation of cell death, mitochondrial protein, apoptosis-inducing factor (AIF), is transported to the nucleus. The mechanism of AIF nuclear translocation, however, is not clear. After protein synthesis, the AIF is originally targetted to the mitochondria, and the nuclear targetting is a secondary event. Therefore, we hypothesised that the nuclear translocation of AIF could be achieved by a novel pathway. RESULTS: By using yeast two-hybrid assay, we identified the human UV excision repair protein RAD23 homolog A (hHR23A) interacts with AIF and their interaction was confirmed by co-immunoprecipitation and fluorescence resonance energy transfer microscopy. Silencing the RAD23A gene expression inhibits the nuclear transportation of AIF and increases cisplatin resistance. Silencing the karyopherin alpha 2 (KPNA2) gene expression, however, did not affect the nuclear import of AIF. Moreover, 2,4-dinitrophenol inhibits staurosporine-induced nuclear translocation of AIF and increases cisplatin resistance. CONCLUSIONS: These results suggest that hHR23A is required for the nuclear translocation of AIF during induction of cell death, and this process is energy dependent, but independent of karyopherins.


Subject(s)
Active Transport, Cell Nucleus , Apoptosis Inducing Factor/metabolism , Cell Death , Cell Nucleus/metabolism , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Mitochondria/metabolism , Humans , Two-Hybrid System Techniques
5.
Neuro Oncol ; 15(10): 1342-52, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24057885

ABSTRACT

BACKGROUND: ATPase-family, AAA domain containing 3A (ATAD3A) is located on human chromosome 1p36.33, and high endogenous expression may associate with radio- and chemosensitivity. This study was conducted to investigate the significance of ATAD3A in glioblastoma multiforme (GBM). METHODS: Clinical significance of ATAD3A expression was assessed by immunohistochemistry in 67 GBM specimens, and prognostic value was assessed in 32 GBM patients statistically. To investigate in vitro phenotypic effects of ATAD3A, cell viability was measured using a clonogenic survival assay under either knockdown or ectopic expression of ATAD3A in GBM cell lines. The effects of ATAD3A knockdown on targeted DNA repair-associated proteins in T98G cells were evaluated using immunofluorescence and Western blotting. RESULTS: Clinically, high expression of ATAD3A was independent of O(6)-DNA methylguanine-methyltransferase methylation status and correlated with worse prognosis. In vitro, high ATAD3A-expressing T98G cells were more resistant to radiation-induced cell death compared with control and low endogenous ATAD3A U87MG cells. After silencing ATAD3A, T98G cells became more sensitive to radiation. On the other hand, enforced ATAD3A expression in U87MG cells exhibited increased radioresistance. ATAD3A may coordinate with aldo-keto reductase genes and participate in bioactivation or detoxication of temozolomide. Surprisingly, deficient DNA repair after irradiation was observed in T98G/ATAD3A knockdown as a result of decreased nuclear ataxia telangiectasia mutated kinase and histones H2AX and H3, which was also evidenced by the sustained elevation of poly (ADP-ribose) polymerase prior to and after radiation treatment. CONCLUSION: Our data suggest that high expression of ATAD3A is an independent biomarker for radioresistance in GBM. ATAD3A could be a potential target for therapy.


Subject(s)
Adenosine Triphosphatases/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Gene Expression Regulation, Neoplastic/radiation effects , Glioblastoma/metabolism , Glioblastoma/pathology , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Radiation Tolerance , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/genetics , Antineoplastic Agents, Alkylating/therapeutic use , Blotting, Western , Brain Neoplasms/radiotherapy , Cell Differentiation , Cell Proliferation , Chemoradiotherapy , DNA Methylation , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Female , Glioblastoma/radiotherapy , Humans , Immunoenzyme Techniques , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Middle Aged , Mitochondria/metabolism , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Neoplasm Staging , O(6)-Methylguanine-DNA Methyltransferase/genetics , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Prognosis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Temozolomide , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...