Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Semin Cell Dev Biol ; 101: 41-50, 2020 05.
Article in English | MEDLINE | ID: mdl-31408699

ABSTRACT

Autophagy is a conserved intracellular degradation process enclosing the bulk of cytosolic components for lysosomal degradation to maintain cellular homeostasis. Accumulating evidences showed that a specialized form of autophagy, known as xenophagy, could serve as an innate immune response to defend against pathogens invading inside the host cells. Correspondingly, infectious pathogens have developed a variety of strategies to disarm xenophagy, leading to a prolonged and persistent intracellular colonization. In this review, we first summarize the current knowledge about the general mechanisms of intracellular bacterial infections and xenophagy. We then focus on the ongoing battle between these two processes.


Subject(s)
Autophagy/immunology , Bacterial Infections/immunology , Animals , Bacterial Infections/pathology , Humans , Immunity, Innate/immunology
2.
J Pathol ; 244(4): 432-444, 2018 04.
Article in English | MEDLINE | ID: mdl-29327342

ABSTRACT

Evasion of autophagy is key for intracellular survival of bacteria in host cells, but its involvement in persistent infection by Helicobacter pylori, a bacterium identified to invade gastric epithelial cells, remains obscure. The aim of this study was to functionally characterize the role of autophagy in H. pylori infection. Autophagy was assayed in H. pylori-infected human gastric epithelium and the functional role of autophagy was determined via genetic or pharmacological ablation of autophagy in mouse and cell line models of H. pylori infection. Here, we showed that H. pylori inhibited lysosomal function and thereby promoted the accumulation of autophagosomes in gastric epithelial cells. Importantly, inhibiting autophagosome formation by pharmacological inhibitors or genetic ablation of BECN1 or ATG5 reduced H. pylori intracellular survival, whereas inhibition of lysosomal functions exerted an opposite effect. Further experiments demonstrated that H. pylori inhibited lysosomal acidification and the retrograde trafficking of mannose-6-phosphate receptors, both of which are known to positively regulate lysosomal function. We conclude that H. pylori subverts autophagy into a pro-survival mechanism through inhibition of lysosomal clearance of autophagosomes. Disruption of autophagosome formation offers a novel strategy to reduce H. pylori colonization in human stomachs. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Autophagosomes/microbiology , Autophagy , Gastric Mucosa/microbiology , Helicobacter Infections/microbiology , Helicobacter pylori/growth & development , Lysosomes/microbiology , Animals , Autophagosomes/pathology , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Beclin-1/genetics , Beclin-1/metabolism , Case-Control Studies , Cell Line , Gastric Mucosa/pathology , Helicobacter Infections/genetics , Helicobacter Infections/pathology , Host-Pathogen Interactions , Humans , Hydrogen-Ion Concentration , Lysosomes/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbial Viability , Protein Transport , Receptor, IGF Type 2/metabolism
3.
J Immunol ; 196(4): 1799-809, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26800870

ABSTRACT

The antimicrobial peptide cathelicidin is critical for protection against different kinds of microbial infection. This study sought to elucidate the protective action of cathelicidin against Helicobacter pylori infection and its associated gastritis. Exogenous cathelicidin was found to inhibit H. pylori growth, destroy the bacteria biofilm, and induce morphological alterations in H. pylori membrane. Additionally, knockdown of endogenous cathelicidin in human gastric epithelial HFE-145 cells markedly increased the intracellular survival of H. pylori. Consistently, cathelicidin knockout mice exhibited stronger H. pylori colonization, higher expression of proinflammatory cytokines IL-6, IL-1ß, and ICAM1, and lower expression of the anti-inflammatory cytokine IL-10 in the gastric mucosa upon H. pylori infection. In wild-type mice, H. pylori infection also stimulated gastric epithelium-derived cathelicidin production. Importantly, pretreatment with bioengineered Lactococcus lactis that actively secretes cathelicidin significantly increased mucosal cathelicidin levels and reduced H. pylori infection and the associated inflammation. Moreover, cathelicidin strengthened the barrier function of gastric mucosa by stimulating mucus synthesis. Collectively, these findings indicate that cathelicidin plays a significant role as a potential natural antibiotic for H. pylori clearance and a therapeutic agent for chronic gastritis.


Subject(s)
Cathelicidins/immunology , Gastric Mucosa/immunology , Gastritis/immunology , Helicobacter Infections/immunology , Animals , Antimicrobial Cationic Peptides , Cell Line , Disease Models, Animal , Fluorescent Antibody Technique , Gastric Mucosa/microbiology , Helicobacter pylori/immunology , Humans , Mice , Mice, Knockout , Microscopy, Electron, Scanning , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Transfection
4.
J Gastroenterol Hepatol ; 32(3): 609-619, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27470075

ABSTRACT

BACKGROUND AND AIM: The preventive effect of intrarectal administration of mouse cathelicidin (mCRAMP) and oral administration of mCRAMP-encoding Lactococcus lactis (N4I) has been shown in murine experimental colitis. It is pivotal to understand the ability of N4I whether it can promote mucosal repair in existing colitis. METHODS: Mice with dextran sulfate sodium-induced ulcerative colitis (UC) were treated orally with L. lactis or its transformed strain with or without nisin induction. The body weight, clinical symptoms, and histological changes of colonic tissues were determined. Sulfasalazine was used as a reference drug. Young adult mouse colon cells were used to further elucidate the direct action and possible mechanisms of mCRAMP to promote colonic wound repair. RESULTS: Results showed that N4I could improve the clinical symptoms, maintain crypt integrity and preserve mucus-secreting layer in colitis animals. The preparation also could prevent cell death and promote cell proliferation. In contrast, effective dose of sulfasalazine only alleviated clinical symptoms but not the mucosal damage and repair in the colon. In vitro study further showed that mCRAMP could directly promote wound repair by accelerating cell migration but not cell proliferation through the GPCR/MAPK pathway. CONCLUSIONS: mCRAMP-encoding L. lactis could be a potential therapeutic preparation better than the traditional anti-inflammatory agent in the treatment of UC.


Subject(s)
Antimicrobial Cationic Peptides/administration & dosage , Antimicrobial Cationic Peptides/pharmacology , Colitis, Ulcerative/drug therapy , Intestinal Mucosa/physiology , Lactococcus lactis , Wound Healing/drug effects , Administration, Oral , Administration, Rectal , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Colitis, Ulcerative/pathology , Colitis, Ulcerative/physiopathology , Colitis, Ulcerative/therapy , Colon/cytology , Epithelial Cells , Intestinal Mucosa/pathology , Male , Mice, Inbred BALB C , Cathelicidins
SELECTION OF CITATIONS
SEARCH DETAIL