Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 669, 2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38253620

ABSTRACT

The role of N6-methyladenosine (m6A) modification of host mRNA during bacterial infection is unclear. Here, we show that Helicobacter pylori infection upregulates host m6A methylases and increases m6A levels in gastric epithelial cells. Reducing m6A methylase activity via hemizygotic deletion of methylase-encoding gene Mettl3 in mice, or via small interfering RNAs targeting m6A methylases, enhances H. pylori colonization. We identify LOX-1 mRNA as a key m6A-regulated target during H. pylori infection. m6A modification destabilizes LOX-1 mRNA and reduces LOX-1 protein levels. LOX-1 acts as a membrane receptor for H. pylori catalase and contributes to bacterial adhesion. Pharmacological inhibition of LOX-1, or genetic ablation of Lox-1, reduces H. pylori colonization. Moreover, deletion of the bacterial catalase gene decreases adhesion of H. pylori to human gastric sections. Our results indicate that m6A modification of host LOX-1 mRNA contributes to protection against H. pylori infection by downregulating LOX-1 and thus reducing H. pylori adhesion.


Subject(s)
Adenosine , Helicobacter Infections , Helicobacter pylori , Scavenger Receptors, Class E , Animals , Humans , Mice , Adenosine/analogs & derivatives , Catalase/metabolism , Helicobacter Infections/metabolism , Helicobacter pylori/metabolism , RNA, Messenger/genetics , Scavenger Receptors, Class E/genetics
2.
Crit Care ; 24(1): 47, 2020 02 10.
Article in English | MEDLINE | ID: mdl-32041659

ABSTRACT

OBJECTIVES: The intestinal epithelium compartmentalizes the sterile bloodstream and the commensal bacteria in the gut. Accumulating evidence suggests that this barrier is impaired in sepsis, aggravating systemic inflammation. Previous studies reported that cathelicidin is differentially expressed in various tissues in sepsis. However, its role in sepsis-induced intestinal barrier dysfunction has not been investigated. DESIGN: To examine the role of cathelicidin in polymicrobial sepsis, cathelicidin wild-(Cnlp+/+) and knockout (Cnlp-/-) mice underwent cecal-ligation and puncture (CLP) followed by the assessment of septic mortality and morbidity as well as histological, biochemical, immunological, and transcriptomic analyses in the ileal tissues. We also evaluated the prophylactic and therapeutic efficacies of vitamin D3 (an inducer of endogenous cathelicidin) in the CLP-induced murine polymicrobial sepsis model. RESULTS: The ileal expression of cathelicidin was increased by three-fold after CLP, peaking at 4 h. Knockout of Cnlp significantly increased 7-day mortality and was associated with a higher murine sepsis score. Alcian-blue staining revealed a reduced number of mucin-positive goblet cells, accompanied by reduced mucin expression. Increased number of apoptotic cells and cleavage of caspase-3 were observed. Cnlp deletion increased intestinal permeability to 4kD fluorescein-labeled dextran and reduced the expression of tight junction proteins claudin-1 and occludin. Notably, circulating bacterial DNA load increased more than two-fold. Transcriptome analysis revealed upregulation of cytokine/inflammatory pathway. Depletion of Cnlp induced more M1 macrophages and neutrophils compared with the wild-type mice after CLP. Mice pre-treated with cholecalciferol (an inactive form of vitamin D3) or treated with 1alpha, 25-dihydroxyvitamin D3 (an active form of VD3) had decreased 7-day mortality and significantly less severe symptoms. Intriguingly, the administration of cholecalciferol after CLP led to worsened 7-day mortality and the associated symptoms. CONCLUSIONS: Endogenous cathelicidin promotes intestinal barrier integrity accompanied by modulating the infiltration of neutrophils and macrophages in polymicrobial sepsis. Our data suggested that 1alpha, 25-dihydroxyvitamin D3 but not cholecalciferol is a potential therapeutic agent for treating sepsis.


Subject(s)
Antimicrobial Cationic Peptides , Intestinal Mucosa , Sepsis , Animals , Antimicrobial Cationic Peptides/physiology , Intestinal Mucosa/metabolism , Macrophages , Male , Mice , Mice, Knockout , Neutrophils , Sepsis/physiopathology , Vitamin D/analogs & derivatives , Vitamin D/pharmacology , Cathelicidins
3.
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
4.
Cell Prolif ; 51(4): e12441, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29484738

ABSTRACT

Tumour necrosis factor alpha (TNFα) and interferon gamma (IFNγ) were originally found to be produced by inflammatory cells and play important roles in the immune system and surveillance of tumour growth. By activating distinct signalling pathways of nuclear factor-κB (NF-κB), mitogen-activated protein kinase (MAPK), and JAK/STAT, TNFα and IFNγ were reported to effectively trigger cell death and perform powerful anti-cancer effects. In this review, we will discuss the new advancements of TNFα and IFNγ in anti-cancer therapy.


Subject(s)
Interferon-gamma/therapeutic use , Neoplasms/drug therapy , Tumor Necrosis Factor-alpha/therapeutic use , Clinical Trials as Topic , Humans , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/metabolism
5.
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
6.
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
7.
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
9.
J Cell Physiol ; 230(10): 2382-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25708728

ABSTRACT

Hepatitis B virus (HBV) and one of its encoded proteins, HBV X protein (HBx), have been shown to induce autophagy in hepatoma cells. Substantial evidence indicates that autophagy is a potent suppressor of inflammation. However, sporadic reports suggest that autophagy could promote pro-inflammatory cytokine expression and inflammation in some biological contexts. Here, we show that overexpression of HBx induces LC3B-positive autophagosome formation, increases autophagic flux and enhances the expression of ATG5, ATG7, and LC3B-II in normal hepatocytes. Abrogation of autophagy by small interfering RNA against ATG5 and ATG7 prevents HBx-induced formation of autophagosomes. Autophagy inhibition also abrogates HBx-induced activation of nuclear factor-κB (NF-κB) and production of interleukin-6 (IL-6), IL-8, and CXCL2. These findings suggest that autophagy is required for HBx-induced NF-κB activation and pro-inflammatory cytokine production and could shed new light on the complex role of autophagy in the modulation of inflammation.


Subject(s)
Autophagy/physiology , Chemokine CXCL2/metabolism , Hepatitis B virus/isolation & purification , Hepatocytes/metabolism , Interleukin-6/biosynthesis , Interleukin-8/biosynthesis , NF-kappa B/metabolism , Cell Line , Gene Expression Regulation/physiology , Humans , Liver Neoplasms/metabolism , Signal Transduction/physiology , Trans-Activators/metabolism , Viral Regulatory and Accessory Proteins
10.
Cancer Res ; 75(4): 754-65, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25503559

ABSTRACT

Chromatin remodeling has emerged as a hallmark of gastric cancer, but the regulation of chromatin regulators other than genetic change is unknown. Helicobacter pylori causes epigenetic dysregulation to promote gastric carcinogenesis, but the roles and functions of microRNAs (miRNA) in this multistage cascade are not fully explored. In this study, miRNA expression in preneoplastic and neoplastic lesions in murine stomachs induced by H. pylori and N-methyl-N-nitrosourea (MNU) was profiled by miRNA expression array. miR-490-3p exhibited progressive downregulation in gastritis, intestinal metaplasia, and adenocarcinoma during H. pylori and MNU-induced gastric carcinogenesis. Significant downregulation of miR-490-3p was confirmed in human gastric cancer tissues in which its regulatory region was found to be hypermethylated. miR-490-3p exerted growth- and metastasis-suppressive effects on gastric cancer cells through directly targeting SMARCD1, a SWItch/Sucrose NonFermentable (SWI/SNF) chromatin remodeling complex subunit. Knockdown of SMARCD1 significantly attenuated the protumorigenic effects of miR-490-3p inhibitor, whereas enforced expression of SMARCD1 promoted in vitro and in vivo oncogenic phenotypes of gastric cancer cells. SMARCD1 was markedly upregulated in gastric cancer in which its high expression was associated with shortened patients' survival independent of TNM staging. In conclusion, hypermethylation-mediated silencing of miR-490-3p reactivates SMARCD1 to confer malignant phenotypes, mechanistically linking H. pylori, chromatin remodeling, and gastric carcinogenesis.


Subject(s)
Adenocarcinoma/genetics , Carcinogenesis , MicroRNAs/genetics , Stomach Neoplasms/genetics , Adenocarcinoma/chemically induced , Adenocarcinoma/pathology , Animals , Cell Line, Tumor , Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Gene Silencing , Helicobacter pylori/pathogenicity , Humans , Metaplasia/chemically induced , Methylnitrosourea/toxicity , Mice , Stomach Neoplasms/chemically induced , Stomach Neoplasms/pathology
11.
J Pathol ; 233(2): 103-12, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24633785

ABSTRACT

Helicobacter pylori and Epstein-Barr virus (EBV) account for roughly 80% and 10%, respectively, of gastric carcinomas worldwide. Autophagy is an evolutionarily conserved and intricately regulated cellular process that involves the sequestration of cytoplasmic proteins and organelles into double-membrane autophagosomes that eventually fuse with lysosomes for degradation of the engulfed content. Emerging evidence indicates that xenophagy, a form of selective autophagy, plays a crucial role in the pathogenesis of H. pylori- and EBV-induced gastric cancer. Xenophagy specifically recognizes intracellular H. pylori and EBV and physically targets these pathogens to the autophagosomal-lysosomal pathway for degradation. In this connection, H. pylori or EBV-induced dysregulation of autophagy may be causally linked to gastric tumourigenesis and therefore can be exploited as therapeutic targets. This review will discuss how H. pylori and EBV infection activate autophagy and how these pathogens evade recognition and degradation by the autophagic pathway. Elucidating the molecular aspects of H. pylori- and EBV-induced autophagy will help us better understand the pathogenesis of gastric cancer and promote the development of autophagy modulators as antimicrobial agents.


Subject(s)
Autophagy , Cell Transformation, Neoplastic/pathology , Epstein-Barr Virus Infections/virology , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Herpesvirus 4, Human/pathogenicity , Stomach Neoplasms/pathology , Animals , Cell Transformation, Viral , Epstein-Barr Virus Infections/complications , Helicobacter Infections/complications , Helicobacter pylori/growth & development , Helicobacter pylori/metabolism , Herpesvirus 4, Human/growth & development , Herpesvirus 4, Human/metabolism , Humans , Stomach Neoplasms/microbiology , Stomach Neoplasms/virology , Virulence Factors/metabolism , Virus Replication
12.
Autophagy ; 9(10): 1500-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23934082

ABSTRACT

Eukaryotes have two major intracellular protein degradation pathways, namely the ubiquitin-proteasome system (UPS) and autophagy. Inhibition of proteasomal activities has been previously shown to induce autophagy, indicating a coordinated and complementary relationship between these two systems. However, little is known about the regulation of the UPS by autophagy. In this study, we showed for the first time that proteasomes were activated in response to pharmacological inhibition of autophagy as well as disruption of autophagy-related genes by RNA interference under nutrient-deficient conditions in cultured human colon cancer cells. The induction was evidenced by the increased proteasomal activities and the upregulation of proteasomal subunits, including the proteasome ß5 subunit, PSMB5. Co-inhibition of the proteasome and autophagy also synergistically increased the accumulation of polyubiquitinated proteins. Collectively, our findings suggest that proteasomes are activated in a compensatory manner for protein degradation upon autophagy inhibition. Our studies unveiled a novel regulatory mechanism between the two protein degradation pathways.


Subject(s)
Autophagy/physiology , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Autophagy/genetics , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Proteasome Inhibitors/metabolism , RNA Interference , Signal Transduction/physiology , Ubiquitin/metabolism
13.
PLoS One ; 8(5): e63641, 2013.
Article in English | MEDLINE | ID: mdl-23700428

ABSTRACT

Host immune peptides, including cathelicidins, have been reported to possess anticancer properties. We previously reported that LL-37, the only cathelicidin in humans, suppresses the development of colon cancer. In this study, the potential anticancer effect of FK-16, a fragment of LL-37 corresponding to residues 17 to 32, on cultured colon cancer cells was evaluated. FK-16 induced a unique pattern of cell death, marked by concurrent activation of caspase-independent apoptosis and autophagy. The former was mediated by the nuclear translocation of AIF and EndoG whereas the latter was characterized by enhanced expression of LC3-I/II, Atg5 and Atg7 and increased formation of LC3-positive autophagosomes. Knockdown of Atg5 or Atg7 attenuated the cytotoxicity of FK-16, indicating FK-16-induced autophagy was pro-death in nature. Mechanistically, FK-16 activated nuclear p53 to upregulate Bax and downregulate Bcl-2. Knockdown of p53, genetic ablation of Bax, or overexpression of Bcl-2 reversed FK-16-induced apoptosis and autophagy. Importantly, abolition of AIF/EndoG-dependent apoptosis enhanced FK-16-induced autophagy while abolition of autophagy augmented FK-16-induced AIF-/EndoG-dependent apoptosis. Collectively, FK-16 induces caspase-independent apoptosis and autophagy through the common p53-Bcl-2/Bax cascade in colon cancer cells. Our study also uncovered previously unknown reciprocal regulation between these two cell death pathways.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Caspases/metabolism , Cathelicidins/pharmacology , Peptide Fragments/pharmacology , Apoptosis Inducing Factor/metabolism , Colonic Neoplasms , Drug Screening Assays, Antitumor , Endodeoxyribonucleases/metabolism , Enzyme Activation , HCT116 Cells , Humans , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism
14.
Crit Rev Oncol Hematol ; 86(3): 251-77, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23287077

ABSTRACT

Multiple intracellular signaling pathways, such as Wnt/ß-catenin signaling, epidermal growth factor receptor/Ras signaling, and p53 signaling are frequently dysregulated in colorectal cancer. Recent evidence also points to the involvement of signaling pathways in the developmental process, including Notch signaling, Hedgehog signaling, and Hippo signaling. Dysregulation of these signaling pathways contribute to the acquisition of malignant phenotypes, including unchecked cell cycle progression, evasion of apoptosis, induction of genetic instability, and enhanced invasiveness and metastasis. Understanding their relative importance and crosstalk will provide a rational basis for anticancer drug development.


Subject(s)
Carcinogenesis/metabolism , Colon/metabolism , Signal Transduction , Carcinogenesis/genetics , Colon/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Humans
15.
Curr Pharm Des ; 19(1): 5-10, 2013.
Article in English | MEDLINE | ID: mdl-22950507

ABSTRACT

Cigarette smoke has always been the single most preventive cause of death in the world. In 2011, over 460,000 died from cigarette smoke-related diseases in US. The detrimental effects of cigarette smoke on human beings are due to the presence of many carcinogens and other components (e.g. nicotine and tar). Nicotine is now accepted as one of the major components responsible for gastrointestinal disorders. Cigarette smoking, nicotine and a nicotine-derived nitrosamine, 4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) are considered as risk factors for gastrointestinal cancer, however, the underlying mechanism remains largely unknown. Previous studies reported that cigarette smoke and nicotine aggravated inflammation not only in the stomach, but also in the colon. The carcinogenic actions of cigarette smoke, nicotine and NNK on gastrointestinal cancers development have been widely studied. The strong association of cyclooxygenase-2 (COX-2) with gastrointestinal diseases has been extensively studied, however, due to the unresolved cardiovascular risk, it is of great importance to develop other new anti-cancer drugs for the treatment of cancers. This current review aims to provide an overview of the effects of cigarette smoke, nicotine and NNK on gastrointestinal inflammation, and also the carcinogenic properties in cancer development (tumor growth, angiogenesis and epithelial-mesenchymal transition). In addition, current studies on nicotinic acetylcholine receptors, adrenergic receptors and miRNAs in nicotine-related cancer pathogenesis are also highlighted.


Subject(s)
Gastrointestinal Diseases/etiology , Nicotine/adverse effects , Smoking/adverse effects , Animals , Cyclooxygenase 2/metabolism , Gastrointestinal Diseases/pathology , Gastrointestinal Neoplasms/etiology , Gastrointestinal Neoplasms/pathology , Humans , Inflammation/etiology , Inflammation/pathology , Nitrosamines/adverse effects , Peptic Ulcer/etiology , Peptic Ulcer/pathology , Risk Factors , Smoking/epidemiology , Stomach Ulcer/etiology , Stomach Ulcer/pathology , United States/epidemiology
16.
Curr Pharm Des ; 19(7): 1301-10, 2013.
Article in English | MEDLINE | ID: mdl-23092341

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression post-transcriptionally. They have been found to be dysregulated in many pathological conditions including cancer and play an important role during the progression of such disease. Recent efforts have been directed in translating the primary findings of miRNAs into clinical uses. This article gives a general overview on the potential of miRNAs as diagnostic and prognostic markers and also as therapeutic targets for gastrointestinal and liver cancers in animals and humans. Other contributors in this special series would focus in discussing the roles of specific miRNAs and their pathogenic mechanisms and therapeutic applications in different types of cancer in the gastrointestinal tract and liver.


Subject(s)
Gastrointestinal Neoplasms/genetics , Liver Neoplasms/genetics , MicroRNAs/physiology , Biomarkers, Tumor/genetics , Humans , MicroRNAs/genetics , Oncogenes , Prognosis
17.
Cancer Res ; 72(24): 6512-23, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23100468

ABSTRACT

Cathelicidins are a family of bacteriocidal polypeptides secreted by macrophages and polymorphonuclear leukocytes (PMN). LL-37, the only human cathelicidin, has been implicated in tumorigenesis, but there has been limited investigation of its expression and function in cancer. Here, we report that LL-37 activates a p53-mediated, caspase-independent apoptotic cascade that contributes to suppression of colon cancer. LL-37 was expressed strongly in normal colon mucosa but downregulated in colon cancer tissues, where in both settings its expression correlated with terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling-positive apoptotic cells. Exposure of colon cancer cells to LL-37 induced phosphatidylserine externalization and DNA fragmentation in a manner independent of caspase activation. Apoptogenic function was mediated by nuclear translocation of the proapoptotic factors, apoptosis-inducing factor (AIF) and endonuclease G (EndoG), through p53-dependent upregulation of Bax and Bak and downregulation of Bcl-2 via a pertussis toxin-sensitive G-protein-coupled receptor (GPCR) pathway. Correspondingly, colonic mucosa of cathelicidin-deficient mice exhibited reduced expression of p53, Bax, and Bak and increased expression of Bcl-2 together with a lower basal level of apoptosis. Cathelicidin-deficient mice exhibited an increased susceptibility to azoxymethane-induced colon tumorigenesis, establishing pathophysiologic relevance in colon cancer. Collectively, our findings show that LL-37 activates a GPCR-p53-Bax/Bak/Bcl-2 signaling cascade that triggers AIF/EndoG-mediated apoptosis in colon cancer cells.


Subject(s)
Adenocarcinoma/prevention & control , Antimicrobial Cationic Peptides/pharmacology , Apoptosis/drug effects , Colonic Neoplasms/prevention & control , Adaptive Immunity/drug effects , Adaptive Immunity/physiology , Adenocarcinoma/immunology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Animals , Antimicrobial Cationic Peptides/metabolism , Antimicrobial Cationic Peptides/physiology , Apoptosis/immunology , Case-Control Studies , Caspases/metabolism , Caspases/physiology , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Down-Regulation/drug effects , Drug Evaluation, Preclinical , Female , HCT116 Cells , HT29 Cells , Humans , Male , Mice , Mice, Knockout , Middle Aged , Cathelicidins
18.
PLoS One ; 7(5): e37572, 2012.
Article in English | MEDLINE | ID: mdl-22679478

ABSTRACT

Hydrogen sulfide (H(2)S) is a gaseous bacterial metabolite that reaches high levels in the large intestine. In the present study, the effect of H(2)S on the proliferation of normal and cancerous colon epithelial cells was investigated. An immortalized colon epithelial cell line (YAMC) and a panel of colon cancer cell lines (HT-29, SW1116, HCT116) were exposed to H(2)S at concentrations similar to those found in the human colon. H(2)S inhibited normal and cancerous colon epithelial cell proliferation as measured by MTT assay. The anti-mitogenic effect of H(2)S was accompanied by G(1)-phase cell cycle arrest and the induction of the cyclin-dependent kinase inhibitor p21(Cip). Moreover, exposure to H(2)S led to features characteristic of autophagy, including increased formation of LC3B(+) autophagic vacuoles and acidic vesicular organelles as determined by immunofluorescence and acridine orange staining, respectively. Abolition of autophagy by RNA interference targeting Vps34 or Atg7 enhanced the anti-proliferative effect of H(2)S. Further mechanistic investigation revealed that H(2)S stimulated the phosphorylation of AMP-activated protein kinase (AMPK) and inhibited the phosphorylation of mammalian target of rapamycin (mTOR) and S6 kinase. Inhibition of AMPK significantly reversed H(2)S-induced autophagy and inhibition of cell proliferation. Collectively, we demonstrate that H(2)S inhibits colon epithelial cell proliferation and induces protective autophagy via the AMPK pathway.


Subject(s)
Autophagy/drug effects , Colon/metabolism , Epithelial Cells/metabolism , Hydrogen Sulfide/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Autophagy-Related Protein 7 , Cell Cycle/drug effects , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Class III Phosphatidylinositol 3-Kinases/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Epithelial Cells/drug effects , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Phagosomes/drug effects , Phagosomes/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Ubiquitin-Activating Enzymes/metabolism
19.
Curr Pharm Des ; 17(16): 1583-6, 2011.
Article in English | MEDLINE | ID: mdl-21548872

ABSTRACT

Defensins and cathelicidins are small cationic peptides produced by neutrophils and epithelial cells. They are highly expressed during infection. The role of constitutive and inducible antibacterial peptides has been extensively studied over the recent years; especially in the gastrointestinal (GI) tract, where the balance between the luminal bacteria and antibacterial peptides is crucial in the maintenance of a healthy GI tract. There are reports showing that the expressions of defensins and cathelicidins in the gut are dysregulated in various disease states. They could participate in the development of different disorders ranging from inflammation to cancer. Experimental findings showed that supplementation with animal cathelicidin promoted gastric ulcer healing in rats and suppressed tumorigenesis of gastric cancer in mice. Mouse cathelicidin could alleviate murine colitis by preserving mucus content and suppression of apoptosis. Other clinical applications for these antibacterial peptides are awaiting for further studies.


Subject(s)
Cathelicidins/physiology , Defensins/physiology , Gastrointestinal Diseases/prevention & control , Animals , Anti-Bacterial Agents , Gastrointestinal Diseases/physiopathology , Humans , Mice
20.
Cancer Sci ; 102(5): 926-33, 2011 May.
Article in English | MEDLINE | ID: mdl-21261791

ABSTRACT

Prostaglandin E (EP) receptor is positively related with COX-2, which is involved in cancer biology. A mechanistic study on how 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) promotes gastric carcinogenesis is lacking. Recently, we found that nicotine promoted tumor growth through upregulation of the COX-2/prostaglandin E(2) pathway. This extended our study on the involvement of EP receptors in gastric carcinogenesis. Both in vitro and in vivo studies showed that NNK promoted cancer cell growth with concomitant EP2 and EP4 upregulation. We found that NNK stimulated vascular endothelial growth factor (VEGF) and angiogenesis, but suppressed apoptosis by increasing Bcl2 and decreasing caspase-3 expressions. Both EP2 and EP4 siRNA significantly impaired these tumorigenic actions of NNK in xenograft tumor. Cell cycle analysis showed that NNK increased S phase entry with increased cyclin D1 and the associated cyclin-dependent kinase 4/6, and downregulation of p21 and p27. The p38 phosphorylation was EP2/4-dependent, and SB203580 (p38 inhibitor) suppressed NNK-induced prostaglandin E(2) , VEGF, and cell proliferation. Antagonists of EP2 or EP4 abolished the elevated VEGF and VEGF receptor-2. These data strongly indicate that EP2/4 are important for NNK-promoted gastric carcinogenesis, thus providing a framework for future evaluation of EP antagonist(s) as anticancer drugs for smokers.


Subject(s)
Carcinogens/toxicity , Nitrosamines/toxicity , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Stomach Neoplasms/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Mice , Mice, Nude , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Stomach Neoplasms/chemically induced , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...