1
|
Daily ZA, Mohammed NB, Mohammed SM, Hussein HM. Correlation Between Periodontal Disease and Oral, Oropharyngeal, and Parapharyngeal Cancers. Clin Cosmet Investig Dent 2025; 17:147-158. [PMID: 40027983 PMCID: PMC11869753 DOI: 10.2147/ccide.s512557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/13/2025] [Indexed: 03/05/2025] Open
Abstract
Background Periodontitis is an inflammatory disease, and contributes to several inflammatory conditions, such as cancer. The relationship between periodontal disorders and different human malignancies is not well understood. The purpose of this study is to assess the association of periodontitis severity with cancers in the oral cavity, oropharyngeal and parapharyngeal regions. Methods The sample comprised 300 participants divided into four case groups: 75 oral, oropharyngeal, or parapharyngeal cancer patients with periodontitis (CA-with-P); 75 oral, oropharyngeal, or parapharyngeal cancer patients without periodontitis (CA-without-P); 75 periodontitis (P) patients without cancers; and a control (C) group of 75 healthy individuals. All participants were subjected to a periodontal examination that considered parameters such as bleeding on probing (BOP), plaque index (PI), probing pocket depth (PPD), and clinical attachment loss (CAL). The type of tumours was identified via a histological analysis of a biopsy sample. Saliva samples were also collected, and an enzyme-linked immunosorbent assay (ELISA) kit was used to determine interleukin 8 (IL-8) and nuclear factor kappa B (NF-κB) levels. Results The research findings indicated a significant increase in the number of sites with clinical observations of BOP (85.11,73.84), PI (87.23.88.14), PPD (8.03,6.82), and CAL (8.67,7.34) in groups CA-with-P and P. The CA-with-P, CA-without-P, and P groups had higher levels of salivary IL-8 (192.03, 121.89,89.22) and NF-κB (10.242, 8.172, 6.324) than the C group. Moreover, there was a significant correlation between the severity of periodontitis and the malignancies in the oral, oropharynx, and parapharyngeal regions. Conclusion This study assessed the mechanisms underlying the correlation between these two disorders, as elucidated by higher levels of salivary IL-8, NF-κB and an increase in clinical periodontal parameters. Periodontal bacteria, which contributes to the development of periodontal disorders, could have a major impact on the onset of oral cancers.
Collapse
Affiliation(s)
- Zina Ali Daily
- Periodontics Department, College of Dentistry, University of Al-Ameed, Karbala, Iraq
| | - Nawres Bahaa Mohammed
- Maxillofacial Surgery Department, Dentistry College, University of Al-Ameed, Karbala, Iraq
| | - Samer Majeed Mohammed
- Maxillofacial Surgery Department, Dentistry College, University of Al-Ameed, Karbala, Iraq
| | - Hashim Mueen Hussein
- Department of Conservative Dentistry, College of Dentistry, Mustansiriyah University, Baghdad, Iraq
| |
Collapse
|
2
|
Chen Q, Chen T, Xiao H, Wang F, Li C, Hu N, Bao L, Tong X, Feng Y, Xu Y, Li C, Zhu JW, Wang D, Li MX. APEX1 in intestinal epithelium triggers neutrophil infiltration and intestinal barrier damage in ulcerative colitis. Free Radic Biol Med 2024; 225:359-373. [PMID: 39389211 DOI: 10.1016/j.freeradbiomed.2024.10.260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024]
Abstract
Ulcerative colitis (UC) can lead to the generation of large amounts of reactive oxygen species and DNA damage. DNA repair caused by base excision repair (BER) enzymes is an important mechanism for maintaining genomic integrity. However, the specific relationship between the function of BER enzymes and UC remains unclear. To address this, we conducted a study on non-cancerous colon tissue from patients with UC, focusing on the role of apurinic/apyrimidinic endonuclease 1 (APEX1) in BER to explore its significance in the progression of UC. Our research found that the expression of APEX1 in epithelium cells was significantly correlated to the severity of inflammatory bowel disease (IBD) and the infiltration and function of neutrophils in human UC and mouse models, particularly in relation to neutrophil extracellular traps (NETs) and the degranulation processes. APEX1 deficiency resulted in decreased production of the chemokines CXCL1 by the NF-κB pathway in epithelium cells, leading to reduced accumulation and activation of neutrophils associated with colitis in colon tissue, as well as decreased levels of IL-1β. Furthermore, APEX1 deficiency reduced symptoms of colitis by decreasing epithelial cell apoptosis and altering the gut microbiome. Studies related to the redox activity of APEX1 have shown that the combination of the redox inhibitor E3330 with 5-aminosalicylic acid (5-ASA) can effectively alleviate colitis, indicating that APEX1 has promising prospects for clinical treatment of IBD. APEX1 is required for interactions between neutrophil and intestinal epithelial cells. This study provided a mechanism demonstrating that APEX1 protein triggered the risk of UC by promoting neutrophil infiltration and compromising intestinal epithelial barrier function.
Collapse
Affiliation(s)
- Qian Chen
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China.
| | - TianYi Chen
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China
| | - He Xiao
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China
| | - Fangjie Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center (Daping Hospital), Army Medical University, Chongqing, 400042, China
| | - ChaoFan Li
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China
| | - Nana Hu
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China; Yu-Yue Pathology Scientific Research Center, Choongqing, China
| | - Lingbo Bao
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China
| | - Xueling Tong
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China
| | - Yan Feng
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China
| | - Yu Xu
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China
| | - ChunXue Li
- Department of General Surgery, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Jian Wu Zhu
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China
| | - Dong Wang
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China.
| | - Meng Xia Li
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China.
| |
Collapse
|
3
|
Xue ZJ, Gong YN, He LH, Sun L, You YH, Fan DJ, Zhang MJ, Yan XM, Zhang JZ. Amino acid deletions at positions 893 and 894 of cytotoxin-associated gene A protein affect Helicobacter pylori gastric epithelial cell interactions. World J Gastroenterol 2024; 30:4449-4460. [PMID: 39534413 PMCID: PMC11551673 DOI: 10.3748/wjg.v30.i41.4449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 09/29/2024] [Accepted: 10/12/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Helicobacter pylori (H. pylori) persistently colonizes the human gastric mucosa in more than 50% of the global population, leading to various gastroduodenal diseases ranging from chronic gastritis to gastric carcinoma. Cytotoxin-associated gene A (CagA) protein, an important oncoprotein, has highly polymorphic Glu-Pro-Ile-Tyr-Ala segments at the carboxyl terminus, which play crucial roles in pathogenesis. Our previous study revealed a significant association between amino acid deletions at positions 893 and 894 and gastric cancer. AIM To investigate the impact of amino acid deletions at positions 893 and 894 on CagA function. METHODS We selected a representative HZT strain from a gastric cancer patient with amino acid deletions at positions 893 and 894. The cagA gene was amplified and mutated into cagA-NT and cagA-NE (sequence characteristics of strains from nongastric cancer patients), cloned and inserted into pAdtrack-CMV, and then transfected into AGS cells. The expression of cagA and its mutants was examined using real-time polymerase chain reaction and Western blotting, cell elongation via cell counting, F-actin cytoskeleton visualization using fluorescence staining, and interleukin-8 (IL-8) secretion via enzyme-linked immunosorbent assay. RESULTS The results revealed that pAdtrack/cagA induced a more pronounced hummingbird phenotype than pAdtrack/cagA-NT and pAdtrack/cagA-NE (40.88 ± 3.10 vs 32.50 ± 3.17, P < 0.001 and 40.88 ± 3.10 vs 32.17 ± 3.00, P < 0.001) at 12 hours after transfection. At 24 hours, pAdtrack/cagA-NE induced significantly fewer hummingbird phenotypes than pAdtrack/cagA and pAdtrack/cagA-NT (46.02 ± 2.12 vs 53.90 ± 2.10, P < 0.001 and 46.02 ± 2.12 vs 51.15 ± 3.74, P < 0.001). The total amount of F-actin caused by pAdtrack/cagA was significantly lower than that caused by pAdtrack/cagA-NT and pAdtrack/cagA-NE (27.54 ± 17.37 vs 41.51 ± 11.90, P < 0.001 and 27.54 ± 17.37 vs 41.39 ± 14.22, P < 0.001) at 12 hours after transfection. Additionally, pAdtrack/cagA induced higher IL-8 secretion than pAdtrack/cagA-NT and pAdtrack/cagA-NE at different times after transfection. CONCLUSION Amino acid deletions at positions 893 and 894 enhance CagA pathogenicity, which is crucial for revealing the pathogenic mechanism of CagA and identifying biomarkers of highly pathogenic H. pylori.
Collapse
Affiliation(s)
- Zhi-Jing Xue
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, Shandong Province, China
| | - Ya-Nan Gong
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Li-Hua He
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Lu Sun
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Yuan-Hai You
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Dong-Jie Fan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Mao-Jun Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Xiao-Mei Yan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Jian-Zhong Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| |
Collapse
|
4
|
Luk CYM, Lee SA, Naidovski N, Liu F, Tay ACY, Wang L, Riordan S, Zhang L. Investigation of Campylobacter concisus gastric epithelial pathogenicity using AGS cells. Front Microbiol 2024; 14:1289549. [PMID: 38274743 PMCID: PMC10808343 DOI: 10.3389/fmicb.2023.1289549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
Campylobacter concisus is an oral bacterium. Recent studies suggest that C. concisus may be involved in human gastric diseases. The mechanisms, however, by which C. concisus causes human gastric diseases have not been investigated. Here we examined the gastric epithelial pathogenicity of C. concisus using a cell culture model. Six C. concisus strains and the human gastric epithelial cell line AGS cells were used. IL-8 produced by AGS cells after incubation with C. concisus was measured using enzyme-linked immunosorbent assay (ELISA), and AGS cell apoptosis was determined by caspase 3/7 activities. The effects of C. concisus on actin arrangement in AGS cells was determined using fluorescence staining. The effects of C. concisus on global gene expression in AGS cells was determined by transcriptomic analysis and quantitative real-time PCR (qRT-PCR). The role of the upregulated CYP1A1 gene in gastric cancer survival was assessed using the Kaplan-Meier method. C. concisus induced production of IL-8 by AGS cells with strain variation. Significantly increased caspase 3/7 activities were observed in AGS cells incubated with C. concisus strains when compared to AGS cells without bacteria. C. concisus induced actin re-arrangement in AGS cells. C. concisus upregulated 30 genes in AGS cells and the upregulation of CYP1A1 gene was confirmed by qRT-PCR. The Kaplan-Meier analysis showed that upregulation of CYP1A1 gene is associated with worse survival in gastric cancer patients. Our findings suggest that C. concisus may play a role in gastric inflammation and the progression of gastric cancer. Further investigation in clinical studies is warranted.
Collapse
Affiliation(s)
- Christopher Yau Man Luk
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Seul A. Lee
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Nicholas Naidovski
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Fang Liu
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Alfred Chin Yen Tay
- Helicobacter Research Laboratory, School of Pathology and Laboratory Medicine, Marshall Centre for Infectious Diseases Research and Training, University of Western Australia, Perth, WA, Australia
| | - Liang Wang
- Laboratory Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- The Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
- Department of Medical Informatics, School of Medical Informatics and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Stephen Riordan
- Gastrointestinal and Liver Unit, Prince of Wales Hospital, University of New South Wales, Sydney, NSW, Australia
| | - Li Zhang
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
5
|
Sahakian L, Robinson AM, Sahakian L, Stavely R, Kelley MR, Nurgali K. APE1/Ref-1 as a Therapeutic Target for Inflammatory Bowel Disease. Biomolecules 2023; 13:1569. [PMID: 38002251 PMCID: PMC10669584 DOI: 10.3390/biom13111569] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/18/2023] [Accepted: 10/22/2023] [Indexed: 11/26/2023] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by chronic relapsing inflammation of the gastrointestinal tract. The prevalence of IBD is increasing with approximately 4.9 million cases reported worldwide. Current therapies are limited due to the severity of side effects and long-term toxicity, therefore, the development of novel IBD treatments is necessitated. Recent findings support apurinic/apyrimidinic endonuclease 1/reduction-oxidation factor 1 (APE1/Ref-1) as a target in many pathological conditions, including inflammatory diseases, where APE1/Ref-1 regulation of crucial transcription factors impacts significant pathways. Thus, a potential target for a novel IBD therapy is the redox activity of the multifunctional protein APE1/Ref-1. This review elaborates on the status of conventional IBD treatments, the role of an APE1/Ref-1 in intestinal inflammation, and the potential of a small molecule inhibitor of APE1/Ref-1 redox activity to modulate inflammation, oxidative stress response, and enteric neuronal damage in IBD.
Collapse
Affiliation(s)
- Lauren Sahakian
- Institute for Health & Sport, Victoria University, Melbourne, VIC 3021, Australia; (L.S.); (A.M.R.)
| | - Ainsley M. Robinson
- Institute for Health & Sport, Victoria University, Melbourne, VIC 3021, Australia; (L.S.); (A.M.R.)
| | - Linda Sahakian
- Department of Medicine Western Health, The University of Melbourne, Melbourne, VIC 3010, Australia; (L.S.); (R.S.)
| | - Rhian Stavely
- Department of Medicine Western Health, The University of Melbourne, Melbourne, VIC 3010, Australia; (L.S.); (R.S.)
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Mark R. Kelley
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kulmira Nurgali
- Institute for Health & Sport, Victoria University, Melbourne, VIC 3021, Australia; (L.S.); (A.M.R.)
- Department of Medicine Western Health, The University of Melbourne, Melbourne, VIC 3010, Australia; (L.S.); (R.S.)
- Regenerative Medicine and Stem Cells Program, Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
| |
Collapse
|
6
|
Klapp V, Álvarez-Abril B, Leuzzi G, Kroemer G, Ciccia A, Galluzzi L. The DNA Damage Response and Inflammation in Cancer. Cancer Discov 2023; 13:1521-1545. [PMID: 37026695 DOI: 10.1158/2159-8290.cd-22-1220] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/27/2023] [Accepted: 02/23/2023] [Indexed: 04/08/2023]
Abstract
Genomic stability in normal cells is crucial to avoid oncogenesis. Accordingly, multiple components of the DNA damage response (DDR) operate as bona fide tumor suppressor proteins by preserving genomic stability, eliciting the demise of cells with unrepairable DNA lesions, and engaging cell-extrinsic oncosuppression via immunosurveillance. That said, DDR sig-naling can also favor tumor progression and resistance to therapy. Indeed, DDR signaling in cancer cells has been consistently linked to the inhibition of tumor-targeting immune responses. Here, we discuss the complex interactions between the DDR and inflammation in the context of oncogenesis, tumor progression, and response to therapy. SIGNIFICANCE Accumulating preclinical and clinical evidence indicates that DDR is intimately connected to the emission of immunomodulatory signals by normal and malignant cells, as part of a cell-extrinsic program to preserve organismal homeostasis. DDR-driven inflammation, however, can have diametrically opposed effects on tumor-targeting immunity. Understanding the links between the DDR and inflammation in normal and malignant cells may unlock novel immunotherapeutic paradigms to treat cancer.
Collapse
Affiliation(s)
- Vanessa Klapp
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
- Tumor Stroma Interactions, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Beatriz Álvarez-Abril
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
- Department of Hematology and Oncology, Hospital Universitario Morales Meseguer, Murcia, Spain
| | - Giuseppe Leuzzi
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, New York
- Herbert Irving Comprehensive Cancer Center, New York, New York
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, New York
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le Cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Alberto Ciccia
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, New York
- Herbert Irving Comprehensive Cancer Center, New York, New York
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, New York
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
- Sandra and Edward Meyer Cancer Center, New York, New York
- Caryl and Israel Englander Institute for Precision Medicine, New York, New York
| |
Collapse
|
7
|
Lv R, Sun N, Mao C, Zheng Z, Lin S. Prevention and potential repair of colitis: Beneficial effects and regulatory mechanisms of food-derived anti-inflammatory peptides. Crit Rev Food Sci Nutr 2023; 64:8184-8202. [PMID: 37017113 DOI: 10.1080/10408398.2023.2197068] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2023]
Abstract
Intestinal inflammatory diseases are increasingly prevalent worldwide, and their pathogenesis is still not fully understood. As of late, studies have discovered that food-derived peptides have specific anti-inflammatory activity and can play a positive role in intestinal health. At the same time, it has broad application prospects in the prevention and treatment of colitis because of its wide source, fast absorption, and high safety. This article reviews the structure-activity and quantity-effect relationships of food-derived peptides for their anti-inflammatory effects. It then discusses their mechanism of action in inhibiting colitis from four aspects. Food-derived anti-inflammatory peptides can delay the progression of the disease by stimulating innate immunity, inhibiting inflammation, and promoting wound healing. Further experiments showed that food-derived anti-inflammatory peptides could prevent and treat colitis through four mechanisms: (a) regulation of inflammatory cytokines; (b) regulation of inflammatory pathways; (c) regulation of intestinal epithelial barrier; (d) regulation of intestinal flora balance. However, due to the treatment of colitis having limitations, there is an urgent to develop food-derived anti-inflammatory peptides as a treatment or adjunctive treatment for colitis. This review highlights the positive effects of food-derived peptides on colitis and anticipates the appearance of mitigating peptides for the therapy of colitis.
Collapse
Affiliation(s)
- Renzhi Lv
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P. R. China
| | - Na Sun
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P. R. China
| | - Chuwen Mao
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P. R. China
| | - Zhihong Zheng
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P. R. China
| | - Songyi Lin
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P. R. China
| |
Collapse
|
8
|
Rios-Covian D, Butcher LD, Ablack AL, den Hartog G, Matsubara MT, Ly H, Oates AW, Xu G, Fisch KM, Ahrens ET, Toden S, Brown CC, Kim K, Le D, Eckmann L, Dhar B, Izumi T, Ernst PB, Crowe SE. A Novel Hypomorphic Apex1 Mouse Model Implicates Apurinic/Apyrimidinic Endonuclease 1 in Oxidative DNA Damage Repair in Gastric Epithelial Cells. Antioxid Redox Signal 2023; 38:183-197. [PMID: 35754343 PMCID: PMC10039277 DOI: 10.1089/ars.2021.0119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 06/06/2022] [Accepted: 06/09/2022] [Indexed: 01/20/2023]
Abstract
Aims: Though best known for its role in oxidative DNA damage repair, apurinic/apyrimidinic endonuclease 1 (APE1) is a multifunctional protein that regulates multiple host responses during oxidative stress, including the reductive activation of transcription factors. As knockout of the APE1-encoding gene, Apex1, is embryonically lethal, we sought to create a viable model with generalized inhibition of APE1 expression. Results: A hypomorphic (HM) mouse with decreased APE1 expression throughout the body was generated using a construct containing a neomycin resistance (NeoR) cassette knocked into the Apex1 site. Offspring were assessed for APE1 expression, breeding efficiency, and morphology with a focused examination of DNA damage in the stomach. Heterozygotic breeding pairs yielded 50% fewer HM mice than predicted by Mendelian genetics. APE1 expression was reduced up to 90% in the lungs, heart, stomach, and spleen. The HM offspring were typically smaller, and most had a malformed tail. Oxidative DNA damage was increased spontaneously in the stomachs of HM mice. Further, all changes were reversed when the NeoR cassette was removed. Primary gastric epithelial cells from HM mice differentiated more quickly and had more evidence of oxidative DNA damage after stimulation with Helicobacter pylori or a chemical carcinogen than control lines from wildtype mice. Innovation: A HM mouse with decreased APE1 expression throughout the body was generated and extensively characterized. Conclusion: The results suggest that HM mice enable studies of APE1's multiple functions throughout the body. The detailed characterization of the stomach showed that gastric epithelial cells from HM were more susceptible to DNA damage. Antioxid. Redox Signal. 38, 183-197.
Collapse
Affiliation(s)
- David Rios-Covian
- Center for Veterinary Sciences and Comparative Medicine, Division of Comparative Pathology and Medicine, Department of Pathology, University of California, San Diego, La Jolla, California, USA
| | - Lindsay D. Butcher
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Amber L. Ablack
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Gerco den Hartog
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Mason T. Matsubara
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Hong Ly
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Andrew W. Oates
- Center for Veterinary Sciences and Comparative Medicine, Division of Comparative Pathology and Medicine, Department of Pathology, University of California, San Diego, La Jolla, California, USA
| | - Guorong Xu
- Center for Computational Biology & Bioinformatics, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Kathleen M. Fisch
- Center for Computational Biology & Bioinformatics, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Eric T. Ahrens
- Department of Radiology, University of California, San Diego, La Jolla, California, USA
| | - Shusuke Toden
- Molecular Stethoscope, Inc., San Diego, California, USA
| | - Corrie C. Brown
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Kenneth Kim
- La Jolla Institute for Immunology, La Jolla, California, USA
| | - Dzung Le
- Center for Veterinary Sciences and Comparative Medicine, Division of Comparative Pathology and Medicine, Department of Pathology, University of California, San Diego, La Jolla, California, USA
| | - Lars Eckmann
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Bithika Dhar
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, USA
| | - Tadahide Izumi
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, USA
| | - Peter B. Ernst
- Center for Veterinary Sciences and Comparative Medicine, Division of Comparative Pathology and Medicine, Department of Pathology, University of California, San Diego, La Jolla, California, USA
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, California, USA
- Department of Immunology, Chiba University, Chiba, Japan
| | - Sheila E. Crowe
- Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
9
|
Anti-Helicobacter pylori, anti-Inflammatory, and Antioxidant Activities of Trunk Bark of Alstonia boonei (Apocynaceae). BIOMED RESEARCH INTERNATIONAL 2022; 2022:9022135. [PMID: 36158881 PMCID: PMC9499789 DOI: 10.1155/2022/9022135] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/25/2022] [Indexed: 12/14/2022]
Abstract
An ulcer is an erosion of the gastric mucosa that occurs following an imbalance between the aggression and protective factors and/or an infection with Helicobacter pylori (H. pylori). About 90-100% of duodenal ulcers and 70-80% of gastric ulcers are caused by H. pylori. The objective of this work was to evaluate in vitro the anti-H. pylori activity and then the anti-inflammatory and antioxidant properties of aqueous and methanol extracts of Alstonia boonei. The anti-H. pylori tests (CMI and antiureasic activity) were determined using the agar well diffusion method, the microbroth dilution method, and the measurement of ammonia production by the indophenol method; the anti-inflammatory properties were evaluated by inhibition of proteinases, denaturation of albumin, production of NO by macrophages, cell viability, and hemolysis of red blood cells by heat; then, the antioxidant properties were evaluated by the FRAP method (ferric reducing antioxidant power) and the DPPH (1,1-diphenyl-2-picrylhydrazyl) test. The results show that the best trapping of the DPPH radical was obtained with the methanol extract (EC50 = 8.91 μg/mL) compared to the aqueous extract (EC50 = 19.86 μg/mL). The methanol extract also showed greater iron-reducing activity than the aqueous extract and vitamin C. Furthermore, at the concentration of 200 μg/mL, the methanol extract showed a percentage (96.34%) strains of H. pylori higher than that of the aqueous extract (88.52%). The MIC90 of the methanol extract was lower than that of the aqueous extract. The methanol extract showed a higher percentage inhibition (85%) of urease than the aqueous extract (73%). The methanol extract at a concentration of 1000 μg/mL showed the greatest ability to inhibit proteinase activity, albumin denaturation, and red blood cell hemolysis; on the other hand, maximum cell viability and greater production of nitrite oxide by macrophages were obtained with the aqueous extract. Aqueous and methanol extracts of Alstonia boonei possess anti-H. pylori which would probably be linked to their antioxidant and anti-inflammatory properties.
Collapse
|
10
|
Old and New Aspects of H. pylori-Associated Inflammation and Gastric Cancer. CHILDREN 2022; 9:children9071083. [PMID: 35884067 PMCID: PMC9322908 DOI: 10.3390/children9071083] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 07/17/2022] [Accepted: 07/18/2022] [Indexed: 12/16/2022]
Abstract
H. pylori is involved in the development of 80% of gastric cancers and 5.5% of all malignant conditions worldwide. Its persistence within the host’s stomach causes chronic inflammation, which is a well-known hallmark of carcinogenesis. A wide range of cytokines was reported to be involved in the initiation and long-term persistence of this local and systemic inflammation. IL-8 was among the first cytokines described to be increased in patients with H. pylori infection. Although, this cytokine was initially identified to exert a chemoattracting effect that represents a trigger for the activation of inflammatory cells within H.-pylori-infected mucosa, more recent studies failed in encountering any association between IL-8 and H. pylori infection. IL-6 is a multifunctional, pleiotropic and multipotent cytokine involved in mediating the interaction between innate and adaptive immunity with a dichotomous role acting as both a proinflammatory and an anti-inflammatory cytokine depending on the signaling pathway. IL-1α functions as a promoter of angiogenesis and vascular endothelial cell proliferation in gastric carcinoma since it is closely related to H.-pylori-induced inflammation in children. IL-1β is an essential trigger and enhancer of inflammation. The association between a low IL-1β level and an increased TNF-α level might be considered a risk factor for peptic ulcer disease in the setting of H. pylori infection. IL-10 downregulates both cytotoxic inflammatory responses and cell-mediated immune responses. H. pylori uses the immunosuppressive role of IL-10 to favor its escape from the host’s immune system. TGFβ is a continuous inflammatory mediator that promotes the adherence of H. pylori to the host’s cells and their subsequent colonization. The role of H.-pylori-induced inflammatory responses in the onset of gastric carcinogenesis seems to represent the missing puzzle piece for designing effective preventive and therapeutic strategies in patients with H.-pylori-associated gastric cancer.
Collapse
|
11
|
Oliveira TT, Coutinho LG, de Oliveira LOA, Timoteo ARDS, Farias GC, Agnez-Lima LF. APE1/Ref-1 Role in Inflammation and Immune Response. Front Immunol 2022; 13:793096. [PMID: 35296074 PMCID: PMC8918667 DOI: 10.3389/fimmu.2022.793096] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 02/07/2022] [Indexed: 12/12/2022] Open
Abstract
Apurinic/apyrimidinic endonuclease 1/redox effector factor 1 (APE1/Ref-1) is a multifunctional enzyme that is essential for maintaining cellular homeostasis. APE1 is the major apurinic/apyrimidinic endonuclease in the base excision repair pathway and acts as a redox-dependent regulator of several transcription factors, including NF-κB, AP-1, HIF-1α, and STAT3. These functions render APE1 vital to regulating cell signaling, senescence, and inflammatory pathways. In addition to regulating cytokine and chemokine expression through activation of redox sensitive transcription factors, APE1 participates in other critical processes in the immune response, including production of reactive oxygen species and class switch recombination. Furthermore, through participation in active chromatin demethylation, the repair function of APE1 also regulates transcription of some genes, including cytokines such as TNFα. The multiple functions of APE1 make it an essential regulator of the pathogenesis of several diseases, including cancer and neurological disorders. Therefore, APE1 inhibitors have therapeutic potential. APE1 is highly expressed in the central nervous system (CNS) and participates in tissue homeostasis, and its roles in neurodegenerative and neuroinflammatory diseases have been elucidated. This review discusses known roles of APE1 in innate and adaptive immunity, especially in the CNS, recent evidence of a role in the extracellular environment, and the therapeutic potential of APE1 inhibitors in infectious/immune diseases.
Collapse
Affiliation(s)
- Thais Teixeira Oliveira
- Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte (UFRN), Natal, Brazil
| | - Leonam Gomes Coutinho
- Instituto Federal de Educação, Ciência e Tecnologia do Rio Grande do Norte (IFRN), São Paulo do Potengi, Brazil
| | | | | | - Guilherme Cavalcanti Farias
- Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte (UFRN), Natal, Brazil
| | - Lucymara Fassarella Agnez-Lima
- Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte (UFRN), Natal, Brazil
- *Correspondence: Lucymara Fassarella Agnez-Lima,
| |
Collapse
|
12
|
Choi MS, Ze EY, Park JY, Shin TS, Kim JG. Helicobacter pylori-derived outer membrane vesicles stimulate interleukin 8 secretion through nuclear factor kappa B activation. Korean J Intern Med 2021; 36:854-867. [PMID: 33242939 PMCID: PMC8273812 DOI: 10.3904/kjim.2019.432] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/19/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND/AIMS Bacteria-derived outer membrane vesicles (OMVs) are commonly associated with various biological activities and functions. Helicobacter pylori-derived OMVs are thought to contribute to pathogenesis. This study aimed to investigate the effects of H. pylori-derived OMVs. METHODS H. pylori strains were isolated from patients with gastritis, gastric ulcer, or gastric cancer using endoscopic biopsy. The U-937, AGS, and MKN-45 cell lines were exposed to H. pylori and H. pylori-derived OMVs. The expression of interleukin 8 (IL-8) messenger RNA (mRNA) was assessed using reverse transcription-polymerase chain reaction (RT-PCR) and real-time RT-PCR, and IL-8 secretion was analyzed using enzyme-linked immunosorbent assay. Nuclear factor kappa B (NF-κB) activation was evaluated by Western blotting. RESULTS H. pylori and H. pylori-derived OMVs induced the expression of IL-8 mRNA and protein. Importantly, the bacteria induced higher IL-8 mRNA and protein expression than the OMVs. IL-8 expression was induced to different levels in response to H. pylori-derived OMVs from hosts with different gastric diseases. Western blotting revealed the increased phosphorylation and reduced degradation of inhibitor of NF-κB alpha in cells exposed to OMVs. CONCLUSION H. pylori-derived OMVs may aid the development of various gastric diseases by inducing IL-8 production and NF-κB activation.
Collapse
Affiliation(s)
- Mun Sun Choi
- Division of Gastroenterology, Department of Internal Medicine, Armed Forces Capital Hospital, Seongnam,
Korea
| | - Eun Young Ze
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul,
Korea
| | - Jae Yong Park
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul,
Korea
| | - Tae-Seop Shin
- Research Institute, Chung-Ang University, Seoul,
Korea
| | - Jae Gyu Kim
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul,
Korea
| |
Collapse
|
13
|
den Hartog G, Butcher LD, Ablack AL, Pace LA, Ablack JNG, Xiong R, Das S, Stappenbeck TS, Eckmann L, Ernst PB, Crowe SE. Apurinic/Apyrimidinic Endonuclease 1 Restricts the Internalization of Bacteria Into Human Intestinal Epithelial Cells Through the Inhibition of Rac1. Front Immunol 2021; 11:553994. [PMID: 33603730 PMCID: PMC7884313 DOI: 10.3389/fimmu.2020.553994] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Pathogenic intestinal bacteria lead to significant disease in humans. Here we investigated the role of the multifunctional protein, Apurinic/apyrimidinic endonuclease 1 (APE1), in regulating the internalization of bacteria into the intestinal epithelium. Intestinal tumor-cell lines and primary human epithelial cells were infected with Salmonella enterica serovar Typhimurium or adherent-invasive Escherichia coli. The effects of APE1 inhibition on bacterial internalization, the regulation of Rho GTPase Rac1 as well as the epithelial cell barrier function were assessed. Increased numbers of bacteria were present in APE1-deficient colonic tumor cell lines and primary epithelial cells. Activation of Rac1 was augmented following infection but negatively regulated by APE1. Pharmacological inhibition of Rac1 reversed the increase in intracellular bacteria in APE1-deficient cells whereas overexpression of constitutively active Rac1 augmented the numbers in APE1-competent cells. Enhanced numbers of intracellular bacteria resulted in the loss of barrier function and a delay in its recovery. Our data demonstrate that APE1 inhibits the internalization of invasive bacteria into human intestinal epithelial cells through its ability to negatively regulate Rac1. This activity also protects epithelial cell barrier function.
Collapse
Affiliation(s)
- Gerco den Hartog
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Lindsay D Butcher
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Amber L Ablack
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Laura A Pace
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Jailal N G Ablack
- Department of Medicine, Division of Rheumatology, University of California San Diego, La Jolla, CA, United States
| | - Richard Xiong
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Soumita Das
- Division of Comparative Pathology and Medicine, Department of Pathology, University of California San Diego, La Jolla, CA, United States
| | | | - Lars Eckmann
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Peter B Ernst
- Division of Comparative Pathology and Medicine, Department of Pathology, University of California San Diego, La Jolla, CA, United States.,Center for Mucosal Immunology, Allergy and Vaccine Development, Department of Pathology, University of California San Diego, La Jolla, CA, United States.,Department of Immunology, Chiba University, Chiba, Japan
| | - Sheila E Crowe
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States.,Division of ImmunoBiology, Washington University, St. Louis, MO, United States
| |
Collapse
|
14
|
Tang Z, Wang Y, Wan Y, Xie Y, Li S, Tao D, Wang C, Wu YZ, Sui JD. Apurinic/apyrimidinic endonuclease 1/reduction-oxidation effector factor-1 (APE1) regulates the expression of NLR family pyrin domain containing 3 (NLRP3) inflammasome through modulating transcription factor NF-κB and promoting the secretion of inflammatory mediators in macrophages. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:145. [PMID: 33569447 PMCID: PMC7867945 DOI: 10.21037/atm-20-7752] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background Inflammatory mediators play an important role in the occurrence, development, and metastasis of tumors. The aim of the present study was to elucidate the effect of apurinic/apyrimidinic endonuclease 1/reduction-oxidation effector factor-1 (APE1) on inflammatory mediator secretion, which is dependent on the APE1-mediated NLR family pyrin domain containing 3 (NLRP3) regulatory mechanism. Methods The human myeloid leukemia mononuclear cell line (THP-1) cells were cultured and polarized to M2 subset macrophages. Enzyme-linked immunosorbent assay was used for determining tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-18, IL-10, and IL-33 levels. Reverse transcription–polymerase chain reaction and western blot were used for evaluating TNF-α, NLR family pyrin domain containing 1 (NLRP1), NLRP3, caspase-1, and apoptosis-associated speck-like protein containing a card expression. Plasmid silencing APE1 gene (APE1shRNA) was synthesized and packaged into lentiviral. For activating inflammasomes, M2-type THP-1 cells were transfected with lentiviral vector APE1shRNA incubated with lipopolysaccharide (LPS) (100 ng/mL)/APE1 inhibitor (E3330, 20 µM) and ATP. Electrophoretic mobility shift assay and dual-luciferase reporter assay were used for determining the interaction between NLRP3 and nuclear factor-κB (NF-κB) molecule. Results APE1 significantly induced LPS-induced pro-inflammatory cytokine production, including TNF-α, IL-1β, and IL18, compared with THP-1 cells without APE1 treatment (P<0.05). APE1 promoted LPS-induced NLRP3 inflammasome activation by modulating the gene transcription of NLRP3-associated molecules. APE1 enhanced LPS-induced NLRP3 inflammasome activation by regulating NLRP3 and caspase-1 protein expression. APE1 improved NLRP3 activity by modulating the interaction between NLRP3 and NF-κB, and the modulation of NF-κB. APE1 promoted LPS-induced NLRP3 inflammasome activation through an NF-κB-dependent pathway. Conclusions APE1 regulates the expression of NLRP3 by modulating transcription factor NF-κB and further promoting the secretion of inflammatory mediators IL-1β and IL-18 in macrophages. The findings of the present study provide theoretical and experimental bases for the design of tumor-associated macrophage (TAM)-targeted therapy, with APE1 as the target molecule.
Collapse
Affiliation(s)
- Zheng Tang
- College of Bioengineering, Chongqing University, Chongqing, China.,Radiation Oncology Center, Chongqing University Cancer Hospital, Chongqing, China.,Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Ying Wang
- Radiation Oncology Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Yue Wan
- Radiation Oncology Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Yue Xie
- Radiation Oncology Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Shujie Li
- Radiation Oncology Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Dan Tao
- Radiation Oncology Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Can Wang
- Radiation Oncology Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Yong-Zhong Wu
- Radiation Oncology Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Jiang-Dong Sui
- Radiation Oncology Center, Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
15
|
Kontizas E, Tastsoglou S, Karamitros T, Karayiannis Y, Kollia P, Hatzigeorgiou AG, Sgouras DN. Impact of Helicobacter pylori Infection and Its Major Virulence Factor CagA on DNA Damage Repair. Microorganisms 2020; 8:microorganisms8122007. [PMID: 33339161 PMCID: PMC7765595 DOI: 10.3390/microorganisms8122007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 01/10/2023] Open
Abstract
Helicobacter pylori infection induces a plethora of DNA damages. Gastric epithelial cells, in order to maintain genomic integrity, require an integrous DNA damage repair (DDR) machinery, which, however, is reported to be modulated by the infection. CagA is a major H. pylori virulence factor, associated with increased risk for gastric carcinogenesis. Its pathogenic activity is partly regulated by phosphorylation on EPIYA motifs. Our aim was to identify effects of H. pylori infection and CagA on DDR, investigating the transcriptome of AGS cells, infected with wild-type, ΔCagA and EPIYA-phosphorylation-defective strains. Upon RNA-Seq-based transcriptomic analysis, we observed that a notable number of DDR genes were found deregulated during the infection, potentially resulting to base excision repair and mismatch repair compromise and an intricate deregulation of nucleotide excision repair, homologous recombination and non-homologous end-joining. Transcriptome observations were further investigated on the protein expression level, utilizing infections of AGS and GES-1 cells. We observed that CagA contributed to the downregulation of Nth Like DNA Glycosylase 1 (NTHL1), MutY DNA Glycosylase (MUTYH), Flap Structure-Specific Endonuclease 1 (FEN1), RAD51 Recombinase, DNA Polymerase Delta Catalytic Subunit (POLD1), and DNA Ligase 1 (LIG1) and, contrary to transcriptome results, Apurinic/Apyrimidinic Endodeoxyribonuclease 1 (APE1) upregulation. Our study accentuates the role of CagA as a significant contributor of H. pylori infection-mediated DDR modulation, potentially disrupting the balance between DNA damage and repair, thus favoring genomic instability and carcinogenesis.
Collapse
Affiliation(s)
- Eleftherios Kontizas
- Laboratory of Medical Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece;
- Department of Genetics and Biotechnology, Faculty of Biology, National and Kapodistrian University of Athens, 15772 Athens, Greece;
- Correspondence: (E.K.); (D.N.S.); Tel.: +30-210-647-8812 (E.K.); +30-210-647-8824 (D.N.S.)
| | - Spyros Tastsoglou
- Department of Electrical and Computer Engineering, University of Thessaly, 38221 Volos, Greece;
- DIANA-Lab, Hellenic Pasteur Institute, 11521 Athens, Greece;
| | - Timokratis Karamitros
- Bioinformatics and Applied Genomics Unit, Hellenic Pasteur Institute, 11521 Athens, Greece;
| | - Yiannis Karayiannis
- Laboratory of Medical Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece;
- Department of Genetics and Biotechnology, Faculty of Biology, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| | - Panagoula Kollia
- Department of Genetics and Biotechnology, Faculty of Biology, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| | - Artemis G. Hatzigeorgiou
- DIANA-Lab, Hellenic Pasteur Institute, 11521 Athens, Greece;
- DIANA-Lab, Department of Computer Science and Biomedical Informatics, University of Thessaly, 35131 Lamia, Greece
| | - Dionyssios N. Sgouras
- Laboratory of Medical Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece;
- Correspondence: (E.K.); (D.N.S.); Tel.: +30-210-647-8812 (E.K.); +30-210-647-8824 (D.N.S.)
| |
Collapse
|
16
|
Ilchmann-Diounou H, Menard S. Psychological Stress, Intestinal Barrier Dysfunctions, and Autoimmune Disorders: An Overview. Front Immunol 2020; 11:1823. [PMID: 32983091 PMCID: PMC7477358 DOI: 10.3389/fimmu.2020.01823] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
Autoimmune disorders (ADs) are multifactorial diseases involving, genetic, epigenetic, and environmental factors characterized by an inappropriate immune response toward self-antigens. In the past decades, there has been a continuous rise in the incidence of ADs, which cannot be explained by genetic factors alone. Influence of psychological stress on the development or the course of autoimmune disorders has been discussed for a long time. Indeed, based on epidemiological studies, stress has been suggested to precede AD occurrence and to exacerbate symptoms. Furthermore, compiling data showed that most of ADs are associated with gastrointestinal symptoms, that is, microbiota dysbiosis, intestinal hyperpermeability, and intestinal inflammation. Interestingly, social stress (acute or chronic, in adult or in neonate) is a well-described intestinal disrupting factor. Taken together, those observations question a potential role of stress-induced defect of the intestinal barrier in the onset and/or the course of ADs. In this review, we aim to present evidences supporting the hypothesis for a role of stress-induced intestinal barrier disruption in the onset and/or the course of ADs. We will mainly focus on autoimmune type 1 diabetes, multiple sclerosis and systemic lupus erythematosus, ADs for which we could find sufficient circumstantial data to support this hypothesis. We excluded gastrointestinal (GI) ADs like coeliac disease to privilege ADs not focused on intestinal disorders to avoid confounding factors. Indeed, GIADs are characterized by antibodies directed against intestinal barrier actors.
Collapse
MESH Headings
- Animals
- Autoimmune Diseases/epidemiology
- Autoimmune Diseases/immunology
- Autoimmune Diseases/metabolism
- Autoimmune Diseases/microbiology
- Autoimmunity
- Diabetes Mellitus, Type 1/epidemiology
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/microbiology
- Dysbiosis
- Gastrointestinal Microbiome
- Host-Pathogen Interactions
- Humans
- Intestinal Mucosa/immunology
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/microbiology
- Lupus Erythematosus, Systemic/epidemiology
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/metabolism
- Lupus Erythematosus, Systemic/microbiology
- Multiple Sclerosis/epidemiology
- Multiple Sclerosis/immunology
- Multiple Sclerosis/metabolism
- Multiple Sclerosis/microbiology
- Permeability
- Risk Factors
- Stress, Psychological/epidemiology
- Stress, Psychological/immunology
- Stress, Psychological/metabolism
- Stress, Psychological/microbiology
Collapse
Affiliation(s)
| | - Sandrine Menard
- Neuro-Gastroenterology and Nutrition Team, Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| |
Collapse
|
17
|
Sahakian L, Filippone RT, Stavely R, Robinson AM, Yan XS, Abalo R, Eri R, Bornstein JC, Kelley MR, Nurgali K. Inhibition of APE1/Ref-1 Redox Signaling Alleviates Intestinal Dysfunction and Damage to Myenteric Neurons in a Mouse Model of Spontaneous Chronic Colitis. Inflamm Bowel Dis 2020; 27:388-406. [PMID: 32618996 PMCID: PMC8287929 DOI: 10.1093/ibd/izaa161] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) associates with damage to the enteric nervous system (ENS), leading to gastrointestinal (GI) dysfunction. Oxidative stress is important for the pathophysiology of inflammation-induced enteric neuropathy and GI dysfunction. Apurinic/apyrimidinic endonuclease 1/redox factor-1 (APE1/Ref-1) is a dual functioning protein that is an essential regulator of the cellular response to oxidative stress. In this study, we aimed to determine whether an APE1/Ref-1 redox domain inhibitor, APX3330, alleviates inflammation-induced oxidative stress that leads to enteric neuropathy in the Winnie murine model of spontaneous chronic colitis. METHODS Winnie mice received APX3330 or vehicle via intraperitoneal injections over 2 weeks and were compared with C57BL/6 controls. In vivo disease activity and GI transit were evaluated. Ex vivo experiments were performed to assess functional parameters of colonic motility, immune cell infiltration, and changes to the ENS. RESULTS Targeting APE1/Ref-1 redox activity with APX3330 improved disease severity, reduced immune cell infiltration, restored GI function ,and provided neuroprotective effects to the enteric nervous system. Inhibition of APE1/Ref-1 redox signaling leading to reduced mitochondrial superoxide production, oxidative DNA damage, and translocation of high mobility group box 1 protein (HMGB1) was involved in neuroprotective effects of APX3330 in enteric neurons. CONCLUSIONS This study is the first to investigate inhibition of APE1/Ref-1's redox activity via APX3330 in an animal model of chronic intestinal inflammation. Inhibition of the redox function of APE1/Ref-1 is a novel strategy that might lead to a possible application of APX3330 for the treatment of IBD.
Collapse
Affiliation(s)
- Lauren Sahakian
- Institute for Health and Sport, Victoria University; Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia
| | - Rhiannon T Filippone
- Institute for Health and Sport, Victoria University; Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia
| | - Rhian Stavely
- Institute for Health and Sport, Victoria University; Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia,Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ainsley M Robinson
- Institute for Health and Sport, Victoria University; Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia
| | - Xu Sean Yan
- Institute for Health and Sport, Victoria University; Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia
| | - Raquel Abalo
- Área de Farmacología y Nutrición y Unidad Asociada al Instituto de Química Médica (IQM) del Consejo Superior de Investigaciones Científicas (CSIC), Universidad Rey Juan Carlos (URJC), Alcorcón, Madrid, Spain,High Performance Research Group in Physiopathology and Pharmacology of the Digestive System at URJC, Alcorcón, Madrid, Spain
| | - Rajaraman Eri
- University of Tasmania, School of Health Sciences, Launceston, Tasmania, Australia
| | - Joel C Bornstein
- Department of Physiology, Melbourne University, Melbourne, Australia
| | - Mark R Kelley
- Indiana University Simon Comprehensive Cancer Center, Departments of Pediatrics, Biochemistry & Molecular Biology and Pharmacology & Toxicology, Program in Pediatric Molecular Oncology & Experimental Therapeutics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine Indianapolis, USA
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University; Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia,Department of Medicine Western Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia,Regenerative Medicine and Stem Cells Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, Victoria, Australia,Address correspondence to: Kulmira Nurgali, Level 4, Research Labs, Western Centre for Health Research & Education, Sunshine Hospital, 176 Furlong Road, St Albans, 3021, VIC, Australia. E-mail:
| |
Collapse
|
18
|
APEX1 Expression as a Potential Diagnostic Biomarker of Clear Cell Renal Cell Carcinoma and Hepatobiliary Carcinomas. J Clin Med 2019; 8:jcm8081151. [PMID: 31375000 PMCID: PMC6723795 DOI: 10.3390/jcm8081151] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 07/29/2019] [Accepted: 07/31/2019] [Indexed: 12/28/2022] Open
Abstract
Apurinic/apyrimidinic endonuclease 1/redox effector factor 1 (APEX1) has been known to play key roles in DNA repair, the regulation of diverse transcriptional activity, and cellular responses to redox activity. This study aimed to examine serum APEX1 (s-APEX1) expression as a possible screening biomarker for clear cell renal cell carcinoma (ccRCC), hepatocellular carcinoma (HCC), and proximal and distal cholangiocarcinoma (CC). A total of 216 frozen serum samples were collected from 39 healthy control cases, 32 patients with ≥58 copies/mL of hepatitis B viral DNA (HBV DNA (+)), 40 ccRCC cases, 59 HCC cases, and 46 CC cases. The serum samples were examined for s-APEX1 concentration by enzyme-linked immunosorbent assay. The association of APEX1 expression with clinicopathological characteristics was also studied by immunohistochemical staining in 106 ccRCC, 131 HCC, and 32 intrahepatic CC cases. The median s-APEX1 concentrations of the HCC, CC, ccRCC, healthy control, and HBV DNA (+) groups were 0.294, 0.710, 0.474, 0.038, and 2.384 ng/mL, respectively (p < 0.001). Univariate and multivariate analyses revealed that increased cytoplasmic APEX1 expression led to a shorter disease-free survival period in HCC and CC cases. We suggest that the s-APEX1 level could be a potential diagnostic biomarker of ccRCC, HCC, and CC. Additionally, cytoplasmic APEX1 expression in cancer cells could be used to predict relapses in patients with HCC or CC.
Collapse
|
19
|
Li H, Zhong C, Wang Q, Chen W, Yuan Y. Curcumin is an APE1 redox inhibitor and exhibits an antiviral activity against KSHV replication and pathogenesis. Antiviral Res 2019; 167:98-103. [PMID: 31034848 DOI: 10.1016/j.antiviral.2019.04.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 01/06/2023]
Abstract
Curcumin, a polyphenol, is the main bioactive compound in dietary spice turmeric curcuma longa. It possesses anti-inflammatory, anti-oxidant and anti-neoplastic properties and shows potentials in treating or preventing particular diseases such as oxidative and inflammatory conditions, metabolic syndrome, arthritis, anxiety, hyperlipidemia and cancers. The diverse range and potential health beneficial effects has generated enthusiasm leading to intensive investigation into the phytochemical. However, a concern has been also raised if curcumin has a promiscuous bioassay profile and is a Pan-Assay INterference compound (PAINS). Here we present evidence indicating that curcumin is not a PAINS, but an inhibitor to APE1 redox function that affects many genes and pathways. This discovery explains the wide range of effects of curcumin on diverse human diseases and predicts a potential application in treatment of viral infection and virus-associated cancer. As a proof-of-concept, we demonstrated that curcumin is able to efficiently block Kaposi's sarcoma-associated herpesvirus replication and inhibit the pathogenic processes of angiogenesis and cell invasion.
Collapse
Affiliation(s)
- He Li
- Department of Microbiology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, 19104, USA; Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, China
| | - Canrong Zhong
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Qian Wang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Weikang Chen
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yan Yuan
- Department of Microbiology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, 19104, USA; Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
20
|
Kidane D. Molecular Mechanisms of H. pylori-Induced DNA Double-Strand Breaks. Int J Mol Sci 2018; 19:ijms19102891. [PMID: 30249046 PMCID: PMC6213211 DOI: 10.3390/ijms19102891] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/11/2018] [Accepted: 09/21/2018] [Indexed: 12/17/2022] Open
Abstract
Infections contribute to carcinogenesis through inflammation-related mechanisms. H. pylori infection is a significant risk factor for gastric carcinogenesis. However, the molecular mechanism by which H. pylori infection contributes to carcinogenesis has not been fully elucidated. H. pylori-associated chronic inflammation is linked to genomic instability via reactive oxygen and nitrogen species (RONS). In this article, we summarize the current knowledge of H. pylori-induced double strand breaks (DSBs). Furthermore, we provide mechanistic insight into how processing of oxidative DNA damage via base excision repair (BER) leads to DSBs. We review recent studies on how H. pylori infection triggers NF-κB/inducible NO synthase (iNOS) versus NF-κB/nucleotide excision repair (NER) axis-mediated DSBs to drive genomic instability. This review discusses current research findings that are related to mechanisms of DSBs and repair during H. pylori infection.
Collapse
Affiliation(s)
- Dawit Kidane
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Dell Pediatric Research Institute, 1400 Barbara Jordan Blvd. R1800, Austin, TX 78723, USA.
| |
Collapse
|
21
|
The Role of Interleukin-8 and Its Mechanism in Patients with Breast Cancer: Its Relation with Oxidative Stress and Estrogen Receptor. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2017. [DOI: 10.5812/ijcm.8791] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
22
|
Abstract
Reduction-oxidation factor 1-apurinic/apyrimidinic endonuclease (Ref-1/APE1) is a critical node in tumor cells, both as a redox regulator of transcription factor activation and as part of the DNA damage response. As a redox signaling protein, Ref-1/APE1 enhances the transcriptional activity of STAT3, HIF-1α, nuclear factor kappa B, and other transcription factors to promote growth, migration, and survival in tumor cells as well as inflammation and angiogenesis in the tumor microenvironment. Ref-1/APE1 is activated in a variety of cancers, including prostate, colon, pancreatic, ovarian, lung and leukemias, leading to increased aggressiveness. Transcription factors downstream of Ref-1/APE1 are key contributors to many cancers, and Ref-1/APE1 redox signaling inhibition slows growth and progression in a number of tumor types. Ref-1/APE1 inhibition is also highly effective when paired with other drugs, including standard-of-care therapies and therapies targeting pathways affected by Ref-1/APE1 redox signaling. Additionally, Ref-1/APE1 plays a role in a variety of other indications, such as retinopathy, inflammation, and neuropathy. In this review, we discuss the functional consequences of activation of the Ref-1/APE1 node in cancer and other diseases, as well as potential therapies targeting Ref-1/APE1 and related pathways in relevant diseases. APX3330, a novel oral anticancer agent and the first drug to target Ref-1/APE1 for cancer is entering clinical trials and will be explored in various cancers and other diseases bringing bench discoveries to the clinic.
Collapse
|
23
|
Pérez S, Taléns-Visconti R, Rius-Pérez S, Finamor I, Sastre J. Redox signaling in the gastrointestinal tract. Free Radic Biol Med 2017; 104:75-103. [PMID: 28062361 DOI: 10.1016/j.freeradbiomed.2016.12.048] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 12/20/2016] [Accepted: 12/31/2016] [Indexed: 12/16/2022]
Abstract
Redox signaling regulates physiological self-renewal, proliferation, migration and differentiation in gastrointestinal epithelium by modulating Wnt/β-catenin and Notch signaling pathways mainly through NADPH oxidases (NOXs). In the intestine, intracellular and extracellular thiol redox status modulates the proliferative potential of epithelial cells. Furthermore, commensal bacteria contribute to intestine epithelial homeostasis through NOX1- and dual oxidase 2-derived reactive oxygen species (ROS). The loss of redox homeostasis is involved in the pathogenesis and development of a wide diversity of gastrointestinal disorders, such as Barrett's esophagus, esophageal adenocarcinoma, peptic ulcer, gastric cancer, ischemic intestinal injury, celiac disease, inflammatory bowel disease and colorectal cancer. The overproduction of superoxide anion together with inactivation of superoxide dismutase are involved in the pathogenesis of Barrett's esophagus and its transformation to adenocarcinoma. In Helicobacter pylori-induced peptic ulcer, oxidative stress derived from the leukocyte infiltrate and NOX1 aggravates mucosal damage, especially in HspB+ strains that downregulate Nrf2. In celiac disease, oxidative stress mediates most of the cytotoxic effects induced by gluten peptides and increases transglutaminase levels, whereas nitrosative stress contributes to the impairment of tight junctions. Progression of inflammatory bowel disease relies on the balance between pro-inflammatory redox-sensitive pathways, such as NLRP3 inflammasome and NF-κB, and the adaptive up-regulation of Mn superoxide dismutase and glutathione peroxidase 2. In colorectal cancer, redox signaling exhibits two Janus faces: On the one hand, NOX1 up-regulation and derived hydrogen peroxide enhance Wnt/β-catenin and Notch proliferating pathways; on the other hand, ROS may disrupt tumor progression through different pro-apoptotic mechanisms. In conclusion, redox signaling plays a critical role in the physiology and pathophysiology of gastrointestinal tract.
Collapse
Affiliation(s)
- Salvador Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Burjasot, 46100 Valencia, Spain
| | - Raquel Taléns-Visconti
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, Faculty of Pharmacy, University of Valencia, Burjasot, 46100 Valencia, Spain
| | - Sergio Rius-Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Burjasot, 46100 Valencia, Spain
| | - Isabela Finamor
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Burjasot, 46100 Valencia, Spain
| | - Juan Sastre
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Burjasot, 46100 Valencia, Spain.
| |
Collapse
|
24
|
den Hartog G, Chattopadhyay R, Ablack A, Hall EH, Butcher LD, Bhattacharyya A, Eckmann L, Harris PR, Das S, Ernst PB, Crowe SE. Regulation of Rac1 and Reactive Oxygen Species Production in Response to Infection of Gastrointestinal Epithelia. PLoS Pathog 2016; 12:e1005382. [PMID: 26761793 PMCID: PMC4711900 DOI: 10.1371/journal.ppat.1005382] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 12/12/2015] [Indexed: 12/15/2022] Open
Abstract
Generation of reactive oxygen species (ROS) during infection is an immediate host defense leading to microbial killing. APE1 is a multifunctional protein induced by ROS and after induction, protects against ROS-mediated DNA damage. Rac1 and NAPDH oxidase (Nox1) are important contributors of ROS generation following infection and associated with gastrointestinal epithelial injury. The purpose of this study was to determine if APE1 regulates the function of Rac1 and Nox1 during oxidative stress. Gastric or colonic epithelial cells (wild-type or with suppressed APE1) were infected with Helicobacter pylori or Salmonella enterica and assessed for Rac1 and NADPH oxidase-dependent superoxide production. Rac1 and APE1 interactions were measured by co-immunoprecipitation, confocal microscopy and proximity ligation assay (PLA) in cell lines or in biopsy specimens. Significantly greater levels of ROS were produced by APE1-deficient human gastric and colonic cell lines and primary gastric epithelial cells compared to control cells after infection with either gastric or enteric pathogens. H. pylori activated Rac1 and Nox1 in all cell types, but activation was higher in APE1 suppressed cells. APE1 overexpression decreased H. pylori-induced ROS generation, Rac1 activation, and Nox1 expression. We determined that the effects of APE1 were mediated through its N-terminal lysine residues interacting with Rac1, leading to inhibition of Nox1 expression and ROS generation. APE1 is a negative regulator of oxidative stress in the gastrointestinal epithelium during bacterial infection by modulating Rac1 and Nox1. Our results implicate APE1 in novel molecular interactions that regulate early stress responses elicited by microbial infections. Helicobacter pylori infection of the gastric mucosa is largely lifelong leading to continued stimulation of immune cells. This results in the generation of reactive oxygen species (ROS) which are produced to kill bacteria, but at the same time ROS regulate cellular events in the host. However, prolonged generation of ROS has been implicated in damage of DNA, which ultimately could lead to the development of cancer. We studied a molecule known as APE-1 in gastric and intestinal cells, which is activated upon encounter of ROS. Our results show that APE1 limits the production of ROS in cells that form the lining of the gastrointestinal tract. APE1 regulates ROS production by inhibiting activation of the molecule Rac1. Inhibition of ROS production by APE1 occurred after infection of gastric cells with Helicobacter pylori and after Salmonella infection of intestinal cells. These data demonstrate that APE1 inhibits production of ROS in cells that line the inside of the digestive tract.
Collapse
Affiliation(s)
- Gerco den Hartog
- Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Ranajoy Chattopadhyay
- Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Amber Ablack
- Department of Pathology, University of California, San Diego, La Jolla, California, United States of America
| | - Emily H. Hall
- Department of Surgery, University of Virginia, Charlottesville, Virginia, United States of America
| | - Lindsay D. Butcher
- Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Asima Bhattacharyya
- National Institute of Science Education and Research (NISER), Bhubaneswar, India
| | - Lars Eckmann
- Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Paul R. Harris
- Division of Pediatrics, Unit of Gastroenterology and Nutrition, School of Medicine, Pontifical Catholic University, Santiago, Chile
| | - Soumita Das
- Department of Pathology, University of California, San Diego, La Jolla, California, United States of America
| | - Peter B. Ernst
- Department of Pathology, University of California, San Diego, La Jolla, California, United States of America
| | - Sheila E. Crowe
- Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
25
|
Thakur S, Sarkar B, Cholia RP, Gautam N, Dhiman M, Mantha AK. APE1/Ref-1 as an emerging therapeutic target for various human diseases: phytochemical modulation of its functions. Exp Mol Med 2014; 46:e106. [PMID: 25033834 PMCID: PMC4119211 DOI: 10.1038/emm.2014.42] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 01/27/2014] [Accepted: 03/05/2014] [Indexed: 12/12/2022] Open
Abstract
Apurinic/apyrimidinic endonuclease 1 (APE1) is a multifunctional enzyme involved in the base excision repair (BER) pathway, which repairs oxidative base damage caused by endogenous and exogenous agents. APE1 acts as a reductive activator of many transcription factors (TFs) and has also been named redox effector factor 1, Ref-1. For example, APE1 activates activator protein-1, nuclear factor kappa B, hypoxia-inducible factor 1α, paired box gene 8, signal transducer activator of transcription 3 and p53, which are involved in apoptosis, inflammation, angiogenesis and survival pathways. APE1/Ref-1 maintains cellular homeostasis (redox) via the activation of TFs that regulate various physiological processes and that crosstalk with redox balancing agents (for example, thioredoxin, catalase and superoxide dismutase) by controlling levels of reactive oxygen and nitrogen species. The efficiency of APE1/Ref-1's function(s) depends on pairwise interaction with participant protein(s), the functions regulated by APE1/Ref-1 include the BER pathway, TFs, energy metabolism, cytoskeletal elements and stress-dependent responses. Thus, APE1/Ref-1 acts as a ‘hub-protein' that controls pathways that are important for cell survival. In this review, we will discuss APE1/Ref-1's versatile nature in various human etiologies, including neurodegeneration, cancer, cardiovascular and other diseases that have been linked with alterations in the expression, subcellular localization and activities of APE/Ref-1. APE1/Ref-1 can be targeted for therapeutic intervention using natural plant products that modulate the expression and functions of APE1/Ref-1. In addition, studies focusing on translational applications based on APE1/Ref-1-mediated therapeutic interventions are discussed.
Collapse
Affiliation(s)
- Shweta Thakur
- Center for Biosciences, School of Basic and Applied Sciences, Central University of Punjab, Punjab, India
| | - Bibekananda Sarkar
- Center for Biosciences, School of Basic and Applied Sciences, Central University of Punjab, Punjab, India
| | - Ravi P Cholia
- Center for Biosciences, School of Basic and Applied Sciences, Central University of Punjab, Punjab, India
| | - Nandini Gautam
- Center for Environmental Science and Technology, School of Environment and Earth Sciences, Central University of Punjab, Punjab, India
| | - Monisha Dhiman
- Center for Genetic Diseases and Molecular Medicine, School of Emerging Life Science Technologies, Central University of Punjab, Punjab, India
| | - Anil K Mantha
- 1] Center for Biosciences, School of Basic and Applied Sciences, Central University of Punjab, Punjab, India [2] Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
26
|
Bhattacharyya A, Chattopadhyay R, Mitra S, Crowe SE. Oxidative stress: an essential factor in the pathogenesis of gastrointestinal mucosal diseases. Physiol Rev 2014; 94:329-54. [PMID: 24692350 DOI: 10.1152/physrev.00040.2012] [Citation(s) in RCA: 1507] [Impact Index Per Article: 137.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Reactive oxygen species (ROS) are generated as by-products of normal cellular metabolic activities. Superoxide dismutase, glutathione peroxidase, and catalase are the enzymes involved in protecting cells from the damaging effects of ROS. ROS are produced in response to ultraviolet radiation, cigarette smoking, alcohol, nonsteroidal anti-inflammatory drugs, ischemia-reperfusion injury, chronic infections, and inflammatory disorders. Disruption of normal cellular homeostasis by redox signaling may result in cardiovascular, neurodegenerative diseases and cancer. ROS are produced within the gastrointestinal (GI) tract, but their roles in pathophysiology and disease pathogenesis have not been well studied. Despite the protective barrier provided by the mucosa, ingested materials and microbial pathogens can induce oxidative injury and GI inflammatory responses involving the epithelium and immune/inflammatory cells. The pathogenesis of various GI diseases including peptic ulcers, gastrointestinal cancers, and inflammatory bowel disease is in part due to oxidative stress. Unraveling the signaling events initiated at the cellular level by oxidative free radicals as well as the physiological responses to such stress is important to better understand disease pathogenesis and to develop new therapies to manage a variety of conditions for which current therapies are not always sufficient.
Collapse
|
27
|
Cesaratto L, Codarin E, Vascotto C, Leonardi A, Kelley MR, Tiribelli C, Tell G. Specific inhibition of the redox activity of ape1/ref-1 by e3330 blocks tnf-α-induced activation of IL-8 production in liver cancer cell lines. PLoS One 2013; 8:e70909. [PMID: 23967134 PMCID: PMC3744539 DOI: 10.1371/journal.pone.0070909] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 06/24/2013] [Indexed: 12/23/2022] Open
Abstract
APE1/Ref-1 is a main regulator of cellular response to oxidative stress via DNA-repair function and co-activating activity on the NF-κB transcription factor. APE1 is central in controlling the oxidative stress-based inflammatory processes through modulation of cytokines expression and its overexpression is responsible for the onset of chemoresistance in different tumors including hepatic cancer. We examined the functional role of APE1 overexpression during hepatic cell damage related to fatty acid accumulation and the role of the redox function of APE1 in the inflammatory process. HepG2 cells were stably transfected with functional and non-functional APE1 encoding plasmids and the protective effect of APE1 overexpression toward genotoxic compounds or FAs accumulation, was tested. JHH6 cells were stimulated with TNF-α in the presence or absence of E3330, an APE1 redox inhibitor. IL-8 promoter activity was assessed by a luciferase reporter assay, gene expression by Real-Time PCR and cytokines (IL-6, IL-8, IL-12) levels measured by ELISA. APE1 over-expression did not prevent cytotoxicity induced by lipid accumulation. E3330 treatment prevented the functional activation of NF-κB via the alteration of APE1 subcellular trafficking and reduced IL-6 and IL-8 expression induced by TNF-α and FAs accumulation through blockage of the redox-mediated activation of NF-κB. APE1 overexpression observed in hepatic cancer cells may reflect an adaptive response to cell damage and may be responsible for further cell resistance to chemotherapy and for the onset of inflammatory response. The efficacy of the inhibition of APE1 redox activity in blocking TNF-α and FAs induced inflammatory response opens new perspectives for treatment of inflammatory-based liver diseases.
Collapse
Affiliation(s)
- Laura Cesaratto
- Dipartimento di Scienze Mediche e Biologiche, Università di Udine, Udine, Italy
- Fondazione Italiana Fegato, AREA Science Park, Basovizza, Trieste, Italy
| | - Erika Codarin
- Dipartimento di Scienze Mediche e Biologiche, Università di Udine, Udine, Italy
| | - Carlo Vascotto
- Dipartimento di Scienze Mediche e Biologiche, Università di Udine, Udine, Italy
| | - Antonio Leonardi
- Dipartimento di Patologia Cellulare e Molecolare ‘L. Califano’ Università “Federico II” di Napoli, Napoli, Italy
| | - Mark R. Kelley
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, Indiana, United States of America
| | - Claudio Tiribelli
- Fondazione Italiana Fegato, AREA Science Park, Basovizza, Trieste, Italy
- Dipartimento Scienze Cliniche, Università di Trieste, Trieste, Italy
| | - Gianluca Tell
- Dipartimento di Scienze Mediche e Biologiche, Università di Udine, Udine, Italy
- * E-mail:
| |
Collapse
|
28
|
Sankpal NV, Fleming TP, Gillanders WE. EpCAM modulates NF-κB signaling and interleukin-8 expression in breast cancer. Mol Cancer Res 2013; 11:418-26. [PMID: 23378578 DOI: 10.1158/1541-7786.mcr-12-0518] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The epithelial cell adhesion molecule (EpCAM) is a 40-kD type I transmembrane protein that is overexpressed in human epithelial cancers and is currently the target of molecular therapy based on its overexpression at the cell surface. Recently, we and others have shown a role for EpCAM in cell signaling and carcinogenesis, and EpCAM expression seems to promote breast cancer invasion. Interleukin-8 (IL-8/CXCL-8) is an inflammatory cytokine that has recently been shown to modulate breast cancer invasion and angiogenesis. In preliminary experiments, we identified a correlation between EpCAM and IL-8 expression in primary human breast cancers. Specific ablation of EpCAM in breast cancer cell lines results in decreased IL-8 expression, and IL-8 contributes to EpCAM-dependent breast cancer invasion. Specific ablation of EpCAM is also associated with decreased NF-κB transcription factor activity, decreased phosphorylation of the NF-κB family member RELA, and increased IκBα protein expression. EpCAM modulates IL-8 expression at baseline, and following IL-1β stimulation, which is known to be a potent inducer of NF-κB in breast cancer. In functional rescue experiments, specific ablation of RELA or forced expression of the NF-κB inhibitor protein IκBα prevented EpCAM-dependent rescue of IL-8 promoter activity. These studies show for the first time that EpCAM can modulate NF-κB transcription factor activity and IL-8 expression in breast cancer and confirm the role of EpCAM signaling in modulating breast cancer invasion. Further study is required to define the molecular mechanism(s) of EpCAM signaling in breast cancer and to direct the rational development of molecular therapies targeting EpCAM.
Collapse
Affiliation(s)
- Narendra V Sankpal
- Department of Surgery, Siteman Cancer Center, Washington University School of Medicine, Saint Louis, Missouri 63110, USA
| | | | | |
Collapse
|
29
|
Eftang LL, Esbensen Y, Tannæs TM, Bukholm IRK, Bukholm G. Interleukin-8 is the single most up-regulated gene in whole genome profiling of H. pylori exposed gastric epithelial cells. BMC Microbiol 2012; 12:9. [PMID: 22248188 PMCID: PMC3292955 DOI: 10.1186/1471-2180-12-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 01/17/2012] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The association between Helicobacter pylori infection and upper gastrointestinal disease is well established. However, only a small fraction of H. pylori carriers develop disease, and there are great geographical differences in disease penetrance. The explanation to this enigma lies in the interaction between the bacterium and the host. H. pylori Outer Membrane Phospholipase A (OMPLA) has been suggested to play a role in the virulence of this bacterium. The aim of this study was to profile the most significant cellular pathways and biological processes affected in gastric epithelial cells during 24 h of H. pylori exposure, and to study the inflammatory response to OMPLA⁺ and OMPLA⁻ H. pylori variants. RESULTS Interleukin-8 was the most significantly up-regulated gene and appears to play a paramount role in the epithelial cell response to H. pylori infection and in the pathological processes leading to gastric disease. MAPK and NF-kappaB cellular pathways were powerfully activated, but did not seem to explain the impressive IL-8 response. There was marked up-regulation of TP53BP2, whose corresponding protein ASPP2 may interact with H. pylori CagA and cause marked p53 suppression of apoptosis. Other regulators of apoptosis also showed abberant regulation. We also identified up-regulation of several oncogenes and down-regulation of tumor suppressor genes as early as during the first 24 h of infection. H. pylori OMPLA phase variation did not seem to influence the inflammatory epithelial cell gene response in this experiment. CONCLUSION In whole genome analysis of the epithelial response to H. pylori exposure, IL-8 demonstrated the most marked up-regulation, and was involved in many of the most important cellular response processes to the infection. There was dysregulation of apoptosis, tumor suppressor genes and oncogenes as early as in the first 24 h of H. pylori infection, which may represent early signs of gastric tumorigenesis. OMPLA⁺/⁻ did not affect the acute inflammatory response to H. pylori.
Collapse
Affiliation(s)
- Lars L Eftang
- Department of Clinical Molecular Biology (Epigen), Institute of Clinical Medicine, University of Oslo, Akershus University Hospital, Lørenskog, Norway
- Department of Gastroenterological Surgery, Akershus University Hospital, Lørenskog, Norway
| | - Ying Esbensen
- Department of Clinical Molecular Biology (Epigen), Institute of Clinical Medicine, University of Oslo, Akershus University Hospital, Lørenskog, Norway
| | - Tone M Tannæs
- Department of Clinical Molecular Biology (Epigen), Akershus University Hospital, Lørenskog, Norway
| | - Ida RK Bukholm
- Department of Gastroenterological Surgery, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Akershus University Hospital, University of Oslo, Lørenskog, Norway
| | - Geir Bukholm
- Institute of Health and Society, University of Oslo, Oslo, Norway
| |
Collapse
|
30
|
da Silva TA, Fontes FL, Coutinho LG, de Souza FRS, de Melo JTA, de Souto JT, Leib SL, Agnez-Lima LF. SNPs in DNA repair genes associated to meningitis and host immune response. Mutat Res 2011; 713:39-47. [PMID: 21651918 DOI: 10.1016/j.mrfmmm.2011.05.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2011] [Revised: 05/11/2011] [Accepted: 05/20/2011] [Indexed: 05/30/2023]
Abstract
In vitro and in animal models, APE1, OGG1, and PARP-1 have been proposed as being involved with inflammatory response. In this work, we have investigated if the SNPs APE1 Asn148Glu, OGG1 Ser326Cys, and PARP-1 Val762Ala are associated to meningitis. The patient genotypes were investigated by PIRA-PCR or PCR-RFLP. DNA damages were detected in genomic DNA by Fpg treatment. IgG and IgA were measured from plasma and the cytokines and chemokines were measured from cerebrospinal fluid samples using Bio-Plex assays. A higher frequency (P<0.05) of APE1 Glu allele in bacterial meningitis (BM) and aseptic meningitis (AM) patients was observed. The genotypes Asn/Asn in control group and Asn/Glu in BM group was also higher. For the SNP OGG1 Ser326Cys, the genotype Cys/Cys was more frequent (P<0.05) in BM group. The frequency of PARP-1 Val/Val genotype was higher in control group (P<0.05). The occurrence of combined SNPs is significantly higher in BM patients, indicating that these SNPs may be associated to the disease. Increasing in sensitive sites to Fpg was observed in carriers of APE1 Glu allele or OGG1 Cys allele, suggesting that SNPs affect DNA repair activity. Alterations in IgG production were observed in the presence of SNPs APE1 Asn148Glu, OGG1 Ser326Cys or PARP-1 Val762Ala. Moreover, reduction in the levels of IL-6, IL-1Ra, MCP-1/CCL2 and IL-8/CXCL8 was observed in the presence of APE1 Glu allele in BM patients. In conclusion, we obtained indications of an effect of SNPs in DNA repair genes on the regulation of immune response in meningitis.
Collapse
Affiliation(s)
- Thayse Azevedo da Silva
- Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte (UFRN), Av. Salgado Filho s/n, 59072-970 Natal, RN, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Xie JY, Li MX, Xiang DB, Mou JH, Qing Y, Zeng LL, Yang ZZ, Guan W, Wang D. Elevated Expression of APE1/Ref-1 and its Regulation on IL-6 and IL-8 in Bone Marrow Stromal Cells of Multiple Myeloma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2010; 10:385-93. [DOI: 10.3816/clml.2010.n.072] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
32
|
Chattopadhyay R, Bhattacharyya A, Crowe SE. Dual regulation by apurinic/apyrimidinic endonuclease-1 inhibits gastric epithelial cell apoptosis during Helicobacter pylori infection. Cancer Res 2010; 70:2799-808. [PMID: 20332233 DOI: 10.1158/0008-5472.can-09-4136] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Human apurinic/apyrimidinic endonuclease-1 (APE-1), a key enzyme involved in repair of oxidative DNA base damage, is an important transcriptional coregulator. We previously reported that Helicobacter pylori infection induces apoptosis and increases APE-1 expression in human gastric epithelial cells (GEC). Although both the DNA repair activity and the acetylation-mediated transcriptional regulation of APE-1 are required to prevent cell death, the mechanisms of APE-1-mediated inhibition of infection-induced apoptosis are unclear. Here, we show that short hairpin RNA-mediated stable suppression of APE-1 results in increased apoptosis in GEC after H. pylori infection. We show that programmed cell death involves both the caspase-9-mediated mitochondrial pathway and the caspase-8-dependent extrinsic pathway by measuring different markers for both the pathways. Overexpression of wild-type APE-1 in APE-1-suppressed GEC reduced apoptosis after infection; however, overexpression of the DNA repair mutant or the nonacetylable mutant of APE-1 alone was unable to reduce apoptosis, suggesting that both DNA repair and acetylation functions of APE-1 modulate programmed cell death. We show for the first time that the DNA repair activity of APE-1 inhibits the mitochondrial pathway, whereas the acetylation function inhibits the extrinsic pathway during H. pylori infection. Thus, our findings establish that the two different functions of APE-1 differentially regulate the intrinsic and the extrinsic pathway of H. pylori-mediated GEC apoptosis. As proapoptotic and antiapoptotic mechanisms determine the development and progression of gastritis, gastric ulceration, and gastric cancer, this dual regulatory role of APE-1 represents one of the important molecular strategies by H. pylori to sustain chronic infection.
Collapse
|
33
|
Lee HM, Yuk JM, Shin DM, Yang CS, Kim KK, Choi DK, Liang ZL, Kim JM, Jeon BH, Kim CD, Lee JH, Jo EK. Apurinic/apyrimidinic endonuclease 1 is a key modulator of keratinocyte inflammatory responses. THE JOURNAL OF IMMUNOLOGY 2009; 183:6839-48. [PMID: 19846872 DOI: 10.4049/jimmunol.0901856] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Apurinic/apyrimidinic endonuclease 1/redox factor-1 (APE1) functions in both DNA repair and redox signaling, making it an attractive emerging therapeutic target. However, the role of APE1 in cutaneous inflammatory responses is largely unknown. In this study, we report that APE1 is a key upstream regulator in TLR2-dependent keratinocyte inflammatory responses. We found that nuclear expression of APE1 in epidermal layers was markedly up-regulated in psoriatic skin. APE1 was essential for the transcriptional activation and nuclear translocation of hypoxia-inducible factor-1alpha and NF-kappaB, both of which are crucial for inflammatory signaling in keratinocytes. Moreover, APE1 played a crucial role in the expression of TLR2-mediated inflammatory mediators, including TNF-alpha, CXCL8, and LL-37, in HaCaT cells and human primary keratinocytes. Silencing of APE1 attenuated cyclin D1/cyclin-dependent kinase 4 expression and phosphorylation of ERK1/2 and Akt, thereby affecting keratinocyte proliferation. Importantly, TLR2-induced generation of reactive oxygen species contributed to the nuclear translocation and expression of APE1, suggesting an autoregulatory circuit in which the subcellular localization of APE1 is associated with the production of APE1 per se through reactive oxygen species-dependent signaling. Taken together, these findings establish a role for APE1 as a master regulator of TLR2-dependent inflammatory responses in human keratinocytes.
Collapse
Affiliation(s)
- Hye-Mi Lee
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Lamb A, Yang XD, Tsang YHN, Li JD, Higashi H, Hatakeyama M, Peek RM, Blanke SR, Chen LF. Helicobacter pylori CagA activates NF-kappaB by targeting TAK1 for TRAF6-mediated Lys 63 ubiquitination. EMBO Rep 2009; 10:1242-9. [PMID: 19820695 DOI: 10.1038/embor.2009.210] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Revised: 08/14/2009] [Accepted: 08/20/2009] [Indexed: 02/06/2023] Open
Abstract
Helicobacter pylori-initiated chronic gastritis is characterized by the cag pathogenicity island-dependent upregulation of proinflammatory cytokines, which is largely mediated by the transcription factor nuclear factor (NF)-kappaB. However, the cag pathogenicity island-encoded proteins and cellular signalling molecules that are involved in H. pylori-induced NF-kappaB activation and inflammatory response remain unclear. Here, we show that H. pylori virulence factor CagA and host protein transforming growth factor-beta-activated kinase 1 (TAK1) are essential for H. pylori-induced activation of NF-kappaB. CagA physically associates with TAK1 and enhances its activity and TAK1-induced NF-kappaB activation through the tumour necrosis factor receptor-associated factor 6-mediated, Lys 63-linked ubiquitination of TAK1. These findings show that polyubiquitination of TAK1 regulates the activation of NF-kappaB, which in turn is used by H. pylori CagA for the H. pylori-induced inflammatory response.
Collapse
Affiliation(s)
- Acacia Lamb
- Department of Biochemistry, College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Recent data from several studies suggest that oxidative stress is involved in the biochemical mechanisms that underlie neuropsychiatric disorders. The present study was designed to investigate oxidative stress status in depressive patients with gastric adenocarcinoma (GA) at TNM stage III. Oxidative stress, depression and expression of specific genes were monitored during a pretreatment period. Serum total antioxidant capacity, catalase, superoxide dismutase concentrations, and antisuperoxide anion capacity (A-ASC) were significantly decreased in depressive patients compared to control subjects, whereas serum malondialdehyde (MDA) levels were significantly increased. Importantly, the formation of 8-hydroxy-deoxyguanosine (8-OHdG) accumulated. Furthermore, SYBR Green real-time PCR revealed that the expression levels of human oxoguanine glycosylase 1 and APEX nuclease 1 (APEX1) were increased in depressive patients. Pearson correlation analysis revealed that depression was positively correlated with SAS, SCL-90, MDA, 8-OHdG and APEX1, but negatively correlated with A-ASC. Thus, this study confirms oxidative imbalance in depressive patients with GA, and oxidative stress may play a role in the onset and exacerbation of depression.
Collapse
|
36
|
Bhattaracharyya A, Chattopadhyay R, Burnette BR, Cross JV, Mitra S, Ernst PB, Bhakat KK, Crowe SE. Acetylation of apurinic/apyrimidinic endonuclease-1 regulates Helicobacter pylori-mediated gastric epithelial cell apoptosis. Gastroenterology 2009; 136:2258-69. [PMID: 19505426 PMCID: PMC2694750 DOI: 10.1053/j.gastro.2009.02.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2008] [Revised: 02/02/2009] [Accepted: 02/10/2009] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Helicobacter pylori-induced gastric epithelial cell (GEC) apoptosis is a complex process that includes activation of the tumor suppressor p53. p53-mediated apoptosis involves p53 activation, bax transcription, and cytochrome c release from mitochondria. Apurinic/apyrimidinic endonuclease-1 (APE-1) regulates transcriptional activity of p53, and H pylori induce APE-1 expression in human GECs. H pylori infection increases intracellular calcium ion concentration [Ca2+]i of GECs, which induces APE-1 acetylation. We investigated the effects of H pylori infection and APE-1 acetylation on GEC apoptosis. METHODS AGS cells (wild-type or with suppressed APE-1), KATO III cells, and cells isolated from gastric biopsy specimens were infected with H pylori. Effects were examined by immunoblotting, real-time reverse-transcription polymerase chain reaction, immunoprecipitation, immunofluorescence microscopy, chromatin immunoprecipitation, mobility shift, DNA binding, and luciferase assays. RESULTS H pylori infection increased [Ca2+]i and acetylation of APE-1 in GECs, but the acetylation status of APE-1 did not affect the transcriptional activity of p53. In GECs, expression of a form of APE-1 that could not be acetylated increased total and mitochondrial levels of Bax and induced release of cytochrome c and fragmentation of DNA; expression of wild-type APE-1 reduced these apoptotic events. We identified a negative calcium response element in the human bax promoter and found that poly (adenosine diphosphate-ribose) polymerase 1 recruited the acetylated APE-1/histone deacetylase-1 repressor complex to bax nCaRE. CONCLUSIONS H pylori-mediated acetylation of APE-1 suppresses Bax expression; this prevents p53-mediated apoptosis when H pylori infect GECs.
Collapse
Affiliation(s)
| | | | - Brent R. Burnette
- Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Janet V. Cross
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Sankar Mitra
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Peter B. Ernst
- Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Kishor K. Bhakat
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Sheila E. Crowe
- Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
37
|
O'Hara AM, Bhattacharyya A, Bai J, Mifflin RC, Ernst PB, Mitra S, Crowe SE. Tumor necrosis factor (TNF)-alpha-induced IL-8 expression in gastric epithelial cells: role of reactive oxygen species and AP endonuclease-1/redox factor (Ref)-1. Cytokine 2009; 46:359-69. [PMID: 19376732 DOI: 10.1016/j.cyto.2009.03.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2008] [Revised: 12/07/2008] [Accepted: 03/18/2009] [Indexed: 12/31/2022]
Abstract
TNF-alpha contributes to oxidative stress via induction of reactive oxygen species (ROS) and pro-inflammatory cytokines. The molecular basis of this is not well understood but it is partly mediated through the inducible expression of IL-8. As redox factor-1 (Ref-1), is an important mediator of redox-regulated gene expression we investigated whether ROS and Ref-1 modulate TNF-alpha-induced IL-8 expression in human gastric epithelial cells. We found that TNF-alpha treatment of AGS cells enhanced nuclear expression of Ref-1 and potently induced IL-8 expression. Overexpression of Ref-1 enhanced IL-8 gene transcription at baseline and after TNF-alpha treatment whereas Ref-1 suppression and antioxidant treatment inhibited TNF-alpha-stimulated IL-8 expression. TNF-alpha-mediated enhancement of other pro-inflammatory chemokines like MIP-3 alpha and Gro-alpha was also regulated by Ref-1. Although TNF-alpha increased DNA binding activity of Ref-1-regulated transcription factors, AP-1 and NF-kappaB, to the IL-8 promoter, promoter activity was mainly mediated by NF-kappaB binding. Silencing of Ref-1 in AGS cells inhibited basal and TNF-alpha-induced AP-1 and NF-kappaB DNA binding activity, but not their nuclear accumulation. Collectively, we provide the first mechanistic evidence of Ref-1 involvement in TNF-alpha-mediated, redox-sensitive induction of IL-8 and other chemokines in human gastric mucosa. This has implications for understanding the pathogenesis of gastrointestinal inflammatory disorders.
Collapse
Affiliation(s)
- Ann M O'Hara
- Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Yuk JM, Yang CS, Shin DM, Kim KK, Lee SK, Song YJ, Lee HM, Cho CH, Jeon BH, Jo EK. A dual regulatory role of apurinic/apyrimidinic endonuclease 1/redox factor-1 in HMGB1-induced inflammatory responses. Antioxid Redox Signal 2009; 11:575-88. [PMID: 18715145 DOI: 10.1089/ars.2008.2196] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Apurinic/apyrimidinic endonuclease 1/Redox factor-1 (APE1) is a multifunctional protein involved in reduction-oxidation regulation. High-mobility group box 1 (HMGB1) is released by necrotic cells and various inflammatory stimuli, acting as an inflammatory marker in sepsis and autoimmune diseases. Here, we report the dual regulatory role of APE1 in inflammatory signaling to extracellular HMGB1 or in the release of endogenous HMGB1 in human monocytes/macrophages. Forced cytoplasmic overexpression of APE1 profoundly attenuated the upregulation of HMGB1-mediated reactive oxygen species generation, cytokine secretion, and cyclooxygenase-2 expression by primary monocytes and macrophage-like THP-1 cell lines. In addition, HMGB1-induced activation of p38 and c-Jun N-terminal kinase (JNK), but not extracellular signal-regulated kinase 1/2, was strongly abrogated by the overexpression of APE1. The activation of apoptosis signal-regulating kinase 1 was required for both the p38 and JNK activation challenge with HMGB1. The extracellular release of HMGB1 by activated macrophages was inhibited by APE1 transfection. Small interfering RNA (siRNA) knockdown of endogenous APE1 impaired HMGB1-mediated cytokine expression and MAPK activation in THP-1 cells. HMGB1 stimulation induced the translocation of APE1 to the nucleus of the cell. In addition, APE1 silencing via siRNA transfection inhibited both the nuclear and cytoplasmic expression of APE1. These data identify APE1 as a novel dual regulator of inflammatory signaling to HMGB1 by human monocytes/macrophages. The modulation of cytosolic APE1 expression might be useful as a potential therapeutic modality for the treatment of inflammatory or autoimmune diseases.
Collapse
Affiliation(s)
- Jae-Min Yuk
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Bhakat KK, Mantha AK, Mitra S. Transcriptional regulatory functions of mammalian AP-endonuclease (APE1/Ref-1), an essential multifunctional protein. Antioxid Redox Signal 2009; 11:621-38. [PMID: 18715144 PMCID: PMC2933571 DOI: 10.1089/ars.2008.2198] [Citation(s) in RCA: 202] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The mammalian AP-endonuclease (APE1/Ref-1) plays a central role in the repair of oxidized and alkylated bases in mammalian genomes via the base excision repair (BER) pathway. However, APE1, unlike its E. coli prototype Xth, has two unique and apparently distinct transcriptional regulatory activities. APE1 functions as a redox effector factor (Ref-1) for several transcription factors including AP-1, HIF1-alpha, and p53. APE1 was also identified as a direct trans-acting factor for repressing human parathyroid hormone (PTH) and renin genes by binding to the negative calcium-response element (nCaRE) in their promoters. We have characterized APE1's post-translational modification, namely, acetylation which modulates its transcriptional regulatory function. Furthermore, stable interaction of APE1 with several other trans-acting factors including HIF-1alpha, STAT3, YB-1, HDAC1, and CBP/p300 and formation of distinct trans-acting complexes support APE1's direct regulatory function for diverse genes. Multiple functions of mammalian APE1, both in DNA repair and gene regulation, warrant extensive analysis of its own regulation and dissection of the mechanisms. In this review, we have discussed APE1's own regulation and its role as a transcriptional coactivator or corepressor by both redox-dependent and redox-independent (acetylation-mediated) mechanisms, and explore the potential utility of targeting these functions for enhancing drug sensitivity of cancer cells.
Collapse
Affiliation(s)
- Kishor K Bhakat
- Department of Biochemistry and Molecular Biology, and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas 77555, USA.
| | | | | |
Collapse
|
40
|
Tell G, Quadrifoglio F, Tiribelli C, Kelley MR. The many functions of APE1/Ref-1: not only a DNA repair enzyme. Antioxid Redox Signal 2009; 11:601-20. [PMID: 18976116 PMCID: PMC2811080 DOI: 10.1089/ars.2008.2194] [Citation(s) in RCA: 365] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
APE1/Ref-1 (APE1), the mammalian ortholog of Escherichia coli Xth, and a multifunctional protein possessing both DNA repair and transcriptional regulatory activities, has a pleiotropic role in controlling cellular response to oxidative stress. APE1 is the main apurinic/apyrimidinic endonuclease in eukaryotic cells, playing a central role in the DNA base excision repair pathway of all DNA lesions (uracil, alkylated and oxidized, and abasic sites), including single-strand breaks, and has also cotranscriptional activity by modulating genes expression directly regulated by either ubiquitous (i.e., AP-1, Egr-1, NFkappa-B, p53, and HIF) and tissue specific (i.e., PEBP-2, Pax-5 and -8, and TTF-1) transcription factors. In addition, it controls the intracellular redox state by inhibiting the reactive oxygen species (ROS) production. At present, information is still inadequate regarding the molecular mechanisms responsible for the coordinated control of its several activities. Both expression and/or subcellular localization are altered in several metabolic and proliferative disorders such as in tumors and aging. Here, we have attempted to coalesce the most relevant information concerning APE1's different functions in order to shed new light and to focus current and future studies to fully understand this unique molecule that is acquiring more and more interest and translational relevance in the field of molecular medicine.
Collapse
Affiliation(s)
- Gianluca Tell
- Department of Biomedical Sciences and Technologies, University of Udine, Udine, Italy.
| | | | | | | |
Collapse
|
41
|
Yang S, Meyskens FL. Apurinic/apyrimidinic endonuclease/redox effector factor-1(APE/Ref-1): a unique target for the prevention and treatment of human melanoma. Antioxid Redox Signal 2009; 11:639-50. [PMID: 18715151 PMCID: PMC2933576 DOI: 10.1089/ars.2008.2226] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Management of melanoma is a growing and challenging public health issue requiring novel and multidisciplinary approaches to achieve more efficient prevention and therapeutic benefits. The aim of this article is to show the critical role of APE/Ref-1 on melanomagenesis and progression. APE/Ref-1 serves as a redox-sensitive node of convergence of various signals as well as a DNA-repair enzyme, and its activation protects melanocytes and melanoma cells from chronic oxidative stress and promotes cell survival via mediation of downstream pathways. APE/Ref-1 is a strong candidate as a potential drug-treatable target for the prevention and treatment of human melanoma. Lead compounds exhibiting inhibitory effects on APE/Ref-1 are also reviewed. We anticipate potential clinical benefit in the future through inhibition of APE/Ref-1 and/or Ref-1-mediated signaling.
Collapse
Affiliation(s)
- Sun Yang
- Chao Family Comprehensive Cancer Center, Department of Medicine, Orange, California, USA
| | | |
Collapse
|
42
|
Futagami S, Hiratsuka T, Shindo T, Horie A, Hamamoto T, Suzuki K, Kusunoki M, Miyake K, Gudis K, Crowe SE, Tsukui T, Sakamoto C. Expression of apurinic/apyrimidinic endonuclease-1 (APE-1) in H. pylori-associated gastritis, gastric adenoma, and gastric cancer. Helicobacter 2008; 13:209-18. [PMID: 18466396 DOI: 10.1111/j.1523-5378.2008.00605.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND AIM Apurinic/apyrimidinic endonuclease-1 (APE-1) is a key enzyme in DNA base excision repair (BER), linked to cancer chemosensitivity. However, little is known about the localization of APE-1 in Helicobacter pylori-infected gastric mucosa or its role in the development of gastric cancer. To investigate the role of APE-1 in the development of gastric cancer, we examined APE-1 expression and localization in cultured cells and gastric biopsies from patients with H. pylori-infected gastritis or gastric adenoma, and from surgically resected gastric cancer. METHODS APE-1 mRNA and protein expression were determined in H. pylori (CagA+) water-extract protein (HPWEP)-stimulated MKN-28 cells, gastric adenocarcinoma cell-line (AGS) cells, and human peripheral macrophages by real-time polymerase chain reaction and Western blot analysis. APE-1 expression and 8-OHdG as a measure of oxidative DNA damage were evaluated by immunostaining. Localization of APE-1 and IkappaBalpha phosphorylation in gastric adenoma and gastric cancer tissues were evaluated by single- and double-label immunohistochemistry. RESULTS In studies in vitro, HPWEP-stimulation significantly increased APE-1 mRNA expression levels in both MKN-28 cells and human peripheral macrophages. Hypo/reoxygenation treatment significantly increased APE-1 protein expression in HPWEP-stimulated MKN-28 cells. HPWEP stimulation significantly increased both APE-1 expression and IkappaBalpha phosphorylation levels in MKN-28 and AGS cells. In human tissues, APE-1 expression in H. pylori-infected gastritis without goblet cell metaplasia was significantly increased as compared to that in tissues from uninfected subjects. Eradication therapy significantly reduced both APE-1 and 8-OHdG expression levels in the gastric mucosa. APE-1 expression was mainly localized in epithelial cells within gastric adenoma and in mesenchymal cells of gastric cancer tissues. APE-1 expression in gastric cancer tissues was significantly reduced compared to that in H. pylori-infected gastric adenoma, while 8-OHdG index and IkappaBalpha phosphorylation levels did not differ between these two neoplastic tissue types. Co-localization of APE-1 and IkappaBalpha phosphorylation was observed not in gastric cancer cells but in gastric adenoma cells. CONCLUSION H. pylori infection is associated with increased APE-1 expression in human cell lines and in gastric tissues from subjects with gastritis and gastric adenomas. The observed distinct expression patterns of APE-1 and 8-OHdG in gastric adenoma and gastric cancer tissues may provide insight into the progression of these conditions and warrants further investigation.
Collapse
Affiliation(s)
- Seiji Futagami
- Division of Gastroenterology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Hisatsune J, Nakayama M, Isomoto H, Kurazono H, Mukaida N, Mukhopadhyay AK, Azuma T, Yamaoka Y, Sap J, Yamasaki E, Yahiro K, Moss J, Hirayama T. Molecular characterization of Helicobacter pylori VacA induction of IL-8 in U937 cells reveals a prominent role for p38MAPK in activating transcription factor-2, cAMP response element binding protein, and NF-kappaB activation. THE JOURNAL OF IMMUNOLOGY 2008; 180:5017-27. [PMID: 18354227 DOI: 10.4049/jimmunol.180.7.5017] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Helicobacter pylori VacA induces multiple effects on susceptible cells, including vacuolation, mitochondrial damage, inhibition of cell growth, and enhanced cyclooxygenase-2 expression. To assess the ability of H. pylori to modulate the production of inflammatory mediators, we examined the mechanisms by which VacA enhanced IL-8 production by promonocytic U937 cells, which demonstrated the greatest VacA-induced IL-8 release of the cells tested. Inhibitors of p38 MAPK (SB203580), ERK1/2 (PD98059), IkappaBalpha ((E)-3-(4-methylphenylsulfonyl)-2-propenenitrile), Ca(2+) entry (SKF96365), and intracellular Ca(2+) channels (dantrolene) blocked VacA-induced IL-8 production. Furthermore, an intracellular Ca(2+) chelator (BAPTA-AM), which inhibited VacA-activated p38 MAPK, caused a dose-dependent reduction in VacA-induced IL-8 secretion by U937 cells, implying a role for intracellular Ca(2+) in mediating activation of MAPK and the canonical NF-kappaB pathway. VacA stimulated translocation of NF-kappaBp65 to the nucleus, consistent with enhancement of IL-8 expression by activation of the NF-kappaB pathway. In addition, small interfering RNA of activating transcription factor (ATF)-2 or CREB, which is a p38MAPK substrate and binds to the AP-1 site of the IL-8 promoter, inhibited VacA-induced IL-8 production. VacA activated an IL-8 promoter containing an NF-IL-6 site, but not a mutated AP-1 or NF-kappaB site, suggesting direct involvement of the ATF-2/CREB binding region or NF-kappaB-binding regions in VacA-induced IL-8 promoter activation. Thus, in U937 cells, VacA directly increases IL-8 production by activation of the p38 MAPK via intracellular Ca(2+) release, leading to activation of the transcription factors, ATF-2, CREB, and NF-kappaB.
Collapse
Affiliation(s)
- Junzo Hisatsune
- Department of Bacteriology, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Helicobacter pylori infects almost 50% of the world population and is the major cause of gastroduodenal diseases. H. pylori colonizes the gastric mucosa, activates Toll-like and Nod-like receptors, and usually elicits a T helper 1 (Th1) type of immune response, fully polarized in peptic ulcer patients. Among several bacterial factors, the neutrophil-activating protein represents a key factor driving Th1 inflammation. A complex and fascinating balance between H. pylori and host factors takes part in the gastric niche and allows the majority of infected individuals to be without any symptom during their entire life. Novel insights into the innate and adaptive responses against H. pylori, dealing with regulatory T cells and cytokines, CTLA-4 molecule, cholesterol glucosylation, and immune evasion have been elucidated during the past year and are discussed for the development of an effective vaccine.
Collapse
|
45
|
Ding SZ, Minohara Y, Fan XJ, Wang J, Reyes VE, Patel J, Dirden-Kramer B, Boldogh I, Ernst PB, Crowe SE. Helicobacter pylori infection induces oxidative stress and programmed cell death in human gastric epithelial cells. Infect Immun 2007; 75:4030-9. [PMID: 17562777 PMCID: PMC1952011 DOI: 10.1128/iai.00172-07] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Helicobacter pylori infection is associated with altered gastric epithelial cell turnover. To evaluate the role of oxidative stress in cell death, gastric epithelial cells were exposed to various strains of H. pylori, inflammatory cytokines, and hydrogen peroxide in the absence or presence of antioxidant agents. Increased intracellular reactive oxygen species (ROS) were detected using a redox-sensitive fluorescent dye, a cytochrome c reduction assay, and measurements of glutathione. Apoptosis was evaluated by detecting DNA fragmentation and caspase activation. Infection with H. pylori or exposure of epithelial cells to hydrogen peroxide resulted in apoptosis and a dose-dependent increase in ROS generation that was enhanced by pretreatment with inflammatory cytokines. Basal levels of ROS were greater in epithelial cells isolated from gastric mucosal biopsy specimens from H. pylori-infected subjects than in cells from uninfected individuals. H. pylori strains bearing the cag pathogenicity island (PAI) induced higher levels of intracellular oxygen metabolites than isogenic cag PAI-deficient mutants. H. pylori infection and hydrogen peroxide exposure resulted in similar patterns of caspase 3 and 8 activation. Antioxidants inhibited both ROS generation and DNA fragmentation by H. pylori. These results indicate that bacterial factors and the host inflammatory response confer oxidative stress to the gastric epithelium during H. pylori infection that may lead to apoptosis.
Collapse
Affiliation(s)
- Song-Ze Ding
- Division of Gastroenterology and Hepatology, Department of Medicine, P.O. Box 800708, Charlottesville, VA 22908-0708, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|