1
|
Reyes VE. Helicobacter pylori and Its Role in Gastric Cancer. Microorganisms 2023; 11:1312. [PMID: 37317287 PMCID: PMC10220541 DOI: 10.3390/microorganisms11051312] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/08/2023] [Accepted: 05/15/2023] [Indexed: 06/16/2023] Open
Abstract
Gastric cancer is a challenging public health concern worldwide and remains a leading cause of cancer-related mortality. The primary risk factor implicated in gastric cancer development is infection with Helicobacter pylori. H. pylori induces chronic inflammation affecting the gastric epithelium, which can lead to DNA damage and the promotion of precancerous lesions. Disease manifestations associated with H. pylori are attributed to virulence factors with multiple activities, and its capacity to subvert host immunity. One of the most significant H. pylori virulence determinants is the cagPAI gene cluster, which encodes a type IV secretion system and the CagA toxin. This secretion system allows H. pylori to inject the CagA oncoprotein into host cells, causing multiple cellular perturbations. Despite the high prevalence of H. pylori infection, only a small percentage of affected individuals develop significant clinical outcomes, while most remain asymptomatic. Therefore, understanding how H. pylori triggers carcinogenesis and its immune evasion mechanisms is critical in preventing gastric cancer and mitigating the burden of this life-threatening disease. This review aims to provide an overview of our current understanding of H. pylori infection, its association with gastric cancer and other gastric diseases, and how it subverts the host immune system to establish persistent infection.
Collapse
Affiliation(s)
- Victor E Reyes
- Department of Pediatrics and Microbiology & Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-0372, USA
| |
Collapse
|
2
|
Innate immune activation and modulatory factors of Helicobacter pylori towards phagocytic and nonphagocytic cells. Curr Opin Immunol 2023; 82:102301. [PMID: 36933362 DOI: 10.1016/j.coi.2023.102301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/17/2023] [Indexed: 03/18/2023]
Abstract
Helicobacter pylori is an intriguing obligate host-associated human pathogen with a specific host interaction biology, which has been shaped by thousands of years of host-pathogen coevolution. Molecular mechanisms of interaction of H. pylori with the local immune cells in the human system are less well defined than epithelial cell interactions, although various myeloid cells, including neutrophils and other phagocytes, are locally present or attracted to the sites of infection and interact with H. pylori. We have recently addressed the question of novel bacterial innate immune stimuli, including bacterial cell envelope metabolites, that can activate and modulate cell responses via the H. pylori Cag type IV secretion system. This review article gives an overview of what is currently known about the interaction modes and mechanisms of H. pylori with diverse human cell types, with a focus on bacterial metabolites and cells of the myeloid lineage including phagocytic and antigen-presenting cells.
Collapse
|
3
|
Liu H, Wang J, Hu X, Tang X, Zhang C. A rapid and high-throughput Helicobacter pylori RPA-CRISPR/Cas12a-based nucleic acid detection system. Clin Chim Acta 2023; 540:117201. [PMID: 36572137 DOI: 10.1016/j.cca.2022.12.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/30/2022] [Accepted: 12/13/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND Helicobacter pylori lives in the human stomach and causes gastric cancer and other gastric diseases. The development of molecular technology has facilitated low-cost, rapid, and high-throughput detection of H. pylori. MATERIALS AND METHODS The combination of isothermal recombinase polymerase amplification (RPA) and CRISPR-Cas12a was used for early diagnosis and monitoring of H. pylori in clinical settings. The UreB genes from 242 H. pylori strains were subjected to cluster analysis, and we designed corresponding RPA primers and screened 2 sets of CRISPR-derived RNAs (crRNAs) for accurate H. pylori recognition. We then performed specificity and sensitivity validation of seven strains using this RPA-CRISPR/Cas12a method. In addition, the cut-off values of this RPA-CRISPR/Cas12a method based on fluorescence values (i.e., RPA-CRISPR/Cas12a-FT) were determined by comparison with quantitative PCR (qPCR), and further experiments comparing different methods were performed using clinical samples. RESULTS We developed a rapid detection system based on the combination of RPA and CRISPR-Cas12a, which was applied to the early diagnosis and monitoring of H. pylori in clinical settings. The RPA-CRISPR/Cas12a system was used to detect the UreB gene. We found that the limit of detection (LOD) for the CRISPR/Cas12a method based on the lateral flow dipstick result (i.e., CRISPR/Cas12a-LFD) was 100 copies, the cut-off value was 1.4; and for CRISPR/Cas12a-FT the LOD was 50 copies. This system was used to assess clinical samples and showed high reproducibility with proof-of-concept sensitivity, and the whole detection process was completed within 40 min. CONCLUSION As a diagnostic method that can detect the UreB gene of H. pylori in gastric tissue samples rapidly, sensitively, visually, and in a high throughput manner, our method provides a new diagnostic option for clinicians. This system is ideal for hospitals or testing sites with limited medical resources.
Collapse
Affiliation(s)
- Hua Liu
- Fundamental Research Center, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Life Sciences and Technology, Tongji University, Shanghai 201619, China
| | - Jinbin Wang
- Institute of Biotechnology Research, Shanghai Academy of Agricultural Sciences, Key Laboratory of Agricultural Genetics and Breeding, Shanghai 201106, China
| | - Xiuwen Hu
- Institute of Biotechnology Research, Shanghai Academy of Agricultural Sciences, Key Laboratory of Agricultural Genetics and Breeding, Shanghai 201106, China
| | - Xueming Tang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Chao Zhang
- Fundamental Research Center, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Life Sciences and Technology, Tongji University, Shanghai 201619, China.
| |
Collapse
|
4
|
Reyes VE. Helicobacter pylori Immune Response in Children Versus Adults. MEDICAL RESEARCH ARCHIVES 2022; 10:3370. [PMID: 37936946 PMCID: PMC10629867 DOI: 10.18103/mra.v10i12.3370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
H. pylori is perhaps the most prevalent human pathogen worldwide and infects almost half of the world's population. Despite the decreasing prevalence of infection overall, it is significant in developing countries. Most infections are acquired in childhood and persist for a lifetime unless treated. Children are often asymptomatic and often develop a tolerogenic immune response that includes T regulatory cells and their products, immunosuppressive cytokines, such as interleukin (IL)-10, and transforming growth factor-β (TGF-β). This contrasts to the gastric immune response seen in H. pylori-infected adults, where the response is mainly inflammatory, with predominant Th1 and Th17 cells, as well as, inflammatory cytokines, such as TNF-α, IFN-γ, IL-1, IL-6, IL-8, and IL-17. Therefore, compared to adults, infected children generally have limited gastric inflammation and peptic ulcer disease. H. pylori surreptitiously subverts immune defenses to persist in the human gastric mucosa for decades. The chronic infection might result in clinically significant diseases in adults, such as peptic ulcer disease, gastric adenocarcinoma, and mucosa-associated lymphoid tissue lymphoma. This review compares the infection in children and adults and highlights the H. pylori virulence mechanisms responsible for the pathogenesis and immune evasion.
Collapse
Affiliation(s)
- Victor E. Reyes
- Department of Pediatrics, Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd. Galveston, TX 77555-0372 USA
| |
Collapse
|
5
|
Zhao S, Wan D, Zhong Y, Xu X. 1α, 25-Dihydroxyvitamin D3 protects gastric mucosa epithelial cells against Helicobacter pylori-infected apoptosis through a vitamin D receptor-dependent c-Raf/MEK/ERK pathway. PHARMACEUTICAL BIOLOGY 2022; 60:801-809. [PMID: 35587225 PMCID: PMC9122358 DOI: 10.1080/13880209.2022.2058559] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 03/17/2022] [Accepted: 03/22/2022] [Indexed: 05/19/2023]
Abstract
CONTEXT Due to the resistance of Helicobacter pylori to antibiotics, it is difficult to eradicate this pathogenic bacterium from the host. The role of 1α, 25-dihydroxyvitamin D3 (1,25-D3) in H. pylori-infected gastric mucosa epithelial cells remains unknown. OBJECTIVE This study investigates the protective property of 1,25-D3 against H. pylori-infected apoptosis in gastric mucosa epithelial cells and its potential molecular mechanisms. MATERIALS AND METHODS GES-1 cells were infected with H. pylori SS1 strain (MOI: 100) and treated with 1,25-D3 at 100, 200, and 300 nM for 24 h. Mice were orally gavaged with 108 CFUs of H. pylori and 25 µg/kg 1,25-D3 every other day for 1 month. CCK-8, LDH assay, TUNEL assay and western blot were used to determine the effect of 1,25-D3 on H. pylori-induced apoptosis. RESULTS H. pylori infection decreased cell viability to 59.2%, while 100-300 nM 1,25-D3 increased cell viability to 62.2%, 78.4% and 87.1%, respectively. Compared with positive control (4.53-fold), 1,25-D3 reduced caspase-3 activity to 4.49-, 2.88- and 1.49-fold, reduced caspase-6 activity to 2.36-, 1.88- and 1.50-fold, reduced caspase-9 activity to 4.55-, 2.91- and 2.01-fold. 1,25-D3 alters Bcl-2 family, caspase protein expression and c-Raf/MEK/ERK phosphorylation levels in vivo and in vitro. Suppression of 1,25-D3 in apoptosis was reliant on binding to vitamin D receptor. The pharmacological inhibition of c-Raf/MEK/ERK phosphorylation blocked the anti-apoptotic effect of 1,25-D3. DISCUSSION AND CONCLUSION 1,25-D3 protected gastric mucosa epithelial cells against H. pylori-infected apoptosis through a VDR-dependent c-Raf/MEK/ERK pathway.
Collapse
Affiliation(s)
- Shuai Zhao
- Department of Gastroenterology, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Pediatric Department of Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Daihong Wan
- Pediatric Department of Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Yaoyao Zhong
- Pediatric Department of Qingdao Women and Children’s Hospital, Qingdao University, Qingdao, China
| | - Xiwei Xu
- Department of Gastroenterology, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- CONTACT Xiwei Xu Department of Gastroenterology, Beijing Children’s Hospital, Capital Medical Universtiy, National Center for Children’s Health, Beijing100045, China
| |
Collapse
|
6
|
Hailemariam S, Zhao S, He Y, Wang J. Urea transport and hydrolysis in the rumen: A review. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2021; 7:989-996. [PMID: 34738029 PMCID: PMC8529027 DOI: 10.1016/j.aninu.2021.07.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 07/02/2021] [Accepted: 07/08/2021] [Indexed: 11/29/2022]
Abstract
Inefficient dietary nitrogen (N) conversion to microbial proteins, and the subsequent use by ruminants, is a major research focus across different fields. Excess bacterial ammonia (NH3) produced due to degradation or hydrolyses of N containing compounds, such as urea, leads to an inefficiency in a host's ability to utilize nitrogen. Urea is a non-protein N containing compound used by ruminants as an ammonia source, obtained from feed and endogenous sources. It is hydrolyzed by ureases from rumen bacteria to produce NH3 which is used for microbial protein synthesis. However, lack of information exists regarding urea hydrolysis in ruminal bacteria, and how urea gets to hydrolysis sites. Therefore, this review describes research on sites of urea hydrolysis, urea transport routes towards these sites, the role and structure of urea transporters in rumen epithelium and bacteria, the composition of ruminal ureolytic bacteria, mechanisms behind urea hydrolysis by bacterial ureases, and factors influencing urea hydrolysis. This review explores the current knowledge on the structure and physiological role of urea transport and ureolytic bacteria, for the regulation of urea hydrolysis and recycling in ruminants. Lastly, underlying mechanisms of urea transportation in rumen bacteria and their physiological importance are currently unknown, and therefore future research should be directed to this subject.
Collapse
Affiliation(s)
- Samson Hailemariam
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
- Dilla University, College of Agriculture and Natural Resource, Dilla P. O. Box 419, Ethiopia
| | - Shengguo Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yue He
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Jiaqi Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| |
Collapse
|
7
|
Abstract
Outer membrane vesicles (OMV) shed by pathogenic bacteria have multifunctional roles in disease initiation and progression. Further, their efficacy as novel vaccines has underscored their importance as potential therapeutics. Consequently, to advance allied research related to their immunogenicity and pathogenicity it is important to separate these vesicular structures from parental cells and demonstrate them to be free from cellular debris and other non-vesicle-related constituents such as protein aggregates. To do so represents a key step in initiating OMV-related studies and the techniques and strategies adopted by the H. pylori community to achieve this will be the focus of this chapter.The key methods used typically to obtain a heterogeneous mixture of OMV (size range: ~20-300 nm in diameter) include growth of bacteria in broth culture followed by differential centrifugation, filtration, and concentration to separate OMV from the intact organisms. Additional measures may be adopted to further size-fractionate the population of OMV including gel filtration or density gradient ultra-centrifugation in order to facilitate differentiation between the activities of small versus large OMV, as recent studies have demonstrated differential modes of entry into host cells as well as size-dependent differences in the OMV proteome (Turner et al., Front Immunol 9:1466, 2018). The OMV from H. pylori harbor many of the virulence factors associated with gastric disease including the CagA oncoprotein, the cytotoxin VacA, and the HtrA protease (Olofsson et al., mBio 5:e00979-14, 2014; Mullaney et al., Proteomics Clin Appl 3:785-96, 2009) and their close association with areas of cell-cell contact and efficient endocytosis supports a role for these complexes in gastric disease (Turkina et al., FEMS Microbiol Lett 362:fnv076, 2015).
Collapse
Affiliation(s)
- Henry J Windle
- Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St James's Hospital, Dublin 8, Ireland.
| |
Collapse
|
8
|
Sharma A, Sarkaraisamy P, Shukla S, Alam SI. Screening of immunogenic proteins from extracellular proteome of C. botulinum type B by immunoproteomic approach. FOOD BIOTECHNOL 2020. [DOI: 10.1080/08905436.2020.1835671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Arti Sharma
- Biotechnology Division, Defence Research & Development Establishment, Gwalior, India
- Government Degree College Prithvipur, Niwari, India
| | | | - Sangeeta Shukla
- Zoology Department, Jiwaji University Gwalior, Gwalior, India
| | - Syed Imtiaz Alam
- Biotechnology Division, Defence Research & Development Establishment, Gwalior, India
| |
Collapse
|
9
|
Valenzuela-Valderrama M, Cerda-Opazo P, Backert S, González MF, Carrasco-Véliz N, Jorquera-Cordero C, Wehinger S, Canales J, Bravo D, Quest AFG. The Helicobacter pylori Urease Virulence Factor Is Required for the Induction of Hypoxia-Induced Factor-1α in Gastric Cells. Cancers (Basel) 2019; 11:cancers11060799. [PMID: 31185594 PMCID: PMC6627347 DOI: 10.3390/cancers11060799] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/24/2019] [Accepted: 04/27/2019] [Indexed: 12/14/2022] Open
Abstract
Chronic Helicobacter pylori infection increases the risk of gastric cancer and induction of hypoxia-induced factor (HIF), which is frequently associated with the development and progression of several types of cancer. We recently showed that H. pylori activation of the PI3K-AKT-mTOR pathway in gastric cells increased HIF-1α expression. Here, we identified the H. pylori virulence factor responsible for HIF-1α induction. A mutant of the H. pylori 84-183 strain was identified with reduced ability to induce HIF-1α. Coomassie blue staining of extracts from these bacteria separated by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) revealed poor expression of urease subunits that correlated with reduced urease activity. This finding was confirmed in the 26695 strain, where urease mutants were unable to induce HIF-1α expression. Of note, HIF-1α induction was also observed in the presence of the urease inhibitor acetohydroxamic acid at concentrations (of 20 mM) that abrogated urease activity in bacterial culture supernatants, suggesting that enzymatic activity of the urease is not required for HIF-1α induction. Finally, the pre-incubation of the human gastric adenocarcinoma cell line AGS with blocking antibodies against Toll-like receptor-2 (TLR2), but not TLR4, prevented HIF-1α induction. In summary, these results reveal a hitherto unexpected role for the urease protein in HIF-1α induction via TLR2 activation following H. pylori infection of gastric cells.
Collapse
Affiliation(s)
- Manuel Valenzuela-Valderrama
- Laboratorio de Microbiología Celular, Instituto de Innovación e Investigación en Salud, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago 8320000, Chile.
- Centro de Estudios Avanzados en Enfermedades Crónicas (ACCDiS), Independencia, Santiago 8380000, Chile.
| | - Paulina Cerda-Opazo
- Laboratorio de Comunicaciones Celulares, Centro de estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Independencia, Santiago 8380000, Chile.
| | - Steffen Backert
- Department of Biology, Division of Microbiology, University of Erlangen-Nuremberg, 91054 Erlangen, Germany.
| | - María Fernanda González
- Laboratorio de Comunicaciones Celulares, Centro de estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Independencia, Santiago 8380000, Chile.
| | - Nicolás Carrasco-Véliz
- Laboratorio de Microbiología Celular, Instituto de Innovación e Investigación en Salud, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago 8320000, Chile.
| | - Carla Jorquera-Cordero
- Laboratorio de Comunicaciones Celulares, Centro de estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Independencia, Santiago 8380000, Chile.
| | - Sergio Wehinger
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca 3460000, Chile.
| | - Jimena Canales
- Laboratorio de Comunicaciones Celulares, Centro de estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Independencia, Santiago 8380000, Chile.
| | - Denisse Bravo
- Centro de Estudios Avanzados en Enfermedades Crónicas (ACCDiS), Independencia, Santiago 8380000, Chile.
- Laboratorio de Microbiología Oral, Departamento de Patología y Medicina Oral, Facultad de Odontología, Universidad de Chile, Independencia, Santiago 8380000, Chile.
| | - Andrew F G Quest
- Centro de Estudios Avanzados en Enfermedades Crónicas (ACCDiS), Independencia, Santiago 8380000, Chile.
- Laboratorio de Comunicaciones Celulares, Centro de estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Independencia, Santiago 8380000, Chile.
| |
Collapse
|
10
|
Zhao H, Wu Y, Xu Z, Ma R, Ding Y, Bai X, Rong Q, Zhang Y, Li B, Ji X. Mechanistic Insight Into the Interaction Between Helicobacter pylori Urease Subunit α and Its Molecular Chaperone Hsp60. Front Microbiol 2019; 10:153. [PMID: 30804917 PMCID: PMC6370633 DOI: 10.3389/fmicb.2019.00153] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/22/2019] [Indexed: 12/11/2022] Open
Abstract
Helicobacter pylori is the etiologic agent in a variety of gastroduodenal diseases. As its key pathogenic factors, both urease and Hsp60 play important roles in the pathogenesis of H. pylori. Previous studies have suggested that there is close relationship between urease and Hsp60, which implied that Hsp60 may act as a chaperone in urease stabilization and assembly. However, how these two proteins interact remains unclear. In this study, the impact of Hsp60 on urease activity of H. pylori lysate was first detected to confirm the interaction between urease and Hsp60. Pull-down assays further indicated that Hsp60 could bind to UreA subunit but not UreB. Then, the 3D structure of Hsp60 was modeled using I-TASSER to simulate the binding complex with UreA by molecular docking. The results showed that UreA is a perfect fit for the cavity of Hsp60. Analysis of the resulting model demonstrated that at least seven residues of UreA, located on two interfaces, participate in the interaction. Site-directed mutagenesis of these potential residues showed reduced affinity with Hsp60 than the wild type UreA through surface plasmon resonance (SPR) experiments, and D68 appears to have an important role in the affinity. Further analysis also showed that mutation of E25 and K26 caused a more rapid association and dissociation than with wild UreA, implying that they have roles in stabilizing the interaction complex. These affinity comparisons suggested that the interfaces predicted by molecular docking are credible. Our study indicated a direct interaction between Hsp60 and urease and revealed the binding interfaces and key residues involved in the interaction. These results provide further evidence for the chaperone activity of Hsp60 toward urease and lay a foundation to better understand the maturation mechanism of urease in H. pylori.
Collapse
Affiliation(s)
- Huilin Zhao
- Department of Pathogenic Biology, School of Basic Medical Sciences, Binzhou Medical University, Yantai, China
| | - Yulong Wu
- Department of Pathogenic Biology, School of Basic Medical Sciences, Binzhou Medical University, Yantai, China
| | - Zheng Xu
- Department of Pathogenic Biology, School of Basic Medical Sciences, Binzhou Medical University, Yantai, China
| | - Ran Ma
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Yunfei Ding
- Department of Pathogenic Biology, School of Basic Medical Sciences, Binzhou Medical University, Yantai, China
| | - Xuelian Bai
- Clinical Medicine Laboratory, Binzhou Medical University Hospital, Binzhou, China
| | - Qianyu Rong
- Department of Pathogenic Biology, School of Basic Medical Sciences, Binzhou Medical University, Yantai, China
| | - Ying Zhang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Binzhou Medical University, Yantai, China
| | - Boqing Li
- Department of Pathogenic Biology, School of Basic Medical Sciences, Binzhou Medical University, Yantai, China
| | - Xiaofei Ji
- Department of Pathogenic Biology, School of Basic Medical Sciences, Binzhou Medical University, Yantai, China
| |
Collapse
|
11
|
Butt J, Varga MG, Blot WJ, Teras L, Visvanathan K, Le Marchand L, Haiman C, Chen Y, Bao Y, Sesso HD, Wassertheil-Smoller S, Ho GY, Tinker LE, Peek RM, Potter JD, Cover TL, Hendrix LH, Huang LC, Hyslop T, Um C, Grodstein F, Song M, Zeleniuch-Jacquotte A, Berndt S, Hildesheim A, Waterboer T, Pawlita M, Epplein M. Serologic Response to Helicobacter pylori Proteins Associated With Risk of Colorectal Cancer Among Diverse Populations in the United States. Gastroenterology 2019; 156:175-186.e2. [PMID: 30296434 PMCID: PMC6309494 DOI: 10.1053/j.gastro.2018.09.054] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/12/2018] [Accepted: 09/27/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Previous studies reported an association of the bacteria Helicobacter pylori, the primary cause of gastric cancer, and risk of colorectal cancer (CRC). However, these findings have been inconsistent, appear to vary with population characteristics, and may be specific for virulence factor VacA. To more thoroughly evaluate the potential association of H pylori antibodies with CRC risk, we assembled a large consortium of cohorts representing diverse populations in the United States. METHODS We used H pylori multiplex serologic assays to analyze serum samples from 4063 incident cases of CRC, collected before diagnosis, and 4063 matched individuals without CRC (controls) from 10 prospective cohorts for antibody responses to 13 H pylori proteins, including virulence factors VacA and CagA. The association of seropositivity to H pylori proteins, as well as protein-specific antibody level, with odds of CRC was determined by conditional logistic regression. RESULTS Overall, 40% of controls and 41% of cases were H pylori-seropositive (odds ratio [OR], 1.09; 95% CI, 0.99-1.20). H pylori VacA-specific seropositivity was associated with an 11% increased odds of CRC (OR, 1.11; 95% CI, 1.01-1.22), and this association was particularly strong among African Americans (OR, 1.45; 95% CI, 1.08-1.95). Additionally, odds of CRC increased with level of VacA antibody in the overall cohort (P = .008) and specifically among African Americans (P = .007). CONCLUSIONS In an analysis of a large consortium of cohorts representing diverse populations, we found serologic responses to H pylori VacA to associate with increased risk of CRC risk, particularly for African Americans. Future studies should seek to understand whether this marker is related to virulent H pylori strains carried in these populations.
Collapse
Affiliation(s)
- Julia Butt
- Infection and Cancer Epidemiology, Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany (; ; )
- Cancer Control and Population Sciences Program, Duke Cancer Institute, and Department of Population Health Sciences, Duke University, 2424 Erwin Road, Suite 602, Durham, NC 27705, USA ()
| | - Matthew G. Varga
- University of North Carolina at Chapel Hill, Department of Epidemiology, Gillings School for Global Public Health and Lineberger Comprehensive Cancer Center, 2102E McGavran Greenberg Hall, Chapel Hill, NC 27599, USA ()
| | - William J. Blot
- Division of Epidemiology, Vanderbilt University Medical Center, 2525 West End Avenue Nashville, TN 37203, USA ()
| | - Lauren Teras
- Behavioral and Epidemiology Research Group, American Cancer Society, 250 Williams St, Atlanta, GA 30303, USA (; )
| | - Kala Visvanathan
- Department of Epidemiology, Johns Hopkins School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205 USA ()
| | - Loïc Le Marchand
- Epidemiology Program, University of Hawai’i Cancer Center, 701 Ilalo Street, Honolulu, HI 96813 USA ()
| | - Christopher Haiman
- University of Southern California and USC Norris Comprehensive Cancer Center, Los Angeles, CA, USA ()
| | - Yu Chen
- Department of Population Health, New York University School of Medicine, 650 First Avenue, New York, NY 10016 USA (; )
| | - Ying Bao
- Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115 USA (; )
| | - Howard D. Sesso
- Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115 USA (; )
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 667 Huntington Avenue, Boston, MA 02115 USA (; )
| | - Sylvia Wassertheil-Smoller
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461 USA ()
| | - Gloria Y.F. Ho
- Department of Occupational Medicine, Epidemiology and Prevention, Feinstein Institute for Medical Research, Northwell Health; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Great Neck, NY 11021 USA ()
| | - Lesley E. Tinker
- Cancer Prevention Program, Division of Public Health Sciences at Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue, Seattle, WA 98109 USA ()
| | - Richard M. Peek
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Avenue, 1030C MRB IV (1025C), Nashville, TN 37232 USA, ()
| | - John D. Potter
- Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue, Seattle WA 98109 USA ()
| | - Timothy L. Cover
- Department of Medicine and Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232 USA; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN USA ()
| | - Laura H. Hendrix
- Department of Biostatistics and Bioinformatics, Duke University, 2424 Erwin Road, Durham, NC 27705 USA (; )
| | - Li-Ching Huang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37203 USA ()
| | - Terry Hyslop
- Cancer Control and Population Sciences Program, Duke Cancer Institute, and Department of Population Health Sciences, Duke University, 2424 Erwin Road, Suite 602, Durham, NC 27705, USA ()
- Department of Biostatistics and Bioinformatics, Duke University, 2424 Erwin Road, Durham, NC 27705 USA (; )
| | - Caroline Um
- Behavioral and Epidemiology Research Group, American Cancer Society, 250 Williams St, Atlanta, GA 30303, USA (; )
| | - Francine Grodstein
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 667 Huntington Avenue, Boston, MA 02115 USA (; )
| | - Mingyang Song
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 667 Huntington Avenue, Boston, MA 02115 USA (; )
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115 USA ()
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115 USA, ()
| | - Anne Zeleniuch-Jacquotte
- Department of Population Health, New York University School of Medicine, 650 First Avenue, New York, NY 10016 USA (; )
| | - Sonja Berndt
- Infections and Immunoepidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Room SG/6E102, Rockville, MD 20850 USA (; )
| | - Allan Hildesheim
- Infections and Immunoepidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Room SG/6E102, Rockville, MD 20850 USA (; )
| | - Tim Waterboer
- Infection and Cancer Epidemiology, Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany (; ; )
| | - Michael Pawlita
- Infection and Cancer Epidemiology, Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany (; ; )
| | - Meira Epplein
- Cancer Control and Population Sciences Program, Duke Cancer Institute, and Department of Population Health Sciences, Duke University, 2424 Erwin Road, Suite 602, Durham, NC 27705, USA ()
| |
Collapse
|
12
|
Pepe S, Pinatel E, Fiore E, Puccio S, Peano C, Brignoli T, Vannini A, Danielli A, Scarlato V, Roncarati D. The Helicobacter pylori Heat-Shock Repressor HspR: Definition of Its Direct Regulon and Characterization of the Cooperative DNA-Binding Mechanism on Its Own Promoter. Front Microbiol 2018; 9:1887. [PMID: 30154784 PMCID: PMC6102357 DOI: 10.3389/fmicb.2018.01887] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 07/27/2018] [Indexed: 12/12/2022] Open
Abstract
The ability of pathogens to perceive environmental conditions and modulate gene expression accordingly is a crucial feature for bacterial survival. In this respect, the heat-shock response, a universal cellular response, allows cells to adapt to hostile environmental conditions and to survive during stress. In the major human pathogen Helicobacter pylori the expression of chaperone-encoding operons is under control of two auto-regulated transcriptional repressors, HrcA and HspR, with the latter acting as the master regulator of the regulatory circuit. To further characterize the HspR regulon in H. pylori, we used global transcriptome analysis (RNA-sequencing) in combination with Chromatin Immunoprecipitation coupled with deep sequencing (ChIP-sequencing) of HspR genomic binding sites. Intriguingly, these analyses showed that HspR is involved in the regulation of different crucial cellular functions through a limited number of genomic binding sites. Moreover, we further characterized HspR-DNA interactions through hydroxyl-radical footprinting assays. This analysis in combination with a nucleotide sequence alignment of HspR binding sites, revealed a peculiar pattern of DNA protection and highlighted sequence conservation with the HAIR motif (an HspR-associated inverted repeat of Streptomyces spp.). Site-directed mutagenesis demonstrated that the HAIR motif is fundamental for HspR binding and that additional nucleotide determinants flanking the HAIR motif are required for complete binding of HspR to its operator sequence spanning over 70 bp of DNA. This finding is compatible with a model in which possibly a dimer of HspR recognizes the HAIR motif overlapping its promoter for binding and in turn cooperatively recruits two additional dimers on both sides of the HAIR motif.
Collapse
Affiliation(s)
- Simona Pepe
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Bologna, Italy
| | - Eva Pinatel
- Institute of Biomedical Technologies, National Research Council, Milan, Italy
| | - Elisabetta Fiore
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Bologna, Italy
| | - Simone Puccio
- Institute of Biomedical Technologies, National Research Council, Milan, Italy.,Humanitas Clinical and Research Center, Milan, Italy
| | - Clelia Peano
- Institute of Biomedical Technologies, National Research Council, Milan, Italy.,Humanitas Clinical and Research Center, Milan, Italy.,Institute of Genetic and Biomedical Research, National Research Council, Milan, Italy
| | - Tarcisio Brignoli
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Bologna, Italy
| | - Andrea Vannini
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Bologna, Italy
| | - Alberto Danielli
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Bologna, Italy
| | - Vincenzo Scarlato
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Bologna, Italy
| | - Davide Roncarati
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Bologna, Italy
| |
Collapse
|
13
|
Noncatalytic Antioxidant Role for Helicobacter pylori Urease. J Bacteriol 2018; 200:JB.00124-18. [PMID: 29866802 DOI: 10.1128/jb.00124-18] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/30/2018] [Indexed: 12/15/2022] Open
Abstract
The well-studied catalytic role of urease, the Ni-dependent conversion of urea into carbon dioxide and ammonia, has been shown to protect Helicobacter pylori against the low pH environment of the stomach lumen. We hypothesized that the abundantly expressed urease protein can play another noncatalytic role in combating oxidative stress via Met residue-mediated quenching of harmful oxidants. Three catalytically inactive urease mutant strains were constructed by single substitutions of Ni binding residues. The mutant versions synthesize normal levels of urease, and the altered versions retained all methionine residues. The three site-directed urease mutants were able to better withstand a hypochlorous acid (HOCl) challenge than a ΔureAB deletion strain. The capacity of purified urease to protect whole cells via oxidant quenching was assessed by adding urease enzyme to nongrowing HOCl-exposed cells. No wild-type cells were recovered with oxidant alone, whereas urease addition significantly aided viability. These results suggest that urease can protect H. pylori against oxidative damage and that the protective ability is distinct from the well-characterized catalytic role. To determine the capability of methionine sulfoxide reductase (Msr) to reduce oxidized Met residues in urease, purified H. pylori urease was exposed to HOCl and a previously described Msr peptide repair mixture was added. Of the 25 methionine residues in urease, 11 were subject to both oxidation and to Msr-mediated repair, as identified by mass spectrometry (MS) analysis; therefore, the oxidant-quenchable Met pool comprising urease can be recycled by the Msr repair system. Noncatalytic urease appears to play an important role in oxidant protection.IMPORTANCE Chronic Helicobacter pylori infection can lead to gastric ulcers and gastric cancers. The enzyme urease contributes to the survival of the bacterium in the harsh environment of the stomach by increasing the local pH. In addition to combating acid, H. pylori must survive host-produced reactive oxygen species to persist in the gastric mucosa. We describe a cyclic amino acid-based antioxidant role of urease, whereby oxidized methionine residues can be recycled by methionine sulfoxide reductase to again quench oxidants. This work expands our understanding of the role of an already acknowledged pathogen virulence factor and specifically expands our knowledge of H. pylori survival mechanisms.
Collapse
|
14
|
Zhang R, Qiao D, Peng X, Duan G, Shi Q, Zhang L, Wang C, Liang W, Chen S, Fan Q. A novel food‐grade lactococcal expression system and its use for secretion and delivery of an oral vaccine antigen. JOURNAL OF CHEMICAL TECHNOLOGY & BIOTECHNOLOGY 2018; 93:1655-1660. [DOI: 10.1002/jctb.5536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Accepted: 11/25/2017] [Indexed: 07/03/2024]
Abstract
AbstractBACKGROUNDFood‐grade bacterial expression systems are relatively rare, and increasing evidence indicates that subcellular location of antigens in bacterial vector vaccines may markedly affect the immune efficacy.RESULTSThis study developed a novel food‐grade secretory expression system for heterologous protein production and oral vaccine delivery. Furthermore, by using the expression system, an engineered L. lactis strain secreting H. pylori UreB was constructed, and used to vaccinate SPF BALB/c mice. As results, the UreB expressed in L. lactis was detected in both cell lysates and culture supernatant of the engineered strain, constituting roughly 50% of the culture supernatant proteins, and recognized by mouse anti‐H. pylori sera. Oral vaccination with the engineered L. lactis produced a significantly elevated anti‐UreB serum antibody level in mice (P < 0.05).CONCLUSIONThese data show a novel food‐grade L. lactis secretory expression system, which may have distinct potential impact on edible and medicinal protein production and oral vaccine development. Moreover, this is the first report on secretory expression of a H. pylori antigen via using a food‐grade lactococcal expression system, and the engineered strain secreting UreB can be a hopeful H. pylori vaccine candidate. © 2017 Society of Chemical Industry
Collapse
Affiliation(s)
- Rongguang Zhang
- Department of Epidemiology and Statistics, College of Public Health Zhengzhou University Zhengzhou China
- Henan Innovation Center of Molecular Diagnosis and Laboratory Medicine Xinxiang Medical University Xinxiang China
| | - Dan Qiao
- Department of Epidemiology and Statistics, College of Public Health Zhengzhou University Zhengzhou China
| | - Xiaoyan Peng
- Department of Epidemiology and Statistics, College of Public Health Zhengzhou University Zhengzhou China
| | - Guangcai Duan
- Department of Epidemiology and Statistics, College of Public Health Zhengzhou University Zhengzhou China
- Henan Innovation Center of Molecular Diagnosis and Laboratory Medicine Xinxiang Medical University Xinxiang China
| | - Qingfeng Shi
- Department of Epidemiology and Statistics, College of Public Health Zhengzhou University Zhengzhou China
| | - Linghan Zhang
- Department of Clinical Medicine Zhengzhou University Zhengzhou China
| | - Chen Wang
- Department of Epidemiology and Statistics, College of Public Health Zhengzhou University Zhengzhou China
| | - Wenjuan Liang
- Department of Epidemiology and Statistics, College of Public Health Zhengzhou University Zhengzhou China
| | - Shuaiyin Chen
- Department of Epidemiology and Statistics, College of Public Health Zhengzhou University Zhengzhou China
| | - Qingtang Fan
- Department of Epidemiology and Statistics, College of Public Health Zhengzhou University Zhengzhou China
| |
Collapse
|
15
|
Jing ZW, Luo M, Jia YY, Li C, Zhou SY, Mei QB, Zhang BL. Anti-Helicobacterpylori effectiveness and targeted delivery performance of amoxicillin-UCCs-2/TPP nanoparticles based on ureido-modified chitosan derivative. Int J Biol Macromol 2018; 115:367-374. [PMID: 29660462 DOI: 10.1016/j.ijbiomac.2018.04.070] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/11/2018] [Accepted: 04/12/2018] [Indexed: 02/07/2023]
Abstract
The amoxicillin-UCCs-2/TPP nanoparticles constructed with ureido-modified chitosan derivative UCCs-2 and sodium tripolyphosphate (TPP) played an important role to deliver drug to achieve more efficacious and specific eradication of Helicobacterpylori (H. pylori) in vitro. In this study, the anti-H. pylori effectiveness in vivo and uptake mechanism was investigated in details, including the effect of temperature, pH values and the addition of competitive substrate urea on uptake. Compared with unmodified nanoparticles, a more efficacious and specific anti-H. pylori activities were obtained in vivo by using this biological chitosan derivative UCCs-2. Histological staining and immunological analysis verified that the amoxicillin-UCCs-2/TPP nanoparticles could diminish the proinflammatory cytokines levels and alleviate the inflammatory damages caused by H. pylori infection. The uredio-modified nanoparticles also have favorable gastric retention property, which is beneficial for the oral drug delivery to targeted eradicate H. pylori infection in stomach. These findings suggest that this targeted drug delivery system may serve for specific treatment of H. pylori infection both in vitro and in vivo, which can also be used as promising nanocarriers for other therapeutic reagents to target H. pylori.
Collapse
Affiliation(s)
- Zi-Wei Jing
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China; The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Min Luo
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Yi-Yang Jia
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Chen Li
- Key Laboratory of Gastrointestinal Pharmacology of the State Administration of Traditional Chinese Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Si-Yuan Zhou
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China; Key Laboratory of Gastrointestinal Pharmacology of the State Administration of Traditional Chinese Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Qi-Bing Mei
- Key Laboratory of Gastrointestinal Pharmacology of the State Administration of Traditional Chinese Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Bang-Le Zhang
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China; Key Laboratory of Gastrointestinal Pharmacology of the State Administration of Traditional Chinese Medicine, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
16
|
Hsu WT, Ho SY, Jian TY, Huang HN, Lin YL, Chen CH, Lin TH, Wu MS, Wu CJ, Chan YL, Liao KW. Helicobacter pylori-derived heat shock protein 60 increases the induction of regulatory T-cells associated with persistent infection. Microb Pathog 2018; 119:152-161. [PMID: 29660522 DOI: 10.1016/j.micpath.2018.04.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/26/2018] [Accepted: 04/10/2018] [Indexed: 02/06/2023]
Abstract
Local Treg responses are involved in Helicobacter pylori-related inflammation and clinical outcomes after infection, and H. pylori-derived HSP60 (HpHSP60) is an important virulence factor associated with gastric carcinogenesis. This study to investigate the role of HpHSP60 in immunosuppression, particularly with regard to whether it could induce the production of Treg cells. For this purpose, human peripheral blood mononuclear cells (PBMCs) were treated with or without HpHSP60 in the presence of an anti-CD3 mAb to determine the effect of HpHSP60 on cell proliferation. In this report, HpHSP60 decreased the expression of CDK4 to significantly arrest the proliferation of mitogen-stimulated T-cells, which correlated with the induction of Treg cells. Moreover, monocytic cells were essential for the induction of HpHSP60-induced Treg cells via the secretion of IL-10 and TGF-β after treatment with HpHSP60. Blockage of HpHSP60 with specific monoclonal antibodies significantly reduced the colonization of H. pylori and the expression of Treg cells in vivo. Overall, our results suggest that HpHSP60 could act on macrophages to trigger the expression of IL-10 and TGF-β, thereby leading to an increase in Treg cells and inhibition of T-cell proliferation.
Collapse
Affiliation(s)
- Wei-Tung Hsu
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan, ROC
| | - Shu-Yi Ho
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan, ROC
| | - Ting-Yan Jian
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan, ROC
| | - Han-Ning Huang
- Department of Food Science and Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung, Taiwan, ROC
| | - Yu-Ling Lin
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan, ROC; Center for Bioinformatics Research, National Chiao Tung University, Hsinchu, Taiwan, ROC
| | - Chia-Hung Chen
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan, ROC
| | - Tsung-Han Lin
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan, ROC
| | - Ming-Shiang Wu
- Department of Internal Medicine, College of Medicine, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Chang-Jer Wu
- Department of Food Science and Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung, Taiwan, ROC; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan, ROC; Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan, ROC; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Yi-Lin Chan
- Department of Life Science, Chinese Culture University, Taipei, Taiwan, ROC
| | - Kuang-Wen Liao
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan, ROC; Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan, ROC; College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan, ROC; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC.
| |
Collapse
|
17
|
Expression and Antigenic Evaluation of Helicobacter pylori UreB Fragment. Jundishapur J Microbiol 2017. [DOI: 10.5812/jjm.41645] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
18
|
Alhussaini MS. Prevalence of Helicobacter pylori among patients with different gastrointestinal disorders in Saudi Arabia. MEDICAL JOURNAL OF INDONESIA 2017. [DOI: 10.13181/mji.v25i4.1442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background: Helicobacter pylori is an important gastrointestinal pathogen associated with gastritis, peptic ulcers, and an increased risk of gastric carcinoma. The present study was carried out to determine the relationship between this organism with different gastrointestinal ailments.Methods: 150 outpatients referrals to Saudi Arabian Medical City, Riyadh, Kingdom of Saudi Arabia was recruited in January to June 2015. Each patient was subjected to endoscopic examination. Biopsy specimens were taken from the stomach for rapid urease test and culture. Suspected H. pylori colonies were subjected to colony morphology identification, microscopical examination and biochemical reactions. The samples were also subjected to PCR to detect ureA subunit of urease gene.Results: The endoscopic examination of patients revealed normal, gastric ulcer, duodenal ulcer, gastritis, and gastric cancer with a rate of 20.7%, 20%, 24%, 33.3%, and 2%, respectively. Direct smear exam revealed that 52% of patients were H. pylori positive while culture and rapid urease test showed a prevalence of 71.33%. Fifty four biopsies (36%) were urease positive after 1 hour at room temperature, 39 (62%) after 1 hour incubation at 37°C and 14 (71.33%) after 24 hours incubation. Isolated H. pylori showed that they were catalase, oxidase, and urease positive. PCR results showed 411-bp fragment, which is indicative for the ureA subunit of urease gene.Conclusion: The prevalence of H. pylori infection was high among tested population. Strong association between H. pylori and duodenal ulcer was noticed. A 411-bp fragment indicative of the ureA subunit of urease gene was detected in all the tested isolates.
Collapse
|
19
|
Sun Y, Zhu DQ, Zhang QX, Pang XH, Sun SR, Liu F, Li AL, Meng XC. The Expression of GroEL Protein Amplified fromBifidobacterium animalissubsp.lactisKLDS 2.0603 and its Role in Competitive Adhesion to Caco-2. FOOD BIOTECHNOL 2016. [DOI: 10.1080/08905436.2016.1244769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
20
|
Enany S, Abdalla S. In vitro antagonistic activity of Lactobacillus casei against Helicobacter pylori. Braz J Microbiol 2015; 46:1201-6. [PMID: 26691482 PMCID: PMC4704617 DOI: 10.1590/s1517-838246420140675] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 02/02/2015] [Indexed: 12/15/2022] Open
Abstract
Helicobacter pylori is one of the most common causes of chronic
infections in humans. Curing H. pylori infection is difficult
because of the habitat of the organism below the mucus adherent layer of gastric
mucosa. Lactobacilli are known as acid-resistant bacteria and can remain in stomach
for a long time than any other organism, we aimed in this study to examine the
efficacy of Lactobacillus casei as a probiotic against H.
pylori in humans. Particularly, L. casei was opted as it
is considered to be one of the widely used probiotics in dairy products. One hundred
and seven strains of H. pylori were isolated from dyspeptic patients
and were tested for their antibiotic susceptibility to metronidazole (MTZ),
clarithromycin (CLR), tetracycline (TET), and amoxicillin (AMX) by the disc diffusion
method. The strains were examined for their susceptibility toward L. casei
- present in fermented milk products - by well diffusion method. It was
found that 74.7% strains were resistant to MTZ; 1.8% to MTZ, TET, and CLR; 3.7% to
MTZ and CLR; 4.6% to MTZ and TET; and 0.9% were resistant to MTZ, TET, and AMX. The
antibacterial activity of L. casei against H.
pylori was determined on all the tested H. pylori
isolates including antibiotic resistant strains with different patterns. Our study
proposed the use of probiotics for the treatment of H. pylori
infection as an effective approach.
Collapse
Affiliation(s)
- Shymaa Enany
- Department of Microbiology and Immunology, Suez Canal University, Ismailia, Egypt
| | - Salah Abdalla
- Department of Microbiology and Immunology, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
21
|
Morphological changes in human gastric epithelial cells induced by nuclear targeting of Helicobacter pylori urease subunit A. J Microbiol 2015; 53:406-14. [PMID: 26025173 DOI: 10.1007/s12275-015-5085-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 05/14/2015] [Accepted: 05/15/2015] [Indexed: 12/20/2022]
Abstract
Nuclear targeting of bacterial proteins and their pathological effects on host cells are an emerging pathogenic mechanism in bacteria. We have previously reported that urease subunit A (UreA) of Helicobacter pylori targets the nuclei of COS-7 cells through nuclear localization signals (NLSs). This study further investigated whether UreA of H. pylori targets the nuclei of gastric epithelial cells and then induces molecular and cellular changes in the host cells. H. pylori 26695 strain produced and secreted outer membrane vesicles (OMVs). UreA was translocated into gastric epithelial AGS cells through outer membrane vesicles (OMVs) and then targeted the nuclei of AGS cells. Nuclear targeting of rUreA did not induce host cell death, but resulted in morphological changes, such as cellular elongation, in AGS cells. In contrast, AGS cells treated with rUreA?NLS proteins did not show this morphological change. Next generation sequencing revealed that nuclear targeting of UreA differentially regulated 102 morphogenesis- related genes, of which 67 and 35 were up-regulated and down-regulated, respectively. Our results suggest that nuclear targeting of H. pylori UreA induces both molecular and cellular changes in gastric epithelial cells.
Collapse
|
22
|
Repetto O, Zanussi S, Casarotto M, Canzonieri V, De Paoli P, Cannizzaro R, De Re V. Differential proteomics of Helicobacter pylori associated with autoimmune atrophic gastritis. Mol Med 2014; 20:57-71. [PMID: 24395566 DOI: 10.2119/molmed.2013.00076] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 12/23/2013] [Indexed: 12/20/2022] Open
Abstract
Atrophic autoimmune gastritis (AAG) is a condition of chronic inflammation and atrophy of stomach mucosa, for which development can be partially triggered by the bacterial pathogen Helicobacter pylori (HP). HP can cause a variety of gastric diseases, such as duodenal ulcer (DU) or gastric cancer (GC). In this study, a comparative proteomic approach was used by two-dimensional fluorescence difference gel electrophoresis (DIGE) to identify differentially expressed proteins of HP strains isolated from patients with AAG, to identify markers of HP strain associated with AAG. Proteome profiles of HP isolated from GC or DU were used as a reference to compare proteomic levels. Proteomics analyses revealed 27 differentially expressed spots in AAG-associated HP in comparison with GC, whereas only 9 differential spots were found in AAG-associated HP profiles compared with DU. Proteins were identified after matrix-assisted laser desorption ionization (MALDI)-TOF and peptide mass fingerprinting. Some AAG-HP differential proteins were common between DU- and GC-HP (peroxiredoxin, heat shock protein 70 [HSP70], adenosine 5'-triphosphate [ATP] synthase subunit α, flagellin A). Our results presented here may suggest that comparative proteomes of HP isolated from AAG and DU share more common protein expression than GC and provide subsets of putative AAG-specific upregulated or downregulated proteins that could be proposed as putative markers of AAG-associated HP. Other comparative studies by two-dimensional maps integrated with functional genomics of candidate proteins will undoubtedly contribute to better decipher the biology of AAG-associated HP strains.
Collapse
Affiliation(s)
- Ombretta Repetto
- Facility of Bio-Proteomics, Centro di Riferimento Oncologico (CRO), Aviano National Cancer Institute, Aviano, Italy
| | - Stefania Zanussi
- Microbiology-Immunology and Virology, Centro di Riferimento Oncologico (CRO), Aviano National Cancer Institute, Aviano, Italy
| | - Mariateresa Casarotto
- Microbiology-Immunology and Virology, Centro di Riferimento Oncologico (CRO), Aviano National Cancer Institute, Aviano, Italy
| | - Vincenzo Canzonieri
- Pathology Unit, Centro di Riferimento Oncologico (CRO), Aviano National Cancer Institute, Aviano, Italy
| | - Paolo De Paoli
- Facility of Bio-Proteomics, Centro di Riferimento Oncologico (CRO), Aviano National Cancer Institute, Aviano, Italy
| | - Renato Cannizzaro
- Gastroenterology Unit, Centro di Riferimento Oncologico (CRO), Aviano National Cancer Institute, Aviano, Italy
| | - Valli De Re
- Facility of Bio-Proteomics, Centro di Riferimento Oncologico (CRO), Aviano National Cancer Institute, Aviano, Italy
| |
Collapse
|
23
|
Kavanaugh DW, O’Callaghan J, Buttó LF, Slattery H, Lane J, Clyne M, Kane M, Joshi L, Hickey RM. Exposure of Bifidobacterium longum subsp. infantis to Milk Oligosaccharides Increases Adhesion to Epithelial Cells and Induces a Substantial Transcriptional Response. PLoS One 2013; 8:e67224. [PMID: 23805302 PMCID: PMC3689703 DOI: 10.1371/journal.pone.0067224] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 05/21/2013] [Indexed: 12/30/2022] Open
Abstract
In this study, we tested the hypothesis that milk oligosaccharides may contribute not only to selective growth of bifidobacteria, but also to their specific adhesive ability. Human milk oligosaccharides (3'sialyllactose and 6'sialyllactose) and a commercial prebiotic (Beneo Orafti P95; oligofructose) were assayed for their ability to promote adhesion of Bifidobacterium longum subsp. infantis ATCC 15697 to HT-29 and Caco-2 human intestinal cells. Treatment with the commercial prebiotic or 3'sialyllactose did not enhance adhesion. However, treatment with 6'sialyllactose resulted in increased adhesion (4.7 fold), while treatment with a mixture of 3'- and 6'-sialyllactose substantially increased adhesion (9.8 fold) to HT-29 intestinal cells. Microarray analyses were subsequently employed to investigate the transcriptional response of B. longum subsp. infantis to the different oligosaccharide treatments. This data correlated strongly with the observed changes in adhesion to HT-29 cells. The combination of 3'- and 6'-sialyllactose resulted in the greatest response at the genetic level (both in diversity and magnitude) followed by 6'sialyllactose, and 3'sialyllactose alone. The microarray data was further validated by means of real-time PCR. The current findings suggest that the increased adherence phenotype of Bifidobacterium longum subsp. infantis resulting from exposure to milk oligosaccharides is multi-faceted, involving transcription factors, chaperone proteins, adhesion-related proteins, and a glycoside hydrolase. This study gives additional insight into the role of milk oligosaccharides within the human intestine and the molecular mechanisms underpinning host-microbe interactions.
Collapse
Affiliation(s)
- Devon W. Kavanaugh
- Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
- Glycoscience Group, National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | - John O’Callaghan
- Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Ludovica F. Buttó
- Department of Microbiology, University College Cork, Cork, Ireland
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | - Helen Slattery
- Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Jonathan Lane
- Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Marguerite Clyne
- Glycoscience Group, National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
- School of Medicine and Medical Science, University College Dublin, Belfield, Dublin, Ireland
| | - Marian Kane
- Glycoscience Group, National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | - Lokesh Joshi
- Glycoscience Group, National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | - Rita M. Hickey
- Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
- Glycoscience Group, National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| |
Collapse
|
24
|
Stent A, Every AL, Sutton P. Helicobacter pylori defense against oxidative attack. Am J Physiol Gastrointest Liver Physiol 2012; 302:G579-87. [PMID: 22194421 DOI: 10.1152/ajpgi.00495.2011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Helicobacter pylori is a microaerophilic, gram-negative pathogen of the human stomach. Despite the chronic active gastritis that develops following colonization, H. pylori is able to persist unharmed in the stomach for decades. Much of the damage caused by gastric inflammation results from the accumulation of reactive oxygen/nitrogen species within the stomach environment, which can induce oxidative damage in a wide range of biological molecules. Without appropriate defenses, this oxidative damage would be able to rapidly kill nearby H. pylori, but the organism employs a range of measures, including antioxidant enzymes, biological repair systems, and inhibitors of oxidant generation, to counter the attack. Despite the variety of measures employed to defend against oxidative injury, these processes are intimately interdependent, and any deficiency within the antioxidant system is generally sufficient to cause substantial impairment of H. pylori viability and persistence. This review provides an overview of the development of oxidative stress during H. pylori gastritis and examines the methods the organism uses to survive the resultant damage.
Collapse
Affiliation(s)
- Andrew Stent
- Centre for Animal Biotechnology, School of Veterinary Science, University of Melbourne, Parkville, Victoria, Australia
| | | | | |
Collapse
|
25
|
CbpA acts as a modulator of HspR repressor DNA binding activity in Helicobacter pylori. J Bacteriol 2011; 193:5629-36. [PMID: 21840971 DOI: 10.1128/jb.05295-11] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The ability of pathogens to cope with disparate environmental stresses is a crucial feature for bacterial survival and for the establishment of a successful infection and colonization of the host; in this respect, chaperones and heat shock proteins (HSPs) play a fundamental role in host-pathogen interactions. In Helicobacter pylori, the expression of the major HSPs is tightly regulated through dedicated transcriptional repressors (named HspR and HrcA), as well as via a GroESL-dependent posttranscriptional feedback control acting positively on the DNA binding affinity of the HrcA regulator itself. In the present work we show that the CbpA chaperone also participates in the posttranscriptional feedback control of the H. pylori heat shock regulatory network. Our experiments suggest that CbpA specifically modulates HspR in vitro binding to DNA without affecting HrcA regulator activity. In particular, CbpA directly interacts with HspR, preventing the repressor from binding to its target operators. This interaction takes place only when HspR is not bound to DNA since CbpA is unable to affect HspR once the repressor is bound to its operator site. Accordingly, in vivo overexpression of CbpA compromises the response kinetics of the regulatory circuit, inducing a failure to restore HspR-dependent transcriptional repression after heat shock. The data presented in this work support a model in which CbpA acts as an important modulator of HspR regulation by fine-tuning the shutoff response of the regulatory circuit that governs HSP expression in H. pylori.
Collapse
|
26
|
Safaei HG, Rahimi E, Zandi A, Rashidipour A. Helicobacter pylori as a zoonotic infection: the detection of H. pylori antigens in the milk and faeces of cows. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2011; 16:184-7. [PMID: 22091229 PMCID: PMC3214301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2010] [Accepted: 11/14/2010] [Indexed: 11/28/2022]
Abstract
BACKGROUND The prevalence of Helicobacter pylori infection, which may increase the risk of gastritis, peptic ulcers, and cancer, has increased worldwide. This number is estimated to be around 70-90% in developing countries and 25-50% in developed countries. It is possible that the bacterium can be transmitted via food and water as well as zoonotically and iatrogenically. Because of high prevalence of this infection in Iran, the aim of this study is to examine whether H. pylori infection might be transmitted from cow's milk and faeces. METHODS The existence of the H. pylori antibody and antigen was investigated in samples of serum, milk, and faeces from 92 lactating Holstein cows in Shahrekord, Iran. The H. pylori antigen and antibody were detected using ELISA and were confirmed by PCR. RESULTS It was found that out of 92 serum specimens, 25 (27%) of the cows were positive for the H. pylori antibody and 67 specimens were negative. From these 25 seropositive cows, 10 (40%) faeces samples and four (16%) milk samples were antigen positive for H. pylori. Four of the antigen-positive milk specimens were also antigen positive for faeces. The existence of the UreC gene was also confirmed in positive samples of milk and faeces. CONCLUSIONS There is a possibility that cow's milk is a transmission mode in H. pylori infection and faecal contamination and inappropriate management processes could transfer H. pylori to humans. The awareness of the H. pylori epidemiology and its method of distribution are necessary for public health measures and controlling the spread of this bacterium. Further investigation with a greater sample number is necessary to verify the ability of H. pylori transmission via milk consumption.
Collapse
Affiliation(s)
- Hajieh Ghasemian Safaei
- Department of Microbiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran., Corresponding Author E-mail:
| | - Ebrahim Rahimi
- Department of Food Hygiene, School of Veterinary Medicine, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Ashkan Zandi
- Department of Food Hygiene, School of Veterinary Medicine, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Alireza Rashidipour
- Department of Food Hygiene, School of Veterinary Medicine, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| |
Collapse
|
27
|
Mullaney E, Brown PA, Smith SM, Botting CH, Yamaoka YY, Terres AM, Kelleher DP, Windle HJ. Proteomic and functional characterization of the outer membrane vesicles from the gastric pathogen Helicobacter pylori. Proteomics Clin Appl 2009; 3:785-96. [DOI: 10.1002/prca.200800192] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2008] [Revised: 01/14/2009] [Accepted: 01/19/2009] [Indexed: 12/28/2022]
|
28
|
Morihara F, Hifumi E, Yamada M, Nishizono A, Uda T. Therapeutic effects of molecularly designed antigen UREB138 for mice infected withHelicobacter pylori. Biotechnol Bioeng 2008; 100:634-43. [DOI: 10.1002/bit.21804] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
29
|
Nayak AK, Rose JB. Detection of Helicobacter pylori in sewage and water using a new quantitative PCR method with SYBR green. J Appl Microbiol 2008; 103:1931-41. [PMID: 17953603 DOI: 10.1111/j.1365-2672.2007.03435.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIMS To demonstrate the application of a new quantitative polymerase chain reaction (qPCR) technique for the determination of Helicobacter pylori concentrations in water, and to use this method to investigate the occurrence of the bacteria in sewage. The other aim was to study the survival capacity and detectability of the bacteria in artificially contaminated groundwater at different temperatures of 4 and 15 degrees C. METHODS AND RESULTS The detection of H. pylori in water was aided by PCR using specific primers designed for the amplification of a fragment within the major vacuolating cytotoxin gene. Conventional culture was compared with conventional PCR and the new real-time (RT) qPCR approach for the quantification of the bacterium. Helicobacter pylori remained culturable for 120 h at 4 degrees C as opposed to only 24 h at 15 degrees C. RT qPCR demonstrated a 100-fold greater sensitivity for the detection of H. pylori DNA in comparison with conventional PCR. Scanning electron microscopic (SEM) observation showed that the normal spiral form changed to a coccoid form after 24 and 72 h at 15 and 4 degrees C, respectively. Helicobacter pylori was found at 2-28 cells ml(-1) in sewage, of the 23 sewage samples - 84% were positive for H. pylori species-specific vacuolating cyctotoxin gene (vacA) by RT qPCR, but were negative by conventional PCR. CONCLUSIONS The RT qPCR assay provided a specific, sensitive and rapid method for the quantitative detection of H. pylori in sewage. This molecular method would be valuable in studying the prevalence of H. pylori as required by the United States Environmental Protection Agency Contaminant Candidate List, particularly in nondisinfected ground waters, in sewage as a source of contamination, and for addressing the possible presence of viable but nonculturable of H. pylori. SIGNIFICANCE AND IMPACT OF THE STUDY The quantitative detection of H. pylori by rapid and less-expensive methods than the TaqMan Assay using SYBR green could be an important tool to monitor infection in community by measuring the concentrations in sewage and to meet the new regulatory and risk-based frameworks for water supplies.
Collapse
Affiliation(s)
- A K Nayak
- Michigan State University, Department of Fisheries and Wildlife, East Lansing, MI 48824, USA
| | | |
Collapse
|
30
|
Sicinschi LA, Correa P, Peek RM, Camargo MC, Delgado A, Piazuelo MB, Romero-Gallo J, Bravo LE, Schneider BG. Helicobacter pylori genotyping and sequencing using paraffin-embedded biopsies from residents of colombian areas with contrasting gastric cancer risks. Helicobacter 2008; 13:135-45. [PMID: 18321303 PMCID: PMC2977907 DOI: 10.1111/j.1523-5378.2008.00554.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND cagA-positive and vacA s1 and m1 genotypes of Helicobacter pylori are associated with an elevated risk of gastric cancer (GC). We determined these genotypes using paraffin-embedded gastric biopsy specimens harvested from infected individuals and compared genotype distributions in two Colombian populations residing in geographic regions with a high and low incidence of GC. METHODS DNA from paraffin-embedded gastric biopsies from 107 adults was amplified using primers specific for cagA, for the cag'empty site', for the s and m alleles of vacA, and for H. pylori 16S rRNA. RESULTS H. pylori infection was detected by molecular assays in 97 (90.7%) biopsies. Complete genotyping of cagA and vacA was achieved in 94 (96.9%) cases. The presence of cagA was detected in 78 of 97 cases (80.4%); when considered separately, cagA and vacA s regions were not significantly associated with a particular geographic area. The vacA m1 allele and s1m1 genotypes were more common in the area of high risk for GC (p = .037 and p = .044, respectively), while the vacA m2 allele and s2m2 genotypes were more prevalent in the low-risk area. The prevalence of the combination of cagA-positive, vacA s1m1 genotypes was 84.3% and 60.5% for high and low risk areas, respectively (p = .011). CONCLUSIONS H. pylori cagA and vacA genotyping from paraffin-embedded gastric biopsies permitted reliable typability and discrimination. The more virulent cagA-positive s1m1 strains, as well as vacA m1 genotype, were more prevalent in high risk than in low risk areas, which may contribute to the difference in GC risk between those two regions.
Collapse
Affiliation(s)
- Liviu A. Sicinschi
- Division of Gastroenterology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Pelayo Correa
- Division of Gastroenterology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Richard M. Peek
- Division of Gastroenterology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - M. Constanza Camargo
- Division of Gastroenterology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Alberto Delgado
- Division of Gastroenterology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - M. Blanca Piazuelo
- Division of Gastroenterology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Judith Romero-Gallo
- Division of Gastroenterology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Luis E. Bravo
- Department of Pathology, School of Medicine, Universidad del Valle, Cali, Colombia
| | - Barbara G. Schneider
- Division of Gastroenterology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
31
|
Abstract
Epidemiological evidence links high-salt diets and Helicobacter pylori infection with increased risk of developing gastric maladies. The mechanism by which elevated sodium chloride content causes these manifestations is unclear. Here we characterize the response of H. pylori to temporal changes in sodium chloride concentration and show that growth, cell morphology, survival, and virulence factor expression are all altered by increased salt concentration.
Collapse
|
32
|
Manente L, Perna A, Buommino E, Altucci L, Lucariello A, Citro G, Baldi A, Iaquinto G, Tufano MA, De Luca A. The Helicobacter pylori's protein VacA has direct effects on the regulation of cell cycle and apoptosis in gastric epithelial cells. J Cell Physiol 2008; 214:582-7. [PMID: 17786942 DOI: 10.1002/jcp.21242] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In this study, we have evaluated the effects on cell cycle regulation of VacA alone and in combination with other two Helicobacter pylori proteins, cytotoxin-associated protein (CagA) and HspB, using the human gastric epithelial cells (AGS). Our results indicate that VacA alone was able to inhibit the G1 to S progression of the cell cycle. The VacA capacity of inhibiting cell progression from G1 to S phase was also observed when cells were co-transfected with CagA or HspB. Moreover, VacA over-expression caused apoptosis in AGS cells through activation of caspase 8 and even more of caspase 9, thus indicating an involvement of both the receptor-mediated and the mitochondrial pathways of apoptosis. Indeed, the two pathways probably can co-operate to execute cell death with a prevalence of the mitochondrial pathways. Our data taken together provide additional information to further enhance our understanding of the molecular mechanism by which H. pylori proteins alter the growth status of human gastric epithelial cells.
Collapse
Affiliation(s)
- L Manente
- Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
De Luca A, De Falco M, Manente L, Dattilo D, Lucariello A, Esposito V, Gnarini M, Citro G, Baldi A, Tufano MA, Iaquinto G. Helicobacter pylori heat shock protein B (HspB) localizes in vivo in the gastric mucosa and MALT lymphoma. J Cell Physiol 2008; 216:78-82. [DOI: 10.1002/jcp.21376] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
34
|
Roncarati D, Danielli A, Spohn G, Delany I, Scarlato V. Transcriptional regulation of stress response and motility functions in Helicobacter pylori is mediated by HspR and HrcA. J Bacteriol 2007; 189:7234-43. [PMID: 17693507 PMCID: PMC2168435 DOI: 10.1128/jb.00626-07] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The hrcA and hspR genes of Helicobacter pylori encode two transcriptional repressor proteins that negatively regulate expression of the groES-groEL and hrcA-grpE-dnaK operons. While HspR was previously shown to bind far upstream of the promoters transcribing these operons, the binding sites of HrcA were not identified. Here, we demonstrate by footprinting analysis that HrcA binds to operator elements similar to the so-called CIRCE sequences overlapping both promoters. Binding of HspR and HrcA to their respective operators occurs in an independent manner, but the DNA binding activity of HrcA is increased in the presence of GroESL, suggesting that the GroE chaperonin system corepresses transcription together with HrcA. Comparative transcriptome analysis of the wild-type strain and hspR and hrcA singly and doubly deficient strains revealed that a set of 14 genes is negatively regulated by the action of one or both regulators, while a set of 29 genes is positively regulated. While both positive and negative regulation of transcription by HspR and/or HrcA could be confirmed by RNA primer extension analyses for two representative genes, binding of either regulator to the promoters could not be detected, indicating that transcriptional regulation at these promoters involves indirect mechanisms. Strikingly, 14 of the 29 genes which were found to be positively regulated by HspR or HrcA code for proteins involved in flagellar biosynthesis. Accordingly, loss of motility functions was observed for HspR and HrcA single or double mutants. The possible regulatory intersections of the heat shock response and flagellar assembly are discussed.
Collapse
Affiliation(s)
- Davide Roncarati
- Department of Biology, University of Bologna, Via Selmi 3, 40126, Bologna, Italy
| | | | | | | | | |
Collapse
|
35
|
Morihara F, Fujii R, Hifumi E, Nishizono A, Uda T. Effects of vaccination by a recombinant antigen ureB138 (a segment of the beta-subunit of urease) against Helicobacter pylori infection. J Med Microbiol 2007; 56:847-853. [PMID: 17510273 DOI: 10.1099/jmm.0.47061-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Helicobacter pylori has to counteract acidity during colonization in the stomach. The most important region for the enzymic activity of H. pylori urease, consisting of 138 aa (ureB138), was determined by a comparison of the homology of amino acid sequences, and a structural analysis, between urease of H. pylori and various other species. This region was expressed in Escherichia coli as a fusion protein with glutathione S-transferase (GST), which was cleaved by PreScission protease between the GST moiety and ureB138. The ureB138 protein was then purified by gel filtration. The polyclonal antibody (pAb) induced by immunization with the purified ureB138 could suppress urease activity by about 50 %, while the pAb against the H. pylori urease did not show any inhibitory effect at all. Immunohistochemical analysis indicated that the ureB138-specific pAb specifically recognized the H. pylori infecting human gastric tissues. The effects of vaccination of recombinant ureB138 against infection by this organism were also examined. Specific IgG and IgA antibodies against H. pylori urease were induced in the serum of mice immunized with ureB138. A reduction in the number of colonizing H. pylori was observed in mice treated with ureB138 compared to ones treated with BSA and infection control mice. In the protected mice, severe gastritis characterized by marked infiltration of mononuclear cells was noted compared with the gastritis observed in unprotected mice. Immunohistochemical staining for IgA in gastric mucosa showed that the number of mice positively stained with IgA was significantly higher in ureB138-vaccinated mice than in non-vaccinated mice. This indicates that local IgA antibody and severe post-immunization gastritis correlate well with the protection of mice against H. pylori infection.
Collapse
Affiliation(s)
- Fumiko Morihara
- CREST of JST (Japan Science and Technology Corporation), 4-1-8 Hon-cho, Kawaguchi-shi, Saitama 332-0012, Japan
- Fukuyama Medical Laboratory Co. Ltd, 1-23-21 Kusado-cho, Fukuyama-shi, Hiroshima 720-8510, Japan
| | - Ryoji Fujii
- Fukuyama Medical Laboratory Co. Ltd, 1-23-21 Kusado-cho, Fukuyama-shi, Hiroshima 720-8510, Japan
| | - Emi Hifumi
- Research Center for Applied Medical Engineering, Oita University, 700 Tannohara, Oita-shi, Oita 870-1192, Japan
- Department of Life Sciences, Faculty of Bioscience and Environment, Prefectural University of Hiroshima, 562 Nanatsuka-cho, Shobara-shi, Hiroshima 727-0023, Japan
- CREST of JST (Japan Science and Technology Corporation), 4-1-8 Hon-cho, Kawaguchi-shi, Saitama 332-0012, Japan
| | - Akira Nishizono
- Department of Infectious Diseases, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu-shi, Oita 879-5593, Japan
- CREST of JST (Japan Science and Technology Corporation), 4-1-8 Hon-cho, Kawaguchi-shi, Saitama 332-0012, Japan
| | - Taizo Uda
- Department of Life Sciences, Faculty of Bioscience and Environment, Prefectural University of Hiroshima, 562 Nanatsuka-cho, Shobara-shi, Hiroshima 727-0023, Japan
- CREST of JST (Japan Science and Technology Corporation), 4-1-8 Hon-cho, Kawaguchi-shi, Saitama 332-0012, Japan
| |
Collapse
|
36
|
Roncarati D, Spohn G, Tango N, Danielli A, Delany I, Scarlato V. Expression, purification and characterization of the membrane-associated HrcA repressor protein of Helicobacter pylori. Protein Expr Purif 2007; 51:267-75. [PMID: 16997572 DOI: 10.1016/j.pep.2006.08.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2006] [Revised: 08/03/2006] [Accepted: 08/06/2006] [Indexed: 11/22/2022]
Abstract
Helicobacter pylori, a microaerophilic, gram-negative bacterium is a human pathogen that colonizes the gastric niche and is associated with several acute and chronic stomach diseases. In order to survive in the gastric environment and become pathogenic, the bacterium relies on a plethora of virulence factors, which also include heat shock proteins. We previously showed that two out of the three operons encoding the major cellular chaperone machineries are transcriptionally repressed by two regulators, HrcA and HspR. Till now, molecular studies aimed at the understanding of the role of each protein in controlling transcription was hampered by toxicity and insolubility of HrcA in heterologous expression systems. Similar problems were encountered by many other groups studying HrcA from different bacteria. In this study, we analyzed the amino acid sequence of HrcA that predicted association of this protein to the inner membrane, which was experimentally verified. Subsequently, we implemented a dedicated induction protocol which enabled the overexpression of the recombinant His-HrcA protein in the soluble fraction of Escherichia coli cells. Moreover, we developed a purification procedure for His-HrcA that allowed us to obtain highly pure preparation of the protein. The functionality of the purified protein was then confirmed with an in vitro DNA-binding assay.
Collapse
Affiliation(s)
- Davide Roncarati
- Department of Biology, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
| | | | | | | | | | | |
Collapse
|
37
|
Pyndiah S, Lasserre JP, Ménard A, Claverol S, Prouzet-Mauléon V, Mégraud F, Zerbib F, Bonneu M. Two-dimensional blue native/SDS gel electrophoresis of multiprotein complexes from Helicobacter pylori. Mol Cell Proteomics 2006; 6:193-206. [PMID: 17092930 DOI: 10.1074/mcp.m600363-mcp200] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The study of protein interactions constitutes an important domain to understand the physiology and pathogenesis of microorganisms. The two-dimensional blue native/SDS-PAGE was initially reported to analyze membrane protein complexes. In this study, both cytoplasmic and membrane complexes of a bacterium, the strain J99 of the gastric pathogen Helicobacter pylori, were analyzed by this method. It was possible to identify 34 different proteins grouped in 13 multiprotein complexes, 11 from the cytoplasm and two from the membrane, either previously reported partially or totally in the literature. Besides complexes involved in H. pylori physiology, this method allowed the description of interactions involving known pathogenic factors such as (i) urease with the heat shock protein GroEL or with the putative ketol-acid reductoisomerase IlvC and (ii) the cag pathogenicity island CagA protein with the DNA gyrase GyrA as well as insight on the partners of TsaA, a peroxide reductase/stress-dependent molecular chaperone. The two-dimensional blue native/SDS-PAGE combined with mass spectrometry is a potential tool to study the differences in complexes isolated in various situations and also to study the interactions between bacterial and eucaryotic cell proteins.
Collapse
|
38
|
Gerrits MM, van Vliet AHM, Kuipers EJ, Kusters JG. Helicobacter pylori and antimicrobial resistance: molecular mechanisms and clinical implications. THE LANCET. INFECTIOUS DISEASES 2006; 6:699-709. [PMID: 17067919 DOI: 10.1016/s1473-3099(06)70627-2] [Citation(s) in RCA: 211] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Helicobacter pylori is an important human pathogen that colonises the stomach of about half of the world's population. The bacterium has now been accepted as the causative agent of several gastroduodenal disorders, ranging from chronic active gastritis and peptic ulcer disease to gastric cancer. The recognition of H pylori as a gastric pathogen has had a substantial effect on gastroenterological practice, since many untreatable gastroduodenal disorders with uncertain cause became curable infectious diseases. Treatment of H pylori infection results in ulcer healing and can reduce the risk of gastric cancer development. Although H pylori is susceptible to many antibiotics in vitro, only a few antibiotics can be used in vivo to cure the infection. The frequent indication for anti-H pylori therapy, together with the limited choice of antibiotics, has resulted in the development of antibiotic resistance in H pylori, which substantially impairs the treatment of H pylori-associated disorders. Antimicrobial resistance in H pylori is widespread, and although the prevalence of antimicrobial resistance shows regional variation per antibiotic, it can be as high as 95%. We focus on the treatment of H pylori infection and on the clinical relevance, mechanisms, and diagnosis of antimicrobial resistance.
Collapse
Affiliation(s)
- Monique M Gerrits
- Department of Gastroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, Rotterdam, Netherlands
| | | | | | | |
Collapse
|
39
|
Abstract
The gastric mucosa separates the underlying tissue from the vast array of antigens that traffic through the stomach lumen. While the extreme pH of this environment is essential in aiding the activation of enzymes and food digestion, it also renders the gastric epithelium free from bacterial colonization, with the exception of one important human pathogen, H pylori. This bacterium has developed mechanisms to survive the harsh environment of the stomach, actively move through the mucosal layer, attach to the epithelium, evade immune responses, and achieve persistent colonization. While a hallmark of this infection is a marked inflammatory response with the infiltration of various immune cells into the infected gastric mucosa, the host immune response is unable to clear the infection and may actually contribute to the associated pathogenesis. Here, we review the host responses involved during infection with H pylori and how they are influenced by this bacterium.
Collapse
|
40
|
Schwartz JT, Allen LAH. Role of urease in megasome formation and Helicobacter pylori survival in macrophages. J Leukoc Biol 2006; 79:1214-25. [PMID: 16543403 PMCID: PMC1868427 DOI: 10.1189/jlb.0106030] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Previous studies have demonstrated that Helicobacter pylori (Hp) delays its entry into macrophages and persists inside megasomes, which are poorly acidified and accumulate early endosome autoantigen 1. Herein, we explored the role of Hp urease in bacterial survival in murine peritoneal macrophages and J774 cells. Plasmid-free mutagenesis was used to replace ureA and ureB with chloramphenicol acetyltransferase in Hp Strains 11637 and 11916. ureAB null Hp lacked detectable urease activity and did not express UreA or UreB as judged by immunoblotting. Deletion of ureAB had no effect on Hp binding to macrophages or the rate or extent of phagocytosis. However, intracellular survival of mutant organisms was impaired significantly. Immunofluorescence microscopy demonstrated that (in contrast to parental organisms) mutant Hp resided in single phagosomes, which were acidic and accumulated the lysosome marker lysosome-associated membrane protein-1 but not early endosome autoantigen 1. A similar phenotype was observed for spontaneous urease mutants derived from Hp Strain 60190. Treatment of macrophages with bafilomycin A1, NH4Cl, or chloroquine prevented acidification of phagosomes containing mutant Hp. However, only ammonium chloride enhanced bacterial viability significantly. Rescue of ureAB null organisms was also achieved by surface adsorption of active urease. Altogether, our data indicate a role for urease and urease-derived ammonia in megasome formation and Hp survival.
Collapse
Affiliation(s)
- Justin T. Schwartz
- Department of Medicine, University of Iowa and the VA Medical Center, Iowa City
- Department of Microbiology, University of Iowa and the VA Medical Center, Iowa City
| | - Lee-Ann H. Allen
- Department of Medicine, University of Iowa and the VA Medical Center, Iowa City
- Department of Microbiology, University of Iowa and the VA Medical Center, Iowa City
- Inflammation Program, University of Iowa and the VA Medical Center, Iowa City
| |
Collapse
|
41
|
Pierzchalski P, Krawiec A, Ptak-Belowska A, Barańska A, Konturek SJ, Pawlik WW. The mechanism of heat-shock protein 70 gene expression abolition in gastric epithelium caused by Helicobacter pylori infection. Helicobacter 2006; 11:96-104. [PMID: 16579839 DOI: 10.1111/j.1523-5378.2006.00383.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND The members of the family of heat shock factors coordinate the inducible transcription of heat shock genes in response to diverse stimuli. Any disturbances in signal transduction may lead to the attenuation of heat shock proteins synthesis and to cell death due to apoptosis. It has been shown by others that different nuclear factors, such as nuclear factor interleukin 6 or signal transducer and activator of transcription 3, co-operate with heat shock factors, mostly enhancing their activator effect on heat shock proteins genes expression. Therefore, we sought to determine whether apoptosis induced in the gastric epithelium exposed to live Helicobacter pylori might occur due to the elimination of HSP70 expression and deregulation of the heat shock response of the cell. MATERIALS AND METHODS Experiments were performed on KATO III gastric epithelial cells exposed to live cagA, vacA expressing Hp over different periods of time. Total cellular RNA, cytoplasmic and nuclear proteins were isolated for polymerase chain reaction, western-blot, electrophoretic mobility shift assay, decoy and co-immunoprecipitation studies. RESULTS We found that in human gastric epithelium exposed to Helicobacter pylori, heat shock factor 1 is bound and restrained in complexes by phosphorylated signal transducer and activator of transcription 3 protein. In consequence, heat shock factor 1 bound up with phosphorylated signal transducer and activator of transcription 3 protein is unable to activate HSP70 protein synthesis in KATO III cells under stress conditions. Helicobacter pylori also causes changes in bax/bcl-2 cellular equilibrium, leading to the induction of apoptosis. CONCLUSIONS The observed phenomenon might be the mechanism whereby gastric epithelium adapts to the infection of Helicobacter pylori, eliminating cells which are damaged or altered by bacterial cytotoxic products from the tissue.
Collapse
Affiliation(s)
- Piotr Pierzchalski
- Jagiellonian University Medical College, Grzegórzecka 16, 31-531 Kraków, Poland.
| | | | | | | | | | | |
Collapse
|
42
|
Bergonzelli GE, Granato D, Pridmore RD, Marvin-Guy LF, Donnicola D, Corthésy-Theulaz IE. GroEL of Lactobacillus johnsonii La1 (NCC 533) is cell surface associated: potential role in interactions with the host and the gastric pathogen Helicobacter pylori. Infect Immun 2006; 74:425-34. [PMID: 16368998 PMCID: PMC1346591 DOI: 10.1128/iai.74.1.425-434.2006] [Citation(s) in RCA: 201] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2005] [Revised: 05/31/2005] [Accepted: 10/13/2005] [Indexed: 11/20/2022] Open
Abstract
Heat shock proteins of the GroEL or Hsp60 class are highly conserved proteins essential to all living organisms. Even though GroEL proteins are classically considered intracellular proteins, they have been found at the surface of several mucosal pathogens and have been implicated in cell attachment and immune modulation. The purpose of the present study was to investigate the GroEL protein of a gram-positive probiotic bacterium, Lactobacillus johnsonii La1 (NCC 533). Its presence at the bacterial surface was demonstrated using a whole-cell enzyme-linked immunosorbent assay and could be detected in bacterial spent culture medium by immunoblotting. To assess binding of La1 GroEL to mucins and intestinal epithelial cells, the La1 GroEL protein was expressed in Escherichia coli. We report here that La1 recombinant GroEL (rGroEL) binds to mucins and epithelial cells and that this binding is pH dependent. Immunomodulation studies showed that La1 rGroEL stimulates interleukin-8 secretion in macrophages and HT29 cells in a CD14-dependent mechanism. This property is common to rGroEL from other gram-positive bacteria but not to the rGroEL of the gastric pathogen Helicobacter pylori. In addition, La1 rGroEL mediates the aggregation of H. pylori but not that of other intestinal pathogens. Our in vitro results suggest that GroEL proteins from La1 and other lactic acid bacteria might play a role in gastrointestinal homeostasis due to their ability to bind to components of the gastrointestinal mucosa and to aggregate H. pylori.
Collapse
|
43
|
Fujita Y, Yamaguchi K, Kamegaya T, Sato H, Semura K, Mutoh K, Kashimoto T, Ohori H, Mukai T. A novel mechanism of autolysis in Helicobacter pylori: possible involvement of peptidergic substances. Helicobacter 2005; 10:567-76. [PMID: 16302982 DOI: 10.1111/j.1523-5378.2005.00364.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Helicobacter pylori survival in a hostile acidic environment is known to be caused by its production of urease, which is not released by known secretion pathways. It has been proposed that H. pylori cells undergo spontaneous autolysis during cultivation and that urease becomes surface-associated only concomitant with bacterial autolysis. The aim of this study was to elucidate mechanisms by which H. pylori cells undergo autolysis during cultivation. MATERIALS AND METHODS Autolysis of H. pylori KZ109 cells was estimated by measuring the turbidity of the culture, by detection of cytoplasmic protein release into the culture supernatant and by scanning electron microscopic observation of H. pylori cells during cultivation. An autolysis-inducing factor (AIF) was partially purified from the culture supernatant by a partition method using ethyl acetate. RESULTS Bacterial turbidity of KZ109 cells was drastically decreased after late-log phase accompanying release of urease and HspB into the extracellular space. Concomitantly, cell lytic activity was detected in the culture supernatant. Scanning electron microscopic observation suggested that partially purified AIF induced cell lysis. It was also shown that the AIF is different from other autolytic enzymes or substances so far reported. CONCLUSIONS This study demonstrated the presence of the peptidergic autolytic substances in the culture supernatant of H. pylori KZ109 cells. The results of this study should be useful for further studies aimed at elucidation of the strategy of survival of H. pylori in the gastric environment and elucidation of the mechanisms of pathogenesis induced by H. pylori.
Collapse
Affiliation(s)
- Yukihiro Fujita
- Laboratory of Biomolecular Function, School of Veterinary Medicine and Animal Sciences, Kitasato University, Towada, Aomori 034-8628, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Berven FS, Karlsen OA, Straume AH, Flikka K, Murrell JC, Fjellbirkeland A, Lillehaug JR, Eidhammer I, Jensen HB. Analysing the outer membrane subproteome of Methylococcus capsulatus (Bath) using proteomics and novel biocomputing tools. Arch Microbiol 2005; 184:362-77. [PMID: 16311759 DOI: 10.1007/s00203-005-0055-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2005] [Revised: 10/13/2005] [Accepted: 11/03/2005] [Indexed: 02/05/2023]
Abstract
High-resolution two-dimensional gel electrophoresis and mass spectrometry has been used to identify the outer membrane (OM) subproteome of the Gram-negative bacterium Methylococcus capsulatus (Bath). Twenty-eight unique polypeptide sequences were identified from protein samples enriched in OMs. Only six of these polypeptides had previously been identified. The predictions from novel bioinformatic methods predicting beta-barrel outer membrane proteins (OMPs) and OM lipoproteins were compared to proteins identified experimentally. BOMP ( http://www.bioinfo.no/tools/bomp ) predicted 43 beta-barrel OMPs (1.45%) from the 2,959 annotated open reading frames. This was a lower percentage than predicted from other Gram-negative proteomes (1.8-3%). More than half of the predicted BOMPs in M. capsulatus were annotated as (conserved) hypothetical proteins with significant similarity to very few sequences in Swiss-Prot or TrEMBL. The experimental data and the computer predictions indicated that the protein composition of the M. capsulatus OM subproteome was different from that of other Gram-negative bacteria studied in a similar manner. A new program, Lipo, was developed that can analyse entire predicted proteomes and give a list of recognised lipoproteins categorised according to their lipo-box similarity to known Gram-negative lipoproteins ( http://www.bioinfo.no/tools/lipo ). This report is the first using a proteomics and bioinformatics approach to identify the OM subproteome of an obligate methanotroph.
Collapse
Affiliation(s)
- Frode S Berven
- Department of Molecular Biology, University of Bergen, Thormøhlensgate 55, N-5020, Bergen, Norway.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Toll-like receptor (TLR) molecules play a frontline role in the defence of the host against infection by microbial pathogens. These molecules, together with the recently described Nod family proteins, have been shown to trigger innate immune responses in host cells via the recognition of highly conserved microbial structures. TLR4, which is the best-characterised of these "pathogen-recognition molecules" (PRMs), was the first to be shown to recognise a specific microbial component: the lipopolysaccharide (LPS) from Gram-negative bacteria. The molecular specificities of the remaining PRMs have, in nearly all cases, now also been elucidated. Host cells belonging to the myeloid cell lineage are known to be particularly responsive to these microbial constituents. Conversely, other cell types such as epithelial cells, were generally thought to be hypo-responsive to stimulation by such molecules. New evidence suggests that these cells are in fact likely to play a fundamental role in host defence against pathogenic micro-organisms. Indeed, epithelial cells afford an initial barrier against the host microflora, and appear to be able to differentiate between pathogenic and commensal micro-organisms. This review article will discuss current knowledge regarding innate immune responses in epithelial and myeloid cells to the model non-invasive pathogen, Helicobacter pylori, which is a major cause of upper gastrointestinal tract disease in humans.
Collapse
Affiliation(s)
- R L Ferrero
- Unité de Pathogénie Bactérienne des Muqueuses, Institut Pasteur, 28, Rue du Dr Roux, 75724 Paris, France.
| |
Collapse
|
46
|
De Luca A, Iaquinto G. Helicobacter pylori and gastric diseases: a dangerous association. Cancer Lett 2004; 213:1-10. [PMID: 15312678 DOI: 10.1016/j.canlet.2004.06.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2004] [Accepted: 06/01/2004] [Indexed: 12/13/2022]
Abstract
The bacterium Helicobacter pylori is linked to the appearance of several gastric diseases and in particular is associated with a progression to gastric cancer. Thistrun -1 bacterium colonizes the gastric mucosa directly interacting with epithelial cells. It is well known that H. pylori is associated with alterations in the gastric epithelial cell cycle, and apoptosis, higher levels of mononuclear and neutrophilic infiltrates, more severe atrophy and intestinal metaplasia. In last years, two mechanisms that interact with each other or not have been proposed: the hyperproliferation of gastric cells and oxidative damage of stomach mucosa. In particular, cell cycle alterations induce mitogenic signals and proto-oncogene expression that may trigger the development of cancer. Contemporary, H. pylori is able to induce polymorphonuclear and mononuclear cells that produce oxygen free radicals that could cause DNA damage to the adjacent cells leading to cancer development. Due to dangerous infection of this bacterium, the scientific community must point out its attention on the development of detection and prevention strategies.
Collapse
Affiliation(s)
- Antonio De Luca
- Department of Medicine and Public Health, Section of Clinical Anatomy, Second University of Naples, Naples, Italy
| | | |
Collapse
|
47
|
Dhar SK, Soni RK, Das BK, Mukhopadhyay G. Molecular mechanism of action of major Helicobacter pylori virulence factors. Mol Cell Biochem 2004; 253:207-15. [PMID: 14619971 DOI: 10.1023/a:1026051530512] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although Helicobacter pylori infects 50% of the total human population, only a small fraction of the infected people suffer from severe diseases like peptic ulcers and gastric adenocarcinoma. H. pylori strains, host genotypes and environmental factors play important role in deciding the extent and severity of the gastroduodenal diseases. The bacteria has developed a unique set of virulence factors to survive in the extreme ecological niche of human stomach. Together these virulence factors make H. pylori one of the most successful human pathogenic bacteria colonizing more than half of the human population. Understanding the mechanism of action of the major H. pylori virulence factors will shed light into the molecular basis of its pathogenicity.
Collapse
Affiliation(s)
- Suman Kumar Dhar
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India.
| | | | | | | |
Collapse
|
48
|
Benoit S, Mehta N, Wang G, Gatlin M, Maier RJ. Requirement of hydD, hydE, hypC and hypE genes for hydrogenase activity in Helicobacter pylori. Microb Pathog 2004; 36:153-7. [PMID: 14726233 DOI: 10.1016/j.micpath.2003.11.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Helicobacter pylori possesses a membrane-bound, nickel containing, hydrogen uptake hydrogenase enzyme; its synthesis requires structural as well as accessory proteins, the latter needed for the complete maturation of the enzyme. Our lab previously characterized mutants in the accessory hyp genes, hypA, hypB, hypD and hypF that were all severely affected for hydrogenase activity, and in some cases (hypA and hypB mutants) also affected for urease activity. This finding prompted us to disrupt the two remaining unstudied hyp genes of H. pylori, hypC and hypE, in order to see if the same pleiotropic effect would be observed. In both mutants hydrogenase activity was abolished but urease activity remained unaffected. Addition of 5 microM nickel into the growth medium partially restored the hydrogenase activity in the hypE mutant and to a lesser extent in the hypC mutant. In addition, we also disrupted the genes HP0634 (referred as hydD in the H. pylori 26695 genome database) and HP0635 (whose function was unknown, referred to here as hydE) to address their possible roles in the hydrogenase synthesis/maturation process. In both cases, hydrogenase activities were abolished and addition of nickel could not restore the activity, suggesting that these proteins are involved in the hydrogenase synthesis process rather than in nickel mobilization/insertion steps.
Collapse
Affiliation(s)
- Stéphane Benoit
- Department of Microbiology, 812 Biological Sciences Building, University of Georgia, Athens, GA 30602, USA.
| | | | | | | | | |
Collapse
|
49
|
Spohn G, Danielli A, Roncarati D, Delany I, Rappuoli R, Scarlato V. Dual control of Helicobacter pylori heat shock gene transcription by HspR and HrcA. J Bacteriol 2004; 186:2956-65. [PMID: 15126455 PMCID: PMC400627 DOI: 10.1128/jb.186.10.2956-2965.2004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The HspR repressor regulates transcription of the groESL, hrcA-grpE-dnaK, and cbpA-hspR-orf operons of Helicobacter pylori. Here we show that two of the HspR-regulated operons, namely, the groESL and dnaK operons, encoding the major cellular chaperone machineries are also regulated by the H. pylori homologue of the HrcA repressor. Similarly to the hspR mutation, deletion of the hrcA gene also leads to complete derepression of the Pgro and Phrc promoters. The presence of both HspR and HrcA is therefore necessary for regulated transcription from these promoters. HrcA binds directly to Pgro and Phrc, likely contacting two inverted repeats with similarity to the CIRCE motif, which are present on both promoters. HrcA regulation is, however, shown to depend on binding of the HspR protein, since deletion of the HspR-binding site of the Pgro promoter leads to loss of heat inducibility of this promoter. In contrast, transcription from the Pcbp promoter is regulated solely by HspR. HspR is also shown to form oligomers in vivo through a stretch of hydrophobic repeats between amino acid positions 66 and 97. The implications of these findings for the elucidation of the networks regulating heat shock gene expression in H. pylori are discussed.
Collapse
Affiliation(s)
- Gunther Spohn
- Biochemistry and Molecular Biology Unit, IRIS, 53100 Siena, Italy
| | | | | | | | | | | |
Collapse
|
50
|
De Luca A, De Falco M, Iaquinto S, Iaquinto G. Effects ofHelicobacter pylori infection on cell cycle progression and the expression of cell cycle regulatory proteins. J Cell Physiol 2004; 200:334-42. [PMID: 15254960 DOI: 10.1002/jcp.20022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Helicobacter pylori lives in the stomach lumen adhering and specifically interacting with gastric epithelial cells. H. pylori infection can cause a broad range of diseases. Although most infected individuals only develop a chronic inflammation of the stomach, some patients progress to chronic gastritis, duodenal ulceration, or, rarely, cancer. H. pylori is able to send and to receive signals from the gastric epithelium, allowing host and bacteria to become linked in a dynamic equilibrium. Several studies have demonstrated that H. pylori infection induces morphological changes of gastric epithelial cells other than cell proliferation, increase of mitosis and mutations. It has also been demonstrated that H. pylori may predispose to cancer by altering gastric epithelial cell turnover acting specifically on transcription factors. Although H. pylori is able to induce several host responses, it specifically perturbs the delicate balance of those factors that usually help to maintain cell homeostasis. The study of mechanisms of interaction between the bacterium and gastric cells will surely help to prevent the increase and diffusion of malignancies all over the world.
Collapse
Affiliation(s)
- Antonio De Luca
- Department of Medicine and Public Health, Section of Clinical Anatomy, Second University of Naples, Naples, Italy.
| | | | | | | |
Collapse
|