1
|
Johnston EL, Guy-Von Stieglitz S, Zavan L, Cross J, Greening DW, Hill AF, Kaparakis-Liaskos M. The effect of altered pH growth conditions on the production, composition, and proteomes of Helicobacter pylori outer membrane vesicles. Proteomics 2024; 24:e2300269. [PMID: 37991474 DOI: 10.1002/pmic.202300269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/03/2023] [Accepted: 10/16/2023] [Indexed: 11/23/2023]
Abstract
Gram-negative bacteria release outer membrane vesicles (OMVs) that contain cargo derived from their parent bacteria. Helicobacter pylori is a Gram-negative human pathogen that produces urease to increase the pH of the surrounding environment to facilitate colonization of the gastric mucosa. However, the effect of acidic growth conditions on the production and composition of H. pylori OMVs is unknown. In this study, we examined the production, composition, and proteome of H. pylori OMVs produced during acidic and neutral pH growth conditions. H. pylori growth in acidic conditions reduced the quantity and size of OMVs produced. Additionally, OMVs produced during acidic growth conditions had increased protein, DNA, and RNA cargo compared to OMVs produced during neutral conditions. Proteomic analysis comparing the proteomes of OMVs to their parent bacteria demonstrated significant differences in the enrichment of beta-lactamases and outer membrane proteins between bacteria and OMVs, supporting that differing growth conditions impacts OMV composition. We also identified differences in the enrichment of proteins between OMVs produced during different pH growth conditions. Overall, our findings reveal that growth of H. pylori at different pH levels is a factor that alters OMV proteomes, which may affect their subsequent functions.
Collapse
Affiliation(s)
- Ella L Johnston
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Bundoora, Australia
- Research Centre for Extracellular Vesicles, La Trobe University, Bundoora, Australia
| | - Sebastian Guy-Von Stieglitz
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Bundoora, Australia
- Research Centre for Extracellular Vesicles, La Trobe University, Bundoora, Australia
| | - Lauren Zavan
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Bundoora, Australia
- Research Centre for Extracellular Vesicles, La Trobe University, Bundoora, Australia
| | - Jonathon Cross
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
| | - David W Greening
- Research Centre for Extracellular Vesicles, La Trobe University, Bundoora, Australia
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
| | - Andrew F Hill
- Research Centre for Extracellular Vesicles, La Trobe University, Bundoora, Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Australia
- Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - Maria Kaparakis-Liaskos
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Bundoora, Australia
- Research Centre for Extracellular Vesicles, La Trobe University, Bundoora, Australia
| |
Collapse
|
2
|
Nim YS, Fong IYH, Deme J, Tsang KL, Caesar J, Johnson S, Pang LTH, Yuen NMH, Ng TLC, Choi T, Wong YYH, Lea SM, Wong KB. Delivering a toxic metal to the active site of urease. SCIENCE ADVANCES 2023; 9:eadf7790. [PMID: 37083535 PMCID: PMC10121161 DOI: 10.1126/sciadv.adf7790] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 03/17/2023] [Indexed: 05/03/2023]
Abstract
Urease is a nickel (Ni) enzyme that is essential for the colonization of Helicobacter pylori in the human stomach. To solve the problem of delivering the toxic Ni ion to the active site without diffusing into the cytoplasm, cells have evolved metal carrier proteins, or metallochaperones, to deliver the toxic ions to specific protein complexes. Ni delivery requires urease to form an activation complex with the urease accessory proteins UreFD and UreG. Here, we determined the cryo-electron microscopy structures of H. pylori UreFD/urease and Klebsiella pneumoniae UreD/urease complexes at 2.3- and 2.7-angstrom resolutions, respectively. Combining structural, mutagenesis, and biochemical studies, we show that the formation of the activation complex opens a 100-angstrom-long tunnel, where the Ni ion is delivered through UreFD to the active site of urease.
Collapse
Affiliation(s)
- Yap Shing Nim
- School of Life Sciences, Centre for Protein Science and Crystallography, State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, China
| | - Ivan Yu Hang Fong
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Justin Deme
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
- Center for Structural Biology, CCR, NCI, Boyles Street, Frederick, MD 21702, USA
| | - Ka Lung Tsang
- School of Life Sciences, Centre for Protein Science and Crystallography, State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, China
| | - Joseph Caesar
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
- Center for Structural Biology, CCR, NCI, Boyles Street, Frederick, MD 21702, USA
| | - Steven Johnson
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
- Center for Structural Biology, CCR, NCI, Boyles Street, Frederick, MD 21702, USA
| | - Longson Tsz Hin Pang
- School of Life Sciences, Centre for Protein Science and Crystallography, State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, China
| | - Nicholas Man Hon Yuen
- School of Life Sciences, Centre for Protein Science and Crystallography, State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, China
| | - Tin Long Chris Ng
- School of Life Sciences, Centre for Protein Science and Crystallography, State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, China
| | - Tung Choi
- School of Life Sciences, Centre for Protein Science and Crystallography, State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, China
| | - Yakie Yat Hei Wong
- School of Life Sciences, Centre for Protein Science and Crystallography, State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, China
| | - Susan M. Lea
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
- Center for Structural Biology, CCR, NCI, Boyles Street, Frederick, MD 21702, USA
| | - Kam-Bo Wong
- School of Life Sciences, Centre for Protein Science and Crystallography, State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
3
|
Zambelli B, Basak P, Hu H, Piccioli M, Musiani F, Broll V, Imbert L, Boisbouvier J, Maroney MJ, Ciurli S. The structure of the high-affinity nickel-binding site in the Ni,Zn-HypA•UreE2 complex. Metallomics 2023; 15:mfad003. [PMID: 36638839 PMCID: PMC10001889 DOI: 10.1093/mtomcs/mfad003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
The maturation pathway for the nickel-dependent enzyme urease utilizes the protein UreE as a metallochaperone to supply Ni(II) ions. In Helicobacter pylori urease maturation also requires HypA and HypB, accessory proteins that are commonly associated with hydrogenase maturation. Herein we report on the characterization of a protein complex formed between HypA and the UreE2 dimer. Nuclear magnetic resonance (NMR) coupled with molecular modelling show that the protein complex apo, Zn-HypA•UreE2, forms between the rigorously conserved Met-His-Glu (MHE motif) Ni-binding N-terminal sequence of HypA and the two conserved His102A and His102B located at the dimer interface of UreE2. This complex forms in the absence of Ni(II) and is supported by extensive protein contacts that include the use of the C-terminal sequences of UreE2 to form additional strands of β-sheet with the Ni-binding domain of HypA. The Ni-binding properties of apo, Zn-HypA•UreE2 and the component proteins were investigated by isothermal titration calorimetry using a global fitting strategy that included all of the relevant equilibria, and show that the Ni,Zn-HypA•UreE2 complex contains a single Ni(II)-binding site with a sub-nanomolar KD. The structural features of this novel Ni(II) site were elucidated using proteins produced with specifically deuterated amino acids, protein point mutations, and the analyses of X-ray absorption spectroscopy, hyperfine shifted NMR features, as well as molecular modeling coupled with quantum-mechanical calculations. The results show that the complex contains a six-coordinate, high-spin Ni(II) site with ligands provided by both component proteins.
Collapse
Affiliation(s)
- Barbara Zambelli
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Priyanka Basak
- Department of Chemistry and Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, MA, USA
| | - Heidi Hu
- Department of Chemistry and Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, MA, USA
| | - Mario Piccioli
- Centre for Magnetic Resonance, Department of Chemistry, University of Florence, Florence Italy
| | - Francesco Musiani
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Valquiria Broll
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Lionel Imbert
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale (IBS), Grenoble, France
| | - Jerome Boisbouvier
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale (IBS), Grenoble, France
| | - Michael J Maroney
- Department of Chemistry and Program in Molecular and Cellular Biology, University of Massachusetts, Amherst, MA, USA
| | - Stefano Ciurli
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
- Centre for Magnetic Resonance, Department of Chemistry, University of Florence, Florence Italy
| |
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
|
Shi J, Zeng Y, Wang H, Niu Y, He P, Chen H. Complete genome sequencing and analysis revealed the nitrogen utilization strategy of a novel Acuticoccus species isolated from surface water of the Indian Ocean. Mar Genomics 2022; 65:100971. [DOI: 10.1016/j.margen.2022.100971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/06/2022] [Accepted: 07/06/2022] [Indexed: 11/24/2022]
|
6
|
Zhang X, Sang S, Guan Q, Tao H, Wang Y, Liu C. Oral Administration of a Shigella 2aT32-Based Vaccine Expressing UreB-HspA Fusion Antigen With and Without Parenteral rUreB-HspA Boost Confers Protection Against Helicobacter pylori in Mice Model. Front Immunol 2022; 13:894206. [PMID: 35769459 PMCID: PMC9234132 DOI: 10.3389/fimmu.2022.894206] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Helicobacter pylori (H. pylori) is a gram-negative pathogen classified as a class I carcinogen. The H. pylori urease B subunit (UreB) and heat shock protein A (HspA) are two important vaccine candidate antigens. In this study, we evaluated the immunogenicity and immunoprotective effect of the attenuated Shigella vector vaccine SH02 expressing the UreB-HspA fusion protein of H. pylori in a mouse model. Oral SH02 with or without subcutaneous injection of rUreB-HspA induced antigen-specific serum IgG, mucosal sIgA, and T cells immune response. Subcutaneous injection of the candidate antigen rUreB-HspA enhanced the level of serum antigen-specific IgG antibodies (p < 0.0001) and the levels of IgG1/IgG2a/IgG2b subtypes. In addition, injection boost also increased the proportion of spleen antigen-specific CD4+CD154+ T cells (p < 0.001), and the proportion of CD4+CD154+ T cells that secrete IFN-γ and IL-17A. Following the H. pylori challenge, the levels of H. pylori colonization in the two experimental groups (Groups A and B) significantly reduced compared with the control group (p < 0.001), indicating that the candidate vaccine yielded a preventive effect of anti-H.pylori infection. Compared with the non-subcutaneous booster injection group (Group A), the subcutaneous booster injection group (Group B) exhibited less gastric inflammation, but there was no significant difference in the level of colonization (p > 0.05). These results lay a foundation for the development of a vaccine against H. pylori and the optimization of immunization methods and procedures to prevent H. pylori infection.
Collapse
Affiliation(s)
- Xin Zhang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
- Department of Pharmacy, Medical Supplies Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Shuli Sang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Qing Guan
- State Key Laboratory of Pathogen and Biosecurity, Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Haoxia Tao
- State Key Laboratory of Pathogen and Biosecurity, Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Yanchun Wang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
- *Correspondence: Chunjie Liu, ; Yanchun Wang,
| | - Chunjie Liu
- State Key Laboratory of Pathogen and Biosecurity, Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
- *Correspondence: Chunjie Liu, ; Yanchun Wang,
| |
Collapse
|
7
|
Calado CRC. Antigenic and conserved peptides from diverse Helicobacter pylori antigens. Biotechnol Lett 2022; 44:535-545. [PMID: 35277779 PMCID: PMC8916697 DOI: 10.1007/s10529-022-03238-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 02/14/2022] [Indexed: 12/20/2022]
Abstract
Since the revolutionary finding of Helicobacter pylori as a common bacterial infection, that a high research effort for its eradication has been conducted. Epitope based-vaccine presents advantages over protein-based, as they can be designed to contain epitopes from diverse proteins, therefore, more easily representing the immune-variability of the bacterial population, while minimizing the toxicity associated to some whole proteins. In the present work, an iterative method, to design antigenic and conserved B-epitopes from diverse virulent factors of H. pylori, was established. The method considered the trade-off between epitopes antigenicity and conservation among the bacterial population. For the method validation, five virulent factors from H. pylori were selected. From each virulent factor, two epitopes were predicted, each with twelve residues of aminoacids. The corresponding ten peptides were synthesised and evaluated by enzyme-linked immunosorbent assay using polyclonal antibodies raised against a specific H. pylori strain. All ten peptides were recognised by the antibodies and were consequently antigenic and conserved. This result could strongly contribute to the design of a multivalent epitope-based vaccine, representing the immunogenetic variability within the bacterial population, leading to a sustained and effective immunogenic protection.
Collapse
Affiliation(s)
- Cecília R C Calado
- CIMOSM - Centro de Investigação em Modelação e Otimização de Sistemas Multifuncionais, ISEL - Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, R. Conselheiro Emídio Navarro 1, 1959-007, Lisboa, Portugal.
| |
Collapse
|
8
|
Nickel as a virulence factor in the Class I bacterial carcinogen, Helicobacter pylori. Semin Cancer Biol 2021; 76:143-155. [PMID: 33865991 DOI: 10.1016/j.semcancer.2021.04.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 04/12/2021] [Indexed: 01/16/2023]
Abstract
Helicobacter pylori is a human bacterial pathogen that causes peptic ulcers and has been designated a Class I carcinogen by the International Agency for Research on Cancer (IARC). Its ability to survive in the acid environment of the stomach, to colonize the stomach mucosa, and to cause cancer, are linked to two enzymes that require nickel-urease and hydrogenase. Thus, nickel is an important virulence factor and the proteins involved in nickel trafficking are potential antibiotic targets. This review summarizes the nickel biochemistry of H. pylori with a focus on the roles of nickel in virulence, nickel homeostasis, maturation of urease and hydrogenase, and the unique nickel trafficking that occurs between the hydrogenase maturation pathway and urease nickel incorporation that is mediated by the metallochaperone HypA and its partner, HypB.
Collapse
|
9
|
Cunha ES, Chen X, Sanz-Gaitero M, Mills DJ, Luecke H. Cryo-EM structure of Helicobacter pylori urease with an inhibitor in the active site at 2.0 Å resolution. Nat Commun 2021; 12:230. [PMID: 33431861 PMCID: PMC7801526 DOI: 10.1038/s41467-020-20485-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/04/2020] [Indexed: 02/08/2023] Open
Abstract
Infection of the human stomach by Helicobacter pylori remains a worldwide problem and greatly contributes to peptic ulcer disease and gastric cancer. Without active intervention approximately 50% of the world population will continue to be infected with this gastric pathogen. Current eradication, called triple therapy, entails a proton-pump inhibitor and two broadband antibiotics, however resistance to either clarithromycin or metronidazole is greater than 25% and rising. Therefore, there is an urgent need for a targeted, high-specificity eradication drug. Gastric infection by H. pylori depends on the expression of a nickel-dependent urease in the cytoplasm of the bacteria. Here, we report the 2.0 Å resolution structure of the 1.1 MDa urease in complex with an inhibitor by cryo-electron microscopy and compare it to a β-mercaptoethanol-inhibited structure at 2.5 Å resolution. The structural information is of sufficient detail to aid in the development of inhibitors with high specificity and affinity.
Collapse
Affiliation(s)
- Eva S. Cunha
- grid.5510.10000 0004 1936 8921Structural Biology and Drug Discovery Group, Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, 0318 Oslo, Norway
| | - Xiaorui Chen
- grid.266093.80000 0001 0668 7243Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697 USA ,grid.506938.10000 0004 0633 8088Present Address: Genomics Research Center, Academia Sinica, 128 Academia Road, Sect. 2, Nankang District, Taipei, Taiwan
| | - Marta Sanz-Gaitero
- grid.5510.10000 0004 1936 8921Structural Biology and Drug Discovery Group, Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, 0318 Oslo, Norway
| | - Deryck J. Mills
- grid.419494.50000 0001 1018 9466Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Hartmut Luecke
- Structural Biology and Drug Discovery Group, Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, 0318, Oslo, Norway. .,Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, 92697, USA. .,Department of Medical Biochemistry, University of Oslo and Oslo University Hospital, 0372, Oslo, Norway. .,Department of Physiology and Biophysics, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
10
|
Liu M, Zhong Y, Chen J, Liu Y, Tang C, Wang X, Zhang Y, Wang P, Logan SM, Chen W, Wei B. Oral immunization of mice with a multivalent therapeutic subunit vaccine protects against Helicobacter pylori infection. Vaccine 2020; 38:3031-3041. [PMID: 32139315 DOI: 10.1016/j.vaccine.2020.02.036] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 02/08/2023]
Abstract
Helicobacter pylori is a human class I carcinogen and no effective prophylactic or therapeutic H. pylori vaccine has yet been marketed. H. pylori can escape the host immune response, but the precise immune protection mechanisms in humans remain unknown. In this study, we developed a multivalent, subunit H. pylori vaccine candidate by formulating three commonly used H. pylori antigens, neutrophil-activating protein (NAP), urease subunit A (UreA) and subunit B (UreB) with the mucosal adjuvant, a double-mutant heat-labile toxin (dmLT) from Escherichia coli, and evaluated its immunogenicity and therapeutic efficacy in a mouse model of H. pylori infection. We found that oral immunization of H. pylori-infected mice significantly reduced gastric bacterial colonization at both 2 and 8 weeks after immunization. The reduction in bacterial burdens was accompanied with significantly increased serum antigen-specific IgG responses and mucosal IgA responses. Moreover, oral immunization also induced Th1/Th17 immune responses, which may play a synergistic role with the specific antibodies in the elimination of H. pylori. Thus, our vaccine candidate appears able to overcome the immune evasion mechanism of H. pylori, restore the suppression of Th2 immune responses with the induction of a strong humoral immune response. These results lay the foundation for the development of an optimized oral therapeutic H. pylori vaccine with increased immunogenicity of UreA and UreB, as well as providing long-term immunity.
Collapse
Affiliation(s)
- Meiying Liu
- National Vaccine and Serum Institute (NVSI), 38 Jinghai 2nd Road, Beijing Economic and Technological Development Zone, Beijing, China
| | - Youxiu Zhong
- National Vaccine and Serum Institute (NVSI), 38 Jinghai 2nd Road, Beijing Economic and Technological Development Zone, Beijing, China
| | - Jing Chen
- National Vaccine and Serum Institute (NVSI), 38 Jinghai 2nd Road, Beijing Economic and Technological Development Zone, Beijing, China
| | - Yu Liu
- National Vaccine and Serum Institute (NVSI), 38 Jinghai 2nd Road, Beijing Economic and Technological Development Zone, Beijing, China
| | - Chongfa Tang
- National Vaccine and Serum Institute (NVSI), 38 Jinghai 2nd Road, Beijing Economic and Technological Development Zone, Beijing, China
| | - Xuewei Wang
- National Vaccine and Serum Institute (NVSI), 38 Jinghai 2nd Road, Beijing Economic and Technological Development Zone, Beijing, China
| | - Yanbin Zhang
- National Vaccine and Serum Institute (NVSI), 38 Jinghai 2nd Road, Beijing Economic and Technological Development Zone, Beijing, China
| | - Ping Wang
- National Vaccine and Serum Institute (NVSI), 38 Jinghai 2nd Road, Beijing Economic and Technological Development Zone, Beijing, China
| | - Susan M Logan
- Human Health Therapeutics (HHT) Research Center, National Research Council Canada, 100 Sussex Drive, Ottawa, ON K1A 0R6, Canada
| | - Wangxue Chen
- Human Health Therapeutics (HHT) Research Center, National Research Council Canada, 100 Sussex Drive, Ottawa, ON K1A 0R6, Canada.
| | - Bo Wei
- National Vaccine and Serum Institute (NVSI), 38 Jinghai 2nd Road, Beijing Economic and Technological Development Zone, Beijing, China.
| |
Collapse
|
11
|
Kataria R, Khatkar A. Lead Molecules for Targeted Urease Inhibition: An Updated Review from 2010 -2018. Curr Protein Pept Sci 2020; 20:1158-1188. [PMID: 30894105 DOI: 10.2174/1389203720666190320170215] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/01/2019] [Accepted: 03/15/2019] [Indexed: 12/14/2022]
Abstract
The field of enzyme inhibition is a tremendous and quickly growing territory of research. Urease a nickel containing metalloenzyme found in bacteria, algae, fungi, and plants brings hydrolysis of urea and plays important role in environmental nitrogen cycle. Apart from this it was found to be responsible for many pathological conditions due to its presence in many microorganisms such as H. Pylori, a ureolytic bacteria having urease which elevates pH of gastric medium by hydrolyzing urea present in alimentary canal and help the bacteria to colonize and spread infection. Due to the infections caused by the various bacterial ureases such as Bacillus pasteurii, Brucella abortus, H. pylori, H. mustelae, Klebsiella aerogenes, Klebsiella tuberculosis, Mycobacterium tuberculosis, Pseudomonas putida, Sporosarcina pasteurii and Yersinia enterocolitica, it has been the current topic of today's research. About a wide range of compounds from the exhaustive literature survey has been discussed in this review which is enveloped into two expansive classes, as Inhibitors from synthetic origin and Inhibitors from natural origin. Moreover active site details of enzyme, mechanism of catalysis of substrate by enzyme, uses of plant urease and its pathogenic behavior has been included in the current review. So, overall, this review article diagrams the current landscape of the developments in the improvements in the thriving field of urease inhibitory movement in medicinal chemistry from year 2010 to 2018, with an emphasis on mechanism of action of inhibitors that may be used for more development of recent and strong urease inhibitors and open up new doors for assist examinations in a standout amongst the most lively and promising regions of research.
Collapse
Affiliation(s)
- Ritu Kataria
- International Institute of Pharmaceutical Sciences, Sonepat, Haryana, India
| | - Anurag Khatkar
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| |
Collapse
|
12
|
Helicobacter pylori Uses the TlpB Receptor To Sense Sites of Gastric Injury. Infect Immun 2019; 87:IAI.00202-19. [PMID: 31262979 DOI: 10.1128/iai.00202-19] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 06/18/2019] [Indexed: 01/24/2023] Open
Abstract
Helicobacter pylori is a pathogen that chronically colonizes the stomachs of approximately half of the world's population and contributes to the development of gastric inflammation. We demonstrated previously in vivo that H. pylori uses motility to preferentially colonize injury sites in the mouse stomach. However, the chemoreceptor responsible for sensing gastric injury has not yet been identified. In this study, we utilized murine gastric organoids (gastroids) and mutant H. pylori strains to investigate the components necessary for H. pylori chemotaxis. High-intensity 730-nm light (two-photon photodamage) was used to cause single-cell damage in gastroids, and repair of the damage was monitored over time; complete repair occurred within ∼10 min in uninfected gastroids. Wild-type H. pylori accumulated at the damage site after gastric damage induction. In contrast, mutants lacking motility (ΔmotB) or chemotaxis (ΔcheY) did not accumulate at the injury site. Using mutants lacking individual chemoreceptors, we found that only TlpB was required for H. pylori accumulation, while TlpA, TlpC, and TlpD were dispensable. All strains that were able to accumulate at the damage site limited repair. When urea (an identified chemoattractant sensed by TlpB) was microinjected into the gastroid lumen, it prevented the accumulation of H. pylori at damage sites. Overall, our findings demonstrate that H. pylori colonizes and limits repair at damage sites via chemotactic motility that requires the TlpB chemoreceptor to sense signals generated by gastric epithelial cells.
Collapse
|
13
|
Abstract
Maturation of urease involves post-translational insertion of nickel ions to form an active site with a carbamylated lysine ligand and is assisted by urease accessory proteins UreD, UreE, UreF and UreG. Here, we review our current understandings on how these urease accessory proteins facilitate the urease maturation. The urease maturation pathway involves the transfer of Ni2+ from UreE → UreG → UreF/UreD → urease. To avoid the release of the toxic metal to the cytoplasm, Ni2+ is transferred from one urease accessory protein to another through specific protein–protein interactions. One central theme depicts the role of guanosine triphosphate (GTP) binding/hydrolysis in regulating the binding/release of nickel ions and the formation of the protein complexes. The urease and [NiFe]-hydrogenase maturation pathways cross-talk with each other as UreE receives Ni2+ from hydrogenase maturation factor HypA. Finally, the druggability of the urease maturation pathway is reviewed.
Collapse
|
14
|
Marinoni C, Ribaldone DG, Rosso C, Astegiano M, Caviglia GP. Diagnosis of Helicobacter pylori infection: a look into molecular aspects of urea breath test. MINERVA BIOTECNOL 2019. [DOI: 10.23736/s1120-4826.19.02555-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
15
|
Fagoonee S, Pellicano R. Helicobacter pylori: molecular basis for colonization and survival in gastric environment and resistance to antibiotics. A short review. Infect Dis (Lond) 2019; 51:399-408. [PMID: 30907202 DOI: 10.1080/23744235.2019.1588472] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Helicobacter pylori is a human-specific pathogen with a strict tropism for the gastric mucosa. This bacterium infects around half of the world population and is the main responsible for gastritis, peptic ulcer and, in some cases, for the pathogenesis of gastric cancer. Nevertheless, disease development in infected subjects depends not only on the bacterium, but also on the host genetic predisposition and on environmental factors. The fascinating question of how the bacterium can survive in the gastric environment has stimulated research in this field. It is now clear that H. pylori is able to colonize and adhere to the gastric epithelium through several mechanisms, including the breakdown of urea with production of the cell-toxic ammonia. The resulting raise in pH neutralizes acidity of the stomach, thereby allowing the bacterium to safely cross the mucus layer to the epithelial surface. Current challenges regard understanding the mechanisms of antibiotic resistance and how to overcome it. Lately, an increasing H. pylori resistance rate to antibiotics has been reported and several molecular bases for this phenomenon described. In this review, we highlight the current knowledge on mechanisms supporting H. pylori resistance to gastric environment and to therapy.
Collapse
Affiliation(s)
- Sharmila Fagoonee
- a Institute for Biostructure and Bioimaging (CNR) c/o Molecular Biotechnology Center , Turin , Italy
| | - Rinaldo Pellicano
- b Unit of Gastroenterology , Molinette-SGAS Hospital , Turin , Italy
| |
Collapse
|
16
|
Arora R, Issar U, Kakkar R. Identification of novel urease inhibitors: pharmacophore modeling, virtual screening and molecular docking studies. J Biomol Struct Dyn 2018; 37:4312-4326. [PMID: 30580662 DOI: 10.1080/07391102.2018.1546620] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Pharmacophore modeling and atom-based three-dimensional quantitative structure-activity relationship (3D-QSAR) have been developed on N-acylglycino- and hippurohydroxamic acid derivatives, which are known potential inhibitors of urease. This is followed by virtual screening and ADMET (absorption, distribution, metabolism, excretion and toxicity) studies on a large library of known drugs in order to get lead molecules as Helicobacter pylori urease inhibitors. A suitable three-featured pharmacophore model comprising one H-bond acceptor and two H-bond donor features (ADD.10) has been found to be the best QSAR model. An external library of compounds (∼3000 molecules), pre-filtered using Lipinski's rule of five, has been further screened using the pharmacophore model ADD.10. By analyzing the fitness of the hits with respect to the pharmacophore model and their binding interaction inside the urease active site, four molecules have been predicted to be extremely good urease inhibitors. Two of these have significant potential and should be taken up for further drug-designing process.
Collapse
Affiliation(s)
- Richa Arora
- Computational Chemistry Laboratory, Department of Chemistry, University of Delhi , Delhi , India
| | - Upasana Issar
- Computational Chemistry Laboratory, Department of Chemistry, University of Delhi , Delhi , India
| | - Rita Kakkar
- Computational Chemistry Laboratory, Department of Chemistry, University of Delhi , Delhi , India
| |
Collapse
|
17
|
Romeo L, Iori R, Rollin P, Bramanti P, Mazzon E. Isothiocyanates: An Overview of Their Antimicrobial Activity against Human Infections. Molecules 2018. [PMID: 29522501 PMCID: PMC6017699 DOI: 10.3390/molecules23030624] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The use of plant-derived products as antimicrobial agents has been investigated in depth. Isothiocyanates (ITCs) are bioactive products resulting from enzymatic hydrolysis of glucosinolates (GLs), the most abundant secondary metabolites in the botanical order Brassicales. Although the antimicrobial activity of ITCs against foodborne and plant pathogens has been well documented, little is known about their antimicrobial properties against human pathogens. This review collects studies that focus on this topic. Particular focus will be put on ITCs’ antimicrobial properties and their mechanism of action against human pathogens for which the current therapeutic solutions are deficient and therefore of prime importance for public health. Our purpose was the evaluation of the potential use of ITCs to replace or support the common antibiotics. Even though ITCs appear to be effective against the most important human pathogens, including bacteria with resistant phenotypes, the majority of the studies did not show comparable results and thus it is very difficult to compare the antimicrobial activity of the different ITCs. For this reason, a standard method should be used and further studies are needed.
Collapse
Affiliation(s)
- Letizia Romeo
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy.
| | - Renato Iori
- Consiglio per la Ricerca in Agricoltura e L'analisi Dell'economia Agraria, Centro di Ricerca Agricoltura e Ambiente (CREA-AA), Via di Corticella 133, 40128 Bologna, Italy.
| | - Patrick Rollin
- Institute of Organic and Analytical Chemistry (ICOA), Université d'Orléans et the French National Center for Scientific Research (CNRS), UMR 7311, BP 6759, F-45067 Orléans, France.
| | - Placido Bramanti
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy.
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy.
| |
Collapse
|
18
|
Scopel-Guerra A, Olivera-Severo D, Staniscuaski F, Uberti AF, Callai-Silva N, Jaeger N, Porto BN, Carlini CR. The Impact of Helicobacter pylori Urease upon Platelets and Consequent Contributions to Inflammation. Front Microbiol 2017; 8:2447. [PMID: 29312166 PMCID: PMC5733092 DOI: 10.3389/fmicb.2017.02447] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 11/24/2017] [Indexed: 12/22/2022] Open
Abstract
Gastric infection by Helicobacter pylori is considered a risk factor for gastric and duodenal cancer, and extragastric diseases. Previous data have shown that, in a non-enzymatic way, H. pylori urease (HPU) activates neutrophils to produce ROS and also induces platelet aggregation, requiring ADP secretion modulated by the 12-lipoxygenase pathway, a signaling cascade also triggered by the physiological agonist collagen. Here we investigated further the effects on platelets of recombinant versions of the holoenzyme HPU, and of its two subunits (HpUreA and HpUreB). Although HpUreA had no aggregating activity on platelets, it partially inhibited collagen-induced aggregation. HpUreB induced platelet aggregation in the nanomolar range, and also interfered dose-dependently on both collagen- and ADP-induced platelet aggregation. HPU-induced platelet aggregation was inhibited by antibodies against glycoprotein VI (GPVI), the main collagen receptor in platelets. Flow cytometry analysis revealed exposure of P-selectin in HPU-activated platelets. Anti-glycoprotein IIbIIIa (GPIIbIIIa) antibodies increased the binding of FITC-labeled HPU to activated platelets, whereas anti-GPVI did not. Evaluation of post-transcriptional events in HPU-activated platelets revealed modifications in the pre-mRNA processing of pro-inflammatory proteins, with increased levels of mRNAs encoding IL-1β and CD14. We concluded that HPU activates platelets probably through its HpUreB subunit. Activation of platelets by HPU turns these cells into a pro-inflammatory phenotype. Altogether, our data suggest that H. pylori urease, besides allowing bacterial survival within the gastric mucosa, may have an important, and so far overlooked, role in gastric inflammation mediated by urease-activated neutrophils and platelets.
Collapse
Affiliation(s)
- Adriele Scopel-Guerra
- Center of Biotechnology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Deiber Olivera-Severo
- Center of Biotechnology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Department of Biology, Universidade Regional Integrada do Alto Uruguai e das Missões, São Luiz Gonzaga, Brazil
| | - Fernanda Staniscuaski
- Center of Biotechnology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Department of Molecular Biology and Biotechnology, Institute of Biosciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Augusto F Uberti
- Center of Biotechnology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Institute of Biology, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Natália Callai-Silva
- Center of Biotechnology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Natália Jaeger
- Institute of Biomedical Research, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Bárbara N Porto
- Institute of Biomedical Research, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Celia R Carlini
- Brain Institute (BRAINS-InsCer), Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
19
|
Stress Responses, Adaptation, and Virulence of Bacterial Pathogens During Host Gastrointestinal Colonization. Microbiol Spectr 2017; 4. [PMID: 27227312 DOI: 10.1128/microbiolspec.vmbf-0007-2015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Invading pathogens are exposed to a multitude of harmful conditions imposed by the host gastrointestinal tract and immune system. Bacterial defenses against these physical and chemical stresses are pivotal for successful host colonization and pathogenesis. Enteric pathogens, which are encountered due to the ingestion of or contact with contaminated foods or materials, are highly successful at surviving harsh conditions to colonize and cause the onset of host illness and disease. Pathogens such as Campylobacter, Helicobacter, Salmonella, Listeria, and virulent strains of Escherichia have evolved elaborate defense mechanisms to adapt to the diverse range of stresses present along the gastrointestinal tract. Furthermore, these pathogens contain a multitude of defenses to help survive and escape from immune cells such as neutrophils and macrophages. This chapter focuses on characterized bacterial defenses against pH, osmotic, oxidative, and nitrosative stresses with emphasis on both the direct and indirect mechanisms that contribute to the survival of each respective stress response.
Collapse
|
20
|
Abdel-Baky RM, Ali MA, Abuo-Rahma GEDAA, AbdelAziz N. Inhibition of Urease Enzyme Production and some Other Virulence Factors Expression in Proteus mirabilis by N-Acetyl Cysteine and Dipropyl Disulphide. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 973:99-113. [PMID: 28190143 DOI: 10.1007/5584_2016_197] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
UNLABELLED Proteus mirabilis is one of the important pathogens that colonize the urinary tract and catheters resulting in various complications, such as blockage of the catheters and the formation of infective stones. PURPOSE In this study we evaluated the effect of N-acetyl cysteine (NAC) and dipropyl disulphide on some virulence factors expressed by a Proteus mirabilis strain isolated from a catheterized patient. METHODS Antibacterial activity of both compounds was determined by broth microdilution method. Their effect on different types of motility was determined by LB medium with variable agar content and sub-MIC of each drug. Their effect on adherence and mature biofilms was tested by tissue culture plate assay. Inhibitory effect on urease production was determined and supported by molecular docking studies. RESULTS The minimum inhibitory concentration (MIC) of NAC and dipropyl disulphide was 25 mM and 100 mM, respectively. Both compounds decreased the swarming ability and biofilm formation of the tested isolate in a dose-dependent manner. NAC had higher urease inhibitory activity (IC50 249 ±0.05 mM) than that shown by dipropyl disulphide (IC50 10±0.2 mM). Results were supported by molecular docking studies which showed that NAC and dipropyl disulphide interacted with urease enzyme with binding free energy of -4.8 and -8.528 kcal/mol, respectively. Docking studies showed that both compounds interacted with Ni ion and several amino acids (His-138, Gly-279, Cysteine-321, Met-366 and His-322) which are essential for the enzyme activity. CONCLUSION NAC and dipropyl disulphide could be used in the control of P. mirabilis urinary tract infections.
Collapse
Affiliation(s)
- Rehab Mahmoud Abdel-Baky
- Department of Microbiology and Immunology, Faculty of Pharmacy, Minia University, 61519, Minia, Egypt
| | - Mohamed Abdullah Ali
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, 61519, Minia, Egypt
| | | | - Neveen AbdelAziz
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ahram Canadian University, 12581, Sixth of October City, Egypt.
| |
Collapse
|
21
|
Fischer F, Robbe-Saule M, Turlin E, Mancuso F, Michel V, Richaud P, Veyrier FJ, De Reuse H, Vinella D. Characterization in Helicobacter pylori of a Nickel Transporter Essential for Colonization That Was Acquired during Evolution by Gastric Helicobacter Species. PLoS Pathog 2016; 12:e1006018. [PMID: 27923069 PMCID: PMC5140060 DOI: 10.1371/journal.ppat.1006018] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 10/21/2016] [Indexed: 12/23/2022] Open
Abstract
Metal acquisition is crucial for all cells and for the virulence of many bacterial pathogens. In particular, nickel is a virulence determinant for the human gastric pathogen Helicobacter pylori as it is the cofactor of two enzymes essential for in vivo colonization, urease and a [NiFe] hydrogenase. To import nickel despite its scarcity in the human body, H. pylori requires efficient uptake mechanisms that are only partially defined. Indeed, alternative ways of nickel entry were predicted to exist in addition to the well-described NixA permease. Using a genetic screen, we identified an ABC transporter, that we designated NiuBDE, as a novel H. pylori nickel transport system. Unmarked mutants carrying deletions of nixA, niuD and/or niuB, were constructed and used to measure (i) tolerance to toxic nickel exposure, (ii) intracellular nickel content by ICP-OES, (iii) transport of radioactive nickel and (iv) expression of a reporter gene controlled by nickel concentration. We demonstrated that NiuBDE and NixA function separately and are the sole nickel transporters in H. pylori. NiuBDE, but not NixA, also transports cobalt and bismuth, a metal currently used in H. pylori eradication therapy. Both NiuBDE and NixA participate in nickel-dependent urease activation at pH 5 and survival under acidic conditions mimicking those encountered in the stomach. However, only NiuBDE is able to carry out this activity at neutral pH and is essential for colonization of the mouse stomach. Phylogenomic analyses indicated that both nixA and niuBDE genes have been acquired via horizontal gene transfer by the last common ancestor of the gastric Helicobacter species. Our work highlights the importance of this evolutionary event for the emergence of Helicobacter gastric species that are adapted to the hostile environment of the stomach where the capacity of Helicobacter to import nickel and thereby activate urease needs to be optimized.
Collapse
Affiliation(s)
- Frédéric Fischer
- Institut Pasteur, Département de Microbiologie, Unité Pathogenèse de Helicobacter, ERL CNRS 3526, Paris, FRANCE
| | - Marie Robbe-Saule
- Institut Pasteur, Département de Microbiologie, Unité Pathogenèse de Helicobacter, ERL CNRS 3526, Paris, FRANCE
| | - Evelyne Turlin
- Institut Pasteur, Département de Microbiologie, Unité Pathogenèse de Helicobacter, ERL CNRS 3526, Paris, FRANCE
| | - Francesco Mancuso
- Institut Pasteur, Département de Microbiologie, Unité Pathogenèse de Helicobacter, ERL CNRS 3526, Paris, FRANCE
| | - Valérie Michel
- Institut Pasteur, Département de Microbiologie, Unité Pathogenèse de Helicobacter, ERL CNRS 3526, Paris, FRANCE
| | - Pierre Richaud
- CEA, DRF, BIAM SBVME and CNRS, UMR 7265, Saint-Paul-lez-Durance, Aix Marseille Université, Marseille, FRANCE
| | - Frédéric J. Veyrier
- INRS-Institut Armand-Frappier, Bacterial Symbionts Evolution, Laval, Quebec, CANADA
| | - Hilde De Reuse
- Institut Pasteur, Département de Microbiologie, Unité Pathogenèse de Helicobacter, ERL CNRS 3526, Paris, FRANCE
- * E-mail:
| | - Daniel Vinella
- Institut Pasteur, Département de Microbiologie, Unité Pathogenèse de Helicobacter, ERL CNRS 3526, Paris, FRANCE
| |
Collapse
|
22
|
Darko R, Yawson AE, Osei V, Owusu-Ansah J, Aluze-Ele S. Changing Patterns of the Prevalence of Helicobacter Pylori Among Patients at a Corporate Hospital in Ghana. Ghana Med J 2016; 49:147-53. [PMID: 26693189 DOI: 10.4314/gmj.v49i3.4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Helicobacter pylori (H. pylori) discovered in 1982, has strongly been associated with multiple clinical disorders of the gastrointestinal tract. This study described the prevalence of H. pylori among large numbers of patients over two different time periods in Accra, Ghana. METHODS It was a retrospective records review on patients attending a quasi-government hospital in Accra, Ghana, during two time periods, 1999 and 2012. A total of 2401 records were reviewed, 1128 in first period and 1273 in second period. Biopsy was taken from the gastric antrum for Rapid Urease Test (RUT) in identifying H. Pylori. Data on patient characteristics, clinical diagnosis and findings upon endoscopy were analyzed by simple descriptive statistics. Associations between categorical outcome variables were determined by Chi square test at 95% significance level. RESULTS H. pylori infection was high in patients with upper gastrointestinal symptoms 69.7% (1999) and 45.2% (2012), and was even higher in patients with gastritis and duodenal ulcer. H. pylori infection however, decreased among patients over the period, 69.7% in 1999 to 45.2% in 2012. Sex differences in H. pylori infection was identified (higher among males) and young adults (21-40 years). Commonest symptom in all patients was non-ulcer dyspepsia, 86.9% in 1999 and 84.2% in 2012, while gastritis and duodenal ulcer were the commonest endoscopic finding in the two periods. CONCLUSION Appropriate management guidelines in West Africa considering the high background H. pylori infection and other co-infections requiring particular antibiotic combination therapy is required.
Collapse
Affiliation(s)
- R Darko
- Department of Surgery, University of Ghana School of Medicine and Dentistry, College of Health Sciences, Korle-Bu, Accra, Ghana
| | - A E Yawson
- Department of Community Health, School of Public Health, College of Health Sciences. University of Ghana, Accra, Ghana
| | - V Osei
- Cocoa Clinic, Accra, Ghana
| | | | - S Aluze-Ele
- Department of Community Health, School of Public Health, College of Health Sciences. University of Ghana, Accra, Ghana
| |
Collapse
|
23
|
Jones MD, Ademi I, Yin X, Gong Y, Zamble DB. Nickel-responsive regulation of two novel Helicobacter pylori NikR-targeted genes. Metallomics 2016; 7:662-73. [PMID: 25521693 DOI: 10.1039/c4mt00210e] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Nickel is an essential transition metal for the survival of Helicobacter pylori in the acidic human stomach. The nickel-responsive transcriptional regulator HpNikR is important for maintaining healthy cytosolic nickel concentrations through the regulation of multiple genes, but its complete regulon and role in nickel homeostasis are not well understood. To investigate potential gene targets of HpNikR, ChIP sequencing was performed using H. pylori grown at neutral pH in nickel-supplemented media and this experiment identified HPG27_866 (frpB2) and HPG27_1499 (ceuE). These two genes are annotated to encode a putative iron transporter and a nickel-binding, periplasmic component of an ABC transporter, respectively. In vitro DNA-binding assays revealed that HpNikR binds both gene promoter sequences in a nickel-responsive manner with affinities on the order of ∼10(-7) M. The recognition sites of HpNikR were identified and loosely correlate with the HpNikR pseudo-consensus sequence (TATTATT-N11-AATAATA). Quantitative PCR experiments revealed that HPG27_866 and HPG27_1499 are transcriptionally repressed following growth of H. pylori G27 in nickel-supplemented media, and that this response is dependent on HpNikR. In contrast, iron supplementation results in activation of HPG27_1499, but no impact on the expression of HPG27_866 was observed. Metal analysis of the Δ866 strain revealed that HPG27_866 has an impact on nickel accumulation. These studies demonstrate that HPG27_866 and HPG27_1499 are both direct targets of HpNikR and that HPG27_866 influences nickel uptake in H. pylori.
Collapse
Affiliation(s)
- M D Jones
- Department of Chemistry, University of Toronto, 80 St. George St., Toronto, ON, M5S 3H6, Canada.
| | | | | | | | | |
Collapse
|
24
|
Hong S, Kim O. Evaluation of assays to detect Helicobacter felis infection in cats. J Biomed Res 2015. [DOI: 10.12729/jbr.2015.16.4.159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
25
|
Srivastava AK, Kumar V, Roy BK. Insights from the molecular docking of curcumin to the virulent factors of Helicobacter pylori. Bioinformation 2015; 11:447-53. [PMID: 26664028 PMCID: PMC4658642 DOI: 10.6026/97320630011447] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 10/01/2015] [Indexed: 12/11/2022] Open
Abstract
The domains of virulent (Ureα/β, VacA-p55, and CagA) factors of Helicobacter pylori play a pivotal role in developmental processes of numerous diseases including gastric cancer. The pharmacological role of curcumin indicates that it could regulate the signaling of virulent factors by interacting with active domains. However, the controlling mechanism of the curcumin interactions and the binding diversity on structural basis of virulent (Ureα/β, VacA-p55, and CagA) factors are unknown. Curcumin as therapeutic agent was filtered by using Lipinski rule׳s five and the druglikeness property for assessment of pharmacological properties. Here outcome of molecular docking presented the 3-D structure of curcumin complex, that interacted with especially conserved residues of target domains. The structure revealed that the curcumin complexation with domains of these proteins provided structural insight into the diverse nature of proteins (Ureα/β, VacA-p55, and CagA) recognition. In silico study elucidated that the broad specificity of curcumin was achieved by multiple binding mode mechanisms such as distinct hydrogen and hydrophobic interactions with involvement of binding energy. The higher score of curcumin in complexation with both subunits Ureα/β showed the stable binding, and less stability with VacA-p55 complexation with lower score. Curcumin exhibited good interaction with these targeted virulent factors, although extensive interactions of curcumin with Ureα/β subunits could have an important implication to prevent survival and colonisation of H. pylori in stomach.
Collapse
Affiliation(s)
| | - Vikas Kumar
- Department of Botany, Banaras Hindu University, Varanasi, 221005, India
| | - Bijoy Krishna Roy
- Department of Botany, Banaras Hindu University, Varanasi, 221005, India
| |
Collapse
|
26
|
Lv X, Song H, Yang J, Li T, Xi T, Xing Y. A multi-epitope vaccine CTB-UE relieves Helicobacter pylori-induced gastric inflammatory reaction via up-regulating microRNA-155 to inhibit Th17 response in C57/BL6 mice model. Hum Vaccin Immunother 2015; 10:3561-9. [PMID: 25483699 DOI: 10.4161/hv.36096] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Vaccination is an effective mean of preventing infectious diseases, including those caused by Helicobacter pylori. Th17 cell responses are critical for the pathogenesis of Helicobacter pylori infection. In view of Th17 responses to multi-epitope vaccine CTB-UE, the IL-17 production in antiserum was examined. CTB-UE immunization decreased IL-17 production, implying that Th17 responses may be inhibited. Furthermore, IL-17 aggravated GES-1 cell injury induced by H. pylori SS1; In contrast, CTB-UE antiserum could alleviate this cell injury, which suggesting that CTB-UE can protect GES-1 cell infected with H. pylori SS1 by inhibiting Th17 responses. Treatment of mice with CTB-UE significantly reduced the H. pylori burden and inflammation in the stomach. On the other hand, the production of IL-17 in the stomach in H. pylori-infected mice was increased; but the production of IL-17 in the stomach was decreased after treatment with CTB-UE. Furthermore, the expression of microRNA-155 in gastric tissue was significantly up-regulated. The results suggested that CTB-UE could relieve the H. pylori-induced gastric inflammatory reaction via up-regulating microRNA-155 to inhibit Th17 responses, implying that the microRNA-155/IL-17 pathway was involved. Further study is required to elucidate the relationship between miRNA-155 and IL-17. We found that the production of IL-17 was significantly increased after the expression of miRNA-155 being down-regulated; however, the production of IL-17 was significantly decreased after the expression of miRNA-155 being upregulated.
Collapse
Key Words
- ATCC, American Type Culture Collection
- CTB, Cholera toxin B subunit
- E. coli, Escherichia coli
- ELISA, Enzyme-linkedimmunosorbent assay
- Epitope vaccine
- H. pylori, Helicobacter pylori
- HD, High-dose group
- Helicobacter pylori
- IL-17
- IL-17, Interleukin-17
- LD, Low-dose group
- Lipo-2000, Lipofectamine 2000
- MC, Model control group
- MD, Middle-dose group
- NC, Normal control group
- OD, Optical density
- PAGE, Polyacrylamide gel electrophoresis
- PCR, Polymerase chain reaction
- RT-PCR, Reverse transcription polymerase chain reaction
- SDS, Sodium dodecyl sulfate
- Th, Helper T lymphocyte
- Th1, Type 1 of helper T lymphocyte
- Th17
- Th17, Type 17 of helper T lymphocyte
- Th2, Type 2 of helper T lymphocytel
- UreA, Urease A subunit
- UreB, Urease B subunit
- cDNA, Complementary DNA
- dNTP, Deoxyribonucleoside triphosphate
- ddH2O, Double distilled water
- miR-155, microRNA-155
- miRNA, microRNA
- microRNA-155
Collapse
Affiliation(s)
- Xiaobo Lv
- a Biotechnology Center; School of Life Science and Technology ; China Pharmaceutical University , Nanjing , China
| | | | | | | | | | | |
Collapse
|
27
|
Farnham KR, Dube DH. A semester-long project-oriented biochemistry laboratory based on Helicobacter pylori urease. BIOCHEMISTRY AND MOLECULAR BIOLOGY EDUCATION : A BIMONTHLY PUBLICATION OF THE INTERNATIONAL UNION OF BIOCHEMISTRY AND MOLECULAR BIOLOGY 2015; 43:333-40. [PMID: 26173574 PMCID: PMC4573817 DOI: 10.1002/bmb.20884] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/09/2015] [Accepted: 05/25/2015] [Indexed: 05/20/2023]
Abstract
Here we present the development of a 13 week project-oriented biochemistry laboratory designed to introduce students to foundational biochemical techniques and then enable students to perform original research projects once they have mastered these techniques. In particular, we describe a semester-long laboratory that focuses on a biomedically relevant enzyme--Helicobacter pylori (Hp) urease--the activity of which is absolutely required for the gastric pathogen Hp to colonize the human stomach. Over the course of the semester, students undertake a biochemical purification of Hp urease, assess the success of their purification, and investigate the activity of their purified enzyme. In the final weeks of the semester, students design and implement their own experiments to study Hp urease. This laboratory provides students with an understanding of the importance of biochemistry in human health while empowering them to engage in an active area of research.
Collapse
Affiliation(s)
- Kate R. Farnham
- Department of Chemistry and Biochemistry, Bowdoin College, 6600 College Station, Brunswick, ME 04011, United States
| | - Danielle H. Dube
- Department of Chemistry and Biochemistry, Bowdoin College, 6600 College Station, Brunswick, ME 04011, United States
- To whom all correspondence about the manuscript should be sent: Danielle H. Dube, Ph.D., Department of Chemistry and Biochemistry, Bowdoin College, Brunswick, ME 04011, TEL 207-798-4326, FAX 207-725-3017,
| |
Collapse
|
28
|
Cáceres-Delpiano J, Teneb J, Mansilla R, García A, Salas-Burgos A. Variations in periplasmic loop interactions determine the pH-dependent activity of the hexameric urea transporter UreI from Helicobacter pylori: a molecular dynamics study. BMC STRUCTURAL BIOLOGY 2015; 15:11. [PMID: 26112768 PMCID: PMC4482100 DOI: 10.1186/s12900-015-0038-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 06/15/2015] [Indexed: 11/17/2022]
Abstract
Background Helicobacter pylori is an important factor in the development of diseases such as ulcer and gastric cancer. This bacterium uses a periplasmic transporter, UreI, to deliver urea to the intracelullar space, where later it is transformed into ammonia by the cytoplasmic enzyme urease to survive the acidic condition of the human stomach. The UreI transporter presents a pH-dependent activity, where this pH-dependence remains unknown at a structural level. Althought the existance of several protonable residues in the periplasmic loops are related to the pH-dependent activity, we find interesting to have a clear view of the conformational changes involved in this phenomena through a molecular dynamic study. Results Molecular dynamic simulations of the UreI transporter at three different pH conditions were performed, revealing two main pH-dependent conformations, which we present as the open and close states. We find that salt bridges between the periplasmic loops are crucial interactions that stabilize these conformations. Besides, a cooperative behaviour exists between the six subunits of the system that is necessary to fulfill the activity of this transporter. Conclusions We found different pH-dependent conformations of the urea transporter UreI from Helicobacter pylori, which are related to salt-bridge interactions in the periplasmic regions. The behaviour of every channel in the system is not independent, given the existance of a cooperative behaviour through the formation of salt-bridges between the subunits of the hexameric system. We believe that our results will be related to the generation of new eradication therapies using this transporter as an attractive target, denoting that the knowledge of the possible pH-dependent conformations adopted for this transporter are important for the development of rational drug design approximations. Electronic supplementary material The online version of this article (doi:10.1186/s12900-015-0038-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Javier Cáceres-Delpiano
- Department of Pharmacology, School of Sciences, University of Concepción, Concepción, Chile.
| | - Jaime Teneb
- Department of Pharmacology, School of Sciences, University of Concepción, Concepción, Chile.
| | - Rodrigo Mansilla
- Department of Pharmacology, School of Sciences, University of Concepción, Concepción, Chile.
| | - Apolinaria García
- Department of Microbiology, School of Sciences, University of Concepción, Concepción, Chile.
| | - Alexis Salas-Burgos
- Department of Pharmacology, School of Sciences, University of Concepción, Concepción, Chile.
| |
Collapse
|
29
|
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
|
30
|
Helicobacter pylori: the balance between a role as colonizer and pathogen. Best Pract Res Clin Gastroenterol 2014; 28:1017-29. [PMID: 25439068 DOI: 10.1016/j.bpg.2014.09.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 08/25/2014] [Accepted: 09/15/2014] [Indexed: 02/07/2023]
Abstract
The isolation of Helicobacter pylori from the human stomach produced significant changes in how gastroenterologists, immunologists, epidemiologists, pathologists and microbiologists have approached gastro-duodenal diseases in the last half of the XX century. However, research of this organism has progressed greatly in the first decade of this century, evidence suggest that H. pylori is associated with disease only in humans older than 40 years, while, the lack of H. pylori colonization is associated with the emergence of new diseases, particularly in younger individuals. These differing effects of H. pylori colonization have created two contrasting concepts: the 'bad' and the 'good' Helicobacter. Following from renewed interest in the normal human microbiome, we need to reconsider our definitions and perhaps recognize that H. pylori might be a normal member of the human gastric microbiome in ancient humans that gradually, as results of the improvement in our environment, is disappearing.
Collapse
|
31
|
Yu XD, Xie JH, Wang YH, Li YC, Mo ZZ, Zheng YF, Su JY, Liang YE, Liang JZ, Su ZR, Huang P. Selective Antibacterial Activity of Patchouli Alcohol Against Helicobacter pylori
Based on Inhibition of Urease. Phytother Res 2014; 29:67-72. [DOI: 10.1002/ptr.5227] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 08/22/2014] [Accepted: 08/26/2014] [Indexed: 11/10/2022]
Affiliation(s)
- Xiao-Dan Yu
- College of Chinese Medicine; Guangzhou University of Chinese Medicine; Guangzhou 510006 PR China
| | - Jian-Hui Xie
- College of Chinese Medicine; Guangzhou University of Chinese Medicine; Guangzhou 510006 PR China
- The Second Affiliated Hospital of Chinese Medicine; Guangzhou University of Chinese Medicine; Guangzhou 510115 PR China
| | - Yong-Hong Wang
- Guangdong Institute of Microbiology; Guangzhou 510006 PR China
| | - Yu-Cui Li
- College of Chinese Medicine; Guangzhou University of Chinese Medicine; Guangzhou 510006 PR China
| | - Zhi-Zhun Mo
- College of Chinese Medicine; Guangzhou University of Chinese Medicine; Guangzhou 510006 PR China
| | - Yi-Feng Zheng
- College of Chinese Medicine; Guangzhou University of Chinese Medicine; Guangzhou 510006 PR China
| | - Ji-Yan Su
- College of Chinese Medicine; Guangzhou University of Chinese Medicine; Guangzhou 510006 PR China
| | - Ye-er Liang
- College of Chinese Medicine; Guangzhou University of Chinese Medicine; Guangzhou 510006 PR China
| | - Jin-Zhi Liang
- College of Chinese Medicine; Guangzhou University of Chinese Medicine; Guangzhou 510006 PR China
| | - Zi-Ren Su
- College of Chinese Medicine; Guangzhou University of Chinese Medicine; Guangzhou 510006 PR China
- Dongguan Mathematical Engineering Academy of Chinese Medicine; Guangzhou University of Chinese Medicine; Dongguan 523000 PR China
| | - Ping Huang
- College of Chinese Medicine; Guangzhou University of Chinese Medicine; Guangzhou 510006 PR China
| |
Collapse
|
32
|
Therapeutic efficacy of the multi-epitope vaccine CTB-UE against Helicobacter pylori infection in a Mongolian gerbil model and its microRNA-155-associated immuno-protective mechanism. Vaccine 2014; 32:5343-52. [PMID: 25093281 DOI: 10.1016/j.vaccine.2014.07.041] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 06/11/2014] [Accepted: 07/09/2014] [Indexed: 12/24/2022]
Abstract
Vaccination is an effective means of preventing infectious diseases, including those caused by Helicobacter pylori. In this study, we constructed a novel multi-epitope vaccine, CTB-UE, composed of the cholera toxin B subunit and tandem copies of the B and Th cell epitopes from the H. pylori urease A and B subunits. We evaluated the therapeutic efficacy of the multi-epitope vaccine CTB-UE against H. pylori infection in a Mongolian gerbil model and studied its immuno-protective mechanisms. The experimental results indicated that urease activity, H. pylori colonisation density, the levels of IL-8 and TNF-α in the serum, and the levels of COX-2 and NAP in gastric tissue were significantly lower and the IgG level in the serum and the IFN-γ level in spleen lymphocytes were significantly higher in the vaccinated group compared with the model control group; additionally, gastric mucosal inflammation was notably alleviated following vaccination. The results showed that CTB-UE had a good therapeutic effect on H. pylori infection. The immuno-protective mechanism was closely related to the immune response mediated by microRNA-155, the expression of which was strongly up-regulated after CTB-UE administration. The expression levels of the microRNA-155 target proteins IFN-γRα, AID, and PU.1 were significantly down-regulated; these results indicated that CTB-UE induced an immune response biased towards Th1 cells by up-regulating microRNA-155 to inhibit IFN-γRα expression and induced a humoral immune response towards B cells by up-regulating microRNA-155 to inhibit PU.1 and AID expression. These results demonstrate that the multi-epitope vaccine CTB-UE may be a promising therapeutic vaccine against H. pylori infection and is a new therapeutic tool for human use.
Collapse
|
33
|
The pH-dependent expression of the urease operon in Streptococcus salivarius is mediated by CodY. Appl Environ Microbiol 2014; 80:5386-93. [PMID: 24951785 DOI: 10.1128/aem.00755-14] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Urease gene expression in Streptococcus salivarius 57.I, a strain of one of the major alkali producers in the mouth, is induced by acidic pH and excess amounts of carbohydrate. Expression is controlled primarily at the transcriptional level from a promoter, pureI. Recent sequencing analysis revealed a CodY box located 2 bases 5' to the -35 element of pureI. Using continuous chemostat culture, transcription from pureI was shown to be repressed by CodY, and at pH 7 the repression was more pronounced than that in cells grown at pH 5.5 under both 20 and 100 mM glucose. The direct binding of CodY to pureI was demonstrated by electrophoretic mobility shift assay and chromatin immunoprecipitation (ChIP)-quantitative real-time PCR (qPCR). The result of ChIP-qPCR also confirmed that the regulation of CodY is indeed modulated by pH and the binding of CodY at neutral pH is further enhanced by a limited supply of glucose (20 mM). In the absence of CodY, the C-terminal domain of the RNA polymerase (RNAP) α subunit interacted with the AT tracks within the CodY box, indicating that CodY and RNAP compete for the same binding region. Such regulation could ensure optimal urease expression when the enzyme is most required, i.e., at an acidic growth pH with an excess amount of carbon nutrients.
Collapse
|
34
|
Abstract
The gastric pathogen Helicobacter pylori possesses a highly active urease to support acid tolerance. Urea hydrolysis occurs inside the cytoplasm, resulting in the production of NH3 that is immediately protonated to form NH4 (+). This ammonium must be metabolized or effluxed because its presence within the cell is counterproductive to the goal of raising pH while maintaining a viable proton motive force (PMF). Two compatible hypotheses for mitigating intracellular ammonium toxicity include (i) the exit of protonated ammonium outward via the UreI permease, which was shown to facilitate diffusion of both urea and ammonium, and/or (ii) the assimilation of this ammonium, which is supported by evidence that H. pylori assimilates urea nitrogen into its amino acid pools. We investigated the second hypothesis by constructing strains with altered expression of the ammonium-assimilating enzymes glutamine synthetase (GS) and glutamate dehydrogenase (GDH) and the ammonium-evolving periplasmic enzymes glutaminase (Ggt) and asparaginase (AsnB). H. pylori strains expressing elevated levels of either GS or GDH are more acid tolerant than the wild type, exhibit enhanced ammonium production, and are able to alkalize the medium faster than the wild type. Strains lacking the genes for either Ggt or AsnB are acid sensitive, have 8-fold-lower urea-dependent ammonium production, and are more acid sensitive than the parent. Additionally, we found that purified H. pylori GS produces glutamine in the presence of Mg(2+) at a rate similar to that of unadenylated Escherichia coli GS. These data reveal that all four enzymes contribute to whole-cell acid resistance in H. pylori and are likely important for assimilation and/or efflux of urea-derived ammonium.
Collapse
|
35
|
Minkara MS, Ucisik MN, Weaver MN, Merz KM. Molecular Dynamics Study of Helicobacter pylori Urease. J Chem Theory Comput 2014; 10:1852-1862. [PMID: 24839409 PMCID: PMC4020587 DOI: 10.1021/ct5000023] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Indexed: 12/21/2022]
Abstract
Helicobacter pylori have been implicated in an array of gastrointestinal disorders including, but not limited to, gastric and duodenal ulcers and adenocarcinoma. This bacterium utilizes an enzyme, urease, to produce copious amounts of ammonia through urea hydrolysis in order to survive the harsh acidic conditions of the stomach. Molecular dynamics (MD) studies on the H. pylori urease enzyme have been employed in order to study structural features of this enzyme that may shed light on the hydrolysis mechanism. A total of 400 ns of MD simulation time were collected and analyzed in this study. A wide-open flap state previously observed in MD simulations on Klebsiella aerogenes [Roberts et al. J. Am. Chem. Soc.2012, 134, 9934] urease has been identified in the H. pylori enzyme that has yet to be experimentally observed. Critical distances between residues on the flap, contact points in the closed state, and the separation between the active site Ni2+ ions and the critical histidine α322 residue were used to characterize flap motion. An additional flap in the active site was elaborated upon that we postulate may serve as an exit conduit for hydrolysis products. Finally we discuss the internal hollow cavity and present analysis of the distribution of sodium ions over the course of the simulation.
Collapse
Affiliation(s)
- Mona S Minkara
- Department of Chemistry, Quantum Theory Project, 2328 New Physics Building, University of Florida , Gainesville, Florida 32611-8435, United States
| | - Melek N Ucisik
- Department of Chemistry, Quantum Theory Project, 2328 New Physics Building, University of Florida , Gainesville, Florida 32611-8435, United States
| | - Michael N Weaver
- Department of Chemistry, Quantum Theory Project, 2328 New Physics Building, University of Florida , Gainesville, Florida 32611-8435, United States
| | - Kenneth M Merz
- Department of Chemistry, Department of Biochemistry and Molecular Biology, Michigan State University , 578 S. Shaw Lane, East Lansing, Michigan 48824-1322, United States
| |
Collapse
|
36
|
Champasa K, Longwell SA, Eldridge AM, Stemmler EA, Dube DH. Targeted identification of glycosylated proteins in the gastric pathogen Helicobacter pylori (Hp). Mol Cell Proteomics 2013; 12:2568-86. [PMID: 23754784 PMCID: PMC3769331 DOI: 10.1074/mcp.m113.029561] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Virulence of the gastric pathogen Helicobacter pylori (Hp) is directly linked to the pathogen's ability to glycosylate proteins; for example, Hp flagellin proteins are heavily glycosylated with the unusual nine-carbon sugar pseudaminic acid, and this modification is absolutely essential for Hp to synthesize functional flagella and colonize the host's stomach. Although Hp's glycans are linked to pathogenesis, Hp's glycome remains poorly understood; only the two flagellin glycoproteins have been firmly characterized in Hp. Evidence from our laboratory suggests that Hp synthesizes a large number of as-yet unidentified glycoproteins. Here we set out to discover Hp's glycoproteins by coupling glycan metabolic labeling with mass spectrometry analysis. An assessment of the subcellular distribution of azide-labeled proteins by Western blot analysis indicated that glycoproteins are present throughout Hp and may therefore serve diverse functions. To identify these species, Hp's azide-labeled glycoproteins were tagged via Staudinger ligation, enriched by tandem affinity chromatography, and analyzed by multidimensional protein identification technology. Direct comparison of enriched azide-labeled glycoproteins with a mock-enriched control by both SDS-PAGE and mass spectrometry-based analyses confirmed the selective enrichment of azide-labeled glycoproteins. We identified 125 candidate glycoproteins with diverse biological functions, including those linked with pathogenesis. Mass spectrometry analyses of enriched azide-labeled glycoproteins before and after cleavage of O-linked glycans revealed the presence of Staudinger ligation-glycan adducts in samples only after beta-elimination, confirming the synthesis of O-linked glycoproteins in Hp. Finally, the secreted colonization factors urease alpha and urease beta were biochemically validated as glycosylated proteins via Western blot analysis as well as by mass spectrometry analysis of cleaved glycan products. These data set the stage for the development of glycosylation-based therapeutic strategies, such as new vaccines based on natively glycosylated Hp proteins, to eradicate Hp infection. Broadly, this report validates metabolic labeling as an effective and efficient approach for the identification of bacterial glycoproteins.
Collapse
Affiliation(s)
- Kanokwan Champasa
- Department of Chemistry and Biochemistry, Bowdoin College, 6600 College Station, Brunswick, Maine 04011, USA
| | | | | | | | | |
Collapse
|
37
|
Fahey JW, Stephenson KK, Wade KL, Talalay P. Urease from Helicobacter pylori is inactivated by sulforaphane and other isothiocyanates. Biochem Biophys Res Commun 2013; 435:1-7. [PMID: 23583386 DOI: 10.1016/j.bbrc.2013.03.126] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 03/27/2013] [Indexed: 02/07/2023]
Abstract
Infections by Helicobacter pylori are very common, causing gastroduodenal inflammation including peptic ulcers, and increasing the risk of gastric neoplasia. The isothiocyanate (ITC) sulforaphane [SF; 1-isothiocyanato-4-(methylsulfinyl)butane] derived from edible crucifers such as broccoli is potently bactericidal against Helicobacter, including antibiotic-resistant strains, suggesting a possible dietary therapy. Gastric H. pylori infections express high urease activity which generates ammonia, neutralizes gastric acidity, and promotes inflammation. The finding that SF inhibits (inactivates) urease (jack bean and Helicobacter) raised the issue of whether these properties might be functionally related. The rates of inactivation of urease activity depend on enzyme and SF concentrations and show first order kinetics. Treatment with SF results in time-dependent increases in the ultraviolet absorption of partially purified Helicobacter urease in the 260-320 nm region. This provides direct spectroscopic evidence for the formation of dithiocarbamates between the ITC group of SF and cysteine thiols of urease. The potencies of inactivation of Helicobacter urease by isothiocyanates structurally related to SF were surprisingly variable. Natural isothiocyanates closely related to SF, previously shown to be bactericidal (berteroin, hirsutin, phenethyl isothiocyanate, alyssin, and erucin), did not inactivate urease activity. Furthermore, SF is bactericidal against both urease positive and negative H. pylori strains. In contrast, some isothiocyanates such as benzoyl-ITC, are very potent urease inactivators, but are not bactericidal. The bactericidal effects of SF and other ITC against Helicobacter are therefore not obligatorily linked to urease inactivation, but may reduce the inflammatory component of Helicobacter infections.
Collapse
Affiliation(s)
- Jed W Fahey
- Lewis B. and Dorothy Cullman Chemoprotection Center, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA.
| | | | | | | |
Collapse
|
38
|
Moon DI, Shin EH, Oh HG, Oh JS, Hong S, Chung Y, Kim O. Usefulness of a Helicobacter pylori stool antigen test for diagnosing H. pylori infected C57BL/6 mice. Lab Anim Res 2013; 29:27-32. [PMID: 23573105 PMCID: PMC3616206 DOI: 10.5625/lar.2013.29.1.27] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 03/01/2013] [Accepted: 03/02/2013] [Indexed: 02/07/2023] Open
Abstract
Among several diagnostic tests, a Helicobacter pylori stool antigen (HpSA) test may offer a useful noninvasive method for diagnosing infection without sacrificing animals. In this study, male C57BL/6 mice (n=6) were infected with H. pylori ATCC 49503 (1×10(8) CFU/mouse) by intragastric inoculation three times at 2-day intervals, and H. pylori infected stool specimens were collected 1, 3, 5, 7, 14, 21 days after infection to assess reliability of the HpSA test. Five of six specimens were positive at 5-21 days after infection, and the sensitivity of the HpSA test was 83.33%. The presence of H. pylori infection was confirmed by the rapid urease test and genomic DNA polymerase chain reaction (PCR), and showed the same results as the HpSA. However, the rapid urease test and genomic DNA PCR are invasive tests and require animal sacrifice to detect H. pylori in gastric biopsy samples. We suggest that an HpSA test kit would be useful and effective for monitoring H. pylori in various laboratory animals, as H. pylori can be easily monitored without sacrificing animals.
Collapse
Affiliation(s)
- Dae-In Moon
- Center for Animal Resources Development, Wonkwang University, Iksan, Korea
- Huvet. Inc., Iksan, Korea
| | | | | | | | - Sunhwa Hong
- Center for Animal Resources Development, Wonkwang University, Iksan, Korea
| | - Yungho Chung
- Department of Companion Animal and Animal Recourses Science, Joongbu University, Kummsan, Korea
| | - Okjin Kim
- Center for Animal Resources Development, Wonkwang University, Iksan, Korea
| |
Collapse
|
39
|
Uberti AF, Olivera-Severo D, Wassermann GE, Scopel-Guerra A, Moraes JA, Barcellos-de-Souza P, Barja-Fidalgo C, Carlini CR. Pro-inflammatory properties and neutrophil activation by Helicobacter pylori urease. Toxicon 2013; 69:240-9. [PMID: 23466444 DOI: 10.1016/j.toxicon.2013.02.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 12/25/2012] [Accepted: 02/05/2013] [Indexed: 01/25/2023]
Abstract
The gastric pathogen Helicobacter pylori produces large amounts of urease, whose enzyme activity enables the bacterium to survive in the stomach. We have previously shown that ureases display enzyme-independent effects in mammalian models, most through lipoxygenases-mediated pathways. Here, we evaluated potential pro-inflammatory properties of H. pylori urease (HPU). Mouse paw edema and activation of human neutrophils were tested using a purified, cell-free, recombinant HPU. rHPU induced paw edema with intense neutrophil infiltration. In vitro 100 nM rHPU was chemotactic to human neutrophils, inducing production of reactive oxygen species. rHPU-activated neutrophils showed increased lifespan, with inhibition of apoptosis accompanied by alterations of Bcl-XL and Bad contents. These effects of rHPU persisted in the absence of enzyme activity. rHPU-induced paw edema, neutrophil chemotaxis and apoptosis inhibition reverted in the presence of the lipoxygenase inhibitors esculetin or AA861. Neutrophils exposed to rHPU showed increased content of lipoxygenase(s) and no alteration of cyclooxygenase(s). Altogether, our data indicate that HPU, besides allowing the bacterial survival in the stomach, could play an important role in the pathogenesis of the gastrointestinal inflammatory disease caused by H. pylori.
Collapse
Affiliation(s)
- Augusto F Uberti
- Graduate Program in Cellular and Molecular Biology, Center of Biotechnology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Kuhns LG, Mahawar M, Sharp JS, Benoit S, Maier RJ. Role of Helicobacter pylori methionine sulfoxide reductase in urease maturation. Biochem J 2013; 450:141-8. [PMID: 23181726 PMCID: PMC3935233 DOI: 10.1042/bj20121434] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The persistence of the gastric pathogen Helicobacter pylori is due in part to urease and Msr (methionine sulfoxide reductase). Upon exposure to relatively mild (21% partial pressure of O2) oxidative stress, a Δmsr mutant showed both decreased urease specific activity in cell-free extracts and decreased nickel associated with the partially purified urease fraction as compared with the parent strain, yet urease apoprotein levels were the same for the Δmsr and wild-type extracts. Urease activity of the Δmsr mutant was not significantly different from the wild-type upon non-stress microaerobic incubation of strains. Urease maturation occurs through nickel mobilization via a suite of known accessory proteins, one being the GTPase UreG. Treatment of UreG with H2O2 resulted in oxidation of MS-identified methionine residues and loss of up to 70% of its GTPase activity. Incubation of pure H2O2-treated UreG with Msr led to reductive repair of nine methionine residues and recovery of up to full enzyme activity. Binding of Msr to both oxidized and non-oxidized UreG was observed by cross-linking. Therefore we conclude Msr aids the survival of H. pylori in part by ensuring continual UreG-mediated urease maturation under stress conditions.
Collapse
Affiliation(s)
- Lisa G. Kuhns
- Department of Microbiology, University of Georgia, Athens, GA 30602, U.S.A
| | - Manish Mahawar
- Department of Microbiology, University of Georgia, Athens, GA 30602, U.S.A
| | - Joshua S. Sharp
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, U.S.A
| | - Stéphane Benoit
- Department of Microbiology, University of Georgia, Athens, GA 30602, U.S.A
| | - Robert J. Maier
- Department of Microbiology, University of Georgia, Athens, GA 30602, U.S.A
| |
Collapse
|
41
|
Prophylactic and therapeutic efficacy of the epitope vaccine CTB-UA against Helicobacter pylori infection in a BALB/c mice model. Appl Microbiol Biotechnol 2012; 95:1437-44. [PMID: 22569640 DOI: 10.1007/s00253-012-4122-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 04/14/2012] [Accepted: 04/17/2012] [Indexed: 12/12/2022]
Abstract
Epitope vaccine based on the enzyme urease of Helicobacter pylori is a promising option for prophylactic and therapeutic vaccination against H. pylori infection. In our previous study, the epitope vaccine CTB-UA, which was composed of the mucosal adjuvant cholera toxin B subunit (CTB) and an epitope (UreA₁₈₃₋₂₀₃) from the H. pylori urease A subunit (UreA) was constructed. This particular vaccine was shown to have good immunogenicity and immunoreactivity and could induce specific neutralizing antibodies, which exhibited effectively inhibitory effects on the enzymatic activity of H. pylori urease. In this study, the prophylactic and therapeutic efficacy of the epitope vaccine CTB-UA was evaluated in a BALB/c mice model. The experimental results indicated that oral prophylactic or therapeutic immunization with CTB-UA significantly decreased H. pylori colonization compared with oral immunization with PBS. The results also revealed that the protection was correlated with antigen-specific IgG, IgA, and mucosal secretory IgA antibody responses. CTB-UA may be a promising vaccine candidate for the control of H. pylori infection.
Collapse
|
42
|
Mohammadi M, Kashani SS, Garoosi YT, Tazehkand SJ. In vivo measurement of Helicobacter pylori infection. Methods Mol Biol 2012; 921:239-256. [PMID: 23015509 DOI: 10.1007/978-1-62703-005-2_26] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Helicobacter pylori is a well-recognized gastroduodenal pathogen (National Institute of Health Consensus Conference, JAMA 272:65-9, 1994) and a class I carcinogen (International Agency for Research on Cancer, IARC Monograph on the Evaluation of Carcinogenic Risk to Humans 61:177-240, 1994) which successfully colonizes the harsh acidic environment of the stomach. H. pylori is the causative factor for peptic ulcer disease (PUD) and an independent risk factor for gastric adenocarcinoma development. Therefore, accurate detection of infection is crucial for devising proper eradication regimens and preventing the more severe GI complications.Detection of H. pylori in the gastric mucosa can be performed via (1) direct detection of the bacterium; culture, histology and polymerase chain reaction (PCR) or (2) indirect detection of its enzymatic products particularly urease as well as serum H. pylori-specific antibody responses, which can be detected by rapid urease test (RUT) and serology, respectively.The accuracy of these diagnostic tests is reported as follows: 98.1% for bacterial culture, 98.1% for histology, 94.3% for PCR, 96.2% for RUT, and 84.9% for serology (Ni et al, J Pediatr 136(6):823-7, 2000).
Collapse
Affiliation(s)
- Marjan Mohammadi
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| | | | | | | |
Collapse
|
43
|
Gray LR, Gu SX, Quick M, Khademi S. Transport kinetics and selectivity of HpUreI, the urea channel from Helicobacter pylori. Biochemistry 2011; 50:8656-63. [PMID: 21877689 DOI: 10.1021/bi200887a] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Helicobacter pylori's unique ability to colonize and survive in the acidic environment of the stomach is critically dependent on uptake of urea through the urea channel, HpUreI. Hence, HpUreI may represent a promising target for the development of specific drugs against this human pathogen. To obtain insight into the structure-function relationship of this channel, we developed conditions for the high-yield expression and purification of stable recombinant HpUreI. Detergent-solubilized HpUreI forms a homotrimer, as determined by chemical cross-linking. Urea dissociation kinetics of purified HpUreI were determined by means of the scintillation proximity assay, whereas urea efflux was measured in HpUreI-containing proteoliposomes using stopped-flow spectrometry to determine the kinetics and selectivity of the urea channel. The kinetic analyses revealed that urea conduction in HpUreI is pH-sensitive and saturable with a half-saturation concentration (or K(0.5)) of ~163 mM. The extent of binding of urea by HpUreI was increased at lower pH; however, the apparent affinity of urea binding (~150 mM) was not significantly pH-dependent. The solute selectivity analysis indicated that HpUreI is highly selective for urea and hydroxyurea. Removing either amino group of urea molecules diminishes their permeability through HpUreI. Similar to urea conduction, diffusion of water through HpUreI is pH-dependent with low water permeability at neutral pH.
Collapse
Affiliation(s)
- Lawrence R Gray
- Department of Biochemistry, University of Iowa, Iowa City, Iowa 52241, United States
| | | | | | | |
Collapse
|
44
|
Identification of Proteins Related to Nickel Homeostasis in Helicobater pylori by Immobilized Metal Affinity Chromatography and Two-Dimensional Gel Electrophoresis. Met Based Drugs 2011; 2008:289490. [PMID: 18288244 PMCID: PMC2225478 DOI: 10.1155/2008/289490] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2007] [Accepted: 10/21/2007] [Indexed: 12/16/2022] Open
Abstract
Helicobacter pylori (H. pylori) is a widespread human pathogen causing peptic ulcers and chronic gastritis. Maintaining nickel homeostasis is crucial for the establishment of
H. pylori infection in humans. We used immobilized-nickel affinity chromatography to isolate Ni-related proteins from H. pylori cell extracts. Two-dimensional gel electrophoresis and mass spectrometry were employed to separate and identify twenty two Ni-interacting proteins in H. pylori. These Ni-interacting proteins can be classified into several general functional categories, including cellular processes (HspA, HspB, TsaA, and NapA), enzymes (Urease, Fumarase, GuaB, Cad, PPase, and DmpI), membrane-associated proteins (OM jhp1427 and HpaA), iron storage protein (Pfr), and hypothetical proteins (HP0271, HP jhp0216, HP jhp0301, HP0721, HP0614, and HP jhp0118). The implication of these proteins in nickel homeostasis is discussed.
Collapse
|
45
|
Wang HE, Chang CH, Ker YB, Peng CC, Chen KC, Peng RY. Fermented soybean liquid alleviated peptic ulcer through the destruction of acidic proton pump rather than suppression of urease of Helicobacter pylori: a kinetic analysis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2011; 59:6730-9. [PMID: 21591741 DOI: 10.1021/jf200770n] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Fermented soybean liquid (FSL) has been well cited for its broad spectrum of biological effects, yet its documented gastropeptic ulcer (GPU) ameliorating effect is still lacking. It was hypothesized that to avoid the injury exerted by gastric fluid, HP has to be sheltered in chyme emulsions immediately on infection. The HP urease (HPU) and the acidic proton pump (PP) may act as the "two-point pH modulator" to maintain an optimum pH between 6 and 7, and FSL is able to destroy such a modulating mechanism. FSL exhibited higher contents of isoflavonoids (2.5-17.3-fold) and essential amino acids (1.5-4.0-fold) than the nonfermented. FSL administered at 1 g/20 mL tid for 3 months eradicated Helicobacter pylori (HP) by 82% in 37 volunteers having GPU (p < 0.20); simultaneously, the plasma conjugated diene and TBARs levels were significantly resumed (p < 0.05). Kinetic analysis based on the conventional "urease theory" revealed that a cluster of 2.0 × 10(9) of HP cells is required for a single attack in the gastric lumen at pH 1.0-2.5. To verify the hypothesis, chyme-shelter testing was conducted in artificial gastric fluid (pH 2.4 ± 0.20). Results showed the HP cell viability was time- and size-dependent. At 20 min of contact time, the viability was 100, 4.2, 31.4, 43.3, 57.2, and 82.6%, respectively, in intact, dispersed, and particulate chymes (mesh sizes 80, 60, 40, and 20). The corresponding data became 96.2, 0.0, 14.5, 18.5, 21.3, and 28.6%, respectively, at a contact time of 40 min. Conclusively, the kinetic analysis and the chyme-shelter testing revealed that direct infection by bare HP cells is unlikely in real status. FSL is beneficial to GPU most probably due to its ability to raise blood alkalinity levels, destroying the PP and its ROS suppressing effect.
Collapse
Affiliation(s)
- Hui-Er Wang
- Department of Food and Applied Technology, Hungkuang University, 34 Chung-Chie Road, Shalu County, Taichung Hsien 43302, Taiwan
| | | | | | | | | | | |
Collapse
|
46
|
Muller C, Bahlawane C, Aubert S, Delay CM, Schauer K, Michaud-Soret I, De Reuse H. Hierarchical regulation of the NikR-mediated nickel response in Helicobacter pylori. Nucleic Acids Res 2011; 39:7564-75. [PMID: 21666253 PMCID: PMC3177205 DOI: 10.1093/nar/gkr460] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Nickel is an essential metal for Helicobacter pylori, as it is the co-factor of two enzymes crucial for colonization, urease and hydrogenase. Nickel is taken up by specific transporters and its intracellular homeostasis depends on nickel-binding proteins to avoid toxicity. Nickel trafficking is controlled by the Ni(II)-dependent transcriptional regulator NikR. In contrast to other NikR proteins, NikR from H. pylori is a pleiotropic regulator that depending on the target gene acts as an activator or a repressor. We systematically quantified the in vivo Ni2+-NikR response of 11 direct NikR targets that encode functions related to nickel metabolism, four activated and seven repressed genes. Among these, four targets were characterized for the first time (hpn, hpn-like, hydA and hspA) and NikR binding to their promoter regions was demonstrated by electrophoretic mobility shift assays. We found that NikR-dependent repression was generally set up at higher nickel concentrations than activation. Kinetics of the regulation revealed a gradual and temporal NikR-mediated response to nickel where activation of nickel-protection mechanisms takes place before repression of nickel uptake. Our in vivo study demonstrates, for the first time, a chronological hierarchy in the NikR-dependent transcriptional response to nickel that is coherent with the control of nickel homeostasis in H. pylori.
Collapse
Affiliation(s)
- Cécile Muller
- Département de Microbiologie, Institut Pasteur, Unité Pathogenèse de Helicobacter, Paris Cedex 15, France
| | | | | | | | | | | | | |
Collapse
|
47
|
Lam R, Romanov V, Johns K, Battaile KP, Wu-Brown J, Guthrie JL, Hausinger RP, Pai EF, Chirgadze NY. Crystal structure of a truncated urease accessory protein UreF from Helicobacter pylori. Proteins 2011; 78:2839-48. [PMID: 20635345 DOI: 10.1002/prot.22802] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Urease plays a central role in the pathogenesis of Helicobacter pylori in humans. Maturation of this nickel metalloenzyme in bacteria requires the participation of the accessory proteins UreD (termed UreH in H. pylori), UreF, and UreG, which form sequential complexes with the urease apoprotein as well as UreE, a metallochaperone. Here, we describe the crystal structure of C-terminal truncated UreF from H. pylori (residues 1-233), the first UreF structure to be determined, at 1.55 A resolution using SAD methods. UreF forms a dimer in vitro and adopts an all-helical fold congruent with secondary structure prediction. On the basis of evolutionary conservation analysis, the structure reveals a probable binding surface for interaction with other urease components as well as key conserved residues of potential functional relevance.
Collapse
Affiliation(s)
- Robert Lam
- Division of Cancer Genomics and Proteomics, Ontario Cancer Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Surface properties of Helicobacter pylori urease complex are essential for persistence. PLoS One 2010; 5:e15042. [PMID: 21124783 PMCID: PMC2993952 DOI: 10.1371/journal.pone.0015042] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Accepted: 10/14/2010] [Indexed: 12/18/2022] Open
Abstract
The enzymatic activity of Helicobacter pylori's urease neutralises stomach acidity, thereby promoting infection by this pathogen. Urease protein has also been found to interact with host cells in vitro, although this property's possible functional importance has not been studied in vivo. To test for a role of the urease surface in the host/pathogen interaction, surface exposed loops that display high thermal mobility were targeted for inframe insertion mutagenesis. H. pylori expressing urease with insertions at four of eight sites tested retained urease activity, which in three cases was at least as stable as was wild-type urease at pH 3. Bacteria expressing one of these four mutant ureases, however, failed to colonise mice for even two weeks, and a second had reduced bacterial titres after longer term (3 to 6 months) colonisation. These results indicate that a discrete surface of the urease complex is important for H. pylori persistence during gastric colonisation. We propose that this surface interacts directly with host components important for the host-pathogen interaction, immune modulation or other actions that underlie H. pylori persistence in its special gastric mucosal niche.
Collapse
|
49
|
|
50
|
Ding H, Czinn SJ, Blanchard TG. Recent advances that favor development of a vaccine for Helicobacter pylori infection. PEDIATRIC HEALTH 2008; 2:539. [PMID: 20151035 PMCID: PMC2819429 DOI: 10.2217/17455111.2.5.539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Affiliation(s)
- Hua Ding
- Tel.: +1 410 706 8778
- University of Maryland School of Medicine, Department of Pediatrics, 655 W. Baltimore Street, Bressler Research Building, 13-043, Baltimore, MD 21201, USA, Tel.: +1 410 706 1772; Fax: +1 410 328 1072
| | - Steven J Czinn
- Department of Pediatrics, 22 South Greene Street, Room N5E17, Baltimore, MD 21201-1595, USA, Tel.: +1 410 328 6777; Fax: +1 410 328 8742
- University of Maryland School of Medicine, Department of Pediatrics, 655 W. Baltimore Street, Bressler Research Building, 13-043, Baltimore, MD 21201, USA, Tel.: +1 410 706 1772; Fax: +1 410 328 1072
| | - Thomas G Blanchard
- University of Maryland School of Medicine, Department of Pediatrics, 655 W. Baltimore Street, Bressler Research Building, 13-043, Baltimore, MD 21201, USA, Tel.: +1 410 706 1772; Fax: +1 410 328 1072
| |
Collapse
|