1
|
Viala JPM, Bouveret E. Protein-Protein Interaction: Tandem Affinity Purification in Bacteria. Methods Mol Biol 2024; 2715:285-297. [PMID: 37930536 DOI: 10.1007/978-1-0716-3445-5_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
The discovery of protein-protein interaction networks can lead to the unveiling of protein complex(es) forming cellular machinerie(s) or reveal component proteins of a specific cellular pathway. Deciphering protein-protein interaction networks therefore contributes to a deeper understanding of how cells function. Here we describe the protocol to perform tandem affinity purification (TAP) in bacteria, which enables the identification of the partners of a bait protein under native conditions. This method consists in two sequential steps of affinity purification using two different tags. For that purpose, the bait protein is translationally fused to the TAP tag, which consists of a calmodulin-binding peptide (CBP) and two immunoglobulin G (IgG)-binding domains of Staphylococcus aureus protein A (ProtA) that are separated by the tobacco etch virus (TEV) protease cleavage site. After the first round of purification based on the binding of ProtA to IgG-coated beads, TEV protease cleavage releases CBP-tagged bait protein along with its partners for a second round of purification on calmodulin affinity resin and leaves behind protein contaminants bound to IgG. Creating the TAP-tag translational fusion at the chromosomal locus allows detection of protein interactions occurring in physiological conditions.
Collapse
Affiliation(s)
- Julie P M Viala
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires (UMR 7255), Institut de Microbiologie de la Méditerranée, Aix-Marseille Univ., CNRS, Marseille, France.
| | - Emmanuelle Bouveret
- Institut Pasteur, Department of Microbiology, Unit Stress, Adaptation and Metabolism in enterobacteria, Université Paris Cité, UMR CNRS 6047, Paris, France
| |
Collapse
|
2
|
Denic M, Turlin E, Zamble DB, Betton JM, Vinella D, De Reuse H. The SlyD metallochaperone targets iron-sulfur biogenesis pathways and the TCA cycle. mBio 2023; 14:e0096723. [PMID: 37584558 PMCID: PMC10653786 DOI: 10.1128/mbio.00967-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/26/2023] [Indexed: 08/17/2023] Open
Abstract
IMPORTANCE Correct folding of proteins represents a crucial step for their functions. Among the chaperones that control protein folding, the ubiquitous PPIases catalyze the cis/trans-isomerization of peptidyl-prolyl bonds. Only few protein targets of PPIases have been reported in bacteria. To fill this knowledge gap, we performed a large-scale two-hybrid screen to search for targets of the Escherichia coli and Helicobacter pylori SlyD PPIase-metallochaperone. SlyD from both organisms interacts with enzymes (i) containing metal cofactors, (ii) from the central metabolism tricarboxylic acid (TCA) cycle, and (iii) involved in the formation of the essential and ancestral Fe-S cluster cofactor. E. coli and H. pylori ∆slyD mutants present similar phenotypes of diminished susceptibility to antibiotics and to oxidative stress. In H. pylori, measurements of the intracellular ATP content, proton motive force, and activity of TCA cycle proteins suggest that SlyD regulates TCA cycle enzymes by controlling the formation of their indispensable Fe-S clusters.
Collapse
Affiliation(s)
- Milica Denic
- Unité Pathogenèse de Helicobacter, Département de Microbiologie, UMR CNRS 6047, Institut Pasteur, Université Paris Cité, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
| | - Evelyne Turlin
- Unité Pathogenèse de Helicobacter, Département de Microbiologie, UMR CNRS 6047, Institut Pasteur, Université Paris Cité, Paris, France
| | - Deborah B. Zamble
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Jean-Michel Betton
- Unité Adaptation au stress et Métabolisme chez les entérobactéries, Département de Microbiologie, UMR CNRS 6047, Institut Pasteur, Université Paris Cité, Paris, France
| | - Daniel Vinella
- Unité Pathogenèse de Helicobacter, Département de Microbiologie, UMR CNRS 6047, Institut Pasteur, Université Paris Cité, Paris, France
| | - Hilde De Reuse
- Unité Pathogenèse de Helicobacter, Département de Microbiologie, UMR CNRS 6047, Institut Pasteur, Université Paris Cité, Paris, France
| |
Collapse
|
3
|
Bacterial Membrane Vesicles as a Novel Strategy for Extrusion of Antimicrobial Bismuth Drug in Helicobacter pylori. mBio 2022; 13:e0163322. [PMID: 36154274 PMCID: PMC9601102 DOI: 10.1128/mbio.01633-22] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Bacterial antibiotic resistance is a major threat to human health. A combination of antibiotics with metals is among the proposed alternative treatments. Only one such combination is successfully used in clinics; it associates antibiotics with the metal bismuth to treat infections by Helicobacter pylori. This bacterial pathogen colonizes the human stomach and is associated with gastric cancer, killing 800,000 individuals yearly. The effect of bismuth in H. pylori treatment is not well understood in particular for sublethal doses such as those measured in the plasma of treated patients. We addressed this question and observed that bismuth induces the formation of homogeneously sized membrane vesicles (MVs) with unique protein cargo content enriched in bismuth-binding proteins, as shown by quantitative proteomics. Purified MVs of bismuth-exposed bacteria were strongly enriched in bismuth as measured by inductively coupled plasma optical emission spectrometry (ICP-OES), unlike bacterial cells from which they originate. Thus, our results revealed a novel function of MVs in bismuth detoxification, where secreted MVs act as tool to discard bismuth from the bacteria. Bismuth also induces the formation of intracellular polyphosphate granules that are associated with changes in nucleoid structure. Nucleoid compaction in response to bismuth was established by immunogold electron microscopy and refined by the first chromosome conformation capture (Hi-C) analysis of H. pylori. Our results reveal that even low doses of bismuth induce profound changes in H. pylori physiology and highlight a novel defense mechanism that involves MV-mediated bismuth extrusion from the bacteria and a probable local DNA protective response where polyphosphate granules are associated with nucleoid compaction.
Collapse
|
4
|
Xia X. Multiple regulatory mechanisms for pH homeostasis in the gastric pathogen, Helicobacter pylori. ADVANCES IN GENETICS 2022; 109:39-69. [PMID: 36334916 DOI: 10.1016/bs.adgen.2022.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Acid-resistance in gastric pathogen Helicobacter pylori requires the coordination of four essential processes to regulate urease activity. Firstly, urease expression above a base level needs to be finely tuned at different ambient pH. Secondly, as nickel is needed to activate urease, nickel homeostasis needs to be maintained by proteins that import and export nickel ions, and sequester, store and release nickel when needed. Thirdly, urease accessary proteins that activate urease activity by nickel insertion need to be expressed. Finally, a reliable source of urea needs to be maintained by both intrinsic and extrinsic sources of urea. Two-component systems (arsRS and flgRS), as well as a nickel response regulator (NikR), sense the change in pH and act on a variety of genes to accomplish the function of acid resistance without causing cellular overalkalization and nickel toxicity. Nickel storage proteins also feature built-in switches to store nickel at neutral pH and release nickel at low pH. This review summarizes the current status of H. pylori research and highlights a number of hypotheses that need to be tested.
Collapse
Affiliation(s)
- Xuhua Xia
- Department of Biology, University of Ottawa, Ottawa, Canada; Ottawa Institute of Systems Biology, Ottawa, Canada.
| |
Collapse
|
5
|
Kumar S, Vinella D, De Reuse H. Nickel, an essential virulence determinant of Helicobacter pylori: Transport and trafficking pathways and their targeting by bismuth. Adv Microb Physiol 2022; 80:1-33. [PMID: 35489790 DOI: 10.1016/bs.ampbs.2022.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Metal acquisition and intracellular trafficking are crucial for all cells and metal ions have been recognized as virulence determinants in bacterial pathogens. Nickel is required for the pathogenicity of H. pylori. This bacterial pathogen colonizes the stomach of about half of the human population worldwide and is associated with gastric cancer that is responsible for 800,000 deaths per year. H. pylori possesses two nickel-enzymes that are essential for in vivo colonization, a [NiFe] hydrogenase and an abundant urease responsible for resistance to gastric acidity. Because of these two enzymes, survival of H. pylori relies on an important supply of nickel, implying tight control strategies to avoid its toxic accumulation or deprivation. H. pylori possesses original mechanisms for nickel uptake, distribution, storage and trafficking that will be discussed in this review. During evolution, acquisition of nickel transporters and specific nickel-binding proteins has been a decisive event to allow Helicobacter species to become able to colonize the stomach. Accordingly, many of the factors involved in these mechanisms are required for mouse colonization by H. pylori. These mechanisms are controlled at different levels including protein interaction networks, transcriptional, post-transcriptional and post-translational regulation. Bismuth is another metal used in combination with antibiotics to efficiently treat H. pylori infections. Although the precise mode of action of bismuth is unknown, many targets have been identified in H. pylori and there is growing evidence that bismuth interferes with the essential nickel pathways. Understanding the metal pathways will help improve treatments against H. pylori and other pathogens.
Collapse
Affiliation(s)
- Sumith Kumar
- Unité Pathogenèse de Helicobacter, CNRS UMR6047, Département de Microbiologie, Institut Pasteur, Paris, France
| | - Daniel Vinella
- Unité Pathogenèse de Helicobacter, CNRS UMR6047, Département de Microbiologie, Institut Pasteur, Paris, France
| | - Hilde De Reuse
- Unité Pathogenèse de Helicobacter, CNRS UMR6047, Département de Microbiologie, Institut Pasteur, Paris, France.
| |
Collapse
|
6
|
Abstract
Hydrogenases and ureases play vital metabolic functions in all three domains of life. However, nickel ions are cytotoxic because they can inactivate enzymes that require less competitive ions (e.g. Mg2+) in the Irving-Williams series to function. Life has evolved elegant mechanisms to solve the problem of delivering the toxic metal to the active site of nickel-containing enzymes inside the cells. Here, we review our current understanding of nickel trafficking along the hydrogenase and urease maturation pathways. Metallochaperones and accessory proteins (SlyD, HypA, HypB, UreD, UreE, UreF, and UreG) form specific protein complexes to allow the transfer of nickel from one protein to another without releasing the toxic metal into the cytoplasm. The role of SlyD is not fully understood, but it can interact with and transfer its nickel to HypB. In the hydrogenase maturation pathway, nickel is transferred from HypB to HypA, which can then deliver its nickel to the hydrogenase large subunit precursor. In Helicobacter pylori, the urease maturation pathway receives its nickel from HypA of the hydrogenase maturation pathway via the formation of a HypA/UreE2 complex. Guanosine triphosphate (GTP) binding promotes the formation of a UreE2G2 complex, where UreG receives a nickel from UreE. In the final step of the urease maturation, nickel/GTP-bound UreG forms an activation complex with UreF, UreD, and apo-urease. Upon GTP hydrolysis, nickel is released from UreG to the urease. Finally, some common themes learned from the hydrogenase-urease maturation pathway are discussed.
Collapse
Affiliation(s)
- Ka Lung Tsang
- School of Life Sciences, Centre for Protein Science and Crystallography, State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Kam-Bo Wong
- School of Life Sciences, Centre for Protein Science and Crystallography, State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| |
Collapse
|
7
|
Denic M, Turlin E, Michel V, Fischer F, Khorasani-Motlagh M, Zamble D, Vinella D, de Reuse H. A novel mode of control of nickel uptake by a multifunctional metallochaperone. PLoS Pathog 2021; 17:e1009193. [PMID: 33444370 PMCID: PMC7840056 DOI: 10.1371/journal.ppat.1009193] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 01/27/2021] [Accepted: 11/26/2020] [Indexed: 01/08/2023] Open
Abstract
Cellular metal homeostasis is a critical process for all organisms, requiring tight regulation. In the major pathogen Helicobacter pylori, the acquisition of nickel is an essential virulence determinant as this metal is a cofactor for the acid-resistance enzyme, urease. Nickel uptake relies on the NixA permease and the NiuBDE ABC transporter. Till now, bacterial metal transporters were reported to be controlled at their transcriptional level. Here we uncovered post-translational regulation of the essential Niu transporter in H. pylori. Indeed, we demonstrate that SlyD, a protein combining peptidyl-prolyl isomerase (PPIase), chaperone, and metal-binding properties, is required for the activity of the Niu transporter. Using two-hybrid assays, we found that SlyD directly interacts with the NiuD permease subunit and identified a motif critical for this contact. Mutants of the different SlyD functional domains were constructed and used to perform in vitro PPIase activity assays and four different in vivo tests measuring nickel intracellular accumulation or transport in H. pylori. In vitro, SlyD PPIase activity is down-regulated by nickel, independently of its C-terminal region reported to bind metals. In vivo, a role of SlyD PPIase function was only revealed upon exposure to high nickel concentrations. Most importantly, the IF chaperone domain of SlyD was shown to be mandatory for Niu activation under all in vivo conditions. These data suggest that SlyD is required for the active functional conformation of the Niu permease and regulates its activity through a novel mechanism implying direct protein interaction, thereby acting as a gatekeeper of nickel uptake. Finally, in agreement with a central role of SlyD, this protein is essential for the colonization of the mouse model by H. pylori. Metal ions are essential for the viability of all living organisms. Indeed, more than one-third of all proteins need metal cofactors for their function. Intracellular metal concentrations require tight control as non-physiological amounts are very toxic. In particular, nickel plays a unique role in Helicobacter pylori, a bacterial pathogen that colonizes the stomach of about half of the human population worldwide and is associated with the development of gastric cancer. Nickel is essential for H. pylori as it is the cofactor of urease, an enzyme indispensable for resistance to the gastric acidity of the stomach and thus for in vivo colonization. To import nickel despite its scarcity in the human body, H. pylori requires efficient uptake mechanisms. Till now, control of nickel uptake was only reported to rely on transcriptional regulators. In the present study, we uncovered a novel mechanism of regulation of nickel acquisition. SlyD, a multifunctional enzyme was found to control, by direct protein interaction, the activity of an essential nickel uptake system in H. pylori. We revealed that the SlyD chaperone activity is mandatory for the active conformation and thus functionality of the nickel permease.
Collapse
Affiliation(s)
- Milica Denic
- Institut Pasteur, Département de Microbiologie, Unité Pathogenèse de Helicobacter, CNRS UMR 2001, Paris, France
- Université de Paris, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
| | - Evelyne Turlin
- Institut Pasteur, Département de Microbiologie, Unité Pathogenèse de Helicobacter, CNRS UMR 2001, Paris, France
| | - Valérie Michel
- Institut Pasteur, Département de Microbiologie, Unité Pathogenèse de Helicobacter, CNRS UMR 2001, Paris, France
| | - Frédéric Fischer
- Génétique Moléculaire, Génomique, Microbiologie, UMR 7156, CNRS, Université de Strasbourg, Institut de Botanique, Strasbourg, France
| | | | - Deborah Zamble
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Daniel Vinella
- Institut Pasteur, Département de Microbiologie, Unité Pathogenèse de Helicobacter, CNRS UMR 2001, Paris, France
- * E-mail: (DV); (HDR)
| | - Hilde de Reuse
- Institut Pasteur, Département de Microbiologie, Unité Pathogenèse de Helicobacter, CNRS UMR 2001, Paris, France
- * E-mail: (DV); (HDR)
| |
Collapse
|
8
|
Zhang P, Liu M, Fu J, Zhong C, Zong G, Cao G. Identification of a mobilizable, multidrug-resistant genomic island in Myroides odoratimimus isolated from Tibetan pasture. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 723:137970. [PMID: 32217404 DOI: 10.1016/j.scitotenv.2020.137970] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/28/2020] [Accepted: 03/14/2020] [Indexed: 06/10/2023]
Abstract
Strains of the environmental bacterium Myroides odoratimimus can cause human infections. However, treating M. odoratimimus infections can be difficult because of multidrug resistance in this organism. In this study, we isolated strain M. odoratimimus G13 from pastureland in Tibet, China. The minimum inhibitory concentration analysis suggested that strain G13 has resistance to multiple antibiotics, with an MIC for tetracycline of 168 mg/L. Whole-genome sequencing and bioinformatic analysis revealed that the genome of G13 was rich in virulence factor-encoding genes and antibiotic resistance genes (ARGs). The mobilizable genomic island MGI1313 was also identified and characterized, and six resistance genes related to four types of antibiotics were annotated in MGI1313. Conjugation assays indicated that MGI1313 could be transferred from G13 to Escherichia coli 25DN by horizontal gene transfer, resulting in multidrug-resistant E. coli conjugants. In conclusion, multidrug-resistant M. odoratimimus G13 and the mobility of MGI1313 raise the risk of difficult-to-treat bacterial infections and should be under close surveillance.
Collapse
Affiliation(s)
- Peipei Zhang
- The State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; Department of Epidemiology, the First Affiliated Hospital of Shandong First Medical University, Jinan 250062, China; Shandong Medicinal Biotechnology Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Meng Liu
- School of Municipal and Environmental Engineering, Shandong Jianzhu University, Jinan 250101, China
| | - Jiafang Fu
- Department of Epidemiology, the First Affiliated Hospital of Shandong First Medical University, Jinan 250062, China; Shandong Medicinal Biotechnology Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China; Key Laboratory for Biotech-Drugs of National Health Commission, Jinan 250062, China
| | - Chuanqing Zhong
- School of Municipal and Environmental Engineering, Shandong Jianzhu University, Jinan 250101, China
| | - Gongli Zong
- Department of Epidemiology, the First Affiliated Hospital of Shandong First Medical University, Jinan 250062, China; Shandong Medicinal Biotechnology Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China; Key Laboratory for Biotech-Drugs of National Health Commission, Jinan 250062, China
| | - Guangxiang Cao
- Department of Epidemiology, the First Affiliated Hospital of Shandong First Medical University, Jinan 250062, China; Shandong Medicinal Biotechnology Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China.
| |
Collapse
|
9
|
Salze M, Muller C, Bernay B, Hartke A, Clamens T, Lesouhaitier O, Rincé A. Study of key RNA metabolism proteins in Enterococcus faecalis. RNA Biol 2020; 17:794-804. [PMID: 32070211 DOI: 10.1080/15476286.2020.1728103] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
The control of mRNA turnover is essential in bacteria to allow rapid adaptation, especially in opportunistic pathogen like Enterococcus faecalis. This mechanism involves RNase and DEAD-box helicases that are key elements in RNA processing and their associations form the degradosome with accessory proteins. In this study, we investigated the function of four RNases (J1, J2, Y and III) and three DEAD-box helicases (CshA, CshB, CshC) present in most Enterococci. The interactions of all these RNA metabolism actors were investigated in vitro, and the results are in accordance with a degradosome structure close to the one of Bacillus subtilis. At the physiological level, we showed that RNase J1 is essential, whereas RNases J2 and III have a role in cold, oxidative and bile salts stress response, and RNase Y in general fitness. Furthermore, RNases J2, Y and III mutants are affected in virulence in the Galleria mellonella infection model. Concerning DEAD-box helicases, all of them are involved in cold shock response. Since the ΔcshA mutant was the most stress impacted strain, we studied this DEAD-box helicase CshA in more detail. This showed that CshA autoregulates its own expression by binding to its mRNA 5'Unstranslated Region. Interestingly, CshC is also involved in the expression control of CshA by a hitherto unprecedented mechanism.
Collapse
Affiliation(s)
- Marine Salze
- Normandie Univ, UNICAEN, Unité De Recherche Risques Microbiens U2RM , Caen, France
| | - Cécile Muller
- Normandie Univ, UNICAEN, Unité De Recherche Risques Microbiens U2RM , Caen, France
| | - Benoit Bernay
- Proteogen Platform, Normandie Univ, UNICAEN, SFR ICORE , Caen, France
| | - Axel Hartke
- Normandie Univ, UNICAEN, Unité De Recherche Risques Microbiens U2RM , Caen, France
| | - Thomas Clamens
- Laboratoire de Microbiologie Signaux et Microenvironnement LMSM, Normandie Univ, University of Rouen , Evreux, France
| | - Olivier Lesouhaitier
- Laboratoire de Microbiologie Signaux et Microenvironnement LMSM, Normandie Univ, University of Rouen , Evreux, France
| | - Alain Rincé
- Normandie Univ, UNICAEN, Unité De Recherche Risques Microbiens U2RM , Caen, France
| |
Collapse
|
10
|
Abstract
Nickel is essential for the survival of many pathogenic bacteria. E. coli and H. pylori require nickel for [NiFe]-hydrogenases. H. pylori also requires nickel for urease. At high concentrations nickel can be toxic to the cell, therefore, nickel concentrations are tightly regulated. Metalloregulators help to maintain nickel concentration in the cell by regulating the expression of the genes associated with nickel import and export. Nickel import into the cell, delivery of nickel to target proteins, and export of nickel from the cell is a very intricate and well-choreographed process. The delivery of nickel to [NiFe]-hydrogenase and urease is complex and involves several chaperones and accessory proteins. A combination of biochemical, crystallographic, and spectroscopic techniques has been utilized to study the structures of these proteins, as well as protein-protein interactions resulting in an expansion of our knowledge regarding how these proteins sense and bind nickel. In this review, recent advances in the field will be discussed, focusing on the metal site structures of nickel bound to metalloregulators and chaperones.
Collapse
|
11
|
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
|
12
|
Abstract
Nickel is an essential cofactor for some pathogen virulence factors. Due to its low availability in hosts, pathogens must efficiently transport the metal and then balance its ready intracellular availability for enzyme maturation with metal toxicity concerns. The most notable virulence-associated components are the Ni-enzymes hydrogenase and urease. Both enzymes, along with their associated nickel transporters, storage reservoirs, and maturation enzymes have been best-studied in the gastric pathogen Helicobacter pylori, a bacterium which depends heavily on nickel. Molecular hydrogen utilization is associated with efficient host colonization by the Helicobacters, which include both gastric and liver pathogens. Translocation of a H. pylori carcinogenic toxin into host epithelial cells is powered by H2 use. The multiple [NiFe] hydrogenases of Salmonella enterica Typhimurium are important in host colonization, while ureases play important roles in both prokaryotic (Proteus mirabilis and Staphylococcus spp.) and eukaryotic (Cryptoccoccus genus) pathogens associated with urinary tract infections. Other Ni-requiring enzymes, such as Ni-acireductone dioxygenase (ARD), Ni-superoxide dismutase (SOD), and Ni-glyoxalase I (GloI) play important metabolic or detoxifying roles in other pathogens. Nickel-requiring enzymes are likely important for virulence of at least 40 prokaryotic and nine eukaryotic pathogenic species, as described herein. The potential for pathogenic roles of many new Ni-binding components exists, based on recent experimental data and on the key roles that Ni enzymes play in a diverse array of pathogens.
Collapse
|
13
|
Complex formation between the Escherichia coli [NiFe]-hydrogenase nickel maturation factors. Biometals 2019; 32:521-532. [DOI: 10.1007/s10534-019-00173-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/18/2019] [Indexed: 11/26/2022]
|
14
|
Hu S, Cao L, Wu Y, Zhou Y, Jiang T, Wang L, Wang Q, Ming D, Chen S, Wang M. Comparative genomic analysis of Myroides odoratimimus isolates. Microbiologyopen 2018; 8:e00634. [PMID: 29797432 PMCID: PMC6391281 DOI: 10.1002/mbo3.634] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 03/05/2018] [Accepted: 03/06/2018] [Indexed: 12/13/2022] Open
Abstract
Myroides odoratimimus is an important nosocomial pathogen. Management of M. odoratimimus infection is difficult owing to the multidrug resistance and the unknown pathogenesis mechanisms. Based on our previous genomic sequencing data of M. odoratimimus PR63039 (isolated from a patient with the urinary tract infection), in this study, we further performed comparative genomic analysis for 10 selected Myroides strains. Our results showed that these Myroides genome contexts were very similar and phylogenetically related. Various prophages were identified in the four clinical isolate genomes, which possibly contributed to the genome evolution among the Myroides strains. CRISPR elements were only detected in the two clinical (PR63039 and CCUG10230) isolates and two environmental (CCUG12700 and H1bi) strains. With more stringent cutoff parameters in CARD analysis, the four clinical M. odoratimimus contained roughly equal antibiotic resistance genes, indicating their similar antibiotic resistance profiles. The three clinical (CCUG10230, CCUG12901, CIP101113) and three environmental (CCUG12700, L41, H1bi) M. odoratimimus strains were speculated to carry the indistinguishable virulent factors (VFs), which may involve in the similar pathogenesis mechanism. Moreover, some VFs might confer to the high capacity of dissemination, attacking tissue cells and induction of autoimmune complications. Our results facilitate the research of antibiotic resistance and the development of therapeutic regimens for the M. odoratimimus infections.
Collapse
Affiliation(s)
- Shaohua Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lin Cao
- School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| | - Yiyin Wu
- College of Computer Science and Technology, Huaqiao University, Xiamen, Fujian, China
| | - Yajun Zhou
- School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| | - Tao Jiang
- School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| | - Liqiang Wang
- School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| | - Qiujing Wang
- Department of Neurosurgery, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Desong Ming
- Department of Clinical Laboratory, Quanzhou First Hospital Affiliated to Fujian Medical University, Fujian, China
| | - Shicheng Chen
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Mingxi Wang
- School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian, China
| |
Collapse
|
15
|
Hu HQ, Huang HT, Maroney MJ. The Helicobacter pylori HypA·UreE 2 Complex Contains a Novel High-Affinity Ni(II)-Binding Site. Biochemistry 2018; 57:2932-2942. [PMID: 29708738 DOI: 10.1021/acs.biochem.8b00127] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Helicobacter pylori is a human pathogen that colonizes the stomach, is the major cause of ulcers, and has been associated with stomach cancers. To survive in the acidic environment of the stomach, H. pylori uses urease, a nickel-dependent enzyme, to produce ammonia for maintenance of cellular pH. The bacteria produce apo-urease in large quantities and activate it by incorporating nickel under acid shock conditions. Urease nickel incorporation requires the urease-specific metallochaperone UreE and the (UreFGH)2 maturation complex. In addition, the H. pylori nickel urease maturation pathway recruits accessory proteins from the [NiFe] hydrogenase maturation pathway, namely, HypA and HypB. HypA and UreE dimers (UreE2) are known to form a protein complex, the role of which in urease maturation is largely unknown. Herein, we examine the nickel-binding properties and protein-protein interactions of HypA and UreE2 using isothermal titration calorimetry and fluorometric methods under neutral and acidic pH conditions to gain insight into the roles played by HypA in urease maturation. The results reveal that HypA and UreE2 form a stable complex with micromolar affinity that protects UreE from hydrolytic degradation. The HypA·UreE2 complex contains a unique high-affinity (nanomolar) Ni2+-binding site that is maintained under conditions designed to mimic acid shock (pH 6.3). The data are interpreted in terms of a proposed mechanism wherein HypA and UreE2 act as co-metallochaperones that target the delivery of Ni2+ to apo-urease with high fidelity.
Collapse
|
16
|
Ratet G, Santecchia I, Fanton d’Andon M, Vernel-Pauillac F, Wheeler R, Lenormand P, Fischer F, Lechat P, Haake DA, Picardeau M, Boneca IG, Werts C. LipL21 lipoprotein binding to peptidoglycan enables Leptospira interrogans to escape NOD1 and NOD2 recognition. PLoS Pathog 2017; 13:e1006725. [PMID: 29211798 PMCID: PMC5764436 DOI: 10.1371/journal.ppat.1006725] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 01/11/2018] [Accepted: 10/31/2017] [Indexed: 12/19/2022] Open
Abstract
Leptospirosis is a widespread zoonosis, potentially severe in humans, caused by spirochetal bacteria, Leptospira interrogans (L. interrogans). Host defense mechanisms involved in leptospirosis are poorly understood. Recognition of lipopolysaccharide (LPS) and lipoproteins by Toll-Like Receptors (TLR)4 and TLR2 is crucial for clearance of leptospires in mice, yet the role of Nucleotide Oligomerization Domain (NOD)-like receptors (NOD)1 and NOD2, recognizing peptidoglycan (PG) fragments has not previously been examined. Here, we show that pathogenic leptospires escape from NOD1 and NOD2 recognition both in vitro and in vivo, in mice. We found that leptospiral PG is resistant to digestion by certain hydrolases and that a conserved outer membrane lipoprotein of unknown function, LipL21, specific for pathogenic leptospires, is tightly bound to the PG. Leptospiral PG prepared from a mutant not expressing LipL21 (lipl21-) was more readily digested than the parental or complemented strains. Muropeptides released from the PG of the lipl21- mutant, or prepared using a procedure to eliminate the LipL21 protein from the PG of the parental strain, were recognized in vitro by the human NOD1 (hNOD1) and NOD2 (hNOD2) receptors, suggesting that LipL21 protects PG from degradation into muropeptides. LipL21 expressed in E. coli also resulted in impaired PG digestion and NOD signaling. We found that murine NOD1 (mNOD1) did not recognize PG of L. interrogans. This result was confirmed by mass spectrometry showing that leptospiral PG was primarily composed of MurTriDAP, the natural agonist of hNOD1, and contained only trace amounts of the tetra muropeptide, the mNOD1 agonist. Finally, in transgenic mice expressing human NOD1 and deficient for the murine NOD1, we showed enhanced clearance of a lipl21- mutant compared to the complemented strain, or to what was observed in NOD1KO mice, suggesting that LipL21 facilitates escape from immune surveillance in humans. These novel mechanisms allowing L. interrogans to escape recognition by the NOD receptors may be important in circumventing innate host responses. Leptospirosis is a widespread zoonosis caused by spirochetal bacteria, Leptospira interrogans (L. interrogans). L. interrogans are primarily extracellular pathogens although some reports suggest they may replicate within macrophages. In humans, leptospirosis can cause mild or severe disease, potentially leading to death, although rats or mice, which constitute the reservoir, are asymptomatic carriers. Host defense mechanisms involved in leptospirosis remain poorly understood. Toll-Like Receptor (TLR)2 and TLR4 are crucial for the clearance of L. interrogans, but the role of the cytosolic NOD receptors in leptospirosis is unknown. Here, we report that pathogenic leptospires escape the sensing of bacterial peptidoglycan through the NOD response. We found that an outer membrane lipoprotein of L. interrogans binds to and protects the peptidoglycan from degradation into muropeptides, thereby blocking signaling through NOD proteins. Moreover, in absence of this lipoprotein, the peptidoglycan of L. interrogans is properly sensed by human NOD1 but not by murine NOD1. This is due to the near absence of muramyl tetrapeptide, the murine NOD1 agonist, in the peptidoglycan of pathogenic leptospires. These novel mechanisms of NOD avoidance may facilitate the escape of leptospires from the innate immune system of their hosts.
Collapse
Affiliation(s)
- Gwenn Ratet
- Institut Pasteur, Unité Biologie et Génétique de la Paroi Bactérienne, Paris, France
- INSERM, équipe Avenir, Paris, France
| | - Ignacio Santecchia
- Institut Pasteur, Unité Biologie et Génétique de la Paroi Bactérienne, Paris, France
- INSERM, équipe Avenir, Paris, France
| | - Martine Fanton d’Andon
- Institut Pasteur, Unité Biologie et Génétique de la Paroi Bactérienne, Paris, France
- INSERM, équipe Avenir, Paris, France
| | - Frédérique Vernel-Pauillac
- Institut Pasteur, Unité Biologie et Génétique de la Paroi Bactérienne, Paris, France
- INSERM, équipe Avenir, Paris, France
| | - Richard Wheeler
- Institut Pasteur, Unité Biologie et Génétique de la Paroi Bactérienne, Paris, France
- INSERM, équipe Avenir, Paris, France
| | | | - Frédéric Fischer
- Institut Pasteur, Unité de pathogenèse de Helicobacter, Paris, France
| | - Pierre Lechat
- Institut Pasteur, Hub Bioinformatique et Biostatistique, C3BI, USR 3756 IP CNRS, Paris, France
| | - David A. Haake
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States of America
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | | | - Ivo G. Boneca
- Institut Pasteur, Unité Biologie et Génétique de la Paroi Bactérienne, Paris, France
- INSERM, équipe Avenir, Paris, France
| | - Catherine Werts
- Institut Pasteur, Unité Biologie et Génétique de la Paroi Bactérienne, Paris, France
- INSERM, équipe Avenir, Paris, France
- * E-mail:
| |
Collapse
|
17
|
Zhang H, Lam KH, Lam WWL, Wong SYY, Chan VSF, Au SWN. A putative spermidine synthase interacts with flagellar switch protein FliM and regulates motility inHelicobacter pylori. Mol Microbiol 2017; 106:690-703. [DOI: 10.1111/mmi.13829] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2017] [Indexed: 12/25/2022]
Affiliation(s)
- Huawei Zhang
- Centre for Protein Science and Crystallography, School of Life Sciences; The Chinese University of Hong Kong; Hong Kong
| | - Kwok Ho Lam
- Centre for Protein Science and Crystallography, School of Life Sciences; The Chinese University of Hong Kong; Hong Kong
| | - Wendy Wai Ling Lam
- Centre for Protein Science and Crystallography, School of Life Sciences; The Chinese University of Hong Kong; Hong Kong
| | - Sandra Yuen Yuen Wong
- Division of Rheumatology & Clinical Immunology, Department of Medicine, Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong
| | - Vera Sau Fong Chan
- Division of Rheumatology & Clinical Immunology, Department of Medicine, Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong
| | - Shannon Wing Ngor Au
- Centre for Protein Science and Crystallography, School of Life Sciences; The Chinese University of Hong Kong; Hong Kong
| |
Collapse
|
18
|
Blum FC, Hu HQ, Servetas SL, Benoit SL, Maier RJ, Maroney MJ, Merrell DS. Structure-function analyses of metal-binding sites of HypA reveal residues important for hydrogenase maturation in Helicobacter pylori. PLoS One 2017; 12:e0183260. [PMID: 28809946 PMCID: PMC5557546 DOI: 10.1371/journal.pone.0183260] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 08/01/2017] [Indexed: 01/18/2023] Open
Abstract
The nickel-containing enzymes of Helicobacter pylori, urease and hydrogenase, are essential for efficient colonization in the human stomach. The insertion of nickel into urease and hydrogenase is mediated by the accessory protein HypA. HypA contains an N-terminal nickel-binding site and a dynamic structural zinc-binding site. The coordination of nickel and zinc within HypA is known to be critical for urease maturation and activity. Herein, we test the hydrogenase activity of a panel of H. pylori mutant strains containing point mutations within the nickel- and zinc-binding sites. We found that the residues that are important for hydrogenase activity are those that were similarly vital for urease activity. Thus, the zinc and metal coordination sites of HypA play similar roles in urease and hydrogenase maturation. In other pathogenic bacteria, deletion of hydrogenase leads to a loss in acid resistance. Thus, the acid resistance of two strains of H. pylori containing a hydrogenase deletion was also tested. These mutant strains demonstrated wild-type levels of acid resistance, suggesting that in H. pylori, hydrogenase does not play a role in acid resistance.
Collapse
Affiliation(s)
- Faith C. Blum
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States of America
| | - Heidi Q. Hu
- Department of Chemistry and Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA, United States of America
| | - Stephanie L. Servetas
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States of America
| | - Stéphane L. Benoit
- Department of Microbiology, University of Georgia, Athens, GA, United States of America
| | - Robert J. Maier
- Department of Microbiology, University of Georgia, Athens, GA, United States of America
| | - Michael J. Maroney
- Department of Chemistry and Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA, United States of America
- * E-mail: (MJM); (DSM)
| | - D. Scott Merrell
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States of America
- * E-mail: (MJM); (DSM)
| |
Collapse
|
19
|
Macegoniuk K, Grela E, Biernat M, Psurski M, Gościniak G, Dziełak A, Mucha A, Wietrzyk J, Berlicki Ł, Grabowiecka A. Aminophosphinates against Helicobacter pylori ureolysis-Biochemical and whole-cell inhibition characteristics. PLoS One 2017; 12:e0182437. [PMID: 28792967 PMCID: PMC5550016 DOI: 10.1371/journal.pone.0182437] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 07/18/2017] [Indexed: 12/14/2022] Open
Abstract
Urease is an important virulence factor from Helicobacter pylori that enables bacterial colonization of human gastric mucosa. Specific inhibition of urease activity can be regarded as a promising adjuvant strategy for eradication of this pathogen. A group of organophosphorus inhibitors of urease, namely, aminophosphinic acid and aminophosphonic acid derivatives, were evaluated in vitro against H. pylori urease. The kinetic characteristics of recombinant enzyme activity demonstrated a competitive reversible mode of inhibition with Ki values ranging from 0.294 to 878 μM. N-n-Hexylaminomethyl-P-aminomethylphosphinic acid and N-methylaminomethyl-P-hydroxymethylphosphinic acid were the most effective inhibitors (Ki = 0.294 μM and 1.032 μM, respectively, compared to Ki = 23 μM for the established urease inhibitor acetohydroxamic acid). The biological relevance of the inhibitors was verified in vitro against a ureolytically active Escherichia coli Rosetta host that expressed H. pylori urease and against a reference strain, H. pylori J99 (CagA+/VacA+). The majority of the studied compounds exhibited urease-inhibiting activity in these whole-cell systems. Bis(N-methylaminomethyl)phosphinic acid was found to be the most effective inhibitor in the susceptibility profile studies of H. pylori J99. The cytotoxicity of nine structurally varied inhibitors was evaluated against four normal human cell lines and was found to be negligible.
Collapse
Affiliation(s)
- Katarzyna Macegoniuk
- Wrocław University of Technology, Faculty of Chemistry, Department of Bioorganic Chemistry, Wrocław, Poland
| | - Ewa Grela
- Wrocław University of Technology, Faculty of Chemistry, Department of Bioorganic Chemistry, Wrocław, Poland
| | - Monika Biernat
- Medical University of Wrocław, Department of Microbiology, Wrocław, Poland
| | - Mateusz Psurski
- Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Grażyna Gościniak
- Medical University of Wrocław, Department of Microbiology, Wrocław, Poland
| | - Anna Dziełak
- Wrocław University of Technology, Faculty of Chemistry, Department of Bioorganic Chemistry, Wrocław, Poland
| | - Artur Mucha
- Wrocław University of Technology, Faculty of Chemistry, Department of Bioorganic Chemistry, Wrocław, Poland
| | - Joanna Wietrzyk
- Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Łukasz Berlicki
- Wrocław University of Technology, Faculty of Chemistry, Department of Bioorganic Chemistry, Wrocław, Poland
| | - Agnieszka Grabowiecka
- Wrocław University of Technology, Faculty of Chemistry, Department of Bioorganic Chemistry, Wrocław, Poland
- * E-mail:
| |
Collapse
|
20
|
Li Q, Zhu Y, Liu J, Yu X, Chen M, Dong N, Gong Y, Yuan Y. HpSlyD inducing CDX2 and VIL1 expression mediated through TCTP protein may contribute to intestinal metaplasia in the stomach. Sci Rep 2017; 7:2278. [PMID: 28536478 PMCID: PMC5442128 DOI: 10.1038/s41598-017-02642-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 04/13/2017] [Indexed: 12/21/2022] Open
Abstract
Helicobacter pylori infection is the most important risk factor for gastric intestinal metaplasia (IM). Our previous study demonstrated that infection with H. pylori HpslyD-positive strains associated with IM. To further investigate the signalling pathway involved in HpSlyD-induced IM, CDX2 and VIL1 expressions were determined before and after HpSlyD application. TCTP was knocked down by siRNA or overexpressed by plasmid transfection. An HpSlyD binding protein was used to block HpSlyD's enzymatic activity. The expression of CDX2 and TCTP in gastric diseases was measured by immunohistochemistry. Our results showed HpSlyD induced CDX2 and VIL1 expressions. TCTP protein expression was markedly increased after application of HpSlyD and in an HpSlyD-expressing stable cell line. Downregulation of TCTP protein led to decreased HpSlyD-induced CDX2 and VIL1. Overexpression of TCTP protein improved the expression of CDX2 and VIL1. Co-application of HpSlyD and FK506 led to significant reductions in CDX2, VIL1, and TCTP expression. Immunohistochemistry demonstrated that CDX2 and TCTP expression was higher in HpslyD-positive specimens compared with HpslyD-negative ones. Expression of CDX2 was positively correlated with TCTP in HpslyD-positive cells. Our study is the first to show that HpSlyD induction of CDX2 and VIL1 expression mediated through TCTP may contribute to IM in the stomach.
Collapse
Affiliation(s)
- Qiuping Li
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Affiliated Hospital of China Medical University, and Key Laboratory of Cancer Etiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang, 110001, China
| | - Yanmei Zhu
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Affiliated Hospital of China Medical University, and Key Laboratory of Cancer Etiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang, 110001, China.,Department of Pathology, Cancer Hospital of China Medical University; Liaoning Cancer Hospital & Institute, Shenyang, 110042, Liaoning Province, China
| | - Jun Liu
- Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV, 26506, USA.,Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, 26506-9229, USA
| | - Xiuwen Yu
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Affiliated Hospital of China Medical University, and Key Laboratory of Cancer Etiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang, 110001, China.,Department of Pathology, Qiqihar Medical College, Qiqihar, Heilongjiang, China
| | - Moye Chen
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Affiliated Hospital of China Medical University, and Key Laboratory of Cancer Etiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang, 110001, China
| | - Nannan Dong
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Affiliated Hospital of China Medical University, and Key Laboratory of Cancer Etiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang, 110001, China
| | - Yuehua Gong
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Affiliated Hospital of China Medical University, and Key Laboratory of Cancer Etiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang, 110001, China.
| | - Yuan Yuan
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Affiliated Hospital of China Medical University, and Key Laboratory of Cancer Etiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang, 110001, China.
| |
Collapse
|
21
|
Zepeda Gurrola RC, Fu Y, Rodríguez Luna IC, Benítez Cardoza CG, López López MDJ, López Vidal Y, Gutíerrez GRA, Rodríguez Pérez MA, Guo X. Novel protein interactions with an actin homolog (MreB) of Helicobacter pylori determined by bacterial two-hybrid system. Microbiol Res 2017; 201:39-45. [PMID: 28602400 DOI: 10.1016/j.micres.2017.04.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/24/2017] [Accepted: 04/22/2017] [Indexed: 02/06/2023]
Abstract
The bacterium Helicobacter pylori infects more than 50% of the world population and causes several gastroduodenal diseases, including gastric cancer. Nevertheless, we still need to explore some protein interactions that may be involved in pathogenesis. MreB, an actin homolog, showed some special characteristics in previous studies, indicating that it could have different functions. Protein functions could be realized via protein-protein interactions. In the present study, the MreB protein from H. pylori 26695 fused with two tags 10×His and GST in tandem was overexpressed and purified from Escherchia coli. The purified recombinant protein was used to perform a pull-down assay with H. pylori 26695 cell lysate. The pulled-down proteins were identified by mass spectrometry (MALDI-TOF), in which the known important proteins related to morphogenesis were absent but several proteins related to pathogenesis process were observed. The bacterial two-hybrid system was further used to evaluate the protein interactions and showed that new interactions of MreB respectively with VacA, UreB, HydB, HylB and AddA were confirmed but the interaction MreB-MreC was not validated. These results indicated that the protein MreB in H. pylori has a distinct interactome, does not participate in cell morphogenesis via MreB-MreC but could be related to pathogenesis.
Collapse
Affiliation(s)
| | - Yajuan Fu
- Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Cd. Reynosa Tamaulipas, Mexico
| | | | | | | | - Yolanda López Vidal
- Facultad de Medicina, División de Investigación, Universidad Nacional Autónoma de Mexico
| | - Germán Rubén Aguilar Gutíerrez
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos, Mexico
| | - Mario A Rodríguez Pérez
- Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Cd. Reynosa Tamaulipas, Mexico
| | - Xianwu Guo
- Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Cd. Reynosa Tamaulipas, Mexico.
| |
Collapse
|
22
|
Xia W, Li H, Sun H. Nickel Metallochaperones: Structure, Function, and Nickel-Binding Properties. THE BIOLOGICAL CHEMISTRY OF NICKEL 2017. [DOI: 10.1039/9781788010580-00284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Nickel-containing enzymes catalyze a series of important biochemical processes in both prokaryotes and eukaryotes. The maturation of the enzymes requires the proper assembly of the nickel-containing active sites, which involves a battery of nickel metallochaperones that exert metal delivery and storage functions. “Cross-talk” also exists between different nickel enzyme maturation processes. This chapter summarizes the updated knowledge about the nickel chaperones based on biochemical and structural biology research, and discusses the possible nickel delivery mechanisms.
Collapse
Affiliation(s)
- Wei Xia
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry Sun Yat-sen University Guangzhou 510275 China
| | - Hongyan Li
- Department of Chemistry, The University of Hong Kong Hong Kong SAR China
| | - Hongzhe Sun
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry Sun Yat-sen University Guangzhou 510275 China
- Department of Chemistry, The University of Hong Kong Hong Kong SAR China
| |
Collapse
|
23
|
Kumari R, Shariq M, Kumar N, Mukhopadhyay G. Biochemical characterization of theHelicobacter pyloriCag-type IV secretion system unique component CagU. FEBS Lett 2017; 591:500-512. [DOI: 10.1002/1873-3468.12564] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Revised: 12/16/2016] [Accepted: 01/10/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Rajesh Kumari
- Special Centre for Molecular Medicine; Jawaharlal Nehru University; New Delhi India
| | - Mohd Shariq
- Special Centre for Molecular Medicine; Jawaharlal Nehru University; New Delhi India
- School of Life Sciences; Jawaharlal Nehru University; New Delhi India
| | - Navin Kumar
- Special Centre for Molecular Medicine; Jawaharlal Nehru University; New Delhi India
- School of Biotechnology; Gautam Buddha University; Uttar Pradesh India
| | | |
Collapse
|
24
|
Viala JPM, Bouveret E. Protein-Protein Interaction: Tandem Affinity Purification in Bacteria. Methods Mol Biol 2017; 1615:221-232. [PMID: 28667616 DOI: 10.1007/978-1-4939-7033-9_18] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The discovery of protein-protein interaction networks can lead to the unveiling of protein complex(es) forming cellular machinerie(s) or reveal component proteins of a specific cellular pathway. Deciphering protein-protein interaction networks therefore contributes to a deeper understanding of how cells function. Here we describe the protocol to perform tandem affinity purification (TAP) in bacteria, which enables the identification of the partners of a bait protein under native conditions. This method consists in two sequential steps of affinity purification using two different tags. For that purpose, the bait protein is translationally fused to the TAP tag, which consists of a calmodulin binding peptide (CBP) and two immunoglobulin G (IgG) binding domains of Staphylococcus aureus protein A (ProtA) that are separated by the tobacco etch virus (TEV) protease cleavage site. After the first round of purification based on the binding of ProtA to IgG coated beads, TEV protease cleavage releases CBP-tagged bait-protein along with its partners for a second round of purification on calmodulin affinity resin and leaves behind protein contaminants bound to IgG. Creating the TAP-tag translational fusion at the chromosomal locus allows detection of protein interactions occurring in physiological conditions.
Collapse
Affiliation(s)
- Julie P M Viala
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires, UMR7255, Institut de Microbiologie de la Méditerranée, Aix-Marseille University-CNRS, 31 Chemin Joseph Aiguier, 13009, Marseille, France.
| | - Emmanuelle Bouveret
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires, UMR7255, Institut de Microbiologie de la Méditerranée, Aix-Marseille University-CNRS, 31 Chemin Joseph Aiguier, 13009, Marseille, France
| |
Collapse
|
25
|
Keasey SL, Natesan M, Pugh C, Kamata T, Wuchty S, Ulrich RG. Cell-free Determination of Binary Complexes That Comprise Extended Protein-Protein Interaction Networks of Yersinia pestis. Mol Cell Proteomics 2016; 15:3220-3232. [PMID: 27489291 DOI: 10.1074/mcp.m116.059337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Indexed: 11/06/2022] Open
Abstract
Binary protein interactions form the basic building blocks of molecular networks and dynamic assemblies that control all cellular functions of bacteria. Although these protein interactions are a potential source of targets for the development of new antibiotics, few high-confidence data sets are available for the large proteomes of most pathogenic bacteria. We used a library of recombinant proteins from the plague bacterium Yersinia pestis to probe planar microarrays of immobilized proteins that represented ∼85% (3552 proteins) of the bacterial proteome, resulting in >77,000 experimentally determined binary interactions. Moderate (KD ∼μm) to high-affinity (KD ∼nm) interactions were characterized for >1600 binary complexes by surface plasmon resonance imaging of microarrayed proteins. Core binary interactions that were in common with other gram-negative bacteria were identified from the results of both microarray methods. Clustering of proteins within the interaction network by function revealed statistically enriched complexes and pathways involved in replication, biosynthesis, virulence, metabolism, and other diverse biological processes. The interaction pathways included many proteins with no previously known function. Further, a large assembly of proteins linked to transcription and translation were contained within highly interconnected subregions of the network. The two-tiered microarray approach used here is an innovative method for detecting binary interactions, and the resulting data will serve as a critical resource for the analysis of protein interaction networks that function within an important human pathogen.
Collapse
Affiliation(s)
- Sarah L Keasey
- From the ‡Molecular and Translational Sciences Division, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, Maryland 21702; §Biological Sciences Department, University of Maryland Baltimore County, Baltimore, Maryland 21250
| | - Mohan Natesan
- From the ‡Molecular and Translational Sciences Division, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, Maryland 21702
| | - Christine Pugh
- From the ‡Molecular and Translational Sciences Division, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, Maryland 21702
| | - Teddy Kamata
- From the ‡Molecular and Translational Sciences Division, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, Maryland 21702
| | - Stefan Wuchty
- ¶National Center for Biotechnology Information, National Institutes of Health, Bethesda, Maryland 20892
| | - Robert G Ulrich
- From the ‡Molecular and Translational Sciences Division, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, Maryland 21702;
| |
Collapse
|
26
|
Abstract
[NiFe]-hydrogenases catalyze the reversible conversion of hydrogen gas into protons and electrons and are vital metabolic components of many species of bacteria and archaea. At the core of this enzyme is a sophisticated catalytic center comprising nickel and iron, as well as cyanide and carbon monoxide ligands, which is anchored to the large hydrogenase subunit through cysteine residues. The production of this multicomponent active site is accomplished by a collection of accessory proteins and can be divided into discrete stages. The iron component is fashioned by the proteins HypC, HypD, HypE, and HypF, which functionalize iron with cyanide and carbon monoxide. Insertion of the iron center signals to the metallochaperones HypA, HypB, and SlyD to selectively deliver the nickel to the active site. A specific protease recognizes the completed metal cluster and then cleaves the C-terminus of the large subunit, resulting in a conformational change that locks the active site in place. Finally, the large subunit associates with the small subunit, and the complete holoenzyme translocates to its final cellular position. Beyond this broad overview of the [NiFe]-hydrogenase maturation process, biochemical and structural studies are revealing the fundamental underlying molecular mechanisms. Here, we review recent work illuminating how the accessory proteins contribute to the maturation of [NiFe]-hydrogenase and discuss some of the outstanding questions that remain to be resolved.
Collapse
Affiliation(s)
- Michael J Lacasse
- Department of Chemistry, University of Toronto , Toronto, Ontario, Canada M5S 3H6
| | - Deborah B Zamble
- Department of Chemistry, University of Toronto , Toronto, Ontario, Canada M5S 3H6.,Department of Biochemistry, University of Toronto , Toronto, Ontario, Canada M5S 1A8
| |
Collapse
|
27
|
|
28
|
Vinella D, Fischer F, Vorontsov E, Gallaud J, Malosse C, Michel V, Cavazza C, Robbe-Saule M, Richaud P, Chamot-Rooke J, Brochier-Armanet C, De Reuse H. Evolution of Helicobacter: Acquisition by Gastric Species of Two Histidine-Rich Proteins Essential for Colonization. PLoS Pathog 2015; 11:e1005312. [PMID: 26641249 PMCID: PMC4671568 DOI: 10.1371/journal.ppat.1005312] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 11/05/2015] [Indexed: 02/07/2023] Open
Abstract
Metal acquisition and intracellular trafficking are crucial for all cells and metal ions have been recognized as virulence determinants in bacterial pathogens. Virulence of the human gastric pathogen Helicobacter pylori is dependent on nickel, cofactor of two enzymes essential for in vivo colonization, urease and [NiFe] hydrogenase. We found that two small paralogous nickel-binding proteins with high content in Histidine (Hpn and Hpn-2) play a central role in maintaining non-toxic intracellular nickel content and in controlling its intracellular trafficking. Measurements of metal resistance, intracellular nickel contents, urease activities and interactomic analysis were performed. We observed that Hpn acts as a nickel-sequestration protein, while Hpn-2 is not. In vivo, Hpn and Hpn-2 form homo-multimers, interact with each other, Hpn interacts with the UreA urease subunit while Hpn and Hpn-2 interact with the HypAB hydrogenase maturation proteins. In addition, Hpn-2 is directly or indirectly restricting urease activity while Hpn is required for full urease activation. Based on these data, we present a model where Hpn and Hpn-2 participate in a common pathway of controlled nickel transfer to urease. Using bioinformatics and top-down proteomics to identify the predicted proteins, we established that Hpn-2 is only expressed by H. pylori and its closely related species Helicobacter acinonychis. Hpn was detected in every gastric Helicobacter species tested and is absent from the enterohepatic Helicobacter species. Our phylogenomic analysis revealed that Hpn acquisition was concomitant with the specialization of Helicobacter to colonization of the gastric environment and the duplication at the origin of hpn-2 occurred in the common ancestor of H. pylori and H. acinonychis. Finally, Hpn and Hpn-2 were found to be required for colonization of the mouse model by H. pylori. Our data show that during evolution of the Helicobacter genus, acquisition of Hpn and Hpn-2 by gastric Helicobacter species constituted a decisive evolutionary event to allow Helicobacter to colonize the hostile gastric environment, in which no other bacteria persistently thrives. This acquisition was key for the emergence of one of the most successful bacterial pathogens, H. pylori. Helicobacter pylori is a bacterium that persistently colonizes the stomach of half of the human population. Infection by H. pylori is associated with gastritis, peptic ulcer disease and adenocarcinoma. To resist gastric acidity and proliferate in the stomach, H. pylori relies on urease, an enzyme that contains a nickel-metallocenter at its active site. Thus, nickel is a virulence determinant for H. pylori. Our aim is to characterize how H. pylori controls the intracellular nickel concentration to avoid toxicity, which protein partners are involved, and how they impact urease activity and virulence. We characterized two H. pylori proteins, Hpn and Hpn-2 that are rich in Histidine residues. We demonstrated that Hpn is involved in nickel sequestration, that the two proteins interact with each other and that their combined activities participate in a nickel transfer pathway to urease. Hpn is only expressed in gastric Helicobacter species able to colonize the stomach and Hpn-2 is restricted to the H. pylori and its close relative H. acinonychis. We found that both proteins are essential for colonization of a mouse model by H. pylori. We conclude that during evolution, the acquisition of Hpn and Hpn-2 by gastric Helicobacter species was decisive for their capacity to colonize the stomach.
Collapse
Affiliation(s)
- Daniel Vinella
- Institut Pasteur, Département de Microbiologie, Unité Pathogenèse de Helicobacter, ERL CNRS 3526, Paris, France
| | - Frédéric Fischer
- Institut Pasteur, Département de Microbiologie, Unité Pathogenèse de Helicobacter, ERL CNRS 3526, Paris, France
| | - Egor Vorontsov
- Institut Pasteur, Département de Biologie Structurale et Chimie, Unité Spectrométrie de Masse Structurale et Protéomique, CNRS UMR 3528, Paris, France
| | - Julien Gallaud
- Institut Pasteur, Département de Microbiologie, Unité Pathogenèse de Helicobacter, ERL CNRS 3526, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
| | - Christian Malosse
- Institut Pasteur, Département de Biologie Structurale et Chimie, Unité Spectrométrie de Masse Structurale et Protéomique, CNRS UMR 3528, Paris, France
| | - Valérie Michel
- 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
| | - Pierre Richaud
- CEA, DSV, IBEB, SBVME and CNRS, UMR 7265 Biol Veget & Microbiol Environ, Saint-Paul-lez-Durance, France and Aix Marseille Université, BVME UMR7265, Marseille, France
| | - Julia Chamot-Rooke
- Institut Pasteur, Département de Biologie Structurale et Chimie, Unité Spectrométrie de Masse Structurale et Protéomique, CNRS UMR 3528, Paris, France
| | - Céline Brochier-Armanet
- Université de Lyon, Université Lyon 1, CNRS, UMR5558, Laboratoire de Biométrie et Biologie Evolutive, Villeurbanne, France
| | - Hilde De Reuse
- Institut Pasteur, Département de Microbiologie, Unité Pathogenèse de Helicobacter, ERL CNRS 3526, Paris, France
- * E-mail:
| |
Collapse
|
29
|
Zhu Y, Chen M, Gong Y, Liu Z, Li A, Kang D, Han F, Liu J, Liu J, Yuan Y. Helicobacter pylori FKBP-type PPIase promotes gastric epithelial cell proliferation and anchorage-independent growth through activation of ERK-mediated mitogenic signaling pathway. FEMS Microbiol Lett 2015; 362:fnv023. [DOI: 10.1093/femsle/fnv023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
30
|
Peters JW, Schut GJ, Boyd ES, Mulder DW, Shepard EM, Broderick JB, King PW, Adams MWW. [FeFe]- and [NiFe]-hydrogenase diversity, mechanism, and maturation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:1350-69. [PMID: 25461840 DOI: 10.1016/j.bbamcr.2014.11.021] [Citation(s) in RCA: 286] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 11/10/2014] [Accepted: 11/16/2014] [Indexed: 11/29/2022]
Abstract
The [FeFe]- and [NiFe]-hydrogenases catalyze the formal interconversion between hydrogen and protons and electrons, possess characteristic non-protein ligands at their catalytic sites and thus share common mechanistic features. Despite the similarities between these two types of hydrogenases, they clearly have distinct evolutionary origins and likely emerged from different selective pressures. [FeFe]-hydrogenases are widely distributed in fermentative anaerobic microorganisms and likely evolved under selective pressure to couple hydrogen production to the recycling of electron carriers that accumulate during anaerobic metabolism. In contrast, many [NiFe]-hydrogenases catalyze hydrogen oxidation as part of energy metabolism and were likely key enzymes in early life and arguably represent the predecessors of modern respiratory metabolism. Although the reversible combination of protons and electrons to generate hydrogen gas is the simplest of chemical reactions, the [FeFe]- and [NiFe]-hydrogenases have distinct mechanisms and differ in the fundamental chemistry associated with proton transfer and control of electron flow that also help to define catalytic bias. A unifying feature of these enzymes is that hydrogen activation itself has been restricted to one solution involving diatomic ligands (carbon monoxide and cyanide) bound to an Fe ion. On the other hand, and quite remarkably, the biosynthetic mechanisms to produce these ligands are exclusive to each type of enzyme. Furthermore, these mechanisms represent two independent solutions to the formation of complex bioinorganic active sites for catalyzing the simplest of chemical reactions, reversible hydrogen oxidation. As such, the [FeFe]- and [NiFe]-hydrogenases are arguably the most profound case of convergent evolution. This article is part of a Special Issue entitled: Fe/S proteins: Analysis, structure, function, biogenesis and diseases.
Collapse
Affiliation(s)
- John W Peters
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA.
| | - Gerrit J Schut
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Eric S Boyd
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59717, USA
| | - David W Mulder
- Biosciences Center, National Renewable Energy Laboratory, Golden, CO 80401, USA
| | - Eric M Shepard
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA
| | - Joan B Broderick
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA
| | - Paul W King
- Biosciences Center, National Renewable Energy Laboratory, Golden, CO 80401, USA
| | - Michael W W Adams
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
31
|
Khalilpour A, Osman S, Yunus MH, Santhanam A, Vellasamy N, Noordin R. Helicobacter pylori recombinant UreG protein: cloning, expression, and assessment of its seroreactivity. BMC Res Notes 2014; 7:809. [PMID: 25406411 PMCID: PMC4246484 DOI: 10.1186/1756-0500-7-809] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 10/31/2014] [Indexed: 01/12/2023] Open
Abstract
Background Helicobacter pylori is a human pathogen and during the process of infection, antigens from the bacterium elicit strong host humoral immune responses. In our previous report, native H. pylori UreG protein showed good reactivity with sera from H. pylori patients. This study was aimed at producing the recombinant form of the protein (rUreG) and determining its seroreactivities. Methods The coding sequence of H. pylori UreG was cloned and the recombinant protein expressed and purified by affinity chromatography using nickel nitrilotriacetic acid (Ni-NTA) resin. The antigenicity of rUreG to detect H. pylori specific antibodies was determined by western blot, using HRP-conjugated anti-human IgG and IgA antibodies as probes. A total of 70 sera, comprising 30 positive and 40 control serum samples, were used. The positive sera were from culture-positive H. pylori-infected patients with duodenal ulcers, gastric ulcers, or gastritis. The control sera comprised three types of samples without detectable H. pylori antibodies, i.e. healthy individuals (with no history of gastric disorders) (n = 10); patients who attended an endoscopy clinic (because of gastrointestinal complaints) but were H. pylori culture negative (n = 20); and people with other diseases (n = 10). Additionally, hyperimmune mice serum against rUreG was raised and tested with the native and recombinant UreG protein. Results The ureG gene fragment was successfully cloned and expressed in both soluble and insoluble forms. Western blots on rUreG protein showed 70% (21/30) and 60% (18/30) reactivity with patients’ sera when probed with HRP-conjugated anti-human IgG and IgA antibodies, respectively; and the combination of the IgG and IgA western blots showed reactivity of 83.3% (25/30). By comparison, 97.5% and 92.5% of the control sera showed no reactivity when probed with HRP-conjugated anti-human IgG and IgA antibodies, respectively. Both the H. pylori lysate antigen and rUreG protein displayed a distinctive band at the expected molecular weight when probed with the hyperimmune mice serum. Conclusion The rUreG protein was successfully cloned and expressed and showed good reactivity with H. pylori culture-positive patients’ sera and no reactivity with most control sera. Thus, the diagnostic potential of this recombinant protein merits further investigation.
Collapse
Affiliation(s)
| | | | | | | | | | - Rahmah Noordin
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia.
| |
Collapse
|
32
|
Rowinska-Zyrek M, Zakrzewska-Czerwinska J, Zawilak-Pawlik A, Kozlowski H. Ni²⁺ chemistry in pathogens--a possible target for eradication. Dalton Trans 2014; 43:8976-8989. [PMID: 24781528 DOI: 10.1039/c4dt00421c] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
The survival of all urease and/or hydrogenase containing pathogens depends on the proper homeostasis of nickel. In the scope of this perspectives paper, details of Ni(2+) metabolism of Helicobacter pylori, a widespread stomach-ulcer causing bacterium, are described. Nickel binding proteins and thermodynamics of such metal complexes are discussed in detail and special focus is given to potential nickel binding sequences in this metal's chaperones and regulators. A list of potential Ni(2+) binding sites in various pathogens is presented, which points out numerous examples of nickel interactions that still need to be understood.
Collapse
|
33
|
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
|
34
|
Häuser R, Ceol A, Rajagopala SV, Mosca R, Siszler G, Wermke N, Sikorski P, Schwarz F, Schick M, Wuchty S, Aloy P, Uetz P. A second-generation protein-protein interaction network of Helicobacter pylori. Mol Cell Proteomics 2014; 13:1318-29. [PMID: 24627523 DOI: 10.1074/mcp.o113.033571] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Helicobacter pylori infections cause gastric ulcers and play a major role in the development of gastric cancer. In 2001, the first protein interactome was published for this species, revealing over 1500 binary protein interactions resulting from 261 yeast two-hybrid screens. Here we roughly double the number of previously published interactions using an ORFeome-based, proteome-wide yeast two-hybrid screening strategy. We identified a total of 1515 protein-protein interactions, of which 1461 are new. The integration of all the interactions reported in H. pylori results in 3004 unique interactions that connect about 70% of its proteome. Excluding interactions of promiscuous proteins we derived from our new data a core network consisting of 908 interactions. We compared our data set to several other bacterial interactomes and experimentally benchmarked the conservation of interactions using 365 protein pairs (interologs) of E. coli of which one third turned out to be conserved in both species.
Collapse
Affiliation(s)
- Roman Häuser
- German Cancer Research Center (Deutsches Krebsforschungszentrum), Technologiepark 3, Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Cheng T, Li H, Yang X, Xia W, Sun H. Interaction of SlyD with HypB of Helicobacter pylori facilitates nickel trafficking. Metallomics 2014; 5:804-7. [PMID: 23708681 DOI: 10.1039/c3mt00014a] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
SlyD from Helicobacter pylori interacts with the [NiFe] hydrogenase accessory protein HypB through its IF domain. HpSlyD delivers Ni(2+) to HpHypB, leading to the enhancement of GTPase activity of HpHypB and implying the facilitation of Ni(2+) delivery from HpHypB to [NiFe] hydrogenase.
Collapse
Affiliation(s)
- Tianfan Cheng
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, P. R. China
| | | | | | | | | |
Collapse
|
36
|
de Reuse H, Vinella D, Cavazza C. Common themes and unique proteins for the uptake and trafficking of nickel, a metal essential for the virulence of Helicobacter pylori. Front Cell Infect Microbiol 2013; 3:94. [PMID: 24367767 PMCID: PMC3856676 DOI: 10.3389/fcimb.2013.00094] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 11/21/2013] [Indexed: 12/22/2022] Open
Abstract
Nickel is a virulence determinant for the human gastric pathogen Helicobacter pylori. Indeed, H. pylori possesses two nickel-enzymes that are essential for in vivo colonization, [NiFe] hydrogenase and urease, an abundant virulence factor that contains 24 nickel ions per active complex. Because of these two enzymes, survival of H. pylori relies on an important supply of nickel, implying a tight control of its distribution and storage. In this review, we will present the pathways of activation of the nickel enzymes as well as original mechanisms found in H. pylori for the uptake, trafficking and distribution of nickel between the two enzymes. These include (i) an outer-membrane nickel uptake system, the FrpB4 TonB-dependent transporter, (ii) overlapping protein complexes and interaction networks involved in nickel trafficking and distribution between urease and hydrogenase and, (iii) Helicobacter specific nickel-binding proteins that are involved in nickel storage and can play the role of metallo-chaperones. Finally, we will discuss the implication of the nickel trafficking partners in virulence and propose them as novel therapeutic targets for treatments against H. pylori infection.
Collapse
Affiliation(s)
- Hilde de Reuse
- Unité Pathogenèse de Helicobacter, Département de Microbiologie, Institut Pasteur, ERL CNRS 3526 Paris, France
| | - Daniel Vinella
- Unité Pathogenèse de Helicobacter, Département de Microbiologie, Institut Pasteur, ERL CNRS 3526 Paris, France
| | - Christine Cavazza
- Metalloproteins Group, Institut de Biologie Structurale Jean-Pierre Ebel, UMR 5075, CEA, CNRS, Université Joseph Fourier-Grenoble 1 Grenoble, France
| |
Collapse
|
37
|
Structure of UreG/UreF/UreH complex reveals how urease accessory proteins facilitate maturation of Helicobacter pylori urease. PLoS Biol 2013; 11:e1001678. [PMID: 24115911 PMCID: PMC3792862 DOI: 10.1371/journal.pbio.1001678] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 08/29/2013] [Indexed: 11/19/2022] Open
Abstract
Structural and biochemical study of urease accessory protein complex provides mechanistic insights into the delivery of nickel to metalloenzyme urease, an enzyme enabling the survival of Helicobacter pylori in the human stomach. Urease is a metalloenzyme essential for the survival of Helicobacter pylori in acidic gastric environment. Maturation of urease involves carbamylation of Lys219 and insertion of two nickel ions at its active site. This process requires GTP hydrolysis and the formation of a preactivation complex consisting of apo-urease and urease accessory proteins UreF, UreH, and UreG. UreF and UreH form a complex to recruit UreG, which is a SIMIBI class GTPase, to the preactivation complex. We report here the crystal structure of the UreG/UreF/UreH complex, which illustrates how UreF and UreH facilitate dimerization of UreG, and assembles its metal binding site by juxtaposing two invariant Cys66-Pro67-His68 metal binding motif at the interface to form the (UreG/UreF/UreH)2 complex. Interaction studies revealed that addition of nickel and GTP to the UreG/UreF/UreH complex releases a UreG dimer that binds a nickel ion at the dimeric interface. Substitution of Cys66 and His68 with alanine abolishes the formation of the nickel-charged UreG dimer. This nickel-charged UreG dimer can activate urease in vitro in the presence of the UreF/UreH complex. Static light scattering and atomic absorption spectroscopy measurements demonstrated that the nickel-charged UreG dimer, upon GTP hydrolysis, reverts to its monomeric form and releases nickel to urease. Based on our results, we propose a mechanism on how urease accessory proteins facilitate maturation of urease. Catalytic activities of many important enzymes depend upon metal cofactors. Ensuring each enzyme acquires the proper type of metal cofactor is essential to life. One such example is urease, which is a nickel containing metalloenzyme catalyzing the hydrolysis of urea to ammonia. The survival of Helicobacter pylori, a stomach ulcer–causing pathogen, in the human stomach depends on the ammonia released to neutralize gastric acid. In this study, we revealed the detail mechanism of how urease accessory proteins UreF, UreH, and UreG cooperate to couple GTP hydrolysis to deliver nickel to urease. UreF/UreH complex interacts with two molecules of GTPase UreG and assembles a metal binding site located at the interface between two UreG molecules. Nickel can induce GTP-dependent dimerization of UreG. This nickel-carrying UreG dimer together with UreF, UreH, and urease assemble into a protein complex. Upon stimulation of UreG GTPase activity by bicarbonate, UreG hydrolyses GTP and releases nickel into urease. Other nickel-delivering NTPases share similar properties with UreG; therefore, the nickel delivery mechanism described here is likely universally shared among these proteins.
Collapse
|
38
|
Abstract
AIM To investigate the biological activity of the H. pylori SlyD in vitro. METHODS Helicobacter pylori (H.pylori) slyD prokaryotic expression vector was carried out in Escherichia coli (E.coli), and recombination SlyD (rSlyD) was purified by immobilized metal affinity chromatography. The proliferation, apoptosis, invasion, transformation effects of rSlyD on AGS cells was detected by CCK-8, cell cycle, caspase-3 activity, matrigel invasion assay, and double-deck soft agar colony forming efficiency. In addition, the expressions of PCNA, KI-67, caspase-3, and MMP-9 were detected by western blot and immunofluorescence assay, respectively. RESULTS The CCK-8 assay revealed that cell proliferation was increased in a time and dose-dependent manner in AGS + rSlyD group compared with that of AGS or AGS + PBS group (p < .05). There are significant difference of PCNA and KI67 expressions among AGS, AGS + PBS, AGS + rSlyD groups (p < .05). Soft agar colony formation assay revealed the colony number (foci>100 μm) in AGS + rSlyD group was 26.3 ± 7.09, whereas 5.6 ± 1.15 in AGS and 5.0 ± 1.0 in AGS + PBS groups, respectively (p < .01). Colorimetric enzyme assay revealed the activity of caspase-3 was decreased to 31.45 ± 0.49 after treatment with rSlyD, whereas 55.5 ± 0.43 in AGS and 55.1 ± 0.25 in AGS + PBS group, respectively (p < .001). Similar caspase-3 expression also was confirmed by Western blot. The number of invasive cells in transwell chambers assay is 196.66 ± 40.41 in AGS + rSlyD group higher than 85 ± 22.9 in AGS or 81.66 ± 15.27 in AGS + PBS group, respectively (p < .001). The MMP-9 expression in AGS + rSlyD group was also higher than that of AGS or AGS + PBS group. CONCLUSION These results suggest that the HpSlyD may play an important role in disturbing cell proliferation, apoptosis, and enhancing cell transformation and invasion in the AGS cell line. HpSlyD might contribute to gastric pathogenicity in H.pylori-associated diseases.
Collapse
Affiliation(s)
- Dan Kang
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | | | | | | | | | | | | |
Collapse
|
39
|
Gaudin M, Krupovic M, Marguet E, Gauliard E, Cvirkaite-Krupovic V, Le Cam E, Oberto J, Forterre P. Extracellular membrane vesicles harbouring viral genomes. Environ Microbiol 2013; 16:1167-75. [DOI: 10.1111/1462-2920.12235] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 07/25/2013] [Accepted: 07/28/2013] [Indexed: 12/31/2022]
Affiliation(s)
- Marie Gaudin
- CNRS UMR 8621; Institut de Génétique et Microbiologie; Univ Paris-Sud; 91405 Orsay cedex France
| | - Mart Krupovic
- Biologie Moléculaire du Gène chez les Extrêmophiles; Département de Microbiologie; Institut Pasteur; 75724 Paris cedex France
| | - Evelyne Marguet
- CNRS UMR 8621; Institut de Génétique et Microbiologie; Univ Paris-Sud; 91405 Orsay cedex France
| | - Emilie Gauliard
- CNRS UMR 8621; Institut de Génétique et Microbiologie; Univ Paris-Sud; 91405 Orsay cedex France
| | | | - Eric Le Cam
- Signalisation; Noyaux et Innovations en Cancérologie; Interactions Moléculaires et Cancer; CNRS UMR 8126; Institut de Cancérologie Gustave Roussy 94805 Villejuif cedex France
| | - Jacques Oberto
- CNRS UMR 8621; Institut de Génétique et Microbiologie; Univ Paris-Sud; 91405 Orsay cedex France
| | - Patrick Forterre
- CNRS UMR 8621; Institut de Génétique et Microbiologie; Univ Paris-Sud; 91405 Orsay cedex France
- Biologie Moléculaire du Gène chez les Extrêmophiles; Département de Microbiologie; Institut Pasteur; 75724 Paris cedex France
| |
Collapse
|
40
|
Dieppedale J, Gesbert G, Ramond E, Chhuon C, Dubail I, Dupuis M, Guerrera IC, Charbit A. Possible links between stress defense and the tricarboxylic acid (TCA) cycle in Francisella pathogenesis. Mol Cell Proteomics 2013; 12:2278-92. [PMID: 23669032 PMCID: PMC3734585 DOI: 10.1074/mcp.m112.024794] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 05/01/2013] [Indexed: 12/16/2022] Open
Abstract
Francisella tularensis is a highly infectious bacterium causing the zoonotic disease tularemia. In vivo, this facultative intracellular bacterium survives and replicates mainly in the cytoplasm of infected cells. We have recently identified a genetic locus, designated moxR that is important for stress resistance and intramacrophage survival of F. tularensis. In the present work, we used tandem affinity purification coupled to mass spectrometry to identify in vivo interacting partners of three proteins encoded by this locus: the MoxR-like ATPase (FTL_0200), and two proteins containing motifs predicted to be involved in protein-protein interactions, bearing von Willebrand A (FTL_0201) and tetratricopeptide (FTL_0205) motifs. The three proteins were designated here for simplification, MoxR, VWA1, and TPR1, respectively. MoxR interacted with 31 proteins, including various enzymes. VWA1 interacted with fewer proteins, but these included the E2 component of 2-oxoglutarate dehydrogenase and TPR1. The protein TPR1 interacted with one hundred proteins, including the E1 and E2 subunits of both oxoglutarate and pyruvate dehydrogenase enzyme complexes, and their common E3 subunit. Remarkably, chromosomal deletion of either moxR or tpr1 impaired pyruvate dehydrogenase and oxoglutarate dehydrogenase activities, supporting the hypothesis of a functional role for the interaction of MoxR and TPR1 with these complexes. Altogether, this work highlights possible links between stress resistance and metabolism in F. tularensis virulence.
Collapse
Affiliation(s)
- Jennifer Dieppedale
- From the ‡Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche. 96 rue Didot 75993 Paris Cedex 14 – France
- §INSERM, U1002, Unité de Pathogénie des Infections Systémiques, Paris, France
| | - Gael Gesbert
- From the ‡Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche. 96 rue Didot 75993 Paris Cedex 14 – France
- §INSERM, U1002, Unité de Pathogénie des Infections Systémiques, Paris, France
| | - Elodie Ramond
- From the ‡Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche. 96 rue Didot 75993 Paris Cedex 14 – France
- §INSERM, U1002, Unité de Pathogénie des Infections Systémiques, Paris, France
| | - Cerina Chhuon
- From the ‡Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche. 96 rue Didot 75993 Paris Cedex 14 – France
- ¶Plateau Protéome Necker, PPN, IFR94, Université Paris-Descartes, Faculté de Médecine René Descartes, Paris 75015 France
| | - Iharilalao Dubail
- From the ‡Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche. 96 rue Didot 75993 Paris Cedex 14 – France
- §INSERM, U1002, Unité de Pathogénie des Infections Systémiques, Paris, France
| | - Marion Dupuis
- From the ‡Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche. 96 rue Didot 75993 Paris Cedex 14 – France
- §INSERM, U1002, Unité de Pathogénie des Infections Systémiques, Paris, France
| | - Ida Chiara Guerrera
- From the ‡Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche. 96 rue Didot 75993 Paris Cedex 14 – France
- ¶Plateau Protéome Necker, PPN, IFR94, Université Paris-Descartes, Faculté de Médecine René Descartes, Paris 75015 France
| | - Alain Charbit
- From the ‡Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche. 96 rue Didot 75993 Paris Cedex 14 – France
- §INSERM, U1002, Unité de Pathogénie des Infections Systémiques, Paris, France
| |
Collapse
|
41
|
Choi HP, Juarez S, Ciordia S, Fernandez M, Bargiela R, Albar JP, Mazumdar V, Anton BP, Kasif S, Ferrer M, Steffen M. Biochemical Characterization of Hypothetical Proteins from Helicobacter pylori. PLoS One 2013; 8:e66605. [PMID: 23825549 PMCID: PMC3688963 DOI: 10.1371/journal.pone.0066605] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 05/08/2013] [Indexed: 12/16/2022] Open
Abstract
The functional characterization of Open Reading Frames (ORFs) from sequenced genomes remains a bottleneck in our effort to understand microbial biology. In particular, the functional characterization of proteins with only remote sequence homology to known proteins can be challenging, as there may be few clues to guide initial experiments. Affinity enrichment of proteins from cell lysates, and a global perspective of protein function as provided by COMBREX, affords an approach to this problem. We present here the biochemical analysis of six proteins from Helicobacter pylori ATCC 26695, a focus organism in COMBREX. Initial hypotheses were based upon affinity capture of proteins from total cellular lysate using derivatized nano-particles, and subsequent identification by mass spectrometry. Candidate genes encoding these proteins were cloned and expressed in Escherichia coli, and the recombinant proteins were purified and characterized biochemically and their biochemical parameters compared with the native ones. These proteins include a guanosine triphosphate (GTP) cyclohydrolase (HP0959), an ATPase (HP1079), an adenosine deaminase (HP0267), a phosphodiesterase (HP1042), an aminopeptidase (HP1037), and new substrates were characterized for a peptidoglycan deacetylase (HP0310). Generally, characterized enzymes were active at acidic to neutral pH (4.0–7.5) with temperature optima ranging from 35 to 55°C, although some exhibited outstanding characteristics.
Collapse
Affiliation(s)
- Han-Pil Choi
- Dept of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
| | - Silvia Juarez
- Proteomic Facility, CNB-National Centre for Biotechnology, CSIC, Darwin 3, Madrid, Spain
| | - Sergio Ciordia
- Proteomic Facility, CNB-National Centre for Biotechnology, CSIC, Darwin 3, Madrid, Spain
| | - Marisol Fernandez
- Proteomic Facility, CNB-National Centre for Biotechnology, CSIC, Darwin 3, Madrid, Spain
| | - Rafael Bargiela
- Spanish National Research Council (CSIC), Institute of Catalysis, Madrid, Spain
| | - Juan P. Albar
- Proteomic Facility, CNB-National Centre for Biotechnology, CSIC, Darwin 3, Madrid, Spain
| | - Varun Mazumdar
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
| | - Brian P. Anton
- New England Biolabs, Ipswich, Massachusetts, United States of America
| | - Simon Kasif
- Dept of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
| | - Manuel Ferrer
- Spanish National Research Council (CSIC), Institute of Catalysis, Madrid, Spain
- * E-mail: (MS); (MF)
| | - Martin Steffen
- Dept of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
- Dept of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail: (MS); (MF)
| |
Collapse
|
42
|
Structure of the UreD-UreF-UreG-UreE complex in Helicobacter pylori: a model study. J Biol Inorg Chem 2013; 18:571-7. [PMID: 23661161 DOI: 10.1007/s00775-013-1002-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 04/23/2013] [Indexed: 10/26/2022]
Abstract
The molecular details of the protein complex formed by UreD, UreF, UreG, and UreE, accessory proteins for urease activation in the carcinogenic bacterium Helicobacter pylori, have been elucidated using computational modeling. The calculated structure of the complex supports the hypothesis of UreF acting as a GTPase activation protein that facilitates GTP hydrolysis by UreG during urease maturation, and provides a rationale for the design of new drugs against infections by ureolytic bacterial pathogens.
Collapse
|
43
|
Correia M, Michel V, Osório H, El Ghachi M, Bonis M, Boneca IG, De Reuse H, Matos AA, Lenormand P, Seruca R, Figueiredo C, Machado JC, Touati E. Crosstalk between Helicobacter pylori and gastric epithelial cells is impaired by docosahexaenoic acid. PLoS One 2013; 8:e60657. [PMID: 23577140 PMCID: PMC3618039 DOI: 10.1371/journal.pone.0060657] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 03/01/2013] [Indexed: 12/12/2022] Open
Abstract
H. pylori colonizes half of the world's population leading to gastritis, ulcers and gastric cancer. H. pylori strains resistant to antibiotics are increasing which raises the need for alternative therapeutic approaches. Docosahexaenoic acid (DHA) has been shown to decrease H. pylori growth and its associated-inflammation through mechanisms poorly characterized. We aimed to explore DHA action on H. pylori-mediated inflammation and adhesion to gastric epithelial cells (AGS) and also to identify bacterial structures affected by DHA. H. pylori growth and metabolism was assessed in liquid cultures. Bacterial adhesion to AGS cells was visualized by transmission electron microscopy and quantified by an Enzyme Linked Immunosorbent Assay. Inflammatory proteins were assessed by immunoblotting in infected AGS cells, previously treated with DHA. Bacterial total and outer membrane protein composition was analyzed by 2-dimensional gel electrophoresis. Concentrations of 100 µM of DHA decreased H. pylori growth, whereas concentrations higher than 250 µM irreversibly inhibited bacteria survival. DHA reduced ATP production and adhesion to AGS cells. AGS cells infected with DHA pre-treated H. pylori showed a 3-fold reduction in Interleukin-8 (IL-8) production and a decrease of COX2 and iNOS. 2D electrophoresis analysis revealed that DHA changed the expression of H. pylori outer membrane proteins associated with stress response and metabolism and modified bacterial lipopolysaccharide phenotype. As conclusions our results show that DHA anti-H. pylori effects are associated with changes of bacteria morphology and metabolism, and with alteration of outer membrane proteins composition, that ultimately reduce the adhesion of bacteria and the burden of H. pylori-related inflammation.
Collapse
Affiliation(s)
- Marta Correia
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Rua Dr. Roberto Frias s/n, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto, Al. Hernani Monteiro, Porto, Portugal
- Institut Pasteur, Unité de Pathogenèse de Helicobacter, Paris, France
| | - Valérie Michel
- Institut Pasteur, Unité de Pathogenèse de Helicobacter, Paris, France
| | - Hugo Osório
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Rua Dr. Roberto Frias s/n, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto, Al. Hernani Monteiro, Porto, Portugal
| | - Meriem El Ghachi
- Institut Pasteur, G5 Biologie et Génétique de la Paroi Bactérienne, Paris, France
- INSERM, Group AVENIR, 75724 Paris, France
| | - Mathilde Bonis
- Institut Pasteur, G5 Biologie et Génétique de la Paroi Bactérienne, Paris, France
- INSERM, Group AVENIR, 75724 Paris, France
| | - Ivo G. Boneca
- Institut Pasteur, G5 Biologie et Génétique de la Paroi Bactérienne, Paris, France
- INSERM, Group AVENIR, 75724 Paris, France
| | - Hilde De Reuse
- Institut Pasteur, Unité de Pathogenèse de Helicobacter, Paris, France
| | - António A. Matos
- Anatomia Patológica, Centro Hospitalar de Lisboa Central, Lisboa, Portugal
- Centro de Estudos do Ambiente e do Mar (CESAM/FCUL) – Faculdade de Ciências da Universidade de Lisboa and Centro de Investigação Interdisciplinar Egas Moniz (CiiEM) Campo Grande, Lisboa, Portugal
| | | | - Raquel Seruca
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Rua Dr. Roberto Frias s/n, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto, Al. Hernani Monteiro, Porto, Portugal
| | - Ceu Figueiredo
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Rua Dr. Roberto Frias s/n, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto, Al. Hernani Monteiro, Porto, Portugal
| | - Jose Carlos Machado
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Rua Dr. Roberto Frias s/n, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto, Al. Hernani Monteiro, Porto, Portugal
| | - Eliette Touati
- Institut Pasteur, Unité de Pathogenèse de Helicobacter, Paris, France
- * E-mail:
| |
Collapse
|
44
|
Abstract
Metalloenzymes often require elaborate metallocenter assembly systems to create functional active sites. The medically important dinuclear nickel enzyme urease provides an excellent model for studying metallocenter assembly. Nickel is inserted into the urease active site in a GTP-dependent process with the assistance of UreD/UreH, UreE, UreF, and UreG. These accessory proteins orchestrate apoprotein activation by delivering the appropriate metal, facilitating protein conformational changes, and possibly providing a requisite post-translational modification. The activation mechanism and roles of each accessory protein in urease maturation are the subject of ongoing studies, with the latest findings presented in this minireview.
Collapse
Affiliation(s)
- Mark A Farrugia
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | | | | |
Collapse
|
45
|
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: 1.0] [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
|
46
|
Gaudin M, Gauliard E, Schouten S, Houel-Renault L, Lenormand P, Marguet E, Forterre P. Hyperthermophilic archaea produce membrane vesicles that can transfer DNA. ENVIRONMENTAL MICROBIOLOGY REPORTS 2013; 5:109-116. [PMID: 23757139 DOI: 10.1111/j.1758-2229.2012.00348.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Thermococcales are hyperthermophilic archaea found in deep-sea hydrothermal vents. They have been recently reported to produce membrane vesicles (MVs) into their culture medium. Here, we have characterized the mode of production and determined the biochemical composition of MVs from two species of Thermococcales, Thermococcus gammatolerans and Thermococcus kodakaraensis. We observed that MVs are produced by a budding process from the cell membrane reminiscent of ectosome (microparticle) formation in eukaryotes. MVs and cell membranes from the same species have a similar protein and lipid composition, confirming that MVs are produced from cell membranes. The major protein present in cell membranes and MVs of both species is the oligopeptide binding protein OppA. This protein is also abundant in MVs from cells grown in minimal medium, suggesting that OppA could be involved in processes other than peptides scavenging. We have previously shown that MVs from Thermococcales harbour DNA and protect DNA against thermodegradation. Here, we show that T. kodakaraensis cells transformed with the shuttle plasmid pLC70 release MVs harbouring this plasmid. Notably, these MVs can be used to transfer pLC70 into plasmid-free cells, suggesting that MVs could be involved in DNA transfer between cells at high temperature.
Collapse
Affiliation(s)
- Marie Gaudin
- Institut de Génétique et Microbiologie, Univ Paris-Sud, CNRS UMR8621, 91405 Orsay Cedex, France.
| | | | | | | | | | | | | |
Collapse
|
47
|
Schleker S, Ananthasubramanian S, Klein‐Seetharaman J, Ganapathiraju MK. Prediction of Intra‐ and Interspecies Protein–Protein Interactions Facilitating Systems Biology Studies. METHODS AND PRINCIPLES IN MEDICINAL CHEMISTRY 2013:21-53. [DOI: 10.1002/9783527648207.ch2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
48
|
Cun S, Lai YT, Chang YY, Sun H. Structure-oriented bioinformatic approach exploring histidine-rich clusters in proteins. Metallomics 2013; 5:904-12. [DOI: 10.1039/c3mt00026e] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
49
|
|
50
|
Redko Y, Aubert S, Stachowicz A, Lenormand P, Namane A, Darfeuille F, Thibonnier M, De Reuse H. A minimal bacterial RNase J-based degradosome is associated with translating ribosomes. Nucleic Acids Res 2012; 41:288-301. [PMID: 23093592 PMCID: PMC3592473 DOI: 10.1093/nar/gks945] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Protein complexes directing messenger RNA (mRNA) degradation are present in all kingdoms of life. In Escherichia coli, mRNA degradation is performed by an RNA degradosome organized by the major ribonuclease RNase E. In bacteria lacking RNase E, the existence of a functional RNA degradosome is still an open question. Here, we report that in the bacterial pathogen Helicobacter pylori, RNA degradation is directed by a minimal RNA degradosome consisting of Hp-RNase J and the only DExD-box RNA helicase of H. pylori, RhpA. We show that the protein complex promotes faster degradation of double-stranded RNA in vitro in comparison with Hp-RNase J alone. The ATPase activity of RhpA is stimulated in the presence of Hp-RNase J, demonstrating that the catalytic capacity of both partners is enhanced upon interaction. Remarkably, both proteins are associated with translating ribosomes and not with individual 30S and 50S subunits. Moreover, Hp-RNase J is not recruited to ribosomes to perform rRNA maturation. Together, our findings imply that in H. pylori, the mRNA-degrading machinery is associated with the translation apparatus, a situation till now thought to be restricted to eukaryotes and archaea.
Collapse
Affiliation(s)
- Yulia Redko
- Département de Microbiologie, Unité Pathogenèse de Helicobacter, Institut Pasteur, 75724 Paris Cedex 15, France
| | | | | | | | | | | | | | | |
Collapse
|