1
|
Davidov Y, Tejman-Yarden N, Robinson A, Rahav G, Nissan I. Enterobactin and salmochelin S4 inhibit the growth of Staphylococcus aureus. Front Cell Infect Microbiol 2025; 15:1456046. [PMID: 40110026 PMCID: PMC11919883 DOI: 10.3389/fcimb.2025.1456046] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 02/12/2025] [Indexed: 03/22/2025] Open
Abstract
There is increasing demand for novel antimicrobial agents to tackle the antimicrobial resistance crisis. Here we report that two Enterobacteriaceae-produced siderophores, enterobactin and salmochelin S4, inhibit the growth of Staphylococcus aureus isolates, including methicillin-resistance S. aureus (MRSA) clinical isolates. The IC50 for different S. aureus isolates were 2-5 µM for salmochelin S4 and 5-10 µM for enterobactin. This inhibitory activity was partially repressed by adding Fe+3. These siderophores also inhibited the growth of Enterococcus strains, including vancomycin-resistant enterococci (VRE) clinical isolates, though less effectively than for S. aureus. The growth of various Gram-negative bacteria was barely affected by these siderophores. These results shed new light on the role of enterobactin and salmochelin in bacterial physiology and ecology and have potential for the development of novel strategies to combat the rapid rise of multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Yaacov Davidov
- Infectious Disease Unit, Sheba Medical Center, Tel Hashomer, Israel
| | - Noa Tejman-Yarden
- Department of Laboratories, Public Health Directorate, Ministry of Health, Jerusalem, Israel
| | - Ari Robinson
- Infectious Disease Unit, Sheba Medical Center, Tel Hashomer, Israel
| | - Galia Rahav
- Infectious Disease Unit, Sheba Medical Center, Tel Hashomer, Israel
- Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Israel Nissan
- Infectious Disease Unit, Sheba Medical Center, Tel Hashomer, Israel
- Department of Avian Diseases, Kimron Veterinary Institute, Beit Dagan, Israel
| |
Collapse
|
2
|
Kumar R, Singh A, Srivastava A. Xenosiderophores: bridging the gap in microbial iron acquisition strategies. World J Microbiol Biotechnol 2025; 41:69. [PMID: 39939429 DOI: 10.1007/s11274-025-04287-w] [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] [Received: 12/03/2024] [Accepted: 02/04/2025] [Indexed: 02/14/2025]
Abstract
Microorganisms acquire iron from surrounding environment through specific iron chelators known as siderophores that can be of self-origin or synthesized by neighboring microbes. The latter are termed as xenosiderophores. The acquired iron supports their growth, survival, and pathogenesis. Various microorganisms possess the ability to utilize xenosiderophores, a mechanism popularly termed as 'siderophore piracy' besides synthesizing their own siderophores. This adaptability allows microorganisms to conserve energy by reducing the load of siderogenesis. Owing to the presence of xenosiderophore transport machinery, these microbial systems can be used for targeting antibiotics-siderophore conjugates to control pathogenesis and combat antimicrobial resistance. This review outlines the significance of xenosiderophore utilization for growth, stress management and virulence. Siderogenesis and the molecular mechanism of its uptake by related organisms have been discussed vividly. It focuses on potential applications like disease diagnostics, drug delivery, and combating antibiotic resistance. In brief, this review highlights the importance of xenosiderophores projecting them beyond their role as mere iron chelators.
Collapse
Affiliation(s)
- Ravinsh Kumar
- Department of Life Science, School of Earth, Biological and Environmental Sciences, Central University of South Bihar, Gaya, Bihar, India
| | - Ashutosh Singh
- Department of Life Science, School of Earth, Biological and Environmental Sciences, Central University of South Bihar, Gaya, Bihar, India
| | - Amrita Srivastava
- Department of Life Science, School of Earth, Biological and Environmental Sciences, Central University of South Bihar, Gaya, Bihar, India.
| |
Collapse
|
3
|
Fernandez-Perez J, Senoo A, Caaveiro JMM, Nakakido M, de Vega S, Nakagawa I, Tsumoto K. Structural basis for the ligand promiscuity of the hydroxamate siderophore binding protein FtsB from Streptococcus pyogenes. Structure 2024; 32:2410-2421.e3. [PMID: 39395422 DOI: 10.1016/j.str.2024.09.018] [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] [Received: 03/05/2024] [Revised: 08/04/2024] [Accepted: 09/17/2024] [Indexed: 10/14/2024]
Abstract
Pathogenic bacteria must secure the uptake of nutritional metals such as iron for their growth, making their import systems attractive targets for the development of new antimicrobial modalities. In the pathogenic bacterium Streptococcus pyogenes, the iron uptake system FtsABCD transports iron encapsulated by siderophores of the hydroxamate class. However, the inability of S. pyogenes to produce these metabolites makes the biological and clinical relevance of this route unresolved. Herein, we demonstrated that the periplasmic binding protein FtsB recognizes not only the hydroxamate siderophore ferrichrome, as previously documented, but also ferrioxamine E (FOE), ferrioxamine B (FOB), and bisucaberin (BIS), each of them with high affinity (nM level). Up to seven aromatic residues in the binding pocket accommodate the variable backbones of the different siderophores through CH-π interactions, explaining ligand promiscuity. Collectively, our observations revealed how S. pyogenes exploits the diverse xenosiderophores produced by other microorganisms as iron sources to secure this precious nutrient.
Collapse
Affiliation(s)
- Jorge Fernandez-Perez
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Akinobu Senoo
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan; Laboratory of Protein Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Jose M M Caaveiro
- Laboratory of Protein Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | - Makoto Nakakido
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Susana de Vega
- Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Ichiro Nakagawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kouhei Tsumoto
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan; Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan; Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.
| |
Collapse
|
4
|
Li P, Schulte J, Wurpts G, Hornef MW, Wolz C, Yazdi AS, Burian M. Transcriptional Profiling of Staphylococcus aureus during the Transition from Asymptomatic Nasal Colonization to Skin Colonization/Infection in Patients with Atopic Dermatitis. Int J Mol Sci 2024; 25:9165. [PMID: 39273114 PMCID: PMC11394835 DOI: 10.3390/ijms25179165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/18/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024] Open
Abstract
Staphylococcus aureus acts both as a colonizing commensal bacterium and invasive pathogen. Nasal colonization is associated with an increased risk of infection caused by the identical strain. In patients with atopic dermatitis (AD), the degree of S. aureus colonization is associated with the severity of the disease. Here, we comparatively analyzed the in vivo transcriptional profile of S. aureus colonizing the nose and non-diseased skin (non-lesional skin) as opposed to the diseased skin (lesional skin-defined here as infection) of 12 patients with AD. The transcriptional profile during the asymptomatic colonization of the nose closely resembled that of the lesional skin samples for many of the genes studied, with an elevated expression of the genes encoding adhesion-related proteins and proteases. In addition, the genes that modify and remodel the cell wall and encode proteins that facilitate immune evasion showed increased transcriptional activity. Notably, in a subgroup of patients, the global virulence regulator Agr (accessory gene regulator) and downstream target genes were inactive during nasal colonization but upregulated in the lesional and non-lesional skin samples. Taken together, our results demonstrate a colonization-like transcriptional profile on diseased skin and suggest a role for the peptide quorum sensing system Agr during the transition from asymptomatic nasal colonization to skin colonization/infection.
Collapse
Affiliation(s)
- Peijuan Li
- Department of Dermatology and Allergology, RWTH University Hospital Aachen, D-5207 Aachen, Germany
| | - Julia Schulte
- Department of Dermatology and Allergology, RWTH University Hospital Aachen, D-5207 Aachen, Germany
| | - Gerda Wurpts
- Department of Dermatology and Allergology, RWTH University Hospital Aachen, D-5207 Aachen, Germany
| | - Mathias W Hornef
- Institute of Medical Microbiology, RWTH University Hospital Aachen, D-52074 Aachen, Germany
| | - Christiane Wolz
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tuebingen, D-72076 Tuebingen, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tuebingen, D-72076 Tuebingen, Germany
| | - Amir S Yazdi
- Department of Dermatology and Allergology, RWTH University Hospital Aachen, D-5207 Aachen, Germany
| | - Marc Burian
- Department of Dermatology and Allergology, RWTH University Hospital Aachen, D-5207 Aachen, Germany
| |
Collapse
|
5
|
Merrick CE, Gulati NM, Wencewicz TA. Siderophore-dependent ferrichelatases. Methods Enzymol 2024; 702:281-315. [PMID: 39155116 DOI: 10.1016/bs.mie.2024.06.015] [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: 08/20/2024]
Abstract
Iron is a crucial secondary metabolite for bacterial proliferation, but its bioavailability under infection conditions is limited by the low solubility of ferric ion and the host's ability to sequester iron by protein chelation. In these iron limiting conditions, bacteria produce and secrete low molecular weight ferric ion chelators, siderophores, to scavenge host iron. Iron bound siderophores are recognized by surface displayed receptors and internalized by active transport preceding the liberation of the iron payload by reduction or cleavage of the siderophore. The traditional paradigms surrounding the interactions between siderophores and their corresponding receptors have relied on canonical protein-ligand binding models that do not accurately reflect the conditions experienced by siderophore binding proteins (SBPs). Research by the Raymond group suggested that a ligand displacement model does not fully describe the role of SBPs in siderophore transport where the ferric ion can be shuttled between siderophore molecules during the transport process. This work inspired further research by the Wencewicz group, which demonstrated that the Staphylococcus aureus SBP FhuD2 can catalyze the transfer of iron from the biological iron source holo-transferrin to a SBP bound iron-free siderophore. The discovery of this ferrichelatase activity represents a novel mechanism of receptor mediated active transport which raises the question: is ferrichelatase activity a unique feature of FhuD2 or a previously unappreciated hallmark of SBPs? This chapter highlights a series of protocols for the general functional characterization of SBPs and methodologies to assay ferrichelatase activity with the hopes of providing the tools to answer this question.
Collapse
Affiliation(s)
- C E Merrick
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, United States
| | - N M Gulati
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, United States
| | - T A Wencewicz
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, United States.
| |
Collapse
|
6
|
Liu S, Laman P, Jensen S, van der Wel NN, Kramer G, Zaat SA, Brul S. Isolation and characterization of persisters of the pathogenic microorganism Staphylococcus aureus. iScience 2024; 27:110002. [PMID: 38868179 PMCID: PMC11166702 DOI: 10.1016/j.isci.2024.110002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/14/2024] [Accepted: 05/14/2024] [Indexed: 06/14/2024] Open
Abstract
The presence of antibiotic persisters is one of the leading causes of recurrent and chronic diseases. One challenge in mechanistic research on persisters is the enrichment of pure persisters. In this work, we validated a proposed method to isolate persisters with notorious Staphylococcus aureus cultures. With this, we analyzed the proteome profile of pure persisters and revealed the distinct mechanisms associated with vancomycin and enrofloxacin induced persisters. Furthermore, morphological and metabolic characterizations were performed, indicating further differences between these two persister populations. Finally, we assessed the effect of ATP repression, protein synthesis inhibition, and reactive oxygen species (ROS) level on persister formation. In conclusion, this work provides a comprehensive understanding of S. aureus vancomycin and enrofloxacin induced persisters, facilitating a better mechanistic understanding of persisters and the development of effective strategies to combat them.
Collapse
Affiliation(s)
- Shiqi Liu
- Department of Molecular Biology and Microbial Food Safety, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, the Netherlands
| | - Paul Laman
- Department of Molecular Biology and Microbial Food Safety, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, the Netherlands
| | - Sean Jensen
- Department of Molecular Biology and Microbial Food Safety, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, the Netherlands
| | - Nicole N. van der Wel
- Department of Medical Biology, Electron Microscopy Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Gertjan Kramer
- Department of Mass Spectrometry of Biomolecules, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, the Netherlands
| | - Sebastian A.J. Zaat
- Department of Medical Microbiology, Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Stanley Brul
- Department of Molecular Biology and Microbial Food Safety, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, the Netherlands
| |
Collapse
|
7
|
Subsomwong P, Teng W, Ishiai T, Narita K, Sukchawalit R, Nakane A, Asano K. Extracellular vesicles from Staphylococcus aureus promote the pathogenicity of Pseudomonas aeruginosa. Microbiol Res 2024; 281:127612. [PMID: 38244256 DOI: 10.1016/j.micres.2024.127612] [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] [Received: 10/13/2023] [Revised: 12/26/2023] [Accepted: 01/09/2024] [Indexed: 01/22/2024]
Abstract
Co-infections with Staphylococcus aureus and Pseudomonas aeruginosa are common in patients with chronic wounds, but little is known about their synergistic effect mediated by extracellular vesicles (EVs). In this study, we investigated the effect of EVs derived from S. aureus (SaEVs) on the pathogenicity of P. aeruginosa. By using lipophilic dye, we could confirm the fusion between SaEV and P. aeruginosa membranes. However, SaEVs did not alter the growth and antibiotic susceptible pattern of P. aeruginosa. Differential proteomic analysis between SaEV-treated and non-treated P. aeruginosa was performed, and the results revealed that lipopolysaccharide (LPS) biosynthesis protein in P. aeruginosa significantly increased after SaEV-treatment. Regarding this result, we also found that SaEVs promoted LPS production, biofilm formation, and expression of polysaccharide polymerization-related genes in P. aeruginosa. Furthermore, invasion of epithelial cells by SaEV-pretreated P. aeruginosa was enhanced. On the other hand, uptake of P. aeruginosa by RAW 264.7 macrophages was impaired after pretreatment P. aeruginosa with SaEVs. Proteomic analysis SaEVs revealed that SaEVs contain the proteins involving in host cell colonization, inhibition of host immune response, anti-phagocytosis of the macrophages, and protein translocation and iron uptake of S. aureus. In conclusion, SaEVs serve as a mediator that promote P. aeruginosa pathogenicity by enhancing LPS biosynthesis, biofilm formation, epithelial cell invasion, and macrophage uptake impairment.
Collapse
Affiliation(s)
- Phawinee Subsomwong
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Wei Teng
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Takahito Ishiai
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Kouji Narita
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan; Institute for Animal Experimentation, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Rojana Sukchawalit
- Laboratory of Biotechnology, Chulabhorn Research Institute, Lak Si, Bangkok, Thailand
| | - Akio Nakane
- Department of Biopolymer and Health Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Krisana Asano
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan; Department of Biopolymer and Health Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan.
| |
Collapse
|
8
|
Hastie JL, Carmichael HL, Werner BM, Dunbar KE, Carlson PE. Clostridioides difficile utilizes siderophores as an iron source and FhuDBGC contributes to ferrichrome uptake. J Bacteriol 2023; 205:e0032423. [PMID: 37971230 PMCID: PMC10729759 DOI: 10.1128/jb.00324-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 10/26/2023] [Indexed: 11/19/2023] Open
Abstract
IMPORTANCE This study is the first example of C. difficile growing with siderophores as the sole iron source and describes the characterization of the ferric hydroxamate uptake ABC transporter (FhuDBGC). This transporter shows specificity to the siderophore ferrichrome. While not required for pathogenesis, this transporter highlights the redundancy in iron acquisition mechanisms that C. difficile uses to compete for iron during an infection.
Collapse
Affiliation(s)
- Jessica L. Hastie
- Food and Drug Administration, Center for Biologics Evaluation and Research, Office of Vaccines Research and Review, Division of Bacterial Parasitic and Allergenic Products, Laboratory of Mucosal Pathogens and Cellular Immunology, Silver Spring, Maryland, USA
| | - Hannah L. Carmichael
- Food and Drug Administration, Center for Biologics Evaluation and Research, Office of Vaccines Research and Review, Division of Bacterial Parasitic and Allergenic Products, Laboratory of Mucosal Pathogens and Cellular Immunology, Silver Spring, Maryland, USA
| | - Bailey M. Werner
- Food and Drug Administration, Center for Biologics Evaluation and Research, Office of Vaccines Research and Review, Division of Bacterial Parasitic and Allergenic Products, Laboratory of Mucosal Pathogens and Cellular Immunology, Silver Spring, Maryland, USA
| | - Kristin E. Dunbar
- Food and Drug Administration, Center for Biologics Evaluation and Research, Office of Vaccines Research and Review, Division of Bacterial Parasitic and Allergenic Products, Laboratory of Mucosal Pathogens and Cellular Immunology, Silver Spring, Maryland, USA
| | - Paul E. Carlson
- Food and Drug Administration, Center for Biologics Evaluation and Research, Office of Vaccines Research and Review, Division of Bacterial Parasitic and Allergenic Products, Laboratory of Mucosal Pathogens and Cellular Immunology, Silver Spring, Maryland, USA
| |
Collapse
|
9
|
Jenul C, Keim KC, Jens JN, Zeiler MJ, Schilcher K, Schurr MJ, Melander C, Phelan VV, Horswill AR. Pyochelin biotransformation by Staphylococcus aureus shapes bacterial competition with Pseudomonas aeruginosa in polymicrobial infections. Cell Rep 2023; 42:112540. [PMID: 37227819 PMCID: PMC10592502 DOI: 10.1016/j.celrep.2023.112540] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/29/2023] [Accepted: 05/03/2023] [Indexed: 05/27/2023] Open
Abstract
Pseudomonas aeruginosa and Staphylococcus aureus are among the most frequently isolated bacterial species from polymicrobial infections of patients with cystic fibrosis and chronic wounds. We apply mass spectrometry guided interaction studies to determine how chemical interaction shapes the fitness and community structure during co-infection of these two pathogens. We demonstrate that S. aureus is equipped with an elegant mechanism to inactivate pyochelin via the yet uncharacterized methyltransferase Spm (staphylococcal pyochelin methyltransferase). Methylation of pyochelin abolishes the siderophore activity of pyochelin and significantly lowers pyochelin-mediated intracellular reactive oxygen species (ROS) production in S. aureus. In a murine wound co-infection model, an S. aureus mutant unable to methylate pyochelin shows significantly lower fitness compared with its parental strain. Thus, Spm-mediated pyochelin methylation is a mechanism to increase S. aureus survival during in vivo competition with P. aeruginosa.
Collapse
Affiliation(s)
- Christian Jenul
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Klara C Keim
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Justin N Jens
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Michael J Zeiler
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Katrin Schilcher
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Michael J Schurr
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Christian Melander
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Vanessa V Phelan
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Alexander R Horswill
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Veterans Affairs, Eastern Colorado Health Care System, Aurora, CO 80045, USA.
| |
Collapse
|
10
|
Ghssein G, Ezzeddine Z. The Key Element Role of Metallophores in the Pathogenicity and Virulence of Staphylococcus aureus: A Review. BIOLOGY 2022; 11:1525. [PMID: 36290427 PMCID: PMC9598555 DOI: 10.3390/biology11101525] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/17/2022] [Accepted: 10/14/2022] [Indexed: 11/05/2022]
Abstract
The ubiquitous bacterium Staphylococcus aureus causes many diseases that sometimes can be fatal due to its high pathogenicity. The latter is caused by the ability of this pathogen to secrete secondary metabolites, enabling it to colonize inside the host causing infection through various processes. Metallophores are secondary metabolites that enable bacteria to sequester metal ions from the surrounding environment since the availability of metal ions is crucial for bacterial metabolism and virulence. The uptake of iron and other metal ions such as nickel and zinc is one of these essential mechanisms that gives this germ its virulence properties and allow it to overcome the host immune system. Additionally, extensive interactions occur between this pathogen and other bacteria as they compete for resources. Staphylococcus aureus has high-affinity metal import pathways including metal ions acquisition, recruitment and metal-chelate complex import. These characteristics give this bacterium the ability to intake metallophores synthesized by other bacteria, thus enabling it to compete with other microorganisms for the limited nutrients. In scarce host conditions, free metal ions are extremely low because they are confined to storage and metabolic molecules, so metal ions are sequestered by metallophores produced by this bacterium. Both siderophores (iron chelating molecules) and staphylopine (wide- spectrum metallophore) are secreted by Staphylococcus aureus giving it infectious properties. The genetic regulation of the synthesis and export together with the import of metal loaded metallophores are well established and are all covered in this review.
Collapse
Affiliation(s)
- Ghassan Ghssein
- Department of Laboratory Sciences, Faculty of Public Health, Islamic University of Lebanon, Khalde P.O. Box 30014, Lebanon
| | - Zeinab Ezzeddine
- Department of Laboratory Sciences, Faculty of Public Health, Islamic University of Lebanon, Khalde P.O. Box 30014, Lebanon
| |
Collapse
|
11
|
Mohammad M, Ali A, Nguyen MT, Götz F, Pullerits R, Jin T. Staphylococcus aureus lipoproteins in infectious diseases. Front Microbiol 2022; 13:1006765. [PMID: 36262324 PMCID: PMC9574248 DOI: 10.3389/fmicb.2022.1006765] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Infections with the Gram-positive bacterial pathogen Staphylococcus aureus remain a major challenge for the healthcare system and demand new treatment options. The increasing antibiotic resistance of S. aureus poses additional challenges, consequently inflicting a huge strain in the society due to enormous healthcare costs. S. aureus expresses multiple molecules, including bacterial lipoproteins (Lpps), which play a role not only in immune response but also in disease pathogenesis. S. aureus Lpps, the predominant ligands of TLR2, are important for bacterial survival as they maintain the metabolic activity of the bacteria. Moreover, Lpps possess many diverse properties that are of vital importance for the bacteria. They also contribute to host cell invasion but so far their role in different staphylococcal infections has not been fully defined. In this review, we summarize the current knowledge about S. aureus Lpps and their distinct roles in various infectious disease animal models, such as septic arthritis, sepsis, and skin and soft tissue infections. The molecular and cellular response of the host to S. aureus Lpp exposure is also a primary focus.
Collapse
Affiliation(s)
- Majd Mohammad
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Abukar Ali
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Minh-Thu Nguyen
- Section of Medical and Geographical Infectiology, Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Friedrich Götz
- Department of Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, Tübingen, Germany
| | - Rille Pullerits
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Tao Jin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
12
|
Liu K, Abouelhassan Y, Zhang Y, Jin S, Huigens Iii RW. Transcript Profiling of Nitroxoline-Treated Biofilms Shows Rapid Up-regulation of Iron Acquisition Gene Clusters. ACS Infect Dis 2022; 8:1594-1605. [PMID: 35830188 PMCID: PMC10549994 DOI: 10.1021/acsinfecdis.2c00206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Bacterial biofilms are surface-attached communities of slow- or non-replicating cells embedded within a protective matrix of biomolecules. Unlike free-floating planktonic bacteria, biofilms are innately tolerant to conventional antibiotics and are prevalent in recurring and chronic infections. Nitroxoline, a broad-spectrum biofilm-eradicating agent, was used to probe biofilm viability. Transcript profiling (RNA-seq) showed that 452 of 2594 genes (17.4%) in methicillin-resistant Staphylococcus aureus (MRSA) biofilms were differentially expressed after a 2 h treatment of nitroxoline. WoPPER analysis and time-course validation (RT-qPCR) revealed that gene clusters involved in iron acquisition (sbn, isd, MW2101, MW0695, fhu, and feo) were rapidly up-regulated following nitroxoline treatment, which is indicative of iron starvation in MRSA biofilms. In addition, genes related to oligopeptide transporters and riboflavin biosynthesis were found to be up-regulated, while genes related to carotenoid biosynthesis and nitrate assimilation were down-regulated. RT-qPCR experiments revealed that iron uptake transcripts were also up-regulated in established Staphylococcus epidermidis and Acinetobacter baumannii biofilms following nitroxoline treatment. Overall, we show RNA-seq to be an ideal platform to define cellular pathways critical for biofilm survival, in addition to demonstrating the need these bacterial communities have for iron.
Collapse
Affiliation(s)
- Ke Liu
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Yasmeen Abouelhassan
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Yanping Zhang
- Interdisciplinary Center for Biotechnology Research (ICBR), Gene Expression and Genotyping, University of Florida, Gainesville, Florida 32610, United States
| | - Shouguang Jin
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - Robert W Huigens Iii
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
13
|
Adolf LA, Heilbronner S. Nutritional Interactions between Bacterial Species Colonising the Human Nasal Cavity: Current Knowledge and Future Prospects. Metabolites 2022; 12:489. [PMID: 35736422 PMCID: PMC9229137 DOI: 10.3390/metabo12060489] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/16/2022] [Accepted: 05/25/2022] [Indexed: 12/10/2022] Open
Abstract
The human nasal microbiome can be a reservoir for several pathogens, including Staphylococcus aureus. However, certain harmless nasal commensals can interfere with pathogen colonisation, an ability that could be exploited to prevent infection. Although attractive as a prophylactic strategy, manipulation of nasal microbiomes to prevent pathogen colonisation requires a better understanding of the molecular mechanisms of interaction that occur between nasal commensals as well as between commensals and pathogens. Our knowledge concerning the mechanisms of pathogen exclusion and how stable community structures are established is patchy and incomplete. Nutrients are scarce in nasal cavities, which makes competitive or mutualistic traits in nutrient acquisition very likely. In this review, we focus on nutritional interactions that have been shown to or might occur between nasal microbiome members. We summarise concepts of nutrient release from complex host molecules and host cells as well as of intracommunity exchange of energy-rich fermentation products and siderophores. Finally, we discuss the potential of genome-based metabolic models to predict complex nutritional interactions between members of the nasal microbiome.
Collapse
Affiliation(s)
- Lea A. Adolf
- Interfaculty Institute for Microbiology and Infection Medicine, Institute for Medical Microbiology and Hygiene, UKT Tübingen, 72076 Tübingen, Germany;
| | - Simon Heilbronner
- Interfaculty Institute for Microbiology and Infection Medicine, Institute for Medical Microbiology and Hygiene, UKT Tübingen, 72076 Tübingen, Germany;
- German Centre for Infection Research (DZIF), Partner Site Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence EXC 2124 Controlling Microbes to Fight Infections, 72076 Tübingen, Germany
| |
Collapse
|
14
|
Overview of structurally homologous flavoprotein oxidoreductases containing the low M r thioredoxin reductase-like fold - A functionally diverse group. Arch Biochem Biophys 2021; 702:108826. [PMID: 33684359 DOI: 10.1016/j.abb.2021.108826] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/23/2021] [Accepted: 02/27/2021] [Indexed: 01/12/2023]
Abstract
Structural studies show that enzymes have a limited number of unique folds, although structurally related enzymes have evolved to perform a large variety of functions. In this review, we have focused on enzymes containing the low molecular weight thioredoxin reductase (low Mr TrxR) fold. This fold consists of two domains, both containing a three-layer ββα sandwich Rossmann-like fold, serving as flavin adenine dinucleotide (FAD) and, in most cases, pyridine nucleotide (NAD(P)H) binding-domains. Based on a search of the Protein Data Bank for all published structures containing the low Mr TrxR-like fold, we here present a comprehensive overview of enzymes with this structural architecture. These range from TrxR-like ferredoxin/flavodoxin NAD(P)+ oxidoreductases, through glutathione reductase, to NADH peroxidase. Some enzymes are solely composed of the low Mr TrxR-like fold, while others contain one or two additional domains. In this review, we give a detailed description of selected enzymes containing only the low Mr TrxR-like fold, however, catalyzing a diversity of chemical reactions. Our overview of this structurally similar, yet functionally distinct group of flavoprotein oxidoreductases highlights the fascinating and increasing number of studies describing the diversity among these enzymes, especially during the last decade(s).
Collapse
|
15
|
Carlson SK, Erickson DL, Wilson E. Staphylococcus aureus metal acquisition in the mastitic mammary gland. Microb Pathog 2020; 144:104179. [DOI: 10.1016/j.micpath.2020.104179] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/23/2020] [Accepted: 03/27/2020] [Indexed: 12/28/2022]
|
16
|
Endicott N, Rivera GSM, Yang J, Wencewicz TA. Emergence of Ferrichelatase Activity in a Siderophore-Binding Protein Supports an Iron Shuttle in Bacteria. ACS CENTRAL SCIENCE 2020; 6:493-506. [PMID: 32341999 PMCID: PMC7181320 DOI: 10.1021/acscentsci.9b01257] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Indexed: 05/08/2023]
Abstract
Siderophores are small-molecule high-affinity multidentate chelators selective for ferric iron that are produced by pathogenic microbes to aid in nutrient sequestration and enhance virulence. In Gram-positive bacteria, the currently accepted paradigm in siderophore-mediated iron acquisition is that effluxed metal-free siderophores extract ferric iron from biological sources and the resulting ferric siderophore complex undergoes diffusion-controlled association with a surface-displayed siderophore-binding protein (SBP) followed by ABC permease-mediated translocation across the cell envelope powered by ATP hydrolysis. Here we show that a more efficient paradigm is possible in Gram-positive bacteria where extracellular metal-free siderophores associate directly with apo-SBPs on the cell surface and serve as non-covalent cofactors that enable the holo-SBPs to non-reductively extract ferric iron directly from host metalloproteins with so-called "ferrichelatase" activity. The resulting SBP-bound ferric siderophore complex is ready for import through an associated membrane permease and enzymatic turnover is achieved through cofactor replacement from the readily available pool of extracellular siderophores. This new "iron shuttle" model closes a major knowledge gap in microbial iron acquisition and defines new roles of the siderophore and SBP as cofactor and enzyme, respectively, in addition to the classically accepted roles as a transport substrate and receptor pair. We propose the formal name "siderophore-dependent ferrichelatases" for this new class of catalytic SBPs.
Collapse
|
17
|
Marchetti M, De Bei O, Bettati S, Campanini B, Kovachka S, Gianquinto E, Spyrakis F, Ronda L. Iron Metabolism at the Interface between Host and Pathogen: From Nutritional Immunity to Antibacterial Development. Int J Mol Sci 2020; 21:E2145. [PMID: 32245010 PMCID: PMC7139808 DOI: 10.3390/ijms21062145] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 02/08/2023] Open
Abstract
Nutritional immunity is a form of innate immunity widespread in both vertebrates and invertebrates. The term refers to a rich repertoire of mechanisms set up by the host to inhibit bacterial proliferation by sequestering trace minerals (mainly iron, but also zinc and manganese). This strategy, selected by evolution, represents an effective front-line defense against pathogens and has thus inspired the exploitation of iron restriction in the development of innovative antimicrobials or enhancers of antimicrobial therapy. This review focuses on the mechanisms of nutritional immunity, the strategies adopted by opportunistic human pathogen Staphylococcus aureus to circumvent it, and the impact of deletion mutants on the fitness, infectivity, and persistence inside the host. This information finally converges in an overview of the current development of inhibitors targeting the different stages of iron uptake, an as-yet unexploited target in the field of antistaphylococcal drug discovery.
Collapse
Affiliation(s)
- Marialaura Marchetti
- Interdepartmental Center Biopharmanet-TEC, University of Parma, 43124 Parma, Italy; (M.M.); (S.B.)
| | - Omar De Bei
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (O.D.B.); (B.C.)
| | - Stefano Bettati
- Interdepartmental Center Biopharmanet-TEC, University of Parma, 43124 Parma, Italy; (M.M.); (S.B.)
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Institute of Biophysics, National Research Council, 56124 Pisa, Italy
- National Institute of Biostructures and Biosystems, 00136 Rome, Italy
| | - Barbara Campanini
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (O.D.B.); (B.C.)
| | - Sandra Kovachka
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (S.K.); (E.G.); (F.S.)
| | - Eleonora Gianquinto
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (S.K.); (E.G.); (F.S.)
| | - Francesca Spyrakis
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (S.K.); (E.G.); (F.S.)
| | - Luca Ronda
- Interdepartmental Center Biopharmanet-TEC, University of Parma, 43124 Parma, Italy; (M.M.); (S.B.)
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Institute of Biophysics, National Research Council, 56124 Pisa, Italy
| |
Collapse
|
18
|
Conroy BS, Grigg JC, Kolesnikov M, Morales LD, Murphy MEP. Staphylococcus aureus heme and siderophore-iron acquisition pathways. Biometals 2019; 32:409-424. [PMID: 30911924 DOI: 10.1007/s10534-019-00188-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/18/2019] [Indexed: 11/24/2022]
Abstract
Staphylococcus aureus is a versatile opportunistic human pathogen. Infection by this bacterium requires uptake of iron from the human host, but iron is highly restricted in this environment. Staphylococcus aureus iron sufficiency is achieved primarily through uptake of heme and high-affinity iron chelators, known as siderophores. Two siderophores (staphyloferrins) are produced and secreted by S. aureus into the extracellular environment to capture iron. Staphylococcus aureus expresses specific uptake systems for staphyloferrins and more general uptake systems for siderophores produced by other microorganisms. The S. aureus heme uptake system uses highly-specific cell surface receptors to extract heme from hemoglobin and hemoglobin-haptoglobin complexes for transport into the cytoplasm where it is degraded to liberate iron. Initially thought to be independent systems, recent findings indicate that these iron uptake pathways intersect. IruO is a reductase that releases iron from heme and some ferric-siderophores. Moreover, multifunctional SbnI produces a precursor for staphyloferrin B biosynthesis, and also binds heme to regulate expression of the staphyloferrin B biosynthesis pathway. Intersection of the S. aureus iron uptake pathways is hypothesized to be important for rapid adaptation to available iron sources. Components of the heme and siderophore uptake systems are currently being targeted in the development of therapeutics against S. aureus.
Collapse
Affiliation(s)
- Brigid S Conroy
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, V6T 1Z3, Canada
| | - Jason C Grigg
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, V6T 1Z3, Canada
| | - Maxim Kolesnikov
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, V6T 1Z3, Canada
| | - L Daniela Morales
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, V6T 1Z3, Canada
| | - Michael E P Murphy
- Department of Microbiology and Immunology, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, V6T 1Z3, Canada.
| |
Collapse
|
19
|
Abstract
Staphylococcus aureus is clearly the most pathogenic member of the Staphylococcaceae. This is in large part due to the acquisition of an impressive arsenal of virulence factors that are coordinately regulated by a series of dedicated transcription factors. What is becoming more and more appreciated in the field is the influence of the metabolic state of S. aureus on the activity of these virulence regulators and their roles in modulating metabolic gene expression. Here I highlight recent advances in S. aureus metabolism as it pertains to virulence. Specifically, mechanisms of nutrient acquisition are outlined including carbohydrate and non-carbohydrate carbon/energy sources as well as micronutrient (Fe, Mn, Zn and S) acquisition. Additionally, energy producing strategies (respiration versus fermentation) are discussed and put in the context of pathogenesis. Finally, transcriptional regulators that coordinate metabolic gene expression are outlined, particularly those that affect the activities of major virulence factor regulators. This chapter essentially connects many recent observations that link the metabolism of S. aureus to its overall pathogenesis and hints that the mere presence of a plethora of virulence factors may not entirely explain the extraordinary pathogenic potential of S. aureus.
Collapse
Affiliation(s)
- Anthony R Richardson
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15219
| |
Collapse
|
20
|
Grandi A, Fantappiè L, Irene C, Valensin S, Tomasi M, Stupia S, Corbellari R, Caproni E, Zanella I, Isaac SJ, Ganfini L, Frattini L, König E, Gagliardi A, Tavarini S, Sammicheli C, Parri M, Grandi G. Vaccination With a FAT1-Derived B Cell Epitope Combined With Tumor-Specific B and T Cell Epitopes Elicits Additive Protection in Cancer Mouse Models. Front Oncol 2018; 8:481. [PMID: 30416985 PMCID: PMC6212586 DOI: 10.3389/fonc.2018.00481] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 10/08/2018] [Indexed: 12/30/2022] Open
Abstract
Human FAT1 is overexpressed on the surface of most colorectal cancers (CRCs) and in particular a 25 amino acid sequence (D8) present in one of the 34 cadherin extracellular repeats carries the epitope recognized by mAb198.3, a monoclonal antibody which partially protects mice from the challenge with human CRC cell lines in xenograft mouse models. Here we present data in immune competent mice demonstrating the potential of the D8-FAT1 epitope as CRC cancer vaccine. We first demonstrated that the mouse homolog of D8-FAT1 (mD8-FAT1) is also expressed on the surface of CT26 and B16F10 murine cell lines. We then engineered bacterial outer membranes vesicles (OMVs) with mD8-FAT1 and we showed that immunization of BALB/c and C57bl6 mice with engineered OMVs elicited anti-mD8-FAT1 antibodies and partially protected mice from the challenge against CT26 and EGFRvIII-B16F10 cell lines, respectively. We also show that when combined with OMVs decorated with the EGFRvIII B cell epitope or with OMVs carrying five tumor-specific CD4+ T cells neoepitopes, mD8-FAT1 OMVs conferred robust protection against tumor challenge in C57bl6 and BALB/c mice, respectively. Considering that FAT1 is overexpressed in both KRAS+ and KRAS− CRCs, these data support the development of anti-CRC cancer vaccines in which the D8-FAT1 epitope is used in combination with other CRC-specific antigens, including mutation-derived neoepitopes.
Collapse
|
21
|
Bartual SG, Alcorlo M, Martínez-Caballero S, Molina R, Hermoso JA. Three-dimensional structures of Lipoproteins from Streptococcus pneumoniae and Staphylococcus aureus. Int J Med Microbiol 2018; 308:692-704. [DOI: 10.1016/j.ijmm.2017.10.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/21/2017] [Indexed: 01/01/2023] Open
|
22
|
Fojcik C, Arnoux P, Ouerdane L, Aigle M, Alfonsi L, Borezée-Durant E. Independent and cooperative regulation of staphylopine biosynthesis and trafficking by Fur and Zur. Mol Microbiol 2018; 108:159-177. [DOI: 10.1111/mmi.13927] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2018] [Indexed: 11/25/2022]
Affiliation(s)
- Clémentine Fojcik
- Micalis Institute, INRA, AgroParisTech; University Paris-Saclay; 78350 Jouy-en-Josas France
| | - Pascal Arnoux
- CEA, DRF, BIAM, Laboratoire de Bioénergétique Cellulaire; Saint-Paul-lez-Durance France
- CNRS, UMR 7265 Biologie Végétale et Microbiologie Environnementales; Saint-Paul-lez-Durance France
- Aix Marseille Université, UMR 7265 Biologie Végétale et Microbiologie Environnementales; Saint Paul-Lez-Durance 13108 France
| | - Laurent Ouerdane
- CNRS-UPPA, Laboratoire de Chimie Analytique Bio-inorganique et Environnement, UMR 5254, Hélioparc, 2; Av. Angot 64053 Pau France
| | - Marina Aigle
- Micalis Institute, INRA, AgroParisTech; University Paris-Saclay; 78350 Jouy-en-Josas France
| | - Laura Alfonsi
- Micalis Institute, INRA, AgroParisTech; University Paris-Saclay; 78350 Jouy-en-Josas France
| | - Elise Borezée-Durant
- Micalis Institute, INRA, AgroParisTech; University Paris-Saclay; 78350 Jouy-en-Josas France
| |
Collapse
|
23
|
In Vivo Analysis of Staphylococcus aureus-Infected Mice Reveals Differential Temporal and Spatial Expression Patterns of fhuD2. Infect Immun 2017; 85:IAI.00270-17. [PMID: 28784927 DOI: 10.1128/iai.00270-17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 07/26/2017] [Indexed: 12/15/2022] Open
Abstract
Staphylococcus aureus is an opportunistic human pathogen and a major cause of invasive infections such as bacteremia, endocarditis, pneumonia, and wound infections. FhuD2 is a staphylococcal lipoprotein involved in the uptake of iron-hydroxymate and is under the control of the iron uptake regulator Fur. This protein is part of an investigational multicomponent vaccine formulation that has shown protective efficacy in several murine models of infection. Even though fhuD2 expression has been shown to be upregulated in murine kidneys infected with S. aureus, it is not known whether the bacterium undergoes increased iron deprivation during prolonged infection. Furthermore, different S. aureus infection niches might provide different environments and levels of iron availability, resulting in different fhuD2 expression patterns among organs of the same host. To address these questions, we characterized the in vitro expression of the fhuD2 gene and confirmed Fur-dependent regulation of its expression. We further investigated its expression in mice infected with a bioluminescent reporter strain of S. aureus expressing the luciferase operon under the control of the fhuD2 promoter. The emission of bioluminescence in different organs was followed over a 7-day time course, and quantitative real-time PCR analysis of the RNA transcribed from the endogenous fhuD2 gene was performed. Using this approach, we were able to show that fhuD2 expression was induced during infection in all organs analyzed and that differences in expression were observed at different time points and in different infected organs. Our data suggest that S. aureus undergoes increased iron deprivation during the progression of infection in diverse host organs and accordingly induces dedicated iron acquisition mechanisms. Since FhuD2 plays a central role in providing the pathogen with the required iron, further knowledge of the patterns of fhuD2 expression in vivo during infection will be instrumental in better defining the role of this antigen in S. aureus pathogenesis and as a vaccine antigen.
Collapse
|
24
|
Silva WM, Carvalho RDDO, Dorella FA, Folador EL, Souza GHMF, Pimenta AMC, Figueiredo HCP, Le Loir Y, Silva A, Azevedo V. Quantitative Proteomic Analysis Reveals Changes in the Benchmark Corynebacterium pseudotuberculosis Biovar Equi Exoproteome after Passage in a Murine Host. Front Cell Infect Microbiol 2017; 7:325. [PMID: 28791255 PMCID: PMC5524672 DOI: 10.3389/fcimb.2017.00325] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/03/2017] [Indexed: 11/13/2022] Open
Abstract
Corynebacterium pseudotuberculosis biovar equi is the etiologic agent of ulcerative lymphangitis. To investigate proteins that could be related to the virulence of this pathogen, we combined an experimental passage process using a murine model and high-throughput proteomics with a mass spectrometry, data-independent acquisition (LC-MSE) approach to identify and quantify the proteins released into the supernatants of strain 258_equi. To our knowledge, this approach allowed characterization of the exoproteome of a C. pseudotuberculosis equi strain for the first time. Interestingly, the recovery of this strain from infected mouse spleens induced a change in its virulence potential, and it became more virulent in a second infection challenge. Proteomic screening performed from culture supernatant of the control and recovered conditions revealed 104 proteins that were differentially expressed between the two conditions. In this context, proteomic analysis of the recovered condition detected the induction of proteins involved in bacterial pathogenesis, mainly related to iron uptake. In addition, KEGG enrichment analysis showed that ABC transporters, bacterial secretion systems and protein export pathways were significantly altered in the recovered condition. These findings show that secretion and secreted proteins are key elements in the virulence and adaptation of C. pseudotuberculosis. Collectively, bacterial pathogenesis-related proteins were identified that contribute to the processes of adherence, intracellular growth and evasion of the immune system. Moreover, this study enhances our understanding of the factors that may influence the pathogenesis of C. pseudotuberculosis.
Collapse
Affiliation(s)
- Wanderson M Silva
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas GeraisBelo Horizonte, Brazil.,Institut National de la Recherche Agronomique (INRA), UMR1253 Science & Technologie du Lait & de l'Oeuf (STLO)Rennes, France.,Agrocampus Ouest, UMR1253 Science & Technologie du Lait & de l'Oeuf (STLO)Rennes, France
| | - Rodrigo D De Oliveira Carvalho
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas GeraisBelo Horizonte, Brazil
| | - Fernanda A Dorella
- Escola de Veterinária, Universidade Federal de Minas GeraisBelo Horizonte, Brazil
| | - Edson L Folador
- Centro de Biotecnologia, Universidade Federal da ParaíbaJoão Pessoa, Brazil
| | - Gustavo H M F Souza
- Waters Corporation, Waters Technologies Brazil, MS Applications LaboratorySão Paulo, Brazil
| | - Adriano M C Pimenta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas GeraisBelo Horizonte, Brazil
| | | | - Yves Le Loir
- Institut National de la Recherche Agronomique (INRA), UMR1253 Science & Technologie du Lait & de l'Oeuf (STLO)Rennes, France.,Agrocampus Ouest, UMR1253 Science & Technologie du Lait & de l'Oeuf (STLO)Rennes, France
| | - Artur Silva
- Instituto de Ciências Biológicas, Universidade Federal do ParáBelém, Brazil
| | - Vasco Azevedo
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas GeraisBelo Horizonte, Brazil
| |
Collapse
|
25
|
Kobylarz MJ, Heieis GA, Loutet SA, Murphy MEP. Iron Uptake Oxidoreductase (IruO) Uses a Flavin Adenine Dinucleotide Semiquinone Intermediate for Iron-Siderophore Reduction. ACS Chem Biol 2017; 12:1778-1786. [PMID: 28463500 DOI: 10.1021/acschembio.7b00203] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Many pathogenic bacteria including Staphylococcus aureus use iron-chelating siderophores to acquire iron. Iron uptake oxidoreductase (IruO), a flavin adenine dinucleotide (FAD)-containing nicotinamide adenine dinucleotide phosphate (NADPH)-dependent reductase from S. aureus, functions as a reductase for IsdG and IsdI, two paralogous heme degrading enzymes. Also, the gene encoding for IruO was shown to be required for growth of S. aureus on hydroxamate siderophores as a sole iron source. Here, we show that IruO binds the hydroxamate-type siderophores desferrioxamine B and ferrichrome A with low micromolar affinity and in the presence of NADPH, Fe(II) was released. Steady-state kinetics of Fe(II) release provides kcat/Km values in the range of 600 to 7000 M-1 s-1 for these siderophores supporting a role for IruO as a siderophore reductase in iron utilization. Crystal structures of IruO were solved in two distinct conformational states mediated by the formation of an intramolecular disulfide bond. A putative siderophore binding site was identified adjacent to the FAD cofactor. This site is partly occluded in the oxidized IruO structure consistent with this form being less active than reduced IruO. This reduction in activity could have a physiological role to limit iron release under oxidative stress conditions. Visible spectroscopy of anaerobically reduced IruO showed that the reaction proceeds by a single electron transfer mechanism through an FAD semiquinone intermediate. From the data, a model for single electron siderophore reduction by IruO using NADPH is described.
Collapse
Affiliation(s)
- Marek J. Kobylarz
- The Department of Microbiology
and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3; Canada
| | - Graham A. Heieis
- The Department of Microbiology
and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3; Canada
| | - Slade A. Loutet
- The Department of Microbiology
and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3; Canada
| | - Michael E. P. Murphy
- The Department of Microbiology
and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3; Canada
| |
Collapse
|
26
|
Endicott NP, Lee E, Wencewicz TA. Structural Basis for Xenosiderophore Utilization by the Human Pathogen Staphylococcus aureus. ACS Infect Dis 2017; 3:542-553. [PMID: 28505405 DOI: 10.1021/acsinfecdis.7b00036] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Staphylococcus aureus produces a cocktail of metallophores (staphylopine, staphyloferrin A, and staphyloferrin B) to scavenge transition metals during infection of a host. In addition, S. aureus displays the extracellular surface lipoproteins FhuD1 and FhuD2 along with the ABC transporter complex FhuCBG to facilitate the use of hydroxamate xenosiderophores such as desferrioxamine B (DFOB) for iron acquisition. DFOB is used as a chelation therapy to treat human iron overload diseases and has been linked to an increased risk of S. aureus infections. We used a panel of synthetic DFOB analogs and a FhuD2-selective trihydroxamate sideromycin to probe xenosiderophore utilization in S. aureus and establish structure-activity relationships for Fe(III) binding, FhuD2 binding, S. aureus growth promotion, and competition for S. aureus cell entry. Fe(III) binding assays and FhuD2 intrinsic fluorescence quenching experiments revealed that diverse chemical modifications of the terminal ends of linear ferrioxamine siderophores influences Fe(III) affinity but not FhuD2 binding. Siderophore-sideromycin competition assays and xenosiderophore growth promotion assays revealed that S. aureus SG511 and ATCC 11632 can distinguish between competing siderophores based exclusively on net charge of the siderophore-Fe(III) complex. Our work provides a roadmap for tuning hydroxamate xenosiderophore scaffolds to suppress (net negative charge) or enhance (net positive or neutral charge) uptake by S. aureus for applications in metal chelation therapy and siderophore-mediated antibiotic delivery, respectively.
Collapse
Affiliation(s)
- Nathaniel P. Endicott
- Department of Chemistry, Washington University in St. Louis, One Brookings Drive, St. Louis, Missouri 63130, United States
| | - Eries Lee
- Department of Chemistry, Washington University in St. Louis, One Brookings Drive, St. Louis, Missouri 63130, United States
| | - Timothy A. Wencewicz
- Department of Chemistry, Washington University in St. Louis, One Brookings Drive, St. Louis, Missouri 63130, United States
| |
Collapse
|
27
|
Identification of some main Streptococcus iniae associated proteins: relationship. Vet Res Commun 2017; 41:85-95. [DOI: 10.1007/s11259-017-9675-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 01/08/2017] [Indexed: 10/20/2022]
|
28
|
Shahmirzadi SV, Nguyen MT, Götz F. Evaluation of Staphylococcus aureus Lipoproteins: Role in Nutritional Acquisition and Pathogenicity. Front Microbiol 2016; 7:1404. [PMID: 27679612 PMCID: PMC5020093 DOI: 10.3389/fmicb.2016.01404] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 08/24/2016] [Indexed: 01/22/2023] Open
Abstract
Bacterial lipoproteins (Lpp) represent a major class of membrane proteins. They are distinguished by a lipid moiety at the N-terminus by which they are anchored either in the outer leaflet of the cytoplasmic membrane or, in Gram-negative bacteria, also in the inner leaflet of the outer membrane. In Gram-positive bacteria Lpp significantly contribute to nutrient transport, Toll-like receptor 2 activation and pathogenicity. Here we examine the Lpp of Staphylococcus aureus USA300, as a prototype for a multiple antibiotic resistant and community-acquired pathogen that is rapidly spreading worldwide. The compiled Lpp were grouped according to the postulated function and dissemination of homologs in the genus Staphylococcus and beyond. Based on this evaluation we also point out Lpp as promising vaccine candidates.
Collapse
Affiliation(s)
- Shideh V Shahmirzadi
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen Tübingen, Germany
| | - Minh-Thu Nguyen
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen Tübingen, Germany
| | - Friedrich Götz
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen Tübingen, Germany
| |
Collapse
|
29
|
Lee AA, Chen YCS, Ekalestari E, Ho SY, Hsu NS, Kuo TF, Wang TSA. Facile and Versatile Chemoenzymatic Synthesis of Enterobactin Analogues and Applications in Bacterial Detection. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201603921] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Albert A. Lee
- Department of Chemistry; National Taiwan University; No. 1, Sec. 4, Roosevelt Road Taipei 10617 Taiwan) (R.O.C
| | - Yi-Chen S. Chen
- Department of Chemistry; National Taiwan University; No. 1, Sec. 4, Roosevelt Road Taipei 10617 Taiwan) (R.O.C
| | - Elisa Ekalestari
- Department of Chemistry & Biochemistry; University of California, Los Angeles; Los Angeles CA 90095 USA
| | - Sheng-Yang Ho
- Department of Chemistry; National Taiwan University; No. 1, Sec. 4, Roosevelt Road Taipei 10617 Taiwan) (R.O.C
| | - Nai-Shu Hsu
- Department of Chemistry; National Taiwan University; No. 1, Sec. 4, Roosevelt Road Taipei 10617 Taiwan) (R.O.C
| | - Tang-Feng Kuo
- Department of Chemistry; National Taiwan University; No. 1, Sec. 4, Roosevelt Road Taipei 10617 Taiwan) (R.O.C
| | - Tsung-Shing Andrew Wang
- Department of Chemistry; National Taiwan University; No. 1, Sec. 4, Roosevelt Road Taipei 10617 Taiwan) (R.O.C
| |
Collapse
|
30
|
Lee AA, Chen YCS, Ekalestari E, Ho SY, Hsu NS, Kuo TF, Wang TSA. Facile and Versatile Chemoenzymatic Synthesis of Enterobactin Analogues and Applications in Bacterial Detection. Angew Chem Int Ed Engl 2016; 55:12338-42. [DOI: 10.1002/anie.201603921] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 07/07/2016] [Indexed: 12/11/2022]
Affiliation(s)
- Albert A. Lee
- Department of Chemistry; National Taiwan University; No. 1, Sec. 4, Roosevelt Road Taipei 10617 Taiwan) (R.O.C
| | - Yi-Chen S. Chen
- Department of Chemistry; National Taiwan University; No. 1, Sec. 4, Roosevelt Road Taipei 10617 Taiwan) (R.O.C
| | - Elisa Ekalestari
- Department of Chemistry & Biochemistry; University of California, Los Angeles; Los Angeles CA 90095 USA
| | - Sheng-Yang Ho
- Department of Chemistry; National Taiwan University; No. 1, Sec. 4, Roosevelt Road Taipei 10617 Taiwan) (R.O.C
| | - Nai-Shu Hsu
- Department of Chemistry; National Taiwan University; No. 1, Sec. 4, Roosevelt Road Taipei 10617 Taiwan) (R.O.C
| | - Tang-Feng Kuo
- Department of Chemistry; National Taiwan University; No. 1, Sec. 4, Roosevelt Road Taipei 10617 Taiwan) (R.O.C
| | - Tsung-Shing Andrew Wang
- Department of Chemistry; National Taiwan University; No. 1, Sec. 4, Roosevelt Road Taipei 10617 Taiwan) (R.O.C
| |
Collapse
|
31
|
Dayan GH, Mohamed N, Scully IL, Cooper D, Begier E, Eiden J, Jansen KU, Gurtman A, Anderson AS. Staphylococcus aureus: the current state of disease, pathophysiology and strategies for prevention. Expert Rev Vaccines 2016; 15:1373-1392. [PMID: 27118628 DOI: 10.1080/14760584.2016.1179583] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Staphylococcus aureus is both a commensal organism and also an important opportunistic human pathogen, causing a variety of community and hospital-associated pathologies, such as bacteremia-sepsis, endocarditis, pneumonia, osteomyelitis, arthritis and skin diseases. The resurgence of S. aureus during the last decade in many settings has been facilitated not only by bacterial antibiotic resistance mechanisms but also by the emergence of new S. aureus clonal types with increased expression of virulence factors and the capacity to neutralize the host immune response. Prevention of the spread of S. aureus infection relies on the use of contact precautions and adequate procedures for infection control that so far have not been fully effective. Prevention using a prophylactic vaccine would complement these processes, having the potential to bring additional, significant progress toward decreasing invasive disease due to S. aureus.
Collapse
Affiliation(s)
- Gustavo H Dayan
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - Naglaa Mohamed
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - Ingrid L Scully
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - David Cooper
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - Elizabeth Begier
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - Joseph Eiden
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - Kathrin U Jansen
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | | | | |
Collapse
|
32
|
Mancini F, Monaci E, Lofano G, Torre A, Bacconi M, Tavarini S, Sammicheli C, Arcidiacono L, Galletti B, Laera D, Pallaoro M, Tuscano G, Fontana MR, Bensi G, Grandi G, Rossi-Paccani S, Nuti S, Rappuoli R, De Gregorio E, Bagnoli F, Soldaini E, Bertholet S. One Dose of Staphylococcus aureus 4C-Staph Vaccine Formulated with a Novel TLR7-Dependent Adjuvant Rapidly Protects Mice through Antibodies, Effector CD4+ T Cells, and IL-17A. PLoS One 2016; 11:e0147767. [PMID: 26812180 PMCID: PMC4727907 DOI: 10.1371/journal.pone.0147767] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 01/07/2016] [Indexed: 12/27/2022] Open
Abstract
A rapidly acting, single dose vaccine against Staphylococcus aureus would be highly beneficial for patients scheduled for major surgeries or in intensive care units. Here we show that one immunization with a multicomponent S. aureus candidate vaccine, 4C-Staph, formulated with a novel TLR7-dependent adjuvant, T7-alum, readily protected mice from death and from bacterial dissemination, both in kidney abscess and peritonitis models, outperforming alum-formulated vaccine. This increased efficacy was paralleled by higher vaccine-specific and α-hemolysin-neutralizing antibody titers and Th1/Th17 cell responses. Antibodies played a crucial protective role, as shown by the lack of protection of 4C-Staph/T7-alum vaccine in B-cell-deficient mice and by serum transfer experiments. Depletion of effector CD4+ T cells not only reduced survival but also increased S. aureus load in kidneys of mice immunized with 4C-Staph/T7-alum. The role of IL-17A in the control of bacterial dissemination in 4C-Staph/T7-alum vaccinated mice was indicated by in vivo neutralization experiments. We conclude that single dose 4C-Staph/T7-alum vaccine promptly and efficiently protected mice against S. aureus through the combined actions of antibodies, CD4+ effector T cells, and IL-17A. These data suggest that inclusion of an adjuvant that induces not only fast antibody responses but also IL-17-producing cell-mediated effector responses could efficaciously protect patients scheduled for major surgeries or in intensive care units.
Collapse
Affiliation(s)
- Francesca Mancini
- Novartis Vaccines and Diagnostics, S.r.l., Research Center, Siena, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Elisabetta Monaci
- Novartis Vaccines and Diagnostics, S.r.l., Research Center, Siena, Italy
| | - Giuseppe Lofano
- Novartis Vaccines and Diagnostics, S.r.l., Research Center, Siena, Italy
- Department of Biology and Biotechnologies “Charles Darwin”, University of Rome “La Sapienza”, Rome, Italy
| | - Antonina Torre
- Novartis Vaccines and Diagnostics, S.r.l., Research Center, Siena, Italy
| | - Marta Bacconi
- Novartis Vaccines and Diagnostics, S.r.l., Research Center, Siena, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Simona Tavarini
- Novartis Vaccines and Diagnostics, S.r.l., Research Center, Siena, Italy
| | - Chiara Sammicheli
- Novartis Vaccines and Diagnostics, S.r.l., Research Center, Siena, Italy
| | | | - Bruno Galletti
- Novartis Vaccines and Diagnostics, S.r.l., Research Center, Siena, Italy
| | - Donatello Laera
- Novartis Vaccines and Diagnostics, S.r.l., Research Center, Siena, Italy
| | - Michele Pallaoro
- Novartis Vaccines and Diagnostics, S.r.l., Research Center, Siena, Italy
| | - Giovanna Tuscano
- Novartis Vaccines and Diagnostics, S.r.l., Research Center, Siena, Italy
| | - Maria Rita Fontana
- Novartis Vaccines and Diagnostics, S.r.l., Research Center, Siena, Italy
| | - Giuliano Bensi
- Novartis Vaccines and Diagnostics, S.r.l., Research Center, Siena, Italy
| | - Guido Grandi
- Novartis Vaccines and Diagnostics, S.r.l., Research Center, Siena, Italy
| | | | - Sandra Nuti
- Novartis Vaccines and Diagnostics, S.r.l., Research Center, Siena, Italy
| | - Rino Rappuoli
- Novartis Vaccines and Diagnostics, S.r.l., Research Center, Siena, Italy
| | - Ennio De Gregorio
- Novartis Vaccines and Diagnostics, S.r.l., Research Center, Siena, Italy
| | - Fabio Bagnoli
- Novartis Vaccines and Diagnostics, S.r.l., Research Center, Siena, Italy
| | - Elisabetta Soldaini
- Novartis Vaccines and Diagnostics, S.r.l., Research Center, Siena, Italy
- * E-mail:
| | - Sylvie Bertholet
- Novartis Vaccines and Diagnostics, S.r.l., Research Center, Siena, Italy
| |
Collapse
|
33
|
Abstract
Staphylococcus aureus is a leading pathogen in surgical site, intensive care unit, and skin infections, as well as healthcare-associated pneumonias. These infections are associated with an enormous burden of morbidity, mortality, and increase of hospital length of stay and patient cost. S. aureus is impressively fast in acquiring antibiotic resistance, and multidrug-resistant strains are a serious threat to human health. Due to resistance or insufficient effectiveness, antibiotics and bundle measures leave a tremendous unmet medical need worldwide. There are no licensed vaccines on the market despite the significant efforts done by public and private initiatives. Indeed, vaccines tested in clinical trials in the last two decades have failed to show efficacy. However, they targeted single antigens and contained no adjuvants and efficacy trials were performed in severely ill subjects. Herein, we provide a comprehensive evaluation of potential target populations for efficacy trials taking into account key factors such as population size, incidence of S. aureus infection, disease outcome, primary endpoints, as well as practical advantages and disadvantages. We describe the whole-blood assay as a potential surrogate of protection, and we show the link between phase III clinical trial data of failed vaccines with their preclinical observations. Finally, we give our perspective on how new vaccine formulations and clinical development approaches may lead to successful S. aureus vaccines.
Collapse
|
34
|
Flannagan RS, Heit B, Heinrichs DE. Antimicrobial Mechanisms of Macrophages and the Immune Evasion Strategies of Staphylococcus aureus. Pathogens 2015; 4:826-68. [PMID: 26633519 PMCID: PMC4693167 DOI: 10.3390/pathogens4040826] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 11/17/2015] [Accepted: 11/24/2015] [Indexed: 12/21/2022] Open
Abstract
Habitually professional phagocytes, including macrophages, eradicate microbial invaders from the human body without overt signs of infection. Despite this, there exist select bacteria that are professional pathogens, causing significant morbidity and mortality across the globe and Staphylococcus aureus is no exception. S. aureus is a highly successful pathogen that can infect virtually every tissue that comprises the human body causing a broad spectrum of diseases. The profound pathogenic capacity of S. aureus can be attributed, in part, to its ability to elaborate a profusion of bacterial effectors that circumvent host immunity. Macrophages are important professional phagocytes that contribute to both the innate and adaptive immune response, however from in vitro and in vivo studies, it is evident that they fail to eradicate S. aureus. This review provides an overview of the antimicrobial mechanisms employed by macrophages to combat bacteria and describes the immune evasion strategies and some representative effectors that enable S. aureus to evade macrophage-mediated killing.
Collapse
Affiliation(s)
- Ronald S Flannagan
- Department of Microbiology and Immunology, the University of Western Ontario, London, ON N6A 5C1, Canada.
| | - Bryan Heit
- Department of Microbiology and Immunology, the University of Western Ontario, London, ON N6A 5C1, Canada.
- Centre for Human Immunology, the University of Western Ontario, London, ON N6A 5C1, Canada.
| | - David E Heinrichs
- Department of Microbiology and Immunology, the University of Western Ontario, London, ON N6A 5C1, Canada.
- Centre for Human Immunology, the University of Western Ontario, London, ON N6A 5C1, Canada.
| |
Collapse
|
35
|
Monaci E, Mancini F, Lofano G, Bacconi M, Tavarini S, Sammicheli C, Arcidiacono L, Giraldi M, Galletti B, Rossi Paccani S, Torre A, Fontana MR, Grandi G, de Gregorio E, Bensi G, Chiarot E, Nuti S, Bagnoli F, Soldaini E, Bertholet S. MF59- and Al(OH)3-Adjuvanted Staphylococcus aureus (4C-Staph) Vaccines Induce Sustained Protective Humoral and Cellular Immune Responses, with a Critical Role for Effector CD4 T Cells at Low Antibody Titers. Front Immunol 2015; 6:439. [PMID: 26441955 PMCID: PMC4561515 DOI: 10.3389/fimmu.2015.00439] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 08/13/2015] [Indexed: 11/13/2022] Open
Abstract
Staphylococcus aureus (S. aureus) is an important opportunistic pathogen that may cause invasive life-threatening infections, like sepsis and pneumonia. Due to the increasing antibiotic resistance, the development of an effective vaccine against S. aureus is needed. Although a correlate of protection against staphylococcal diseases is not yet established, several findings suggest that both antibodies and CD4 T cells might contribute to optimal immunity. In this study, we show that adjuvanting a multivalent vaccine (4C-Staph) with MF59, an oil-in-water emulsion licensed in human vaccines, further potentiated antigen-specific IgG titers and CD4 T-cell responses compared to alum and conferred protection in the peritonitis model of S. aureus infection. Moreover, we showed that MF59- and alum-adjuvanted 4C-Staph vaccines induced persistent antigen-specific humoral and T-cell responses, and protected mice from infection up to 4 months after immunization. Furthermore, 4C-Staph formulated with MF59 was used to investigate which immune compartment is involved in vaccine-induced protection. Using CD4 T cell-depleted mice or B cell-deficient mice, we demonstrated that both T and B-cell responses contributed to 4C-Staph vaccine-mediated protective immunity. However, the role of CD4 T cells seemed more evident in the presence of low-antibody responses. This study provides preclinical data further supporting the use of the adjuvanted 4C-Staph vaccines against S. aureus diseases, and provides critical insights on the correlates of protective immunity necessary to combat this pathogen.
Collapse
Affiliation(s)
- Elisabetta Monaci
- Research Center, Novartis Vaccines and Diagnostics S.r.l. , Siena , Italy
| | - Francesca Mancini
- Research Center, Novartis Vaccines and Diagnostics S.r.l. , Siena , Italy ; Department of Biomedical Sciences, University of Padua , Padua , Italy
| | - Giuseppe Lofano
- Research Center, Novartis Vaccines and Diagnostics S.r.l. , Siena , Italy ; Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome , Rome , Italy
| | - Marta Bacconi
- Research Center, Novartis Vaccines and Diagnostics S.r.l. , Siena , Italy ; Department of Biotechnology, Chemistry and Pharmacy, University of Siena , Siena , Italy
| | - Simona Tavarini
- Research Center, Novartis Vaccines and Diagnostics S.r.l. , Siena , Italy
| | - Chiara Sammicheli
- Research Center, Novartis Vaccines and Diagnostics S.r.l. , Siena , Italy
| | | | - Monica Giraldi
- Research Center, Novartis Vaccines and Diagnostics S.r.l. , Siena , Italy
| | - Bruno Galletti
- Research Center, Novartis Vaccines and Diagnostics S.r.l. , Siena , Italy
| | | | - Antonina Torre
- Research Center, Novartis Vaccines and Diagnostics S.r.l. , Siena , Italy
| | - Maria Rita Fontana
- Research Center, Novartis Vaccines and Diagnostics S.r.l. , Siena , Italy
| | - Guido Grandi
- Research Center, Novartis Vaccines and Diagnostics S.r.l. , Siena , Italy
| | - Ennio de Gregorio
- Research Center, Novartis Vaccines and Diagnostics S.r.l. , Siena , Italy
| | - Giuliano Bensi
- Research Center, Novartis Vaccines and Diagnostics S.r.l. , Siena , Italy
| | - Emiliano Chiarot
- Research Center, Novartis Vaccines and Diagnostics S.r.l. , Siena , Italy
| | - Sandra Nuti
- Research Center, Novartis Vaccines and Diagnostics S.r.l. , Siena , Italy
| | - Fabio Bagnoli
- Research Center, Novartis Vaccines and Diagnostics S.r.l. , Siena , Italy
| | | | - Sylvie Bertholet
- Research Center, Novartis Vaccines and Diagnostics S.r.l. , Siena , Italy
| |
Collapse
|
36
|
Madsen JLH, Johnstone TC, Nolan EM. Chemical Synthesis of Staphyloferrin B Affords Insight into the Molecular Structure, Iron Chelation, and Biological Activity of a Polycarboxylate Siderophore Deployed by the Human Pathogen Staphylococcus aureus. J Am Chem Soc 2015; 137:9117-27. [DOI: 10.1021/jacs.5b04557] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Julie L. H. Madsen
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Timothy C. Johnstone
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Elizabeth M. Nolan
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
37
|
Hannauer M, Arifin AJ, Heinrichs DE. Involvement of reductases IruO and NtrA in iron acquisition byStaphylococcus aureus. Mol Microbiol 2015; 96:1192-210. [DOI: 10.1111/mmi.13000] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2015] [Indexed: 12/20/2022]
Affiliation(s)
- Mélissa Hannauer
- Department of Microbiology and Immunology; University of Western Ontario; London ON Canada N6A 5C1
| | - Andrew J. Arifin
- Department of Microbiology and Immunology; University of Western Ontario; London ON Canada N6A 5C1
| | - David E. Heinrichs
- Department of Microbiology and Immunology; University of Western Ontario; London ON Canada N6A 5C1
- Centre for Human Immunology; University of Western Ontario; London ON Canada N6A 5C1
| |
Collapse
|
38
|
Sheldon JR, Heinrichs DE. Recent developments in understanding the iron acquisition strategies of gram positive pathogens. FEMS Microbiol Rev 2015; 39:592-630. [DOI: 10.1093/femsre/fuv009] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2015] [Indexed: 12/26/2022] Open
|
39
|
Vaccine composition formulated with a novel TLR7-dependent adjuvant induces high and broad protection against Staphylococcus aureus. Proc Natl Acad Sci U S A 2015; 112:3680-5. [PMID: 25775551 DOI: 10.1073/pnas.1424924112] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Both active and passive immunization strategies against Staphylococcus aureus have thus far failed to show efficacy in humans. With the attempt to develop an effective S. aureus vaccine, we selected five conserved antigens known to have different roles in S. aureus pathogenesis. They include the secreted factors α-hemolysin (Hla), ess extracellular A (EsxA), and ess extracellular B (EsxB) and the two surface proteins ferric hydroxamate uptake D2 and conserved staphylococcal antigen 1A. The combined vaccine antigens formulated with aluminum hydroxide induced antibodies with opsonophagocytic and functional activities and provided consistent protection in four mouse models when challenged with a panel of epidemiologically relevant S. aureus strains. The importance of antibodies in protection was demonstrated by passive transfer experiments. Furthermore, when formulated with a toll-like receptor 7-dependent (TLR7) agonist recently designed and developed in our laboratories (SMIP.7-10) adsorbed to alum, the five antigens provided close to 100% protection against four different staphylococcal strains. The new formulation induced not only high antibody titers but also a Th1 skewed immune response as judged by antibody isotype and cytokine profiles. In addition, low frequencies of IL-17-secreting T cells were also observed. Altogether, our data demonstrate that the rational selection of mixtures of conserved antigens combined with Th1/Th17 adjuvants can lead to promising vaccine formulations against S. aureus.
Collapse
|
40
|
Arifin AJ, Hannauer M, Welch I, Heinrichs DE. Deferoxamine mesylate enhances virulence of community-associated methicillin resistant Staphylococcus aureus. Microbes Infect 2014; 16:967-72. [PMID: 25251026 DOI: 10.1016/j.micinf.2014.09.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 09/04/2014] [Accepted: 09/09/2014] [Indexed: 11/16/2022]
Abstract
Staphylococcus aureus is a leading cause of bacterial infections. Strains of community-associated methicillin-resistant S. aureus (CA-MRSA), such as USA300, display enhanced virulence and fitness. Patients suffering from iron overload diseases often undergo iron chelation therapy with deferoxamine mesylate (DFO). Here, we show that USA300 uses this drug to acquire iron. We further demonstrate that mice administered DFO I.P., versus those not administered DFO, had significantly higher bacterial burden in livers and kidneys after I.V. challenge with USA300, associated with increased abscess formation and tissue destruction. The virulence of USA300 mutants defective for DFO uptake was not affected by DFO treatment.
Collapse
Affiliation(s)
- Andrew J Arifin
- Department of Microbiology & Immunology, University of Western Ontario, London N6A 5C1, Ontario, Canada
| | - Mélissa Hannauer
- Department of Microbiology & Immunology, University of Western Ontario, London N6A 5C1, Ontario, Canada
| | - Ian Welch
- Department of Animal Care and Veterinary Services, University of Western Ontario, London N6A 5C1, Ontario, Canada
| | - David E Heinrichs
- Department of Microbiology & Immunology, University of Western Ontario, London N6A 5C1, Ontario, Canada; Centre for Human Immunology, University of Western Ontario, London N6A 5C1, Ontario, Canada.
| |
Collapse
|
41
|
Fukushima T, Allred BE, Raymond KN. Direct evidence of iron uptake by the Gram-positive siderophore-shuttle mechanism without iron reduction. ACS Chem Biol 2014; 9:2092-100. [PMID: 25007174 PMCID: PMC4168784 DOI: 10.1021/cb500319n] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 07/09/2014] [Indexed: 11/29/2022]
Abstract
Iron is an essential element for all organisms, and microorganisms produce small molecule iron-chelators, siderophores, to efficiently acquire Fe(III). Gram-positive bacteria possess lipoprotein siderophore-binding proteins (SBPs) on the membrane. Some of the SBPs bind both apo-siderophores (iron-free) and Fe-siderophore (iron-chelated) and only import Fe-siderophores. When the SBP initially binds an apo-siderophore, the SBP uses the Gram-positive siderophore-shuttle mechanism (the SBPs exchange Fe(III) from a Fe-siderophore to the apo-siderophore bound to the protein) and/or displacement mechanism (the apo-siderophore bound to the SBP is released and a Fe-siderophore is then bound to the protein) to import the Fe-siderophore. Previously, we reported that the Bacillus cereus SBP, YxeB, exchanges Fe(III) from a ferrioxamine B (FO) to a desferrioxamine B (DFO) bound to YxeB using the siderophore-shuttle mechanism although the iron exchange was indirectly elucidated. Synthetic Cr-DFO (inert metal FO analog) and Ga-DFO (nonreducible FO analog) are bound to YxeB and imported via YxeB and the corresponding permeases and ATPase. YxeB exchanges Fe(III) from FO and Ga(III) from Ga-DFO to DFO bound to the protein, indicating that the metal-exchange occurs without metal reduction. YxeB also binds DFO derivatives including acetylated DFO (apo-siderophore) and acetylated FO (AcFO, Fe-siderophore). The iron from AcFO is transferred to DFO when bound to YxeB, giving direct evidence of iron exchange. Moreover, YxeB also uses the displacement mechanism when ferrichrome (Fch) is added to the DFO:YxeB complex. Uptake by the displacement mechanism is a minor pathway compared to the shuttle mechanism.
Collapse
Affiliation(s)
| | | | - Kenneth N. Raymond
- Department of Chemistry, University of
California, Berkeley, California 94720-1460, United States
| |
Collapse
|
42
|
Zheng T, Nolan EM. Enterobactin-mediated delivery of β-lactam antibiotics enhances antibacterial activity against pathogenic Escherichia coli. J Am Chem Soc 2014; 136:9677-91. [PMID: 24927110 PMCID: PMC4353011 DOI: 10.1021/ja503911p] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Indexed: 02/08/2023]
Abstract
The design, synthesis, and characterization of enterobactin-antibiotic conjugates, hereafter Ent-Amp/Amx, where the β-lactam antibiotics ampicillin (Amp) and amoxicillin (Amx) are linked to a monofunctionalized enterobactin scaffold via a stable poly(ethylene glycol) linker are reported. Under conditions of iron limitation, these siderophore-modified antibiotics provide enhanced antibacterial activity against Escherichia coli strains, including uropathogenic E. coli CFT073 and UTI89, enterohemorrhagic E. coli O157:H7, and enterotoxigenic E. coli O78:H11, compared to the parent β-lactams. Studies with E. coli K-12 derivatives defective in ferric enterobactin transport reveal that the enhanced antibacterial activity observed for this strain requires the outer membrane ferric enterobactin transporter FepA. A remarkable 1000-fold decrease in minimum inhibitory concentration (MIC) value is observed for uropathogenic E. coli CFT073 relative to Amp/Amx, and time-kill kinetic studies demonstrate that Ent-Amp/Amx kill this strain more rapidly at 10-fold lower concentrations than the parent antibiotics. Moreover, Ent-Amp and Ent-Amx selectively kill E. coli CFT073 co-cultured with other bacterial species such as Staphylococcus aureus, and Ent-Amp exhibits low cytotoxicity against human T84 intestinal cells in both the apo and iron-bound forms. These studies demonstrate that the native enterobactin platform provides a means to effectively deliver antibacterial cargo across the outer membrane permeability barrier of Gram-negative pathogens utilizing enterobactin for iron acquisition.
Collapse
Affiliation(s)
- Tengfei Zheng
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Elizabeth M. Nolan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
43
|
Stentzel S, Vu HC, Weyrich AM, Jehmlich N, Schmidt F, Salazar MG, Steil L, Völker U, Bröker BM. Altered immune proteome ofStaphylococcus aureusunder iron-restricted growth conditions. Proteomics 2014; 14:1857-67. [DOI: 10.1002/pmic.201300512] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 04/16/2014] [Accepted: 05/22/2014] [Indexed: 11/09/2022]
Affiliation(s)
- Sebastian Stentzel
- Department of Immunology; Institute of Immunology and Transfusion Medicine; University Medicine Greifswald; Greifswald Germany
| | - Hai Chi Vu
- Department of Immunology; Institute of Immunology and Transfusion Medicine; University Medicine Greifswald; Greifswald Germany
| | - Anna Maria Weyrich
- Department of Immunology; Institute of Immunology and Transfusion Medicine; University Medicine Greifswald; Greifswald Germany
| | - Nico Jehmlich
- Interfaculty Institute of Genetics and Functional Genomics; University Medicine Greifswald; Greifswald Germany
| | - Frank Schmidt
- Interfaculty Institute of Genetics and Functional Genomics; University Medicine Greifswald; Greifswald Germany
- ZIK-FunGene Junior Research Group “Applied Proteomics”; University Medicine Greifswald; Greifswald Germany
| | - Manuela Gesell Salazar
- Interfaculty Institute of Genetics and Functional Genomics; University Medicine Greifswald; Greifswald Germany
| | - Leif Steil
- Interfaculty Institute of Genetics and Functional Genomics; University Medicine Greifswald; Greifswald Germany
| | - Uwe Völker
- Interfaculty Institute of Genetics and Functional Genomics; University Medicine Greifswald; Greifswald Germany
| | - Barbara M. Bröker
- Department of Immunology; Institute of Immunology and Transfusion Medicine; University Medicine Greifswald; Greifswald Germany
| |
Collapse
|
44
|
Vinés ED, Speziali CD, Heinrichs DE. Demonstration of the functional role of conserved Glu-Arg residues in the Staphylococcus aureus ferrichrome transporter. Biometals 2013; 27:143-53. [DOI: 10.1007/s10534-013-9695-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 12/12/2013] [Indexed: 11/30/2022]
|
45
|
Caza M, Kronstad JW. Shared and distinct mechanisms of iron acquisition by bacterial and fungal pathogens of humans. Front Cell Infect Microbiol 2013; 3:80. [PMID: 24312900 PMCID: PMC3832793 DOI: 10.3389/fcimb.2013.00080] [Citation(s) in RCA: 191] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Accepted: 10/30/2013] [Indexed: 12/12/2022] Open
Abstract
Iron is the most abundant transition metal in the human body and its bioavailability is stringently controlled. In particular, iron is tightly bound to host proteins such as transferrin to maintain homeostasis, to limit potential damage caused by iron toxicity under physiological conditions and to restrict access by pathogens. Therefore, iron acquisition during infection of a human host is a challenge that must be surmounted by every successful pathogenic microorganism. Iron is essential for bacterial and fungal physiological processes such as DNA replication, transcription, metabolism, and energy generation via respiration. Hence, pathogenic bacteria and fungi have developed sophisticated strategies to gain access to iron from host sources. Indeed, siderophore production and transport, iron acquisition from heme and host iron-containing proteins such as hemoglobin and transferrin, and reduction of ferric to ferrous iron with subsequent transport are all strategies found in bacterial and fungal pathogens of humans. This review focuses on a comparison of these strategies between bacterial and fungal pathogens in the context of virulence and the iron limitation that occurs in the human body as a mechanism of innate nutritional defense.
Collapse
Affiliation(s)
| | - James W. Kronstad
- The Michael Smith Laboratories, Department of Microbiology and Immunology, University of British ColumbiaVancouver, BC, Canada
| |
Collapse
|
46
|
Fukushima T, Allred BE, Sia AK, Nichiporuk R, Andersen UN, Raymond KN. Gram-positive siderophore-shuttle with iron-exchange from Fe-siderophore to apo-siderophore by Bacillus cereus YxeB. Proc Natl Acad Sci U S A 2013; 110:13821-6. [PMID: 23924612 PMCID: PMC3752266 DOI: 10.1073/pnas.1304235110] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Small molecule iron-chelators, siderophores, are very important in facilitating the acquisition of Fe(III), an essential element for pathogenic bacteria. Many Gram-negative outer-membrane transporters and Gram-positive lipoprotein siderophore-binding proteins have been characterized, and the binding ability of outer-membrane transporters and siderophore-binding proteins for Fe-siderophores has been determined. However, there is little information regarding the binding ability of these proteins for apo-siderophores, the iron-free chelators. Here we report that Bacillus cereus YxeB facilitates iron-exchange from Fe-siderophore to apo-siderophore bound to the protein, the first Gram-positive siderophore-shuttle system. YxeB binds ferrioxamine B (FO, Fe-siderophore)/desferrioxamine B (DFO, apo-siderophore) in vitro. Disc-diffusion assays and growth assays using the yxeB mutant reveal that YxeB is responsible for importing the FO. Cr-DFO (a FO analog) is bound by YxeB in vitro and B. cereus imports or binds Cr-DFO in vivo. In vivo uptake assays using Cr-DFO and FO and growth assays using DFO and Cr-DFO show that B. cereus selectively imports and uses FO when DFO is present. Moreover, in vitro competition assays using Cr-DFO and FO clearly demonstrate that YxeB binds only FO, not Cr-DFO, when DFO is bound to the protein. Iron-exchange from FO to DFO bound to YxeB must occur when DFO is initially bound by YxeB. Because the metal exchange rate is generally first order in replacement ligand concentration, protein binding of the apo-siderophore acts to dramatically enhance the iron exchange rate, a key component of the Gram-positive siderophore-shuttle mechanism.
Collapse
Affiliation(s)
| | | | - Allyson K. Sia
- Department of Chemistry, University of California, Berkeley, CA 94720-1460
| | - Rita Nichiporuk
- Department of Chemistry, University of California, Berkeley, CA 94720-1460
| | - Ulla N. Andersen
- Department of Chemistry, University of California, Berkeley, CA 94720-1460
| | - Kenneth N. Raymond
- Department of Chemistry, University of California, Berkeley, CA 94720-1460
| |
Collapse
|
47
|
Structural and functional characterization of the Staphylococcus aureus virulence factor and vaccine candidate FhuD2. Biochem J 2013; 449:683-93. [PMID: 23113737 DOI: 10.1042/bj20121426] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Staphylococcus aureus is a human pathogen causing globally significant morbidity and mortality. The development of antibiotic resistance in S. aureus highlights the need for a preventive vaccine. In the present paper we explore the structure and function of FhuD2 (ferric-hydroxamate uptake D2), a staphylococcal surface lipoprotein mediating iron uptake during invasive infection, recently described as a promising vaccine candidate. Differential scanning fluorimetry and calorimetry studies revealed that FhuD2 is stabilized by hydroxamate siderophores. The FhuD2-ferrichrome interaction was of nanomolar affinity in surface plasmon resonance experiments and fully iron(III)-dependent. We determined the X-ray crystallographic structure of ligand-bound FhuD2 at 1.9 Å (1 Å=0.1 nm) resolution, revealing the bilobate fold of class III SBPs (solute-binding proteins). The ligand, ferrichrome, occupies a cleft between the FhuD2 N- and C-terminal lobes. Many FhuD2-siderophore interactions enable the specific recognition of ferrichrome. Biochemical data suggest that FhuD2 does not undergo significant conformational changes upon siderophore binding, supporting the hypothesis that the ligand-bound complex is essential for receptor engagement and uptake. Finally, immunizations with FhuD2 alone or FhuD2 formulated with hydroxamate siderophores were equally protective in a murine staphylococcal infection model, confirming the suitability and efficacy of apo-FhuD2 as a protective antigen, and suggesting that other class III SBPs might also be exploited as vaccine candidates.
Collapse
|
48
|
Mishra RPN, Mariotti P, Fiaschi L, Nosari S, Maccari S, Liberatori S, Fontana MR, Pezzicoli A, De Falco MG, Falugi F, Altindis E, Serruto D, Grandi G, Bagnoli F. Staphylococcus aureus FhuD2 is involved in the early phase of staphylococcal dissemination and generates protective immunity in mice. J Infect Dis 2012; 206:1041-9. [PMID: 22829645 DOI: 10.1093/infdis/jis463] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Iron availability plays an essential role in staphylococcal pathogenesis. We selected FhuD2, a lipoprotein involved in iron-hydroxamate uptake, as a novel vaccine candidate against Staphylococcus aureus. Unprecedented for staphylococcal lipoproteins, the protein was demonstrated to have a discrete, punctate localization on the bacterial surface. FhuD2 vaccination generated protective immunity against diverse clinical S. aureus isolates in murine infection models. Protection appeared to be associated with functional antibodies that were shown to mediate opsonophagocytosis, to be effective in passive transfer experiments, and to potentially block FhuD2-mediated siderophore uptake. Furthermore, the protein was found to be up-regulated in infected tissues and was required for staphylococcal dissemination and abscess formation. Herein we show that the staphylococcal iron-hydroxamate uptake system is important in invasive infection and functions as an efficacious vaccine target.
Collapse
|
49
|
Sheldon JR, Heinrichs DE. The iron-regulated staphylococcal lipoproteins. Front Cell Infect Microbiol 2012; 2:41. [PMID: 22919632 PMCID: PMC3417571 DOI: 10.3389/fcimb.2012.00041] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 03/13/2012] [Indexed: 01/01/2023] Open
Abstract
Lipoproteins fulfill diverse roles in antibiotic resistance, adhesion, protein secretion, signaling and sensing, and many also serve as the substrate binding protein (SBP) partner to ABC transporters for the acquisition of a diverse array of nutrients including peptides, sugars, and scarcely abundant metals. In the staphylococci, the iron-regulated SBPs are significantly upregulated during iron starvation and function to sequester and deliver iron into the bacterial cell, enabling staphylococci to circumvent iron restriction imposed by the host environment. Accordingly, this subset of lipoproteins has been implicated in staphylococcal pathogenesis and virulence. Lipoproteins also activate the host innate immune response, triggered through Toll-like receptor-2 (TLR2) and, notably, the iron-regulated subset of lipoproteins are particularly immunogenic. In this review, we discuss the iron-regulated staphylococcal lipoproteins with regard to their biogenesis, substrate specificity, and impact on the host innate immune response.
Collapse
Affiliation(s)
- Jessica R Sheldon
- Department of Microbiology and Immunology, Western University, London ON, Canada
| | | |
Collapse
|
50
|
DeDent A, Kim HK, Missiakas D, Schneewind O. Exploring Staphylococcus aureus pathways to disease for vaccine development. Semin Immunopathol 2012; 34:317-33. [PMID: 22130613 PMCID: PMC3539746 DOI: 10.1007/s00281-011-0299-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Accepted: 10/28/2011] [Indexed: 12/21/2022]
Abstract
Staphylococcus aureus is a commensal of the human skin or nares and a pathogen that frequently causes skin and soft tissue infections as well as bacteremia and sepsis. Recent efforts in understanding the molecular mechanisms of pathogenesis revealed key virulence strategies of S. aureus in host tissues: bacterial scavenging of iron, induction of coagulation pathways to promote staphylococcal agglutination in the vasculature, and suppression of innate and adaptive immune responses. Advances in all three areas have been explored for opportunities in vaccine design in an effort to identify the critical protective antigens of S. aureus. Human clinical trials with specific subunit vaccines have failed, yet provide important insights for the design of future trials that must address the current epidemic of S. aureus infections with drug-resistant isolates (MRSA, methicillin-resistant S. aureus).
Collapse
Affiliation(s)
- Andrea DeDent
- Department of Microbiology, University of Chicago, 920 East 58th Street, Chicago, IL 60637, USA
| | | | | | | |
Collapse
|