1
|
Durand BARN, Daher R, Grenga L, Morsli M, Armengaud J, Lavigne JP, Dunyach-Remy C. Interactions between Helcococcus kunzii and Staphylococcus aureus: How a commensal bacterium modulates the virulence and metabolism of a pathogen in a chronic wound in vitro model. BMC Microbiol 2024; 24:406. [PMID: 39394082 PMCID: PMC11468182 DOI: 10.1186/s12866-024-03520-0] [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: 05/17/2024] [Accepted: 09/13/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Staphylococcus aureus is the predominant pathogen isolated in diabetic foot infections. Recently, the skin commensal bacterium, Helcococcus kunzii, was found to modulate the virulence of this pathogen in an in vivo model. This study aims to elucidate the molecular mechanisms underlying the interaction between these two bacterial species, using a proteomic approach. RESULTS Our results reveal that H. kunzii can coexist and proliferate alongside S. aureus in a Chronic Wound Media (CWM), thereby mimicking an in vitro chronic wound environment. We noted that the secreted proteome of H. kunzii induced a transcriptional effect on S. aureus virulence, resulting in a decrease in the expression level of agrA, a gene involved in quorum sensing. The observed effect could be ascribed to specific proteins secreted by H. kunzii including polysaccharide deacetylase, peptidoglycan DD-metalloendopeptidase, glyceraldehyde-3-phosphate dehydrogenase, trypsin-like peptidase, and an extracellular solute-binding protein. These proteins potentially interact with the agr system, influencing S. aureus virulence. Additionally, the virulence of S. aureus was notably affected by modifications in iron-related pathways and components of cell wall architecture in the presence of H. kunzii. Furthermore, the overall metabolism of S. aureus was reduced when cocultured with H. kunzii. CONCLUSION Future research will focus on elucidating the role of these excreted factors in modulating virulence.
Collapse
Affiliation(s)
- Benjamin A R N Durand
- Department of Microbiology and Hospital Hygiene, CHU Nîmes, VBIC, INSERM U1047, Univ Montpellier, Nîmes, France
| | - Riham Daher
- Department of Microbiology and Hospital Hygiene, CHU Nîmes, VBIC, INSERM U1047, Univ Montpellier, Nîmes, France
| | - Lucia Grenga
- Département Médicaments et Technologies pour la Santé (DMTS), Université Paris-Saclay, CEA, INRAE, Bagnols-sur-Cèze, SPI, France
| | - Madjid Morsli
- Department of Microbiology and Hospital Hygiene, CHU Nîmes, VBIC, INSERM U1047, Univ Montpellier, Nîmes, France
| | - Jean Armengaud
- Département Médicaments et Technologies pour la Santé (DMTS), Université Paris-Saclay, CEA, INRAE, Bagnols-sur-Cèze, SPI, France
| | - Jean-Philippe Lavigne
- Department of Microbiology and Hospital Hygiene, CHU Nîmes, VBIC, INSERM U1047, Univ Montpellier, Nîmes, France
| | - Catherine Dunyach-Remy
- Department of Microbiology and Hospital Hygiene, CHU Nîmes, VBIC, INSERM U1047, Univ Montpellier, Nîmes, France.
| |
Collapse
|
2
|
Schalk IJ. Bacterial siderophores: diversity, uptake pathways and applications. Nat Rev Microbiol 2024:10.1038/s41579-024-01090-6. [PMID: 39251840 DOI: 10.1038/s41579-024-01090-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2024] [Indexed: 09/11/2024]
Abstract
Iron is an essential nutrient for the growth, survival and virulence of almost all bacteria. To access iron, many bacteria produce siderophores, molecules with a high affinity for iron. Research has highlighted substantial diversity in the chemical structure of siderophores produced by bacteria, as well as remarkable variety in the molecular mechanisms involved in strategies for acquiring iron through these molecules. The metal-chelating properties of siderophores, characterized by their high affinity for iron and ability to chelate numerous other metals (albeit with lower affinity compared with iron), have also generated interest in diverse fields. Siderophores find applications in the environment, such as in bioremediation and agriculture, in which emerging and innovative strategies are being developed to address pollution and enhance nutrient availability for plants. Moreover, in medicine, siderophores could be used as a tool for novel antimicrobial therapies and medical imaging, as well as in haemochromatosis, thalassemia or cancer treatments. This Review offers insights into the diversity of siderophores, highlighting their potential applications in environmental and medical contexts.
Collapse
|
3
|
Duarte M, Pedrosa SS, Khusial PR, Madureira AR. Exploring the interplay between stress mediators and skin microbiota in shaping age-related hallmarks: A review. Mech Ageing Dev 2024; 220:111956. [PMID: 38906383 DOI: 10.1016/j.mad.2024.111956] [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: 02/16/2024] [Revised: 05/27/2024] [Accepted: 06/14/2024] [Indexed: 06/23/2024]
Abstract
Psychological stress is a major contributing factor to several health problems (e.g., depression, cardiovascular disease). Around 35 % of the world's population suffers from it, including younger generations. Physiologically, stress manifests through neuroendocrine pathways (Hypothalamic-Pituitary-Adrenal (HPA) axis and Sympathetic-Adrenal-Medullary (SAM) system) which culminate in the production of stress mediators like cortisol, epinephrine and norepinephrine. Stress and its mediators have been associated to body aging, through molecular mechanisms such as telomere attrition, mitochondrial dysfunction, cellular senescence, chronic inflammation, and dysbiosis, among others. Regarding its impact in the skin, stress impacts its structural integrity and physiological function. Despite this review focusing on several hallmarks of aging, emphasis was placed on skin microbiota dysbiosis. In this line, several studies, comprising different age groups, demographic contexts and body sites, have reported skin microbiota alterations associated with aging, and some effects of stress mediators on skin microbiota have also been reviewed in this paper. From a different perspective, since it is not a "traditional" stress mediator, oxytocin, a cortisol antagonist, has been related to glucorticoids inhibition and to display positive effects on cellular aging. This hormone dysregulation has been associated to psychological issues such as depression, whereas its upregulation has been linked to positive social interaction.
Collapse
Affiliation(s)
- Marco Duarte
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto 4169-005, Portugal
| | - Sílvia Santos Pedrosa
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto 4169-005, Portugal
| | - P Raaj Khusial
- Amyris Biotech INC, 5885 Hollis St Ste 100, Emeryville, CA 94608-2405, USA
| | - Ana Raquel Madureira
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto 4169-005, Portugal.
| |
Collapse
|
4
|
Reyes Ruiz VM, Freiberg JA, Weiss A, Green ER, Jobson ME, Felton E, Shaw LN, Chazin WJ, Skaar EP. Coordinated adaptation of Staphylococcus aureus to calprotectin-dependent metal sequestration. mBio 2024; 15:e0138924. [PMID: 38920392 PMCID: PMC11253595 DOI: 10.1128/mbio.01389-24] [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: 05/07/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
The host protein calprotectin inhibits the growth of a variety of bacterial pathogens through metal sequestration in a process known as "nutritional immunity." Staphylococcus aureus growth is inhibited by calprotectin in vitro, and calprotectin is localized in vivo to staphylococcal abscesses during infection. However, the staphylococcal adaptations that provide defense against nutritional immunity and the role of metal-responsive regulators are not fully characterized. In this work, we define the transcriptional response of S. aureus and the role of the metal-responsive regulators, Zur, Fur, and MntR, in response to metal limitation by calprotectin exposure. Additionally, we identified genes affecting the fitness of S. aureus during metal limitation through a Transposon sequencing (Tn-seq) approach. Loss of function mutations in clpP, which encodes a proteolytic subunit of the ATP-dependent Clp protease, demonstrate reduced fitness of S. aureus to the presence of calprotectin. ClpP contributes to pathogenesis in vivo in a calprotectin-dependent manner. These studies establish a critical role for ClpP to combat metal limitation by calprotectin and reveal the genes required for S. aureus to outcompete the host for metals. IMPORTANCE Staphylococcus aureus is a leading cause of skin and soft tissue infections, bloodstream infections, and endocarditis. Antibiotic treatment failures during S. aureus infections are increasingly prevalent, highlighting the need for novel antimicrobial agents. Metal chelator-based therapeutics have tremendous potential as antimicrobials due to the strict requirement for nutrient metals exhibited by bacterial pathogens. The high-affinity transition metal-binding properties of calprotectin represents a potential therapeutic strategy that functions through metal chelation. Our studies provide a foundation to define mechanisms by which S. aureus combats nutritional immunity and may be useful for the development of novel therapeutics to counter the ability of S. aureus to survive in a metal-limited environment.
Collapse
Affiliation(s)
- Valeria M. Reyes Ruiz
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jeffrey A. Freiberg
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Andy Weiss
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Erin R. Green
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Mary-Elizabeth Jobson
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| | - Emily Felton
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| | - Lindsey N. Shaw
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| | - Walter J. Chazin
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Eric P. Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
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
|
Gannesen AV, Schelkunov MI, Ziganshin RH, Ovcharova MA, Sukhacheva MV, Makarova NE, Mart'yanov SV, Loginova NA, Mosolova AM, Diuvenji EV, Nevolina ED, Plakunov VK. Proteomic and transcriptomic analyses of Cutibacterium acnes biofilms and planktonic cultures in presence of epinephrine. AIMS Microbiol 2024; 10:363-390. [PMID: 38919714 PMCID: PMC11194618 DOI: 10.3934/microbiol.2024019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
Transcriptomic and proteomic analysis were performed on 72 h biofilms of the acneic strain Cutibacterium acnes and planktonic cultures in the presence of epinephrine. Epinephrine predominantly downregulated genes associated with various transporter proteins. No correlation was found between proteomic and transcriptomic profiles. In control samples, the expression of 51 proteins differed between planktonic cultures and biofilms. Addition of 5 nM epinephrine reduced this number, and in the presence of 5 µM epinephrine, the difference in proteomic profiles between planktonic cultures and biofilms disappeared. According to the proteomic profiling, epinephrine itself was more effective in the case of C. acnes biofilms and potentially affected the tricarboxylic acid cycle (as well as alpha-ketoglutarate decarboxylase Kgd), biotin synthesis, cell division, and transport of different compounds in C. acnes cells. These findings are consistent with recent research on Micrococcus luteus, suggesting that the effects of epinephrine on actinobacteria may be universal.
Collapse
Affiliation(s)
- AV Gannesen
- Federal Research Centre “Fundamentals of Biotechnology” of Russian Academy of Sciences, Moscow 119071, Russia
| | - MI Schelkunov
- Skolkovo Institute of Science and Technology, Moscow 121205, Russia
- Institute for Information Transmission Problems of Russian Academy of Sciences, Moscow 127051, Russia
| | - RH Ziganshin
- Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - MA Ovcharova
- Federal Research Centre “Fundamentals of Biotechnology” of Russian Academy of Sciences, Moscow 119071, Russia
| | - MV Sukhacheva
- Federal Research Centre “Fundamentals of Biotechnology” of Russian Academy of Sciences, Moscow 119071, Russia
| | - NE Makarova
- Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - SV Mart'yanov
- Federal Research Centre “Fundamentals of Biotechnology” of Russian Academy of Sciences, Moscow 119071, Russia
| | - NA Loginova
- Federal Research Centre “Fundamentals of Biotechnology” of Russian Academy of Sciences, Moscow 119071, Russia
| | - AM Mosolova
- Federal Research Centre “Fundamentals of Biotechnology” of Russian Academy of Sciences, Moscow 119071, Russia
- Russian Biotechnological University, Moscow 125080, Russia
| | - EV Diuvenji
- Federal Research Centre “Fundamentals of Biotechnology” of Russian Academy of Sciences, Moscow 119071, Russia
| | - ED Nevolina
- Federal Research Centre “Fundamentals of Biotechnology” of Russian Academy of Sciences, Moscow 119071, Russia
| | - VK Plakunov
- Federal Research Centre “Fundamentals of Biotechnology” of Russian Academy of Sciences, Moscow 119071, Russia
| |
Collapse
|
7
|
Zhao Y, Bitzer A, Power JJ, Belikova D, Torres Salazar BO, Adolf LA, Gerlach D, Krismer B, Heilbronner S. Nasal commensals reduce Staphylococcus aureus proliferation by restricting siderophore availability. THE ISME JOURNAL 2024; 18:wrae123. [PMID: 38987933 PMCID: PMC11296517 DOI: 10.1093/ismejo/wrae123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/19/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024]
Abstract
The human microbiome is critically associated with human health and disease. One aspect of this is that antibiotic-resistant opportunistic bacterial pathogens, such as methicillin-resistant Staphylococcus aureus, can reside within the nasal microbiota, which increases the risk of infection. Epidemiological studies of the nasal microbiome have revealed positive and negative correlations between non-pathogenic species and S. aureus, but the underlying molecular mechanisms remain poorly understood. The nasal cavity is iron-limited, and bacteria are known to produce iron-scavenging siderophores to proliferate in such environments. Siderophores are public goods that can be consumed by all members of a bacterial community. Accordingly, siderophores are known to mediate bacterial competition and collaboration, but their role in the nasal microbiome is unknown. Here, we show that siderophore acquisition is crucial for S. aureus nasal colonization in vivo. We screened 94 nasal bacterial strains from seven genera for their capacity to produce siderophores as well as to consume the siderophores produced by S. aureus. We found that 80% of the strains engaged in siderophore-mediated interactions with S. aureus. Non-pathogenic corynebacterial species were found to be prominent consumers of S. aureus siderophores. In co-culture experiments, consumption of siderophores by competitors reduced S. aureus growth in an iron-dependent fashion. Our data show a wide network of siderophore-mediated interactions between the species of the human nasal microbiome and provide mechanistic evidence for inter-species competition and collaboration impacting pathogen proliferation. This opens avenues for designing nasal probiotics to displace S. aureus from the nasal cavity of humans.
Collapse
Affiliation(s)
- Yanfeng Zhao
- Department of Infection Biology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076 Tübingen, Germany
- Laboratory Medicine Center, The Second Affiliated Hospital, Nanjing Medical University, 210011 Nanjing, P. R. China
| | - Alina Bitzer
- Department of Infection Biology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence EXC 2124 Controlling Microbes to Fight Infections, 72076 Tübingen, Germany
| | - Jeffrey John Power
- Department of Infection Biology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence EXC 2124 Controlling Microbes to Fight Infections, 72076 Tübingen, Germany
| | - Darya Belikova
- Department of Infection Biology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence EXC 2124 Controlling Microbes to Fight Infections, 72076 Tübingen, Germany
- Interfaculty Institute of Microbiology and Infection Medicine, Institute for Medical Microbiology and Hygiene, UKT Tübingen, 72076 Tübingen, Germany
| | - Benjamin Orlando Torres Salazar
- Department of Infection Biology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence EXC 2124 Controlling Microbes to Fight Infections, 72076 Tübingen, Germany
| | - Lea Antje Adolf
- Department of Infection Biology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence EXC 2124 Controlling Microbes to Fight Infections, 72076 Tübingen, Germany
- Interfaculty Institute of Microbiology and Infection Medicine, Institute for Medical Microbiology and Hygiene, UKT Tübingen, 72076 Tübingen, Germany
| | - David Gerlach
- Ludwig-Maximilians-Universität München, Faculty of Biology, Microbiology, 82152 Martinsried, Germany
| | - Bernhard Krismer
- Department of Infection Biology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076 Tübingen, Germany
- Cluster of Excellence EXC 2124 Controlling Microbes to Fight Infections, 72076 Tübingen, Germany
| | - Simon Heilbronner
- Cluster of Excellence EXC 2124 Controlling Microbes to Fight Infections, 72076 Tübingen, Germany
- Interfaculty Institute of Microbiology and Infection Medicine, Institute for Medical Microbiology and Hygiene, UKT Tübingen, 72076 Tübingen, Germany
- Ludwig-Maximilians-Universität München, Faculty of Biology, Microbiology, 82152 Martinsried, Germany
- German Center for Infection Research “DZIF” partnersite Tübingen, Germany
| |
Collapse
|
8
|
Hsieh RC, Liu R, Burgin DJ, Otto M. Understanding mechanisms of virulence in MRSA: implications for antivirulence treatment strategies. Expert Rev Anti Infect Ther 2023; 21:911-928. [PMID: 37501364 DOI: 10.1080/14787210.2023.2242585] [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: 05/15/2023] [Accepted: 07/26/2023] [Indexed: 07/29/2023]
Abstract
INTRODUCTION Methicillin-resistant Staphylococcus aureus (MRSA) is a widespread pathogen, often causing recurrent and deadly infections in the hospital and community. Many S. aureus virulence factors have been suggested as potential targets for antivirulence therapy to decrease the threat of diminishing antibiotic availability. Antivirulence methods hold promise due to their adjunctive and prophylactic potential and decreased risk for selective pressure. AREAS COVERED This review describes the dominant virulence mechanisms exerted by MRSA and antivirulence therapeutics that are currently undergoing testing in clinical or preclinical stages. We also discuss the advantages and downsides of several investigational antivirulence approaches, including the targeting of bacterial transporters, host-directed therapy, and quorum-sensing inhibitors. For this review, a systematic search of literature on PubMed, Google Scholar, and Web of Science for relevant search terms was performed in April and May 2023. EXPERT OPINION Vaccine and antibody strategies have failed in clinical trials and could benefit from more basic science-informed approaches. Antivirulence-targeting approaches need to be set up better to meet the requirements of drug development, rather than only providing limited results to provide 'proof-of-principle' translational value of pathogenesis research. Nevertheless, there is great potential of such strategies and potential particular promise for novel probiotic approaches.
Collapse
Affiliation(s)
- Roger C Hsieh
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Ryan Liu
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Dylan J Burgin
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
9
|
Venkateswaran P, Vasudevan S, David H, Shaktivel A, Shanmugam K, Neelakantan P, Solomon AP. Revisiting ESKAPE Pathogens: virulence, resistance, and combating strategies focusing on quorum sensing. Front Cell Infect Microbiol 2023; 13:1159798. [PMID: 37457962 PMCID: PMC10339816 DOI: 10.3389/fcimb.2023.1159798] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 06/08/2023] [Indexed: 07/18/2023] Open
Abstract
The human-bacterial association is long-known and well-established in terms of both augmentations of human health and attenuation. However, the growing incidents of nosocomial infections caused by the ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter sp.) call for a much deeper understanding of these organisms. Adopting a holistic approach that includes the science of infection and the recent advancements in preventing and treating infections is imperative in designing novel intervention strategies against ESKAPE pathogens. In this regard, this review captures the ingenious strategies commissioned by these master players, which are teamed up against the defenses of the human team, that are equally, if not more, versatile and potent through an analogy. We have taken a basketball match as our analogy, dividing the human and bacterial species into two teams playing with the ball of health. Through this analogy, we make the concept of infectious biology more accessible.
Collapse
Affiliation(s)
- Parvathy Venkateswaran
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Sahana Vasudevan
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Helma David
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Adityan Shaktivel
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Karthik Shanmugam
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Prasanna Neelakantan
- Division of Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| |
Collapse
|
10
|
Murdoch CC, Skaar EP. Nutritional immunity: the battle for nutrient metals at the host-pathogen interface. Nat Rev Microbiol 2022; 20:657-670. [PMID: 35641670 PMCID: PMC9153222 DOI: 10.1038/s41579-022-00745-6] [Citation(s) in RCA: 183] [Impact Index Per Article: 91.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2022] [Indexed: 12/21/2022]
Abstract
Trace metals are essential micronutrients required for survival across all kingdoms of life. From bacteria to animals, metals have critical roles as both structural and catalytic cofactors for an estimated third of the proteome, representing a major contributor to the maintenance of cellular homeostasis. The reactivity of metal ions engenders them with the ability to promote enzyme catalysis and stabilize reaction intermediates. However, these properties render metals toxic at high concentrations and, therefore, metal levels must be tightly regulated. Having evolved in close association with bacteria, vertebrate hosts have developed numerous strategies of metal limitation and intoxication that prevent bacterial proliferation, a process termed nutritional immunity. In turn, bacterial pathogens have evolved adaptive mechanisms to survive in conditions of metal depletion or excess. In this Review, we discuss mechanisms by which nutrient metals shape the interactions between bacterial pathogens and animal hosts. We explore the cell-specific and tissue-specific roles of distinct trace metals in shaping bacterial infections, as well as implications for future research and new therapeutic development.
Collapse
Affiliation(s)
- Caitlin C Murdoch
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Eric P Skaar
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
11
|
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: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [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
|
12
|
Zhang Y, Martin JE, Edmonds KA, Winkler ME, Giedroc DP. SifR is an Rrf2-family quinone sensor associated with catechol iron uptake in Streptococcus pneumoniae D39. J Biol Chem 2022; 298:102046. [PMID: 35597283 PMCID: PMC9218516 DOI: 10.1016/j.jbc.2022.102046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/29/2022] [Accepted: 05/03/2022] [Indexed: 01/15/2023] Open
Abstract
Streptococcus pneumoniae (pneumococcus) is a Gram-positive commensal and human respiratory pathogen. How this bacterium satisfies its nutritional iron (Fe) requirement in the context of endogenously produced hydrogen peroxide is not well understood. Here, we characterize a novel virulence-associated Rrf2-family transcriptional repressor that we term SifR (streptococcal IscR-like family transcriptional repressor) encoded by spd_1448 and conserved in Streptococci. Global transcriptomic analysis of a ΔsifR strain defines the SifR regulon as genes encoding a candidate catechol dioxygenase CatE, an uncharacterized oxidoreductase YwnB, a candidate flavin-dependent ferric reductase YhdA, a candidate heme-based ferric reductase domain-containing protein and the Piu (pneumococcus iron uptake) Fe transporter (piuBCDA). Previous work established that membrane-anchored PiuA binds FeIII-bis-catechol or monocatechol complexes with high affinity, including the human catecholamine stress hormone, norepinephrine. We demonstrate that SifR senses quinone via a single conserved cysteine that represses its regulon when in the reduced form. Upon reaction with catechol-derived quinones, we show that SifR dissociates from the DNA leading to regulon derepression, allowing the pneumococcus to access a catechol-derived source of Fe while minimizing reactive electrophile stress induced by quinones. Consistent with this model, we show that CatE is an FeII-dependent 2,3-catechol dioxygenase with broad substrate specificity, YwnB is an NAD(P)H-dependent quinone reductase capable of reducing the oxidized and cyclized norepinephrine, adrenochrome, and YhdA is capable of reducing a number of FeIII complexes, including PiuA-binding transport substrates. These findings are consistent with a model where FeIII-catechol complexes serve as significant nutritional Fe sources in the host.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA; Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, Indiana, USA
| | - Julia E Martin
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA; Department of Biological Sciences, Idaho State University, Pocatello, Idaho, USA
| | | | - Malcolm E Winkler
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, Indiana, USA; Department of Biology, Indiana University, Bloomington, Indiana, USA
| | - David P Giedroc
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA; Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, Indiana, USA.
| |
Collapse
|
13
|
In vivo growth of Staphylococcus lugdunensis is facilitated by the concerted function of heme and non-heme iron acquisition mechanisms. J Biol Chem 2022; 298:101823. [PMID: 35283192 PMCID: PMC9052147 DOI: 10.1016/j.jbc.2022.101823] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/04/2022] [Accepted: 03/07/2022] [Indexed: 12/23/2022] Open
Abstract
Staphylococcus lugdunensis has increasingly been recognized as a pathogen that can cause serious infection indicating this bacterium overcomes host nutritional immunity. Despite this, there exists a significant knowledge gap regarding the iron acquisition mechanisms employed by S. lugdunensis, especially during infection of the mammalian host. Here we show that S. lugdunensis can usurp hydroxamate siderophores and staphyloferrin A and B from Staphylococcus aureus. These transport activities all required a functional FhuC ATPase. Moreover, we show that the acquisition of catechol siderophores and catecholamine stress hormones by S. lugdunensis required the presence of the sst-1 transporter-encoding locus, but not the sst-2 locus. Iron-dependent growth in acidic culture conditions necessitated the ferrous iron transport system encoded by feoAB. Heme iron was acquired via expression of the iron-regulated surface determinant (isd) locus. During systemic infection of mice, we demonstrated that while S. lugdunensis does not cause overt illness, it does colonize and proliferate to high numbers in the kidneys. By combining mutations in the various iron acquisition loci (isd, fhuC, sst-1, and feo), we demonstrate that only a strain deficient for all of these systems was attenuated in its ability to proliferate to high numbers in the murine kidney. We propose the concerted action of heme and non-heme iron acquisition systems also enable S. lugdunensis to cause human infection.
Collapse
|
14
|
Naclerio GA, Onyedibe KI, Karanja CW, Aryal UK, Sintim HO. Comparative Studies to Uncover Mechanisms of Action of N-(1,3,4-Oxadiazol-2-yl)benzamide Containing Antibacterial Agents. ACS Infect Dis 2022; 8:865-877. [PMID: 35297603 PMCID: PMC9188027 DOI: 10.1021/acsinfecdis.1c00613] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Drug-resistant bacterial pathogens still cause high levels of mortality annually despite the availability of many antibiotics. Methicillin-resistant Staphylococcus aureus (MRSA) is especially problematic, and the rise in resistance to front-line treatments like vancomycin and linezolid calls for new chemical modalities to treat chronic and relapsing MRSA infections. Halogenated N-(1,3,4-oxadiazol-2-yl)benzamides are an interesting class of antimicrobial agents, which have been described by multiple groups to be effective against different bacterial pathogens. The modes of action of a few N-(1,3,4-oxadiazol-2-yl)benzamides have been elucidated. For example, oxadiazoles KKL-35 and MBX-4132 have been described as inhibitors of trans-translation (a ribosome rescue pathway), while HSGN-94 was shown to inhibit lipoteichoic acid (LTA). However, other similarly halogenated N-(1,3,4-oxadiazol-2-yl)benzamides neither inhibit trans-translation nor LTA biosynthesis but are potent antimicrobial agents. For example, HSGN-220, -218, and -144 are N-(1,3,4-oxadiazol-2-yl)benzamides that are modified with OCF3, SCF3, or SF5 and have remarkable minimum inhibitory concentrations ranging from 1 to 0.06 μg/mL against MRSA clinical isolates and show a low propensity to develop resistance to MRSA over 30 days. The mechanism of action of these highly potent oxadiazoles is however unknown. To provide insights into how these halogenated N-(1,3,4-oxadiazol-2-yl)benzamides inhibit bacterial growth, we performed global proteomics and RNA expression analysis of some essential genes of S. aureus treated with HSGN-220, -218, and -144. These studies revealed that the oxadiazoles HSGN-220, -218, and -144 are multitargeting antibiotics that regulate menaquinone biosynthesis and other essential proteins like DnaX, Pol IIIC, BirA, LexA, and DnaC. In addition, these halogenated N-(1,3,4-oxadiazol-2-yl)benzamides were able to depolarize bacterial membranes and regulate siderophore biosynthesis and heme regulation. Iron starvation appears to be part of the mechanism of action that led to bacterial killing. This study demonstrates that N-(1,3,4-oxadiazol-2-yl)benzamides are indeed privileged scaffolds for the development of antibacterial agents and that subtle modifications lead to changes to the mechanism of action.
Collapse
Affiliation(s)
- George A. Naclerio
- Chemistry Department, Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Kenneth I. Onyedibe
- Chemistry Department, Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue Institute for Inflammation, Immunology, and Infectious Diseases, West Lafayette, Indiana 47907, United States
| | - Caroline W. Karanja
- Chemistry Department, Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Uma K. Aryal
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, Indiana 47907, United States
| | - Herman O. Sintim
- Chemistry Department, Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue Institute for Inflammation, Immunology, and Infectious Diseases, West Lafayette, Indiana 47907, United States
| |
Collapse
|
15
|
Oliveira F, Lima T, Correia A, Silva AM, Soares C, Morais S, Weißelberg S, Vilanova M, Rohde H, Cerca N. Involvement of the Iron-Regulated Loci hts and fhuC in Biofilm Formation and Survival of Staphylococcus epidermidis within the Host. Microbiol Spectr 2022; 10:e0216821. [PMID: 35019768 PMCID: PMC8754135 DOI: 10.1128/spectrum.02168-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/05/2021] [Indexed: 12/19/2022] Open
Abstract
Staphylococcus epidermidis is a major nosocomial pathogen with a remarkable ability to persist on indwelling medical devices through biofilm formation. Nevertheless, it remains intriguing how this process is efficiently achieved under the host's harsh conditions, where the availability of nutrients, such as essential metals, is scarce. Following our previous identification of two iron-regulated loci putatively involved in iron transport, hts and fhuC, we assessed here their individual contribution to both bacterial physiology and interaction with host immune cells. Single deletions of the hts and fhuC loci led to marked changes in the cell iron content, which were partly detrimental for planktonic growth and strongly affected biofilm formation under iron-restricted conditions. Deletion of each of these two loci did not lead to major changes in S. epidermidis survival within human macrophages or in an ex vivo human blood model of bloodstream infection. However, the lack of either hts or fhuC loci significantly impaired bacterial survival in vivo in a murine model of bacteremia. Collectively, this study establishes, for the first time, the pivotal role of the iron-regulated loci hts and fhuC in S. epidermidis biofilm formation and survival within the host, providing relevant information for the development of new targeted therapeutics against this pathogen. IMPORTANCE Staphylococcus epidermidis is one of the most important nosocomial pathogens and a major cause of central line-associated bloodstream infections. Once in the bloodstream, this bacterium must surpass severe iron restriction in order to survive and establish infection. Surprisingly, very little is known about the iron acquisition mechanisms in this species. This study represents the first report on the involvement of the S. epidermidis iron-regulated loci hts and fhuC in biofilm formation under host relevant conditions and, most importantly, in survival within the host. Ultimately, these findings highlight iron acquisition and these loci in particular, as potential targets for future therapeutic strategies against biofilm-associated S. epidermidis infections.
Collapse
Affiliation(s)
- Fernando Oliveira
- Centre of Biological Engineering, LIBRO – Laboratory of Research in Biofilms Rosário Oliveira, University of Minho, Braga, Portugal
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Tânia Lima
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Alexandra Correia
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Ana Margarida Silva
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Cristina Soares
- REQUIMTE-LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Porto, Portugal
| | - Simone Morais
- REQUIMTE-LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Porto, Portugal
| | - Samira Weißelberg
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel Vilanova
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS-UP, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Holger Rohde
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Nuno Cerca
- Centre of Biological Engineering, LIBRO – Laboratory of Research in Biofilms Rosário Oliveira, University of Minho, Braga, Portugal
| |
Collapse
|
16
|
Maldonado-Hernández J, Román-Ponce B, Arroyo-Herrera I, Guevara-Luna J, Ramos-Garza J, Embarcadero-Jiménez S, Estrada de Los Santos P, Wang ET, Vásquez-Murrieta MS. Metallophores production by bacteria isolated from heavy metal-contaminated soil and sediment at Lerma-Chapala Basin. Arch Microbiol 2022; 204:180. [PMID: 35175407 DOI: 10.1007/s00203-022-02780-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 12/14/2021] [Accepted: 01/28/2022] [Indexed: 11/02/2022]
Abstract
Environmental pollution as a result of heavy metals (HMs) is a worldwide problem and the implementation of eco-friendly remediation technologies is thus required. Metallophores, low molecular weight compounds, could have important biotechnological applications in the fields of agriculture, medicine, and bioremediation. This study aimed to isolate HM-resistant bacteria from soils and sediments of the Lerma-Chapala Basin and evaluated their abilities to produce metallophores and to promote plant growth. Bacteria from the Lerma-Chapala Basin produced metallophores for all the tested metal ions, presented a greater production of As3+ metallophores, and showed high HM resistance especially to Zn2+, As5+, and Ni2+. A total of 320 bacteria were isolated with 170 strains showing siderophores synthesis. Members of the Delftia and Pseudomonas genera showed above 92 percent siderophore units (psu) during siderophores production and hydroxamate proved to be the most common functional group among the analyzed siderophores. Our results provided evidence that Lerma-Chapala Basin bacteria and their metallophores could potentially be employed in bioremediation processes or may even have potential for applications in other biotechnological fields.
Collapse
Affiliation(s)
- Jessica Maldonado-Hernández
- Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Prolongación Carpio y Plan de Ayala S/N, Col. Santo Tomás, Alcaldía Miguel Hidalgo, C.P. 11340, Mexico City, Mexico.,Universidad del Valle de México, Campus Chapultepec, Laboratorio 314, Observatorio No. 400, Col. 16 de Septiembre, Del. Miguel Hidalgo, C.P. 11810, Mexico City, Mexico
| | - Brenda Román-Ponce
- Universidad Politécnica del Estado de Morelos, Boulevard Cuauhnáhuac 556, Lomas del Texcal, 62550, Jiutepec, Morelos, Mexico
| | - Ivan Arroyo-Herrera
- Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Prolongación Carpio y Plan de Ayala S/N, Col. Santo Tomás, Alcaldía Miguel Hidalgo, C.P. 11340, Mexico City, Mexico
| | - Joseph Guevara-Luna
- Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Prolongación Carpio y Plan de Ayala S/N, Col. Santo Tomás, Alcaldía Miguel Hidalgo, C.P. 11340, Mexico City, Mexico
| | - Juan Ramos-Garza
- Universidad del Valle de México, Campus Chapultepec, Laboratorio 314, Observatorio No. 400, Col. 16 de Septiembre, Del. Miguel Hidalgo, C.P. 11810, Mexico City, Mexico
| | - Salvador Embarcadero-Jiménez
- Instituto Mexicano del Petróleo, Eje Central Lázaro Cárdenas 152, Col. San Bartolo Atepehuacan, 07730, Mexico City, Mexico
| | - Paulina Estrada de Los Santos
- Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Prolongación Carpio y Plan de Ayala S/N, Col. Santo Tomás, Alcaldía Miguel Hidalgo, C.P. 11340, Mexico City, Mexico
| | - En Tao Wang
- Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Prolongación Carpio y Plan de Ayala S/N, Col. Santo Tomás, Alcaldía Miguel Hidalgo, C.P. 11340, Mexico City, Mexico
| | - María Soledad Vásquez-Murrieta
- Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Prolongación Carpio y Plan de Ayala S/N, Col. Santo Tomás, Alcaldía Miguel Hidalgo, C.P. 11340, Mexico City, Mexico.
| |
Collapse
|
17
|
Mular A, Shanzer A, Kozłowski H, Hubmann I, Misslinger M, Krzywik J, Decristoforo C, Gumienna-Kontecka E. Cyclic Analogs of Desferrioxamine E Siderophore for 68Ga Nuclear Imaging: Coordination Chemistry and Biological Activity in Staphylococcus aureus. Inorg Chem 2021; 60:17846-17857. [PMID: 34783539 PMCID: PMC8653149 DOI: 10.1021/acs.inorgchem.1c02453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
As multidrug-resistant
bacteria are an emerging problem and threat
to humanity, novel strategies for treatment and diagnostics are actively
sought. We aim to utilize siderophores, iron-specific strong chelating
agents produced by microbes, as gallium ion carriers for diagnosis,
applying that Fe(III) can be successfully replaced by Ga(III) without
losing biological properties of the investigated complex, which allows
molecular imaging by positron emission tomography (PET). Here, we
report synthesis, full solution chemistry, thermodynamic characterization,
and the preliminary biological evaluation of biomimetic derivatives
(FOX) of desferrioxamine E (FOXE) siderophore, radiolabeled with 68Ga for possible applications in PET imaging of S.
aureus. From a series of six biomimetic analogs, which differ
from FOXE with cycle length and position of hydroxamic and amide groups,
the highest Fe(III) and Ga(III) stability was determined for the most
FOXE alike compounds–FOX 2-4 and FOX 2-5; we have also established
the stability constant of the Ga-FOXE complex. For this purpose, spectroscopic
and potentiometric titrations, together with the Fe(III)–Ga(III)
competition method, were used. [68Ga]Ga-FOXE derivatives
uptake and microbial growth promotion studies conducted on S. aureus were efficient for compounds with a larger cavity,
i.e., FOX 2-5, 2-6, and 3-5. Even though showing low uptake values,
Fe-FOX 2-4 seems to be also a good Fe-source to support the growth
of S. aureus. Overall, proposed derivatives may hold
potential as inert and stable carrier agents for radioactive Ga(III)
ions for diagnostic medical applications or interesting starting compounds
for further modifications. In this work,
the authors have investigated a set of novel
ferrioxamine E analogs as potential Ga-68 chelators and tools for
infection imaging.
Collapse
Affiliation(s)
- Andrzej Mular
- Faculty of Chemistry, University of Wrocław, 50-383 Wrocław, Poland
| | - Abraham Shanzer
- Department of Organic Chemistry, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Henryk Kozłowski
- Faculty of Chemistry, University of Wrocław, 50-383 Wrocław, Poland.,Department of Health Sciences, University of Opole, 45-060 Opole, Poland
| | - Isabella Hubmann
- Department of Nuclear Medicine, Medical University Innsbruck, A-6020 Innsbruck, Austria
| | - Matthias Misslinger
- Institute of Molecular Biology, Medical University Innsbruck, A-6020 Innsbruck, Austria
| | - Julia Krzywik
- TriMen Chemicals, Piłsudskiego 141, 92-318 Łódź, Poland
| | - Clemens Decristoforo
- Department of Nuclear Medicine, Medical University Innsbruck, A-6020 Innsbruck, Austria
| | | |
Collapse
|
18
|
Oliveira F, Rohde H, Vilanova M, Cerca N. Fighting Staphylococcus epidermidis Biofilm-Associated Infections: Can Iron Be the Key to Success? Front Cell Infect Microbiol 2021; 11:798563. [PMID: 34917520 PMCID: PMC8670311 DOI: 10.3389/fcimb.2021.798563] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 11/11/2021] [Indexed: 12/29/2022] Open
Abstract
Staphylococcus epidermidis is one of the most important commensal microorganisms of human skin and mucosae. However, this bacterial species is also the cause of severe infections in immunocompromised patients, specially associated with the utilization of indwelling medical devices, that often serve as a scaffold for biofilm formation. S. epidermidis strains are often multidrug resistant and its association with biofilm formation makes these infections hard to treat. Their remarkable ability to form biofilms is widely regarded as its major pathogenic determinant. Although a significant amount of knowledge on its biofilm formation mechanisms has been achieved, we still do not understand how the species survives when exposed to the host harsh environment during invasion. A previous RNA-seq study highlighted that iron-metabolism associated genes were the most up-regulated bacterial genes upon contact with human blood, which suggested that iron acquisition plays an important role in S. epidermidis biofilm development and escape from the host innate immune system. In this perspective article, we review the available literature on the role of iron metabolism on S. epidermidis pathogenesis and propose that exploiting its dependence on iron could be pursued as a viable therapeutic alternative.
Collapse
Affiliation(s)
- Fernando Oliveira
- Centre of Biological Engineering, Laboratory of Research in Biofilms Rosário Oliveira (LIBRO), University of Minho, Braga, Portugal
| | - Holger Rohde
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel Vilanova
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto (ICBAS-UP), Porto, Portugal
| | - Nuno Cerca
- Centre of Biological Engineering, Laboratory of Research in Biofilms Rosário Oliveira (LIBRO), University of Minho, Braga, Portugal
| |
Collapse
|
19
|
Heme-Dependent Siderophore Utilization Promotes Iron-Restricted Growth of the Staphylococcus aureus hemB Small-Colony Variant. J Bacteriol 2021; 203:e0045821. [PMID: 34606375 DOI: 10.1128/jb.00458-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Respiration-deficient Staphylococcus aureus small-colony variants (SCVs) frequently cause persistent infections, which necessitates they acquire iron, yet how SCVs obtain iron remains unknown. To address this, we created a stable hemB mutant from S. aureus USA300 strain LAC. The hemB SCV utilized exogenously supplied hemin but was attenuated for growth under conditions of iron starvation. Transcriptome sequencing (RNA-seq) showed that both wild-type (WT) S. aureus and the hemB mutant sense and respond to iron starvation; however, growth assays show that the hemB mutant is defective for siderophore-mediated iron acquisition. Indeed, the hemB SCV demonstrated limited utilization of endogenous staphyloferrin B or exogenously provided staphyloferrin A, deferoxamine mesylate (Desferal), and epinephrine. Direct measurement of intracellular ATP in hemB and WT S. aureus revealed that both strains can generate comparable levels of ATP during exponential growth, suggesting defects in ATP production cannot account for the inability to efficiently utilize siderophores. Defective siderophore utilization by hemB bacteria was also evident in vivo, as administration of Desferal failed to promote hemB bacterial growth in every organ analyzed except for the kidneys. In support of the hypothesis that S. aureus accesses heme in kidney abscesses, in vitro analyses revealed that increased hemin availability enables hemB bacteria to utilize siderophores for growth when iron availability is restricted. Taken together, our data support the conclusion that hemin is used not only as an iron source itself but also as a nutrient that promotes utilization of siderophore-iron complexes. IMPORTANCE S. aureus small-colony variants (SCVs) are associated with chronic recurrent infection and worsened clinical outcome. SCVs persist within the host despite administration of antibiotics. This study yields insight into how S. aureus SCVs acquire iron, which during infection of a host is a difficult-to-acquire metal nutrient. Under hemin-limited conditions, hemB S. aureus is impaired for siderophore-dependent growth, and in agreement, murine infection indicates that hemin-deficient SCVs meet their nutritional requirement for iron through utilization of hemin. Importantly, we demonstrate that hemB SCVs rely upon hemin as a nutrient to promote siderophore utilization. Therefore, perturbation of heme biosynthesis and/or utilization represents a viable to strategy to mitigate the ability of SCV bacteria to acquire siderophore-bound iron during infection.
Collapse
|
20
|
Monteith AJ, Skaar EP. The impact of metal availability on immune function during infection. Trends Endocrinol Metab 2021; 32:916-928. [PMID: 34483037 PMCID: PMC8516721 DOI: 10.1016/j.tem.2021.08.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 12/16/2022]
Abstract
Nutrient transition metals are required cofactors for many proteins to perform functions necessary for life. As such, the concentration of nutrient metals is carefully maintained to retain critical biological processes while limiting toxicity. During infection, invading bacterial pathogens must acquire essential metals, such as zinc, manganese, iron, and copper, from the host to colonize and cause disease. To combat this, the host exploits the essentiality and toxicity of nutrient metals by producing factors that limit metal availability, thereby starving pathogens or accumulating metals in excess to intoxicate the pathogen in a process termed 'nutritional immunity'. As a result of inflammation, a heterogeneous environment containing both metal-replete and -deplete niches is created, in which nutrient metal availability may have an underappreciated role in regulating immune cell function during infection. How the host manipulates nutrient metal availability during infection, and the downstream effects that nutrient metals and metal-sequestering proteins have on immune cell function, are discussed in this review.
Collapse
Affiliation(s)
- Andrew J Monteith
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eric P Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Institute for Infection, Immunology, & Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
21
|
Ribeiro M, Sousa CA, Simões M. Harnessing microbial iron chelators to develop innovative therapeutic agents. J Adv Res 2021; 39:89-101. [PMID: 35777919 PMCID: PMC9263657 DOI: 10.1016/j.jare.2021.10.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/24/2021] [Accepted: 10/27/2021] [Indexed: 01/19/2023] Open
Abstract
Microbial iron chelators as a new route to develop inspiring antimicrobials. Siderophore-mimicking antibiotics as a pathogen-targeted strategy. Effectiveness of iron chelators on antibiotic-resistant Gram-negative bacteria. Iron chelators and the treatment of iron overload diseases. Iron chelators as powerful tools for cancer therapy.
Background Aim of Review Key Scientific Concepts of Review
Collapse
|
22
|
Barber CC, Zhang W. Small molecule natural products in human nasal/oral microbiota. J Ind Microbiol Biotechnol 2021; 48:6129854. [PMID: 33945611 PMCID: PMC8210680 DOI: 10.1093/jimb/kuab010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/07/2020] [Indexed: 12/26/2022]
Abstract
Small molecule natural products are a chemically diverse class of biomolecules that fulfill myriad biological functions, including autoregulation, communication with microbial neighbors and the host, interference competition, nutrient acquisition, and resistance to oxidative stress. Human commensal bacteria are increasingly recognized as a potential source of new natural products, which may provide insight into the molecular ecology of many different human body sites as well as novel scaffolds for therapeutic development. Here, we review the scientific literature on natural products derived from residents of the human nasal/oral cavity, discuss their discovery, biosynthesis, and ecological roles, and identify key questions in the study of these compounds.
Collapse
Affiliation(s)
- Colin Charles Barber
- Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley 94720, USA
| | - Wenjun Zhang
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley 94720, USA.,Chan-Zuckerberg Biohub, San Francisco 94158, USA
| |
Collapse
|
23
|
Aubourg M, Gravey F, Dhalluin A, Giard JC. Identification of the iron-limitation stimulon in Staphylococcus lugdunensis. Arch Microbiol 2021; 203:3687-3694. [PMID: 33983488 DOI: 10.1007/s00203-021-02342-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 10/21/2022]
Abstract
During the infectious process, pathogens such as Staphylococcus lugdunensis have to cope with the condition of host-induced iron-limitation. Using the RNAseq approach, we performed the first global transcriptomic analysis of S. lugdunensis cells incubated in the absence and presence of iron chelator. One hundred and seventy-five genes were identified as members of the iron-limitation stimulon (127 up- and 48 downregulated). Six gene clusters known or likely required for the acquisition of iron have been identified. Among them, a novel Energy-Coupling Factor type transporter (ECF), homologous to the lhaSTA operon, has been found into a 13-gene putative operon and strongly overexpressed under iron-limitation condition. Moreover, the transcription of genes involved in resistance to oxidative stress (including catalase), virulence, transcriptional regulation, and hemin detoxification were also modified. These data provide some answers on the cellular response to the iron-limitation stress that is important for the opportunistic behavior of this pathogen.
Collapse
Affiliation(s)
- Marion Aubourg
- EA4655 U2RM (équipe "Antibio-résistance"), CHU de Caen, Université de Caen Normandie, Caen, France
| | - François Gravey
- Groupe de Recherche sur l'Adaptation Microbienne (GRAM 2.0), Normandie Univ, Unicaen, Unirouen, GRAM 2.0, 14000, Caen, France
| | - Anne Dhalluin
- EA4655 U2RM (équipe "Antibio-résistance"), CHU de Caen, Université de Caen Normandie, Caen, France
| | - Jean-Christophe Giard
- EA4655 U2RM (équipe "Antibio-résistance"), CHU de Caen, Université de Caen Normandie, Caen, France.
| |
Collapse
|
24
|
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: 6.7] [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
|
25
|
Samsonov SA, Zsila F, Maszota-Zieleniak M. Acute phase α 1-acid glycoprotein as a siderophore-capturing component of the human plasma: A molecular modeling study. J Mol Graph Model 2021; 105:107861. [PMID: 33640788 DOI: 10.1016/j.jmgm.2021.107861] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 11/26/2022]
Abstract
Siderophores are ferric ion-specific organic compounds that are used by bacteria and fungi to secure their iron supply when infecting target organisms. There are a few proteins in the human body, named siderocalins, which bind these important virulence factors and so starve microorganisms of iron. In this study, we analyzed in silico if serum α1-acid glycoprotein (AAG), the major acute phase lipocalin component of the human plasma, could functionally belong to this group. The real biological function of AAG is elusive and its concentration substantially increases in response to pathological stimuli, including bacterial infections. We computationally evaluated the potential binding of nine microbial siderophores into the β-barrel cavity of AAG and compared the results with the corresponding experimental data reported for siderophore-neutrophil gelatinase-associated lipocalin complexes. According to the results, petrobactin and Fe-BisHaCam are putative candidates to be recognized by this protein. It is proposed that AAG may function as a siderophore capturing component of the innate immune system being able to neutralize bacterial iron chelators not recognized by other siderocalins.
Collapse
Affiliation(s)
- Sergey A Samsonov
- Faculty of Chemistry, University of Gdańsk, ul. Wita Stwosza 63, 80-308, Gdańsk, Poland
| | - Ferenc Zsila
- Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, H-1117 Budapest, Magyar tudósok körútja 2, Hungary.
| | | |
Collapse
|
26
|
Pidwill GR, Gibson JF, Cole J, Renshaw SA, Foster SJ. The Role of Macrophages in Staphylococcus aureus Infection. Front Immunol 2021; 11:620339. [PMID: 33542723 PMCID: PMC7850989 DOI: 10.3389/fimmu.2020.620339] [Citation(s) in RCA: 144] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/02/2020] [Indexed: 12/23/2022] Open
Abstract
Staphylococcus aureus is a member of the human commensal microflora that exists, apparently benignly, at multiple sites on the host. However, as an opportunist pathogen it can also cause a range of serious diseases. This requires an ability to circumvent the innate immune system to establish an infection. Professional phagocytes, primarily macrophages and neutrophils, are key innate immune cells which interact with S. aureus, acting as gatekeepers to contain and resolve infection. Recent studies have highlighted the important roles of macrophages during S. aureus infections, using a wide array of killing mechanisms. In defense, S. aureus has evolved multiple strategies to survive within, manipulate and escape from macrophages, allowing them to not only subvert but also exploit this key element of our immune system. Macrophage-S. aureus interactions are multifaceted and have direct roles in infection outcome. In depth understanding of these host-pathogen interactions may be useful for future therapeutic developments. This review examines macrophage interactions with S. aureus throughout all stages of infection, with special emphasis on mechanisms that determine infection outcome.
Collapse
Affiliation(s)
- Grace R. Pidwill
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
| | - Josie F. Gibson
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Joby Cole
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Stephen A. Renshaw
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Simon J. Foster
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
27
|
Tahernia M, Plotkin-Kaye E, Mohammadifar M, Gao Y, Oefelein MR, Cook LC, Choi S. Characterization of Electrogenic Gut Bacteria. ACS OMEGA 2020; 5:29439-29446. [PMID: 33225175 PMCID: PMC7676329 DOI: 10.1021/acsomega.0c04362] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 10/29/2020] [Indexed: 05/13/2023]
Abstract
While electrogenic, or electricity-producing, Gram-negative bacteria predominantly found in anaerobic habitats have been intensively explored, the potential of Gram-positive microbial electrogenic capability residing in a similar anoxic environment has not been considered. Because Gram-positive bacteria contain a thick non-conductive cell wall, they were previously believed to be very weak exoelectrogens. However, with the recent discovery of electrogenicity by Gram-positive pathogens and elucidation of their electron-transfer pathways, significant and accelerated attention has been given to the discovery and characterization of these pathways in the members of gut microbiota. The discovery of electrogenic bacteria present in the human gut and the understanding of their electrogenic capacity opens up possibilities of bacterial powered implantable batteries and provide a novel biosensing platform to monitor human gastrointestinal health. In this work, we characterized microbial extracellular electron-transfer capabilities and capacities of five gut bacteria: Staphylococcus aureus, Enterococcus faecalis, Streptococcus agalactiae, Lactobacillus reuteri, and Lactobacillus rhamnosus. A 21-well paper-based microbial fuel cell array with enhanced sensitivity was developed as a powerful yet simple screening method to accurately and simultaneously characterize bacterial electrogenicity. S. aureus, E. faecalis, and S. agalactiae exhibited distinct electrogenic capabilities, and their power generations were comparable to that of the well-known Gram-negative exoelectrogen, Shewanella oneidensis. Importantly, this system was used to begin a large-scale transposon screen to examine the genes involved in electrogenicity by the human pathobiont S. aureus.
Collapse
Affiliation(s)
- Mehdi Tahernia
- Bioelectronics
& Microsystems Laboratory, Department of Electrical & Computer
Engineering, State University of New York-Binghamton, Binghamton, New York 13902-6000, United States
| | - Ellie Plotkin-Kaye
- Department
of Biological Science, State University
of New York-Binghamton, Binghamton, New York 13902-6000, United States
| | - Maedeh Mohammadifar
- Bioelectronics
& Microsystems Laboratory, Department of Electrical & Computer
Engineering, State University of New York-Binghamton, Binghamton, New York 13902-6000, United States
| | - Yang Gao
- Bioelectronics
& Microsystems Laboratory, Department of Electrical & Computer
Engineering, State University of New York-Binghamton, Binghamton, New York 13902-6000, United States
| | - Melissa R. Oefelein
- Department
of Biological Science, State University
of New York-Binghamton, Binghamton, New York 13902-6000, United States
| | - Laura C. Cook
- Department
of Biological Science, State University
of New York-Binghamton, Binghamton, New York 13902-6000, United States
| | - Seokheun Choi
- Bioelectronics
& Microsystems Laboratory, Department of Electrical & Computer
Engineering, State University of New York-Binghamton, Binghamton, New York 13902-6000, United States
| |
Collapse
|
28
|
How Severe Anaemia Might Influence the Risk of Invasive Bacterial Infections in African Children. Int J Mol Sci 2020; 21:ijms21186976. [PMID: 32972031 PMCID: PMC7555399 DOI: 10.3390/ijms21186976] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/04/2020] [Accepted: 09/15/2020] [Indexed: 12/21/2022] Open
Abstract
Severe anaemia and invasive bacterial infections are common causes of childhood sickness and death in sub-Saharan Africa. Accumulating evidence suggests that severely anaemic African children may have a higher risk of invasive bacterial infections. However, the mechanisms underlying this association remain poorly described. Severe anaemia is characterized by increased haemolysis, erythropoietic drive, gut permeability, and disruption of immune regulatory systems. These pathways are associated with dysregulation of iron homeostasis, including the downregulation of the hepatic hormone hepcidin. Increased haemolysis and low hepcidin levels potentially increase plasma, tissue and intracellular iron levels. Pathogenic bacteria require iron and/or haem to proliferate and have evolved numerous strategies to acquire labile and protein-bound iron/haem. In this review, we discuss how severe anaemia may mediate the risk of invasive bacterial infections through dysregulation of hepcidin and/or iron homeostasis, and potential studies that could be conducted to test this hypothesis.
Collapse
|
29
|
Zhang Y, Edmonds KA, Raines DJ, Murphy BA, Wu H, Guo C, Nolan EM, VanNieuwenhze MS, Duhme-Klair AK, Giedroc DP. The Pneumococcal Iron Uptake Protein A (PiuA) Specifically Recognizes Tetradentate Fe IIIbis- and Mono-Catechol Complexes. J Mol Biol 2020; 432:5390-5410. [PMID: 32795535 DOI: 10.1016/j.jmb.2020.08.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/02/2020] [Accepted: 08/05/2020] [Indexed: 12/15/2022]
Abstract
Streptococcus pneumoniae (Spn) is an important Gram-positive human pathogen that causes millions of infections worldwide with an increasing occurrence of antibiotic resistance. Fe acquisition is a crucial virulence determinant in Spn; further, Spn relies on exogenous FeIII-siderophore scavenging to meet nutritional Fe needs. Recent studies suggest that the human catecholamine stress hormone, norepinephrine (NE), facilitates Fe acquisition in Spn under conditions of transferrin-mediated Fe starvation. Here we show that the solute binding lipoprotein PiuA from the piu Fe acquisition ABC transporter PiuBCDA, previously described as an Fe-hemin binding protein, binds tetradentate catechol FeIII complexes, including NE and the hydrolysis products of enterobactin. Two protein-derived ligands (H238, Y300) create a coordinately saturated FeIII complex, which parallel recent studies in the Gram-negative intestinal pathogen Campylobacter jejuni. Our in vitro studies using NMR spectroscopy and 54Fe LC-ICP-MS confirm the FeIII can move from transferrin to apo-PiuA in an NE-dependent manner. Structural analysis of PiuA FeIII-bis-catechol and GaIII-bis-catechol and GaIII-(NE)2 complexes by NMR spectroscopy reveals only localized structural perturbations in PiuA upon ligand binding, largely consistent with recent descriptions of other solute binding proteins of type II ABC transporters. We speculate that tetradentate FeIII complexes formed by mono- and bis-catechol species are important Fe sources in Gram-positive human pathogens, since PiuA functions in the same way as SstD from Staphylococcus aureus.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102, USA; Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN 47405, USA
| | - Katherine A Edmonds
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102, USA
| | - Daniel J Raines
- Department of Chemistry, University of York, Heslington, York YO10 5DD, United Kingdom
| | - Brennan A Murphy
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102, USA
| | - Hongwei Wu
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102, USA
| | - Chuchu Guo
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Elizabeth M Nolan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Anne-K Duhme-Klair
- Department of Chemistry, University of York, Heslington, York YO10 5DD, United Kingdom
| | - David P Giedroc
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102, USA; Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN 47405, USA.
| |
Collapse
|
30
|
Racine PJ, Janvier X, Clabaut M, Catovic C, Souak D, Boukerb AM, Groboillot A, Konto-Ghiorghi Y, Duclairoir-Poc C, Lesouhaitier O, Orange N, Chevalier S, Feuilloley MGJ. Dialog between skin and its microbiota: Emergence of "Cutaneous Bacterial Endocrinology". Exp Dermatol 2020; 29:790-800. [PMID: 32682345 DOI: 10.1111/exd.14158] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/30/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022]
Abstract
Microbial endocrinology is studying the response of microorganisms to hormones and neurohormones and the microbiota production of hormones-like molecules. Until now, it was mainly applied to the gut and revealed that the intestinal microbiota should be considered as a real organ in constant and bilateral interactions with the whole human body. The skin harbours the second most abundant microbiome and contains an abundance of nerve terminals and capillaries, which in addition to keratinocytes, fibroblasts, melanocytes, dendritic cells and endothelial cells, release a huge diversity of hormones and neurohormones. In the present review, we will examine recent experimental data showing that, in skin, molecules such as substance P, calcitonin gene-related peptide, natriuretic peptides and catecholamines can directly affect the physiology and virulence of common skin-associated bacteria. Conversely, bacteria are able to synthesize and release compounds including histamine, glutamate and γ-aminobutyric acid or peptides showing partial homology with neurohormones such as α-melanocyte-stimulating hormone (αMSH). The more surprising is that some viruses can also encode neurohormones mimicking proteins. Taken together, these elements demonstrate that there is also a cutaneous microbial endocrinology and this emerging concept will certainly have important consequences in dermatology.
Collapse
Affiliation(s)
- Pierre-Jean Racine
- Laboratory of Microbiology Signals and Microenvironment LMSM EA4312, University of Rouen Normandy, Normandie Université, Evreux, France
| | - Xavier Janvier
- Laboratory of Microbiology Signals and Microenvironment LMSM EA4312, University of Rouen Normandy, Normandie Université, Evreux, France
| | - Maximilien Clabaut
- Laboratory of Microbiology Signals and Microenvironment LMSM EA4312, University of Rouen Normandy, Normandie Université, Evreux, France
| | - Chloe Catovic
- Laboratory of Microbiology Signals and Microenvironment LMSM EA4312, University of Rouen Normandy, Normandie Université, Evreux, France
| | - Djouhar Souak
- Laboratory of Microbiology Signals and Microenvironment LMSM EA4312, University of Rouen Normandy, Normandie Université, Evreux, France
| | - Amine M Boukerb
- Laboratory of Microbiology Signals and Microenvironment LMSM EA4312, University of Rouen Normandy, Normandie Université, Evreux, France
| | - Anne Groboillot
- Laboratory of Microbiology Signals and Microenvironment LMSM EA4312, University of Rouen Normandy, Normandie Université, Evreux, France
| | - Yoan Konto-Ghiorghi
- Laboratory of Microbiology Signals and Microenvironment LMSM EA4312, University of Rouen Normandy, Normandie Université, Evreux, France
| | - Cécile Duclairoir-Poc
- Laboratory of Microbiology Signals and Microenvironment LMSM EA4312, University of Rouen Normandy, Normandie Université, Evreux, France
| | - Olivier Lesouhaitier
- Laboratory of Microbiology Signals and Microenvironment LMSM EA4312, University of Rouen Normandy, Normandie Université, Evreux, France
| | - Nicole Orange
- Laboratory of Microbiology Signals and Microenvironment LMSM EA4312, University of Rouen Normandy, Normandie Université, Evreux, France
| | - Sylvie Chevalier
- Laboratory of Microbiology Signals and Microenvironment LMSM EA4312, University of Rouen Normandy, Normandie Université, Evreux, France
| | - Marc G J Feuilloley
- Laboratory of Microbiology Signals and Microenvironment LMSM EA4312, University of Rouen Normandy, Normandie Université, Evreux, France
| |
Collapse
|
31
|
Zhang Y, Sen S, Giedroc DP. Iron Acquisition by Bacterial Pathogens: Beyond Tris-Catecholate Complexes. Chembiochem 2020; 21:1955-1967. [PMID: 32180318 PMCID: PMC7367709 DOI: 10.1002/cbic.201900778] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/06/2020] [Indexed: 12/11/2022]
Abstract
Sequestration of the essential nutrient iron from bacterial invaders that colonize the vertebrate host is a central feature of nutritional immunity and the "fight over transition metals" at the host-pathogen interface. The iron quota for many bacterial pathogens is large, as iron enzymes often make up a significant share of the metalloproteome. Iron enzymes play critical roles in respiration, energy metabolism, and other cellular processes by catalyzing a wide range of oxidation-reduction, electron transfer, and oxygen activation reactions. In this Concept article, we discuss recent insights into the diverse ways that bacterial pathogens acquire this essential nutrient, beyond the well-characterized tris-catecholate FeIII complexes, in competition and cooperation with significant host efforts to cripple these processes. We also discuss pathogen strategies to adapt their metabolism to less-than-optimal iron concentrations, and briefly speculate on what might be an integrated adaptive response to the concurrent limitation of both iron and zinc in the infected host.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102, USA
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN 47405-7102, USA
| | - Sambuddha Sen
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102, USA
| | - David P Giedroc
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102, USA
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN 47405-7102, USA
| |
Collapse
|
32
|
Rusu IG, Suharoschi R, Vodnar DC, Pop CR, Socaci SA, Vulturar R, Istrati M, Moroșan I, Fărcaș AC, Kerezsi AD, Mureșan CI, Pop OL. Iron Supplementation Influence on the Gut Microbiota and Probiotic Intake Effect in Iron Deficiency-A Literature-Based Review. Nutrients 2020; 12:E1993. [PMID: 32635533 PMCID: PMC7400826 DOI: 10.3390/nu12071993] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 12/15/2022] Open
Abstract
Iron deficiency in the human body is a global issue with an impact on more than two billion individuals worldwide. The most important functions ensured by adequate amounts of iron in the body are related to transport and storage of oxygen, electron transfer, mediation of oxidation-reduction reactions, synthesis of hormones, the replication of DNA, cell cycle restoration and control, fixation of nitrogen, and antioxidant effects. In the case of iron deficiency, even marginal insufficiencies may impair the proper functionality of the human body. On the other hand, an excess in iron concentration has a major impact on the gut microbiota composition. There are several non-genetic causes that lead to iron deficiencies, and thus, several approaches in their treatment. The most common methods are related to food fortifications and supplements. In this review, following a summary of iron metabolism and its health implications, we analyzed the scientific literature for the influence of iron fortification and supplementation on the gut microbiome and the effect of probiotics, prebiotics, and/or synbiotics in iron absorption and availability for the organism.
Collapse
Affiliation(s)
- Ioana Gabriela Rusu
- Department of Food Science, University of Agricultural Science and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (I.G.R.); (R.S.); (D.C.V.); (C.R.P.); (S.A.S.); (A.C.F.); (A.D.K.); (C.I.M.)
| | - Ramona Suharoschi
- Department of Food Science, University of Agricultural Science and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (I.G.R.); (R.S.); (D.C.V.); (C.R.P.); (S.A.S.); (A.C.F.); (A.D.K.); (C.I.M.)
| | - Dan Cristian Vodnar
- Department of Food Science, University of Agricultural Science and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (I.G.R.); (R.S.); (D.C.V.); (C.R.P.); (S.A.S.); (A.C.F.); (A.D.K.); (C.I.M.)
| | - Carmen Rodica Pop
- Department of Food Science, University of Agricultural Science and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (I.G.R.); (R.S.); (D.C.V.); (C.R.P.); (S.A.S.); (A.C.F.); (A.D.K.); (C.I.M.)
| | - Sonia Ancuța Socaci
- Department of Food Science, University of Agricultural Science and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (I.G.R.); (R.S.); (D.C.V.); (C.R.P.); (S.A.S.); (A.C.F.); (A.D.K.); (C.I.M.)
| | - Romana Vulturar
- Department of Molecular Sciences, University of Medicine and Pharmacy Iuliu Hatieganu, 400349 Cluj-Napoca, Romania;
- Cognitive Neuroscience Laboratory, University Babes-Bolyai, 400327 Cluj-Napoca, Romania
| | - Magdalena Istrati
- Regional Institute of Gastroenterology and Hepatology “Prof. Dr. Octavian Fodor”, 400158 Cluj-Napoca, Romania;
| | - Ioana Moroșan
- Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400349 Cluj-Napoca, Romania;
| | - Anca Corina Fărcaș
- Department of Food Science, University of Agricultural Science and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (I.G.R.); (R.S.); (D.C.V.); (C.R.P.); (S.A.S.); (A.C.F.); (A.D.K.); (C.I.M.)
| | - Andreea Diana Kerezsi
- Department of Food Science, University of Agricultural Science and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (I.G.R.); (R.S.); (D.C.V.); (C.R.P.); (S.A.S.); (A.C.F.); (A.D.K.); (C.I.M.)
| | - Carmen Ioana Mureșan
- Department of Food Science, University of Agricultural Science and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (I.G.R.); (R.S.); (D.C.V.); (C.R.P.); (S.A.S.); (A.C.F.); (A.D.K.); (C.I.M.)
| | - Oana Lelia Pop
- Department of Food Science, University of Agricultural Science and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (I.G.R.); (R.S.); (D.C.V.); (C.R.P.); (S.A.S.); (A.C.F.); (A.D.K.); (C.I.M.)
| |
Collapse
|
33
|
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: 1] [Impact Index Per Article: 0.3] [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]
|
34
|
Jochim A, Adolf L, Belikova D, Schilling NA, Setyawati I, Chin D, Meyers S, Verhamme P, Heinrichs DE, Slotboom DJ, Heilbronner S. An ECF-type transporter scavenges heme to overcome iron-limitation in Staphylococcus lugdunensis. eLife 2020; 9:e57322. [PMID: 32515736 PMCID: PMC7299338 DOI: 10.7554/elife.57322] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/09/2020] [Indexed: 01/03/2023] Open
Abstract
Energy-coupling factor type transporters (ECF) represent trace nutrient acquisition systems. Substrate binding components of ECF-transporters are membrane proteins with extraordinary affinity, allowing them to scavenge trace amounts of ligand. A number of molecules have been described as substrates of ECF-transporters, but an involvement in iron-acquisition is unknown. Host-induced iron limitation during infection represents an effective mechanism to limit bacterial proliferation. We identified the iron-regulated ECF-transporter Lha in the opportunistic bacterial pathogen Staphylococcus lugdunensis and show that the transporter is specific for heme. The recombinant substrate-specific subunit LhaS accepted heme from diverse host-derived hemoproteins. Using isogenic mutants and recombinant expression of Lha, we demonstrate that its function is independent of the canonical heme acquisition system Isd and allows proliferation on human cells as sources of nutrient iron. Our findings reveal a unique strategy of nutritional heme acquisition and provide the first example of an ECF-transporter involved in overcoming host-induced nutritional limitation.
Collapse
Affiliation(s)
- Angelika Jochim
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of TübingenTübingenGermany
| | - Lea Adolf
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of TübingenTübingenGermany
| | - Darya Belikova
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of TübingenTübingenGermany
| | | | - Inda Setyawati
- Groningen Biomolecular Sciences and Biotechnology Institute, University of GroningenGroningenNetherlands
| | - Denny Chin
- Department of Microbiology and Immunology, University of Western OntarioLondonCanada
| | | | | | - David E Heinrichs
- Department of Microbiology and Immunology, University of Western OntarioLondonCanada
| | - Dirk J Slotboom
- Groningen Biomolecular Sciences and Biotechnology Institute, University of GroningenGroningenNetherlands
| | - Simon Heilbronner
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of TübingenTübingenGermany
- German Centre for Infection Research (DZIF), Partner Site TübingenTübingenGermany
- (DFG) Cluster of Excellence EXC 2124 Controlling Microbes to Fight InfectionsTübingenGermany
| |
Collapse
|
35
|
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: 6] [Impact Index Per Article: 1.5] [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
|
36
|
Zsila F, Beke-Somfai T. Human host-defense peptide LL-37 targets stealth siderophores. Biochem Biophys Res Commun 2020; 526:780-785. [PMID: 32265033 DOI: 10.1016/j.bbrc.2020.03.162] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 03/28/2020] [Indexed: 02/02/2023]
Abstract
A growing number of evidence shows that human-associated microbiota is an important contributor in health and disease. However, much of the complexity of host-microbiota interaction remains to be elucidated both at cellular and molecular levels. Siderophores are chemically diverse, ferric-specific chelators synthesized and secreted by microbes to secure their iron acquisition. The host defense peptide LL-37 is ubiquitously produced at epithelial surfaces modulating microbial communities and suppressing pathogenic strains. The present work demonstrates that LL-37 binds tightly siderocalin-resistant stealth siderophores which are important contributors to the virulence of several pathogens. As indicated by circular dichroism spectroscopic experiments, addition of aerobactin and rhizoferrin increases the membrane active α-helical conformation of the partially folded peptide. The cationic nature of LL-37 (+6 net charge at pH 7.4) and the multiple carboxylate groups present in siderophores refer to the dominant contribution of electrostatic interactions in the stabilization of peptide-chelator adducts. It is proposed that aside siderocalin proteins, LL-37 may be a complementary, less specific component of the siderophore scavenging repertoire of the innate immune system.
Collapse
Affiliation(s)
- Ferenc Zsila
- Biomolecular Self-Assembly Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, P.O. Box 286, H-1519, Budapest, Hungary.
| | - Tamás Beke-Somfai
- Biomolecular Self-Assembly Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, P.O. Box 286, H-1519, Budapest, Hungary
| |
Collapse
|
37
|
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: 42] [Impact Index Per Article: 10.5] [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
|
38
|
Dahyot S, Oxaran V, Niepceron M, Dupart E, Legris S, Destruel L, Didi J, Clamens T, Lesouhaitier O, Zerdoumi Y, Flaman JM, Pestel-Caron M. Role of the LytSR Two-Component Regulatory System in Staphylococcus lugdunensis Biofilm Formation and Pathogenesis. Front Microbiol 2020; 11:39. [PMID: 32038604 PMCID: PMC6993578 DOI: 10.3389/fmicb.2020.00039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 01/09/2020] [Indexed: 11/21/2022] Open
Abstract
Staphylococcus lugdunensis is a coagulase negative Staphylococcus recognized as a virulent pathogen. It is responsible for a wide variety of infections, some of which are associated with biofilm production, such as implanted medical device infections or endocarditis. However, little is known about S. lugdunensis regulation of virulence factor expression. Two-component regulatory systems (TCS) play a critical role in bacterial adaptation, survival, and virulence. Among them, LytSR is widely conserved but has variable roles in different organisms, all connected to metabolism or cell death and lysis occurring during biofilm development. Therefore, we investigated here the functions of LytSR in S. lugdunensis pathogenesis. Deletion of lytSR in S. lugdunensis DSM 4804 strain did not alter either susceptibility to Triton X-100 induced autolysis or death induced by antibiotics targeting cell wall synthesis. Interestingly, ΔlytSR biofilm was characterized by a lower biomass, a lack of tower structures, and a higher rate of dead cells compared to the wild-type strain. Virulence toward Caenorhabditis elegans using a slow-killing assay was significantly reduced for the mutant compared to the wild-type strain. By contrast, the deletion of lytSR had no effect on the cytotoxicity of S. lugdunensis toward the human keratinocyte cell line HaCaT. Transcriptional analyses conducted at mid- and late-exponential phases showed that lytSR deletion affected the expression of 286 genes. Most of them were involved in basic functions such as the metabolism of amino acids, carbohydrates, and nucleotides. Furthermore, LytSR appeared to be involved in the regulation of genes encoding known or putative virulence and colonization factors, including the fibrinogen-binding protein Fbl, the major autolysin AtlL, and the type VII secretion system. Overall, our data suggest that the LytSR TCS is implicated in S. lugdunensis pathogenesis, through its involvement in biofilm formation and potentially by the control of genes encoding putative virulence factors.
Collapse
Affiliation(s)
- Sandrine Dahyot
- Groupe de Recherche sur l’Adaptation Microbienne (GRAM 2.0), Department of Bacteriology, Rouen University Hospital, Normandie University, UNIROUEN, UNICAEN, Rouen, France
| | - Virginie Oxaran
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, United States
| | - Maïté Niepceron
- Groupe de Recherche sur l’Adaptation Microbienne (GRAM 2.0), Normandie University, UNIROUEN, UNICAEN, Rouen, France
| | - Eddy Dupart
- Groupe de Recherche sur l’Adaptation Microbienne (GRAM 2.0), Normandie University, UNIROUEN, UNICAEN, Rouen, France
| | - Stéphanie Legris
- Groupe de Recherche sur l’Adaptation Microbienne (GRAM 2.0), Normandie University, UNIROUEN, UNICAEN, Rouen, France
| | - Laurie Destruel
- Groupe de Recherche sur l’Adaptation Microbienne (GRAM 2.0), Normandie University, UNIROUEN, UNICAEN, Rouen, France
| | - Jennifer Didi
- Groupe de Recherche sur l’Adaptation Microbienne (GRAM 2.0), Normandie University, UNIROUEN, UNICAEN, Rouen, France
| | - Thomas Clamens
- Laboratory of Microbiology Signals and Microenvironment (LMSM), Normandie University, UNIROUEN, Evreux, France
| | - Olivier Lesouhaitier
- Laboratory of Microbiology Signals and Microenvironment (LMSM), Normandie University, UNIROUEN, Evreux, France
| | - Yasmine Zerdoumi
- INSERM U1245, Normandy Centre for Genomic and Personalized Medicine, Rouen University Hospital, Normandie University, UNIROUEN, Rouen, France
| | - Jean-Michel Flaman
- INSERM U1245, Normandy Centre for Genomic and Personalized Medicine, Rouen University Hospital, Normandie University, UNIROUEN, Rouen, France
| | - Martine Pestel-Caron
- Groupe de Recherche sur l’Adaptation Microbienne (GRAM 2.0), Department of Bacteriology, Rouen University Hospital, Normandie University, UNIROUEN, UNICAEN, Rouen, France
| |
Collapse
|
39
|
Crosby HA, Tiwari N, Kwiecinski JM, Xu Z, Dykstra A, Jenul C, Fuentes EJ, Horswill AR. The Staphylococcus aureus ArlRS two-component system regulates virulence factor expression through MgrA. Mol Microbiol 2020; 113:103-122. [PMID: 31618469 PMCID: PMC7175635 DOI: 10.1111/mmi.14404] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The Gram-positive bacterium, Staphylococcus aureus, is a versatile pathogen that can sense and adapt to a wide variety of environments within the human host, in part through its 16 two-component regulatory systems. The ArlRS two-component system has been shown to affect many cellular processes in S. aureus, including autolysis, biofilm formation, capsule synthesis and virulence. Yet the molecular details of this regulation remained largely unknown. We used RNA sequencing to identify the ArlRS regulon, and found 70% overlap with that of the global regulator MgrA. These genes included cell wall-anchored adhesins (ebh, sdrD), polysaccharide and capsule synthesis genes, cell wall remodeling genes (lytN, ddh), the urease operon, genes involved in metal transport (feoA, mntH, sirA), anaerobic metabolism genes (adhE, pflA, nrdDG) and a large number of virulence factors (lukSF, lukAB, nuc, gehB, norB, chs, scn and esxA). We show that ArlR directly activates expression of mgrA and identify a probable ArlR-binding site (TTTTCTCAT-N4 -TTTTAATAA). A highly similar sequence is also found in the spx P2 promoter, which was recently shown to be regulated by ArlRS. We also demonstrate that ArlS has kinase activity toward ArlR in vitro, although it has slower kinetics than other similar histidine kinases.
Collapse
Affiliation(s)
- Heidi A. Crosby
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | - Nitija Tiwari
- Department of Biochemistry, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Jakub M. Kwiecinski
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | - Zhen Xu
- Department of Biochemistry, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Allison Dykstra
- Department of Biochemistry, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Christian Jenul
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | - Ernesto J Fuentes
- Department of Biochemistry, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Alexander R. Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
- Department of Veterans Affairs Eastern Colorado Health Care System, Denver, CO
| |
Collapse
|
40
|
Abstract
Staphylococcus aureus is an important pathogen responsible for nosocomial and community-acquired infections in humans, and methicillin-resistant S. aureus (MRSA) infections have continued to increase despite widespread preventative measures. S. aureus can colonize the female vaginal tract, and reports have suggested an increase in MRSA infections in pregnant and postpartum women as well as outbreaks in newborn nurseries. Currently, little is known about specific factors that promote MRSA vaginal colonization and subsequent infection. To study S. aureus colonization of the female reproductive tract in a mammalian system, we developed a mouse model of S. aureus vaginal carriage and demonstrated that both hospital-associated and community-associated MRSA isolates can colonize the murine vaginal tract. Immunohistochemical analysis revealed an increase in neutrophils in the vaginal lumen during MRSA colonization. Additionally, we observed that a mutant lacking fibrinogen binding adhesins exhibited decreased persistence within the mouse vagina. To further identify novel factors that promote vaginal colonization, we performed RNA sequencing to determine the transcriptome of MRSA growing in vivo during vaginal carriage at 5 h, 1 day, and 3 days postinoculation. Over 25% of the bacterial genes were differentially regulated at all time points during colonization compared to laboratory cultures. The most highly induced genes were those involved in iron acquisition, including the Isd system and siderophore transport systems. Mutants deficient in these pathways did not persist as well during in vivo colonization. These results reveal that fibrinogen binding and the capacity to overcome host nutritional limitation are important determinants of MRSA vaginal colonization.IMPORTANCE Staphylococcus aureus is an opportunistic pathogen able to cause a wide variety of infections in humans. Recent reports have suggested an increasing prevalence of MRSA in pregnant and postpartum women, coinciding with the increased incidence of MRSA infections in neonatal intensive care units (NICUs) and newborn nurseries. Vertical transmission from mothers to infants at delivery is a likely route of MRSA acquisition by the newborn; however, essentially nothing is known about host and bacterial factors that influence MRSA carriage in the vagina. Here, we established a mouse model of vaginal colonization and observed that multiple MRSA strains can persist in the vaginal tract. Additionally, we determined that MRSA interactions with fibrinogen and iron uptake can promote vaginal persistence. This study is the first to identify molecular mechanisms which govern vaginal colonization by MRSA, the critical initial step preceding infection and neonatal transmission.
Collapse
|
41
|
Inbaraj S, Sejian V, Ramasamy S. Role of environmental stressor-host immune system–pathogen interactions in development of infectious disease in farm animals. BIOL RHYTHM RES 2019. [DOI: 10.1080/09291016.2019.1695084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Sophia Inbaraj
- Division of Bacteriology and Mycology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Veerasamy Sejian
- Animal Physiology Division, ICAR-National Institute Animal Nutrition and Physiology, Bengaluru, India
| | - Santhamani Ramasamy
- Department of microbiology and immunology, Post-doctoral research fellow, Albert Einstein College of Medicine, New York, NY, USA
| |
Collapse
|
42
|
Dynamic Relay of Protein-Bound Lipoic Acid in Staphylococcus aureus. J Bacteriol 2019; 201:JB.00446-19. [PMID: 31451544 DOI: 10.1128/jb.00446-19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 08/21/2019] [Indexed: 12/19/2022] Open
Abstract
Staphylococcus aureus competes for myriad essential nutrients during host infection. One of these nutrients is the organosulfur compound lipoic acid, a cofactor required for the activity of several metabolic enzyme complexes. In S. aureus, these include the E2 subunits of three α-ketoacid dehydrogenases and two H proteins, GcvH of the glycine cleavage system and its paralog, GcvH-L. We previously determined that the S. aureus amidotransferase LipL is required for lipoylation of the E2 subunits of pyruvate dehydrogenase (PDH) and branched-chain 2-oxoacid dehydrogenase (BCODH) complexes. The results from this study, coupled with those from Bacillus subtilis, suggested that LipL catalyzes lipoyl transfer from H proteins to E2 subunits. However, to date, the range of LipL targets, the extent of LipL-dependent lipoic acid shuttling between lipoyl domain-containing proteins, and the importance of lipoyl relay in pathogenesis remain unknown. Here, we demonstrate that LipL uses both lipoyl-H proteins as the substrates for lipoyl transfer to all E2 subunits. Moreover, LipL facilitates lipoyl relay between E2 subunits and between H proteins, a property that potentially constitutes an adaptive response to nutrient scarcity in the host, as LipL is required for virulence during infection. Together, these observations support a role for LipL in facilitating flexible lipoyl relay between proteins and highlight the complexity of protein lipoylation in S. aureus IMPORTANCE Protein lipoylation is a posttranslational modification that is evolutionarily conserved from bacteria to humans. Lipoic acid modifications are found on five proteins in S. aureus, four of which are components of major metabolic enzymes. In some bacteria, the amidotransferase LipL is critical for the attachment of lipoic acid to these proteins, and yet it is unclear to what extent LipL facilitates the transfer of this cofactor. We find that S. aureus LipL flexibly shuttles lipoic acid among metabolic enzyme subunits, alluding to a dynamic redistribution mechanism within the bacterial cell. This discovery exemplifies a potential means by which bacteria optimize the use of scarce nutrients when resources are limited.
Collapse
|
43
|
Staphylococcus aureus exhibits heterogeneous siderophore production within the vertebrate host. Proc Natl Acad Sci U S A 2019; 116:21980-21982. [PMID: 31611408 PMCID: PMC6825271 DOI: 10.1073/pnas.1913991116] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Siderophores, iron-scavenging small molecules, are fundamental to bacterial nutrient metal acquisition and enable pathogens to overcome challenges imposed by nutritional immunity. Multimodal imaging mass spectrometry allows visualization of host−pathogen iron competition, by mapping siderophores within infected tissue. We have observed heterogeneous distributions of Staphylococcus aureus siderophores across infectious foci, challenging the paradigm that the vertebrate host is a uniformly iron-depleted environment to invading microbes.
Collapse
|
44
|
Verstraete MM, Morales LD, Kobylarz MJ, Loutet SA, Laakso HA, Pinter TB, Stillman MJ, Heinrichs DE, Murphy MEP. The heme-sensitive regulator SbnI has a bifunctional role in staphyloferrin B production by Staphylococcus aureus. J Biol Chem 2019; 294:11622-11636. [PMID: 31197035 DOI: 10.1074/jbc.ra119.007757] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 06/12/2019] [Indexed: 01/30/2023] Open
Abstract
Staphylococcus aureus infection relies on iron acquisition from its host. S. aureus takes up iron through heme uptake by the iron-responsive surface determinant (Isd) system and by the production of iron-scavenging siderophores. Staphyloferrin B (SB) is a siderophore produced by the 9-gene sbn gene cluster for SB biosynthesis and efflux. Recently, the ninth gene product, SbnI, was determined to be a free l-serine kinase that produces O-phospho-l-serine (OPS), a substrate for SB biosynthesis. Previous studies have also characterized SbnI as a DNA-binding regulatory protein that senses heme to control sbn gene expression for SB synthesis. Here, we present crystal structures at 1.9-2.1 Å resolution of a SbnI homolog from Staphylococcus pseudintermedius (SpSbnI) in both apo form and in complex with ADP, a product of the kinase reaction; the latter confirmed the active-site location. The structures revealed that SpSbnI forms a dimer through C-terminal domain swapping and a dimer of dimers through intermolecular disulfide formation. Heme binding had only a modest effect on SbnI enzymatic activity, suggesting that its two functions are independent and structurally distinct. We identified a heme-binding site and observed catalytic heme transfer between a heme-degrading protein of the Isd system, IsdI, and SbnI. These findings support the notion that SbnI has a bifunctional role contributing precursor OPS to SB synthesis and directly sensing heme to control expression of the sbn locus. We propose that heme transfer from IsdI to SbnI enables S. aureus to control iron source preference according to the sources available in the environment.
Collapse
Affiliation(s)
- Meghan M Verstraete
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - L Daniela Morales
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Marek J Kobylarz
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Slade A Loutet
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Holly A Laakso
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Tyler B Pinter
- Department of Chemistry, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Martin J Stillman
- Department of Chemistry, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - David E Heinrichs
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Michael E P Murphy
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
45
|
Abstract
Community-acquired pneumonia (CAP) is a leading cause of morbidity and mortality worldwide. Despite broad literature including basic and translational scientific studies, many gaps in our understanding of host-pathogen interactions remain. In this review, pathogen virulence factors that drive lung infection and injury are discussed in relation to their associated host immune pathways. CAP epidemiology is considered, with a focus on Staphylococcus aureus and Streptococcus pneumoniae as primary pathogens. Bacterial factors involved in nasal colonization and subsequent virulence are illuminated. A particular emphasis is placed on bacterial pore-forming toxins, host cell death, and inflammasome activation. Identified host-pathogen interactions are then examined by linking pathogen factors to aberrant host response pathways in the context of acute lung injury in both primary and secondary infection. While much is known regarding bacterial virulence and host immune responses, CAP management is still limited to mostly supportive care. It is likely that improvements in therapy will be derived from combinatorial targeting of both pathogen virulence factors and host immunomodulation.
Collapse
|
46
|
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: 7.4] [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
|
47
|
Dopamine Is a Siderophore-Like Iron Chelator That Promotes Salmonella enterica Serovar Typhimurium Virulence in Mice. mBio 2019; 10:mBio.02624-18. [PMID: 30723125 PMCID: PMC6428752 DOI: 10.1128/mbio.02624-18] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We have recently shown that the catecholamine dopamine regulates cellular iron homeostasis in macrophages. As iron is an essential nutrient for microbes, and intracellular iron availability affects the growth of intracellular bacteria, we studied whether dopamine administration impacts the course of Salmonella infections. Dopamine was found to promote the growth of Salmonella both in culture and within bone marrow-derived macrophages, which was dependent on increased bacterial iron acquisition. Dopamine administration to mice infected with Salmonella enterica serovar Typhimurium resulted in significantly increased bacterial burdens in liver and spleen, as well as reduced survival. The promotion of bacterial growth by dopamine was independent of the siderophore-binding host peptide lipocalin-2. Rather, dopamine enhancement of iron uptake requires both the histidine sensor kinase QseC and bacterial iron transporters, in particular SitABCD, and may also involve the increased expression of bacterial iron uptake genes. Deletion or pharmacological blockade of QseC reduced but did not abolish the growth-promoting effects of dopamine. Dopamine also modulated systemic iron homeostasis by increasing hepcidin expression and depleting macrophages of the iron exporter ferroportin, which enhanced intracellular bacterial growth. Salmonella lacking all central iron uptake pathways failed to benefit from dopamine treatment. These observations are potentially relevant to critically ill patients, in whom the pharmacological administration of catecholamines to improve circulatory performance may exacerbate the course of infection with siderophilic bacteria.IMPORTANCE Here we show that dopamine increases bacterial iron incorporation and promotes Salmonella Typhimurium growth both in vitro and in vivo These observations suggest the potential hazards of pharmacological catecholamine administration in patients with bacterial sepsis but also suggest that the inhibition of bacterial iron acquisition might provide a useful approach to antimicrobial therapy.
Collapse
|
48
|
Verstraete MM, Perez-Borrajero C, Brown KL, Heinrichs DE, Murphy MEP. SbnI is a free serine kinase that generates O -phospho-l-serine for staphyloferrin B biosynthesis in Staphylococcus aureus. J Biol Chem 2018; 293:6147-6160. [PMID: 29483190 DOI: 10.1074/jbc.ra118.001875] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 02/15/2018] [Indexed: 12/22/2022] Open
Abstract
Staphyloferrin B (SB) is an iron-chelating siderophore produced by Staphylococcus aureus in invasive infections. Proteins for SB biosynthesis and export are encoded by the sbnABCDEFGHI gene cluster, in which SbnI, a member of the ParB/Srx superfamily, acts as a heme-dependent transcriptional regulator of the sbn locus. However, no structural or functional information about SbnI is available. Here, a crystal structure of SbnI revealed striking structural similarity to an ADP-dependent free serine kinase, SerK, from the archaea Thermococcus kodakarensis We found that features of the active sites are conserved, and biochemical assays and 31P NMR and HPLC analyses indicated that SbnI is also a free serine kinase but uses ATP rather than ADP as phosphate donor to generate the SB precursor O-phospho-l-serine (OPS). SbnI consists of two domains, and elevated B-factors in domain II were consistent with the open-close reaction mechanism previously reported for SerK. Mutagenesis of Glu20 and Asp58 in SbnI disclosed that they are required for kinase activity. The only known OPS source in bacteria is through the phosphoserine aminotransferase activity of SerC within the serine biosynthesis pathway, and we demonstrate that an S. aureus serC mutant is a serine auxotroph, consistent with a function in l-serine biosynthesis. However, the serC mutant strain could produce SB when provided l-serine, suggesting that SbnI produces OPS for SB biosynthesis in vivo These findings indicate that besides transcriptionally regulating the sbn locus, SbnI also has an enzymatic role in the SB biosynthetic pathway.
Collapse
Affiliation(s)
| | - Cecilia Perez-Borrajero
- the Genome Sciences and Technology Program Life Sciences Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada and
| | | | - David E Heinrichs
- the Department of Microbiology and Immunology, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | | |
Collapse
|
49
|
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: 4.2] [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
|
50
|
Oliveira F, França Â, Cerca N. Staphylococcus epidermidis is largely dependent on iron availability to form biofilms. Int J Med Microbiol 2017; 307:552-563. [DOI: 10.1016/j.ijmm.2017.08.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 07/25/2017] [Accepted: 08/23/2017] [Indexed: 10/19/2022] Open
|