1
|
Basta DW, Campbell IW, Sullivan EJ, Hotinger JA, Hullahalli K, Garg M, Waldor MK. Inducible transposon mutagenesis identifies bacterial fitness determinants during infection in mice. Nat Microbiol 2025; 10:1171-1183. [PMID: 40148565 PMCID: PMC12055562 DOI: 10.1038/s41564-025-01975-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 03/03/2025] [Indexed: 03/29/2025]
Abstract
Transposon insertion sequencing (Tn-seq) is a powerful method for genome-scale forward genetics in bacteria. However, inefficient transposon delivery or stochastic loss of mutants due to population bottlenecks can limit its effectiveness. Here we have developed 'InducTn-seq', where an arabinose-inducible Tn5 transposase enables temporal control of mini-Tn5 transposition. InducTn-seq generated up to 1.2 million transposon mutants from a single colony of enterotoxigenic Escherichia coli, Salmonella typhimurium, Shigella flexneri and Citrobacter rodentium. This mutant diversity enabled more sensitive detection of subtle fitness defects and measurement of quantitative fitness effects for essential and non-essential genes. Applying InducTn-seq to C. rodentium in a mouse model of infectious colitis bypassed a highly restrictive host bottleneck, generating a diverse population of >5 × 105 unique transposon mutants compared to 10-102 recovered by traditional Tn-seq. This in vivo screen revealed that the C. rodentium type I-E CRISPR system is required to suppress a toxin otherwise activated during gut colonization. Our findings highlight the potential of InducTn-seq for genome-scale forward genetic screens in bacteria.
Collapse
Affiliation(s)
- David W Basta
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Ian W Campbell
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Emily J Sullivan
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Julia A Hotinger
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Karthik Hullahalli
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Mehek Garg
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Matthew K Waldor
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Microbiology, Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
2
|
Murray AK, Stanton IC, Tipper HJ, Wilkinson H, Schmidt W, Hart A, Singer AC, Gaze WH. A critical meta-analysis of predicted no effect concentrations for antimicrobial resistance selection in the environment. WATER RESEARCH 2024; 266:122310. [PMID: 39217643 DOI: 10.1016/j.watres.2024.122310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Antimicrobial resistance (AMR) is one of the greatest threats to human health with a growing body of evidence demonstrating that selection for AMR can occur at environmental antimicrobial concentrations. Understanding the concentrations at which selection for resistance may occur is critical to help inform environmental risk assessments and highlight where mitigation strategies are required. A variety of experimental and data approaches have been used to determine these concentrations. However, there is minimal standardisation of existing approaches and no consensus on the relative merits of different methods. We conducted a semi-systematic literature review to collect and critically appraise available minimal selective concentration (MSC) and predicted no effect concentration for resistance (PNECR) data and the approaches used to derive them. There were 21 relevant articles providing 331 selective concentrations, ranging from 0.00087 µg/L (ciprofloxacin) to 2000 µg/L (carbenicillin). Meta-analyses of these data found that selective concentrations are highly compound-dependent, and only a subset of all antimicrobials have been the focus of most of the research. The variety of approaches that have been used, knowledge gaps and future research priorities were identified, as well as recommendations for those considering the selective risks of antimicrobials in the environment.
Collapse
Affiliation(s)
- Aimee K Murray
- European Centre for Environment and Human Health, University of Exeter Medical School, Faculty of Health and Life Sciences, Environment & Sustainability Institute, Penryn Campus, Cornwall TR10 9FE, United Kingdom.
| | - Isobel C Stanton
- UK Centre for Ecology and Hydrology, Wallingford OX10 8BB, United Kingdom
| | - Holly J Tipper
- UK Centre for Ecology and Hydrology, Wallingford OX10 8BB, United Kingdom
| | - Helen Wilkinson
- Chief Scientist's Group, Environment Agency, Bristol BS1 5AH, United Kingdom
| | - Wiebke Schmidt
- Chief Scientist's Group, Environment Agency, Bristol BS1 5AH, United Kingdom
| | - Alwyn Hart
- Chief Scientist's Group, Environment Agency, Bristol BS1 5AH, United Kingdom
| | - Andrew C Singer
- UK Centre for Ecology and Hydrology, Wallingford OX10 8BB, United Kingdom
| | - William H Gaze
- European Centre for Environment and Human Health, University of Exeter Medical School, Faculty of Health and Life Sciences, Environment & Sustainability Institute, Penryn Campus, Cornwall TR10 9FE, United Kingdom
| |
Collapse
|
3
|
Tooke AK, Hodges RE, Pyrah JF, Bayles KW, Renshaw SA, Foster SJ. Tetracycline and Oxacillin Act Synergistically on Biofilms and Display Increased Efficacy In Vivo Against Staphylococcus aureus. Curr Microbiol 2024; 81:447. [PMID: 39505760 PMCID: PMC11541413 DOI: 10.1007/s00284-024-03959-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 10/18/2024] [Indexed: 11/08/2024]
Abstract
Oxacillin (bactericidal) and tetracycline (bacteriostatic) are clinically relevant antibiotics that are routinely prescribed to treat Staphylococcus aureus infections but not conventionally used in combination. There is an urgent need for treatment regimens that can act upon biofilms during infection, associated with chronic infections on indwelling devices, as well as acute planktonic (systemic) infection. Here we show that in an in vitro model oxacillin and tetracycline act synergistically against S. aureus UAMS-1 biofilms, reducing the concentration of both antibiotics necessary to eradicate an established biofilm. Using an in vivo zebrafish larval infection model with S. aureus NewHG, they display improved bacterial clearance compared to each drug alone and can counteract a loss of host phagocytes, an important innate defence against S. aureus. In these cases, the bacteriostatic nature of tetracycline enhances rather than dampens the bactericidal action of oxacillin, although an exact mechanism remains to be elucidated. We suggest a dual therapy could be of clinical use against biofilm-forming S. aureus and has a potential use in patients with a compromised immune system.
Collapse
Affiliation(s)
- Amy K Tooke
- School of Biosciences, University of Sheffield, Sheffield, S10 2TH, UK
- Microbes in Health and Disease Theme, Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Rebecca E Hodges
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Josie F Pyrah
- School of Biosciences, University of Sheffield, Sheffield, S10 2TH, UK
| | - Kenneth W Bayles
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Stephen A Renshaw
- The Bateson Centre, University of Sheffield, Western Bank, Sheffield, S10 2TH, UK
- School of Medicine and Population Health, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Simon J Foster
- School of Biosciences, University of Sheffield, Sheffield, S10 2TH, UK.
- Florey Institute, University of Sheffield, Sheffield, S10 2TH, UK.
| |
Collapse
|
4
|
Adedeji-Olulana AF, Wacnik K, Lafage L, Pasquina-Lemonche L, Tinajero-Trejo M, Sutton JAF, Bilyk B, Irving SE, Portman Ross CJ, Meacock OJ, Randerson SA, Beattie E, Owen DS, Florence J, Durham WM, Hornby DP, Corrigan RM, Green J, Hobbs JK, Foster SJ. Two codependent routes lead to high-level MRSA. Science 2024; 386:573-580. [PMID: 39480932 DOI: 10.1126/science.adn1369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 06/13/2024] [Accepted: 08/30/2024] [Indexed: 11/02/2024]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA), in which acquisition of mecA [which encodes the cell wall peptidoglycan biosynthesis component penicillin-binding protein 2a (PBP2a)] confers resistance to β-lactam antibiotics, is of major clinical concern. We show that, in the presence of antibiotics, MRSA adopts an alternative mode of cell division and shows an altered peptidoglycan architecture at the division septum. PBP2a can replace the transpeptidase activity of the endogenous and essential PBP2 but not that of PBP1, which is responsible for the distinctive native septal peptidoglycan architecture. Successful division without PBP1 activity requires the alternative division mode and is enabled by several possible chromosomal potentiator (pot) mutations. MRSA resensitizing agents differentially interfere with the two codependent mechanisms required for high-level antibiotic resistance, which provides opportunities for new interventions.
Collapse
Affiliation(s)
| | - Katarzyna Wacnik
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute, University of Sheffield, Sheffield, UK
| | - Lucia Lafage
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute, University of Sheffield, Sheffield, UK
| | - Laia Pasquina-Lemonche
- School of Mathematical and Physical Sciences, University of Sheffield, Sheffield, UK
- The Florey Institute, University of Sheffield, Sheffield, UK
| | - Mariana Tinajero-Trejo
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute, University of Sheffield, Sheffield, UK
| | - Joshua A F Sutton
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute, University of Sheffield, Sheffield, UK
| | - Bohdan Bilyk
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute, University of Sheffield, Sheffield, UK
| | - Sophie E Irving
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute, University of Sheffield, Sheffield, UK
| | - Callum J Portman Ross
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute, University of Sheffield, Sheffield, UK
| | - Oliver J Meacock
- School of Mathematical and Physical Sciences, University of Sheffield, Sheffield, UK
| | - Sam A Randerson
- School of Mathematical and Physical Sciences, University of Sheffield, Sheffield, UK
| | - Ewan Beattie
- School of Mathematical and Physical Sciences, University of Sheffield, Sheffield, UK
| | - David S Owen
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - James Florence
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute, University of Sheffield, Sheffield, UK
| | - William M Durham
- School of Mathematical and Physical Sciences, University of Sheffield, Sheffield, UK
| | - David P Hornby
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute, University of Sheffield, Sheffield, UK
| | - Rebecca M Corrigan
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute, University of Sheffield, Sheffield, UK
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Jeffrey Green
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute, University of Sheffield, Sheffield, UK
| | - Jamie K Hobbs
- School of Mathematical and Physical Sciences, University of Sheffield, Sheffield, UK
| | - Simon J Foster
- School of Biosciences, University of Sheffield, Sheffield, UK
- The Florey Institute, University of Sheffield, Sheffield, UK
| |
Collapse
|
5
|
Su M, Hoang KL, Penley M, Davis MH, Gresham JD, Morran LT, Read TD. Host and antibiotic jointly select for greater virulence in Staphylococcus aureus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.31.610628. [PMID: 39257827 PMCID: PMC11383984 DOI: 10.1101/2024.08.31.610628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Widespread antibiotic usage has resulted in the rapid evolution of drug-resistant bacterial pathogens and poses significant threats to public health. Resolving how pathogens respond to antibiotics under different contexts is critical for understanding disease emergence and evolution going forward. The impact of antibiotics has been demonstrated most directly through in vitro pathogen passaging experiments. Independent from antibiotic selection, interactions with hosts have also altered the evolutionary trajectories and fitness landscapes of pathogens, shaping infectious disease outcomes. However, it is unclear how interactions between hosts and antibiotics impact the evolution of pathogen virulence. Here, we evolved and re-sequenced Staphylococcus aureus, a major bacterial pathogen, varying exposure to host and antibiotics to tease apart the contributions of these selective pressures on pathogen adaptation. After 12 passages, S. aureus evolving in Caenorhabditis elegans nematodes exposed to a sub-minimum inhibitory concentration of antibiotic (oxacillin) became highly virulent, regardless of whether the ancestral pathogen was methicillin-resistant (MRSA) or methicillin-sensitive (MSSA). Host and antibiotic exposure selected for reduced drug susceptibility in MSSA lineages while increasing MRSA total growth outside hosts. We identified mutations in genes involved in complex regulatory networks linking virulence and metabolism, including codY , agr , and gdpP , suggesting that rapid adaptation to infect hosts may have pleiotropic effects. In particular, MSSA populations under selection from host and antibiotic accumulated mutations in the global regulator gene codY , which controls biofilm formation in S. aureus. These populations had indeed evolved more robust biofilms-a trait linked to both virulence and antibiotic resistance-suggesting evolution of one trait can confer multiple adaptive benefits. Despite evolving in similar environments, MRSA and MSSA populations proceeded on divergent evolutionary paths, with MSSA populations exhibiting more similarities across replicate populations. Our results underscore the importance of considering multiple and concurrent selective pressures as drivers of pervasive pathogen traits.
Collapse
|
6
|
Alves J, Vrieling M, Ring N, Yebra G, Pickering A, Prajsnar TK, Renshaw SA, Fitzgerald JR. Experimental evolution of Staphylococcus aureus in macrophages: dissection of a conditional adaptive trait promoting intracellular survival. mBio 2024; 15:e0034624. [PMID: 38682911 PMCID: PMC11237485 DOI: 10.1128/mbio.00346-24] [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: 02/05/2024] [Accepted: 03/26/2024] [Indexed: 05/01/2024] Open
Abstract
Staphylococcus aureus is a major pathogen associated with important diseases in humans and animals. Macrophages are a key component of the innate immune response to S. aureus infection and play a major role in disease outcomes. To investigate the adaptive evolution of S. aureus in response to macrophages, we developed an experimental infection assay. S. aureus strains representing major human epidemic clones were passaged many times in a macrophage cell line, accumulating mutations in an array of genomic loci. Phenotypic analysis revealed the emergence of a lineage exhibiting increased survival in macrophages and human blood, and resistance to vancomycin. The evolved lineage exhibited a previously undescribed small colony variant (SCV) phenotype characterized by hyper-pigmentation, which resulted from a missense mutation in rsbW. Notably, the novel SCV was a conditional adaptive trait that was unstable in nutrient-replete conditions in vitro, rapidly converting from hyper-pigmented SCV to a non-pigmented large colony variant via spontaneous sigB deletion events. Importantly, we identified similar deletions in the genome sequences of a limited number of clinical S. aureus isolates from public databases, indicating that related events may occur during clinical infection. Experimental infection of zebrafish did not reveal a difference in virulence between parent and novel SCV but demonstrated an in vivo fitness cost for the compensatory sigB deletion events. Taken together, we report an experimental evolutionary approach for investigating bacterial innate immune cell interactions, revealing a conditional adaptation that promotes S. aureus survival in macrophages and resistance to vancomycin. IMPORTANCE Staphylococcus aureus is an important human bacterial pathogen. The host response to S. aureus involves the production of innate immune cells such as macrophages which are important for fighting infection. Here we report a new model of experimental evolution for studying how S. aureus can evade killing by macrophages. We identified a novel adaptive phenotype that promotes survival in macrophages and blood and resistance to antibiotics. The phenotype is lost rapidly upon growth in nutrient-rich conditions via disruption of the alternative sigma factor sigB, revealing a conditional niche-specific fitness advantage. Genomic analysis of clinical isolates suggests similar adaptations may occur during human infections. Our model may be used broadly to identify adaptations of S. aureus to the innate immune response.
Collapse
Affiliation(s)
- Joana Alves
- The Roslin Institute, University of Edinburgh, Easter Bush, Midlothian, Edinburgh, United Kingdom
| | - Manouk Vrieling
- The Roslin Institute, University of Edinburgh, Easter Bush, Midlothian, Edinburgh, United Kingdom
| | - Natalie Ring
- The Roslin Institute, University of Edinburgh, Easter Bush, Midlothian, Edinburgh, United Kingdom
| | - Gonzalo Yebra
- The Roslin Institute, University of Edinburgh, Easter Bush, Midlothian, Edinburgh, United Kingdom
| | - Amy Pickering
- The Roslin Institute, University of Edinburgh, Easter Bush, Midlothian, Edinburgh, United Kingdom
| | - Tomasz K. Prajsnar
- Florey Institute, Bateson Centre and Division of Clinical Medicine, School of Medicine and Population Health, Sheffield, United Kingdom
| | - Stephen A. Renshaw
- Florey Institute, Bateson Centre and Division of Clinical Medicine, School of Medicine and Population Health, Sheffield, United Kingdom
| | - J. Ross Fitzgerald
- The Roslin Institute, University of Edinburgh, Easter Bush, Midlothian, Edinburgh, United Kingdom
| |
Collapse
|
7
|
Basta DW, Campbell IW, Sullivan EJ, Hotinger JA, Hullahalli K, Waldor MK. Inducible transposon mutagenesis for genome-scale forward genetics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595064. [PMID: 38826325 PMCID: PMC11142078 DOI: 10.1101/2024.05.21.595064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Transposon insertion sequencing (Tn-seq) is a powerful method for genome-scale functional genetics in bacteria. However, its effectiveness is often limited by a lack of mutant diversity, caused by either inefficient transposon delivery or stochastic loss of mutants due to population bottlenecks. Here, we introduce "InducTn-seq", which leverages inducible mutagenesis for temporal control of transposition. InducTn-seq generates millions of transposon mutants from a single colony, enabling the sensitive detection of subtle fitness defects and transforming binary classifications of gene essentiality into a quantitative fitness measurement across both essential and non-essential genes. Using a mouse model of infectious colitis, we show that InducTn-seq bypasses a highly restrictive host bottleneck to generate a diverse transposon mutant population from the few cells that initiate infection, revealing the role of oxygen-related metabolic plasticity in pathogenesis. Overall, InducTn-seq overcomes the limitations of traditional Tn-seq, unlocking new possibilities for genome-scale forward genetic screens in bacteria.
Collapse
Affiliation(s)
- David W. Basta
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ian W. Campbell
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Emily J. Sullivan
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Julia A Hotinger
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Karthik Hullahalli
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Matthew K. Waldor
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| |
Collapse
|
8
|
Tooke AK, Foster SJ. Using the Zebrafish Larval Model of Infection to Investigate Antibiotic Efficacy and Combination Treatments Against Staphylococcus aureus. Methods Mol Biol 2024; 2833:1-10. [PMID: 38949695 DOI: 10.1007/978-1-0716-3981-8_1] [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: 07/02/2024]
Abstract
There is an increasing need for new treatment regimens to combat antibiotic-resistant strains of bacteria. Staphylococcus aureus is a clinically important, opportunist pathogen that has developed resistance to a range of antibiotics. The zebrafish larval model of systemic disease has been increasingly utilized to elucidate S. aureus virulence mechanisms and host-pathogen interactions. Here, we outline how this model can be used to investigate the effects of different antibiotics alone and in combination against S. aureus.
Collapse
Affiliation(s)
- Amy K Tooke
- School of Biosciences, University of Sheffield, Sheffield, UK
- Newcastle University Biosciences Institute, Newcastle upon Tyne, UK
| | - Simon J Foster
- School of Biosciences, University of Sheffield, Sheffield, UK.
- Florey Institute, University of Sheffield, Sheffield, UK.
- Krebs Institute, University of Sheffield, Sheffield, UK.
| |
Collapse
|
9
|
Wang Z, Wu A, Cheng W, Li Y, Li D, Wang L, Zhang X, Xiao Y. Adoptive macrophage directed photodynamic therapy of multidrug-resistant bacterial infection. Nat Commun 2023; 14:7251. [PMID: 37945555 PMCID: PMC10636156 DOI: 10.1038/s41467-023-43074-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023] Open
Abstract
Multidrug-resistant (MDR) bacteria cause severe clinical infections and a high mortality rate of over 40% in patients with immunodeficiencies. Therefore, more effective, broad-spectrum, and accurate treatment for severe cases of infection is urgently needed. Here, we present an adoptive transfer of macrophages loaded with a near-infrared photosensitizer (Lyso700D) in lysosomes to boost innate immunity and capture and eliminate bacteria through a photodynamic effect. In this design, the macrophages can track and capture bacteria into the lysosomes through innate immunity, thereby delivering the photosensitizer to the bacteria within a single lysosome, maximizing the photodynamic effect and minimizing the side effects. Our results demonstrate that this therapeutic strategy eliminated MDR Staphylococcus aureus (MRSA) and Acinetobacter baumannii (AB) efficiently and cured infected mice in both two models with 100% survival compared to 10% in the control groups. Promisingly, in a rat model of central nervous system bacterial infection, we performed the therapy using bone marrow-divided macrophages and implanted glass fiber to conduct light irradiation through the lumbar cistern. 100% of infected rats survived while none of the control group survived. Our work proposes an efaficient and safe strategy to cure MDR bacterial infections, which may benefit the future clinical treatment of infection.
Collapse
Affiliation(s)
- Zehui Wang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Anhua Wu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110055, China
| | - Wen Cheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110055, China
| | - Yuhe Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110055, China
| | - Dingxuan Li
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Lai Wang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Xinfu Zhang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China.
| | - Yi Xiao
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| |
Collapse
|
10
|
Howden BP, Giulieri SG, Wong Fok Lung T, Baines SL, Sharkey LK, Lee JYH, Hachani A, Monk IR, Stinear TP. Staphylococcus aureus host interactions and adaptation. Nat Rev Microbiol 2023; 21:380-395. [PMID: 36707725 PMCID: PMC9882747 DOI: 10.1038/s41579-023-00852-y] [Citation(s) in RCA: 286] [Impact Index Per Article: 143.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2023] [Indexed: 01/28/2023]
Abstract
Invasive Staphylococcus aureus infections are common, causing high mortality, compounded by the propensity of the bacterium to develop drug resistance. S. aureus is an excellent case study of the potential for a bacterium to be commensal, colonizing, latent or disease-causing; these states defined by the interplay between S. aureus and host. This interplay is multidimensional and evolving, exemplified by the spread of S. aureus between humans and other animal reservoirs and the lack of success in vaccine development. In this Review, we examine recent advances in understanding the S. aureus-host interactions that lead to infections. We revisit the primary role of neutrophils in controlling infection, summarizing the discovery of new immune evasion molecules and the discovery of new functions ascribed to well-known virulence factors. We explore the intriguing intersection of bacterial and host metabolism, where crosstalk in both directions can influence immune responses and infection outcomes. This Review also assesses the surprising genomic plasticity of S. aureus, its dualism as a multi-mammalian species commensal and opportunistic pathogen and our developing understanding of the roles of other bacteria in shaping S. aureus colonization.
Collapse
Affiliation(s)
- Benjamin P. Howden
- grid.1008.90000 0001 2179 088XCentre for Pathogen Genomics, The University of Melbourne, Melbourne, Victoria Australia ,grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria Australia ,grid.410678.c0000 0000 9374 3516Department of Infectious Diseases, Austin Health, Heidelberg, Victoria Australia ,grid.416153.40000 0004 0624 1200Microbiology Department, Royal Melbourne Hospital, Melbourne, Victoria Australia
| | - Stefano G. Giulieri
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria Australia ,grid.416153.40000 0004 0624 1200Victorian Infectious Diseases Service, Royal Melbourne Hospital, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria Australia
| | - Tania Wong Fok Lung
- grid.21729.3f0000000419368729Department of Paediatrics, Columbia University, New York, NY USA
| | - Sarah L. Baines
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria Australia
| | - Liam K. Sharkey
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria Australia
| | - Jean Y. H. Lee
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria Australia ,grid.419789.a0000 0000 9295 3933Department of Infectious Diseases, Monash Health, Clayton, Victoria Australia
| | - Abderrahman Hachani
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria Australia
| | - Ian R. Monk
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria Australia
| | - Timothy P. Stinear
- grid.1008.90000 0001 2179 088XCentre for Pathogen Genomics, The University of Melbourne, Melbourne, Victoria Australia ,grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria Australia
| |
Collapse
|
11
|
Pidwill GR, Pyrah JF, Sutton JAF, Best A, Renshaw SA, Foster SJ. Clonal population expansion of Staphylococcus aureus occurs due to escape from a finite number of intraphagocyte niches. Sci Rep 2023; 13:1188. [PMID: 36681703 PMCID: PMC9867732 DOI: 10.1038/s41598-023-27928-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 01/10/2023] [Indexed: 01/22/2023] Open
Abstract
Staphylococcus aureus is a human commensal and also an opportunist pathogen causing life threatening infections. During S. aureus disease, the abscesses that characterise infection can be clonal, whereby a large bacterial population is founded by a single or few organisms. Our previous work has shown that macrophages are responsible for restricting bacterial growth such that a population bottleneck occurs and clonality can emerge. A subset of phagocytes fail to control S. aureus resulting in bacterial division, escape and founding of microabscesses that can seed other host niches. Here we investigate the basis for clonal microabscess formation, using in vitro and in silico models of S. aureus macrophage infection. Macrophages that fail to control S. aureus are characterised by formation of intracellular bacterial masses, followed by cell lysis. High-resolution microscopy reveals that most macrophages had internalised only a single S. aureus, providing a conceptual framework for clonal microabscess generation, which was supported by a stochastic individual-based, mathematical model. Once a threshold of masses was reached, increasing the number of infecting bacteria did not result in greater mass numbers, despite enhanced phagocytosis. This suggests a finite number of permissive, phagocyte niches determined by macrophage associated factors. Increased understanding of the parameters of infection dynamics provides avenues for development of rational control measures.
Collapse
Affiliation(s)
- Grace R Pidwill
- School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK
- Florey Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Josie F Pyrah
- School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK
- Florey Institute, University of Sheffield, Sheffield, S10 2TN, UK
- The Bateson Centre, University of Sheffield, Sheffield, S10 2TN, UK
| | - Joshua A F Sutton
- School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK
- Florey Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Alex Best
- School of Mathematics & Statistics, University of Sheffield, Sheffield, S3 7RH, UK.
| | - Stephen A Renshaw
- Florey Institute, University of Sheffield, Sheffield, S10 2TN, UK.
- The Bateson Centre, University of Sheffield, Sheffield, S10 2TN, UK.
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, S10 2RX, UK.
| | - Simon J Foster
- School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK.
- Florey Institute, University of Sheffield, Sheffield, S10 2TN, UK.
| |
Collapse
|
12
|
Abdelaziz R, Tartor YH, Barakat AB, EL-Didamony G, Gado MM, Berbecea A, Radulov HDI. Bioactive metabolites of Streptomyces misakiensis display broad-spectrum antimicrobial activity against multidrug-resistant bacteria and fungi. Front Cell Infect Microbiol 2023; 13:1162721. [PMID: 37168394 PMCID: PMC10165089 DOI: 10.3389/fcimb.2023.1162721] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/07/2023] [Indexed: 05/13/2023] Open
Abstract
Background Antimicrobial resistance is a serious threat to public health globally. It is a slower-moving pandemic than COVID-19, so we are fast running out of treatment options. Purpose Thus, this study was designed to search for an alternative biomaterial with broad-spectrum activity for the treatment of multidrug-resistant (MDR) bacterial and fungal pathogen-related infections. Methods We isolated Streptomyces species from soil samples and identified the most active strains with antimicrobial activity. The culture filtrates of active species were purified, and the bioactive metabolite extracts were identified by thin-layer chromatography (TLC), preparative high-performance liquid chromatography (HPLC), nuclear magnetic resonance (NMR) spectroscopy, and gas chromatography-mass spectrometry (GC-MS). The minimum inhibitory concentrations (MICs) of the bioactive metabolites against MDR bacteria and fungi were determined using the broth microdilution method. Results Preliminary screening revealed that Streptomyces misakiensis and S. coeruleorubidus exhibited antimicrobial potential. The MIC50 and MIC90 of S. misakiensis antibacterial bioactive metabolite (ursolic acid methyl ester) and antifungal metabolite (tetradecamethylcycloheptasiloxane) against all tested bacteria and fungi were 0.5 μg/ml and 1 μg/mL, respectively, versus S. coeruleorubidus metabolites: thiocarbamic acid, N,N-dimethyl, S-1,3-diphenyl-2-butenyl ester against bacteria (MIC50: 2 μg/ml and MIC90: 4 μg/mL) and fungi (MIC50: 4 μg/ml and MIC90: 8 μg/mL). Ursolic acid methyl ester was active against ciprofloxacin-resistant strains of Streptococcus pyogenes, S. agalactiae, Escherichia coli, Klebsiella pneumoniae, and Salmonella enterica serovars, colistin-resistant Aeromonas hydrophila and K. pneumoniae, and vancomycin-resistant Staphylococcus aureus. Tetradecamethylcycloheptasiloxane was active against azole- and amphotericin B-resistant Candida albicans, Cryptococcus neoformans, C. gattii, Aspergillus flavus, A. niger, and A. fumigatus. Ursolic acid methyl ester was applied in vivo for treating S. aureus septicemia and K. pneumoniae pneumonia models in mice. In the septicemia model, the ursolic acid methyl ester-treated group had a significant 4.00 and 3.98 log CFU/g decrease (P < 0.05) in liver and spleen tissue compared to the infected, untreated control group. Lung tissue in the pneumonia model showed a 2.20 log CFU/g significant decrease in the ursolic acid methyl ester-treated group in comparison to the control group. The haematological and biochemical markers in the ursolic acid methyl ester-treated group did not change in a statistically significant way. Moreover, no abnormalities were found in the histopathology of the liver, kidneys, lungs, and spleen of ursolic acid methyl ester-treated mice in comparison with the control group. Conclusion S. misakiensis metabolite extracts are broad-spectrum antimicrobial biomaterials that can be further investigated for the potential against MDR pathogen infections. Hence, it opens up new horizons for exploring alternative drugs for current and reemerging diseases.
Collapse
Affiliation(s)
- Rewan Abdelaziz
- Department of Microbiology, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Yasmine H. Tartor
- Department of Microbiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
- *Correspondence: Yasmine H. Tartor, ;
| | - Ahmed B. Barakat
- Department of Microbiology, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Gamal EL-Didamony
- Department of Botany and Microbiology, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Marwa M. Gado
- Department of Microbiology, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Adina Berbecea
- Department of Soil Science, University of Life Science”King Mihai I” from, Timioara, Romania
| | | |
Collapse
|
13
|
Antibiotics Limit Adaptation of Drug-Resistant Staphylococcus aureus to Hypoxia. Antimicrob Agents Chemother 2022; 66:e0092622. [PMID: 36409116 PMCID: PMC9765076 DOI: 10.1128/aac.00926-22] [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] [Indexed: 11/23/2022] Open
Abstract
Bacterial pathogens are confronted with a range of challenges at the site of infection, including exposure to antibiotic treatment and harsh physiological conditions, that can alter the fitness benefits and costs of acquiring antibiotic resistance. Here, we develop an experimental system to recapitulate resistance gene acquisition by Staphylococcus aureus and test how the subsequent evolution of the resistant bacterium is modulated by antibiotic treatment and oxygen levels, both of which are known to vary extensively at sites of infection. We show that acquiring tetracycline resistance was costly, reducing competitive growth against the isogenic strain without the resistance gene in the absence of the antibiotic, for S. aureus under hypoxic but not normoxic conditions. Treatment with tetracycline or doxycycline drove the emergence of enhanced resistance through mutations in an RluD-like protein-encoding gene and duplications of tetL, encoding the acquired tetracycline-specific efflux pump. In contrast, evolutionary adaptation by S. aureus to hypoxic conditions, which evolved in the absence of antibiotics through mutations affecting gyrB, was impeded by antibiotic treatment. Together, these data suggest that the horizontal acquisition of a new resistance mechanism is merely a starting point for the emergence of high-level resistance under antibiotic selection but that antibiotic treatment constrains pathogen adaptation to other important environmental selective forces such as hypoxia, which in turn could limit the survival of these highly resistant but poorly adapted genotypes after antibiotic treatment is ended.
Collapse
|
14
|
NO-Stressed Y. pseudotuberculosis Has Decreased Cell Division Rates in the Mouse Spleen. Infect Immun 2022; 90:e0016722. [PMID: 35862700 PMCID: PMC9387282 DOI: 10.1128/iai.00167-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Fluorescence dilution approaches can detect bacterial cell division events and can detect if there are differential rates of cell division across individual cells within a population. This approach typically involves inducing expression of a fluorescent protein and then tracking partitioning of fluorescence into daughter cells. However, fluorescence can be diluted very quickly within a rapidly replicating population, such as pathogenic bacterial populations replicating within host tissues. To overcome this limitation, we have generated two revTetR reporter constructs, where either mCherry or yellow fluorescent protein (YFP) is constitutively expressed and repressed by addition of tetracyclines, resulting in fluorescence dilution within defined time frames. We show that fluorescent signals are diluted in replicating populations and that signal accumulates in growth-inhibited populations, including during nitric oxide (NO) exposure. Furthermore, we show that tetracyclines can be delivered to the mouse spleen during Yersinia pseudotuberculosis infection and defined a drug concentration that results in even exposure of cells to tetracyclines. We then used this system to visualize bacterial cell division within defined time frames postinfection. revTetR-mCherry allowed us to detect slow-growing cells in response to NO in culture; however, this strain had a growth defect within mouse tissues, which complicated results. To address this issue, we constructed revTetR-YFP using the less toxic YFP and showed that heightened NO exposure correlated with heightened YFP signal, indicating decreased cell division rates within this subpopulation in vivo. This revTetR reporter will provide a critical tool for future studies to identify and isolate slowly replicating bacterial subpopulations from host tissues.
Collapse
|
15
|
Sudan S, Li J. Dispersal of Pathogen-associated Multispecies Biofilm by Novel Probiotic Bacillus subtilis in a Contact Dependent Manner. J Appl Microbiol 2022; 133:2501-2515. [PMID: 35858688 DOI: 10.1111/jam.15721] [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: 02/16/2022] [Revised: 06/20/2022] [Accepted: 06/27/2022] [Indexed: 11/28/2022]
Abstract
AIMS Biofilms are involved in pathogenesis of various bacterial infections. Treatment of biofilm related bacterial infection remains a major challenge due to the reduced efficacy of antibiotics and associated antibiotic resistance. Given the increasing prevalence of Enterotoxic Escherichia. coli (ETEC), Salmonella Typhimurium (S. Typhimurium), and Methicillin-resistant Staphylococcus aureus (MRSA) related infections and associated drug resistance, it is imperative to develop alternative strategies for treatment and prevention. The current study investigated antibiofilm activity of a recently isolated Bacillus subtilis (B. subtilis-9) against these pathogens. METHODS AND RESULTS Crystal violet staining showed that treatment with B. subtilis-9 significantly reduced biofilm biomass of ETEC (60-80%), S. Typhimurium (68-73%) and MRSA (66-82%). In addition, B. subtilis-9 significantly reduced pre-formed biofilm biomass of ETEC (59%), S. Typhimurium (62%), MRSA (65%) and multispecies (58%). Fluorescence microscopy revealed that B. subtilis-9 treatment significantly reduced the thickness of biofilm and viability of the embedded bacteria. Additionally, B. subtilis-9 significantly reduced planktonic cell growth of ETEC (92%), S. Typhimurium (94%) and MRSA (93%). Interestingly, transwell assay showed that B. subtilis-9 exhibited antibiofilm properties in a cell-to-cell contact dependant manner and significantly reduced mRNA expression of biofilm related genes, bssS, luxS and ihfB in ETEC. CONCLUSION Novel B. subtilis-9 exhibits a strong inhibitory activity against ETEC, S. Typhimurium and MRSA biofilm formation and adhesion to abiotic surfaces. With further investigations, our study could bring forward a novel Bacillus based probiotic intervention strategy to combat pathogenic biofilms, in clinical and agricultural settings. SIGNIFICANCE AND IMPACT OF THE STUDY Probiotic bacteria propose a potential alternative in combating biofilm related infections, however, data on the efficacy and strain selection is limited. Data from this study is critical in further developing a Bacillus-based novel probiotic applications that may reduce the use of antibiotics in biofilm related infections in humans and animals.
Collapse
Affiliation(s)
- Sudhanshu Sudan
- Department of Animal Biosciences, University of Guelph, Guelph, Ontario, Canada
| | - Julang Li
- Department of Animal Biosciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
16
|
Giulieri SG, Guérillot R, Duchene S, Hachani A, Daniel D, Seemann T, Davis JS, Tong SYC, Young BC, Wilson DJ, Stinear TP, Howden BP. Niche-specific genome degradation and convergent evolution shaping Staphylococcus aureus adaptation during severe infections. eLife 2022; 11:e77195. [PMID: 35699423 PMCID: PMC9270034 DOI: 10.7554/elife.77195] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
During severe infections, Staphylococcus aureus moves from its colonising sites to blood and tissues and is exposed to new selective pressures, thus, potentially driving adaptive evolution. Previous studies have shown the key role of the agr locus in S. aureus pathoadaptation; however, a more comprehensive characterisation of genetic signatures of bacterial adaptation may enable prediction of clinical outcomes and reveal new targets for treatment and prevention of these infections. Here, we measured adaptation using within-host evolution analysis of 2590 S. aureus genomes from 396 independent episodes of infection. By capturing a comprehensive repertoire of single nucleotide and structural genome variations, we found evidence of a distinctive evolutionary pattern within the infecting populations compared to colonising bacteria. These invasive strains had up to 20-fold enrichments for genome degradation signatures and displayed significantly convergent mutations in a distinctive set of genes, linked to antibiotic response and pathogenesis. In addition to agr-mediated adaptation, we identified non-canonical, genome-wide significant loci including sucA-sucB and stp1. The prevalence of adaptive changes increased with infection extent, emphasising the clinical significance of these signatures. These findings provide a high-resolution picture of the molecular changes when S. aureus transitions from colonisation to severe infection and may inform correlation of infection outcomes with adaptation signatures.
Collapse
Affiliation(s)
- Stefano G Giulieri
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneAustralia
- Department of Infectious Diseases, Austin HealthHeidelbergAustralia
- Victorian Infectious Diseases Service, Royal Melbourne HospitalMelbourneAustralia
| | - Romain Guérillot
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneAustralia
| | - Sebastian Duchene
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneAustralia
| | - Abderrahman Hachani
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneAustralia
| | - Diane Daniel
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneAustralia
- Microbiological Diagnostic Unit Public Health Laboratory, The University of Melbourne at the Doherty Institute for Infection and ImmunityMelbourneAustralia
| | - Torsten Seemann
- Microbiological Diagnostic Unit Public Health Laboratory, The University of Melbourne at the Doherty Institute for Infection and ImmunityMelbourneAustralia
| | - Joshua S Davis
- Department of Infectious Diseases, John Hunter HospitalNewcastle, New South WalesAustralia
- Menzies School of Health Research, Charles Darwin UniversityCasuarina, Northern TerritoryAustralia
| | - Steven YC Tong
- Menzies School of Health Research, Charles Darwin UniversityCasuarina, Northern TerritoryAustralia
- Victorian Infectious Disease Service, Royal Melbourne Hospital, and University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
| | | | - Daniel J Wilson
- Big Data Institute, Nuffield Department of Population Health, Li Ka Shing Centre for Health Information and Discovery, Old Road Campus, University of OxfordOxfordUnited Kingdom
| | - Timothy P Stinear
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneAustralia
| | - Benjamin P Howden
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneAustralia
- Department of Infectious Diseases, Austin HealthHeidelbergAustralia
- Microbiological Diagnostic Unit Public Health Laboratory, The University of Melbourne at the Doherty Institute for Infection and ImmunityMelbourneAustralia
| |
Collapse
|
17
|
Grijmans BJM, van der Kooij SB, Varela M, Meijer AH. LAPped in Proof: LC3-Associated Phagocytosis and the Arms Race Against Bacterial Pathogens. Front Cell Infect Microbiol 2022; 11:809121. [PMID: 35047422 PMCID: PMC8762105 DOI: 10.3389/fcimb.2021.809121] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/10/2021] [Indexed: 01/05/2023] Open
Abstract
Cells of the innate immune system continuously patrol the extracellular environment for potential microbial threats that are to be neutralized by phagocytosis and delivery to lysosomes. In addition, phagocytes employ autophagy as an innate immune mechanism against pathogens that succeed to escape the phagolysosomal pathway and invade the cytosol. In recent years, LC3-associated phagocytosis (LAP) has emerged as an intermediate between phagocytosis and autophagy. During LAP, phagocytes target extracellular microbes while using parts of the autophagic machinery to label the cargo-containing phagosomes for lysosomal degradation. LAP contributes greatly to host immunity against a multitude of bacterial pathogens. In the pursuit of survival, bacteria have developed elaborate strategies to disarm or circumvent the LAP process. In this review, we will outline the nature of the LAP mechanism and discuss recent insights into its interplay with bacterial pathogens.
Collapse
Affiliation(s)
| | | | - Monica Varela
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | | |
Collapse
|
18
|
Investigating Pathogenicity and Virulence of Staphylococcus pettenkoferi: An Emerging Pathogen. Int J Mol Sci 2021; 22:ijms222413614. [PMID: 34948410 PMCID: PMC8704685 DOI: 10.3390/ijms222413614] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 01/22/2023] Open
Abstract
Staphylococcus pettenkoferi is a coagulase-negative Staphylococcus identified in 2002 that has been implicated in human diseases as an opportunistic pathogenic bacterium. Its multiresistant character is becoming a major health problem, yet the pathogenicity of S. pettenkoferi is poorly characterized. In this study, the pathogenicity of a S. pettenkoferi clinical isolate from diabetic foot osteomyelitis was compared with a Staphylococcus aureus strain in various in vitro and in vivo experiments. Growth kinetics were compared against S. aureus, and bacteria survival was assessed in the RAW 264.7 murine macrophage cell line, the THP-1 human leukemia monocytic cell line, and the HaCaT human keratinocyte cell line. Ex vivo analysis was performed in whole blood survival assays and in vivo assays via the infection model of zebrafish embryos. Moreover, whole-genome analysis was performed. Our results show that S. pettenkoferi was able to survive in human blood, human keratinocytes, murine macrophages, and human macrophages. S. pettenkoferi demonstrated its virulence by causing substantial embryo mortality in the zebrafish model. Genomic analysis revealed virulence factors such as biofilm-encoding genes (e.g., icaABCD; rsbUVW) and regulator-encoding genes (e.g., agr, mgrA, sarA, saeS) well characterized in S. aureus. This study thus advances the knowledge of this under-investigated pathogen and validates the zebrafish infection model for this bacterium.
Collapse
|
19
|
Helekal D, Ledda A, Volz E, Wyllie D, Didelot X. Bayesian inference of clonal expansions in a dated phylogeny. Syst Biol 2021; 71:1073-1087. [PMID: 34893904 PMCID: PMC9366454 DOI: 10.1093/sysbio/syab095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 11/23/2021] [Accepted: 11/29/2021] [Indexed: 11/16/2022] Open
Abstract
Microbial population genetics models often assume that all lineages are constrained by the same population size dynamics over time. However, many neutral and selective events can invalidate this assumption and can contribute to the clonal expansion of a specific lineage relative to the rest of the population. Such differential phylodynamic properties between lineages result in asymmetries and imbalances in phylogenetic trees that are sometimes described informally but which are difficult to analyze formally. To this end, we developed a model of how clonal expansions occur and affect the branching patterns of a phylogeny. We show how the parameters of this model can be inferred from a given dated phylogeny using Bayesian statistics, which allows us to assess the probability that one or more clonal expansion events occurred. For each putative clonal expansion event, we estimate its date of emergence and subsequent phylodynamic trajectory, including its long-term evolutionary potential which is important to determine how much effort should be placed on specific control measures. We demonstrate the applicability of our methodology on simulated and real data sets. Inference under our clonal expansion model can reveal important features in the evolution and epidemiology of infectious disease pathogens. [Clonal expansion; genomic epidemiology; microbial population genomics; phylodynamics.]
Collapse
Affiliation(s)
- David Helekal
- Centre for Doctoral Training in Mathematics for Real-World Systems, University of Warwick, United Kingdom
| | - Alice Ledda
- Healthcare Associated Infections and Antimicrobial Resistance Division, National Infection Service, Public Health England, United Kingdom
| | - Erik Volz
- Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, United Kingdom
| | - David Wyllie
- Field Service, East of England, National Infection Service, Public Health England, Cambridge, United Kingdom
| | - Xavier Didelot
- School of Life Sciences and Department of Statistics, University of Warwick, United Kingdom
| |
Collapse
|
20
|
Gibson JF, Pidwill GR, Carnell OT, Surewaard BGJ, Shamarina D, Sutton JAF, Jeffery C, Derré-Bobillot A, Archambaud C, Siggins MK, Pollitt EJG, Johnston SA, Serror P, Sriskandan S, Renshaw SA, Foster SJ. Commensal bacteria augment Staphylococcus aureus infection by inactivation of phagocyte-derived reactive oxygen species. PLoS Pathog 2021; 17:e1009880. [PMID: 34529737 PMCID: PMC8478205 DOI: 10.1371/journal.ppat.1009880] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/28/2021] [Accepted: 08/09/2021] [Indexed: 12/22/2022] Open
Abstract
Staphylococcus aureus is a human commensal organism and opportunist pathogen, causing potentially fatal disease. The presence of non-pathogenic microflora or their components, at the point of infection, dramatically increases S. aureus pathogenicity, a process termed augmentation. Augmentation is associated with macrophage interaction but by a hitherto unknown mechanism. Here, we demonstrate a breadth of cross-kingdom microorganisms can augment S. aureus disease and that pathogenesis of Enterococcus faecalis can also be augmented. Co-administration of augmenting material also forms an efficacious vaccine model for S. aureus. In vitro, augmenting material protects S. aureus directly from reactive oxygen species (ROS), which correlates with in vivo studies where augmentation restores full virulence to the ROS-susceptible, attenuated mutant katA ahpC. At the cellular level, augmentation increases bacterial survival within macrophages via amelioration of ROS, leading to proliferation and escape. We have defined the molecular basis for augmentation that represents an important aspect of the initiation of infection. S. aureus is a commensal inhabitant of the human skin and nares. However, it can cause serious diseases if it is able to breach our protective barriers such as the skin, often via wounds or surgery. If infection occurs via a wound, this initial inoculum contains both the pathogen, other members of the microflora and also wider environmental microbes. We have previously described “augmentation”, whereby this other non-pathogenic material can enhance the ability of S. aureus to lead to a serious disease outcome. Here we have determined the breadth of augmenting material and elucidated the cellular and molecular basis for its activity. Augmentation occurs via shielding of S. aureus from the direct bactericidal effects of reactive oxygen species produced by macrophages. This initial protection enables the effective establishment of S. aureus infection. Understanding augmentation not only explains an important facet of the interaction of S. aureus with our innate immune system, but also provides a platform for the development of novel prophylaxis approaches.
Collapse
Affiliation(s)
- Josie F. Gibson
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Grace R. Pidwill
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
| | - Oliver T. Carnell
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
| | - Bas G. J. Surewaard
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Daria Shamarina
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
| | - Joshua A. F. Sutton
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
| | - Charlotte Jeffery
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
| | | | - Cristel Archambaud
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Matthew K. Siggins
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Eric J. G. Pollitt
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
| | - Simon A. Johnston
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, 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
| | - Pascale Serror
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Shiranee Sriskandan
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Stephen A. Renshaw
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, 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
- * E-mail: (SAR); (SJF)
| | - Simon J. Foster
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
- * E-mail: (SAR); (SJF)
| |
Collapse
|
21
|
Zebrafish larvae as experimental model to expedite the search for new biomarkers and treatments for neonatal sepsis. J Clin Transl Sci 2021; 5:e140. [PMID: 34422320 PMCID: PMC8358844 DOI: 10.1017/cts.2021.803] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/02/2021] [Accepted: 06/09/2021] [Indexed: 12/22/2022] Open
Abstract
Neonatal sepsis is a major cause of death and disability in newborns. Commonly used biomarkers for diagnosis and evaluation of treatment response lack sufficient sensitivity or specificity. Additionally, new targets to treat the dysregulated immune response are needed, as are methods to effectively screen drugs for these targets. Available research methods have hitherto not yielded the breakthroughs required to significantly improve disease outcomes, we therefore describe the potential of zebrafish (Danio rerio) larvae as preclinical model for neonatal sepsis. In biomedical research, zebrafish larvae combine the complexity of a whole organism with the convenience and high-throughput potential of in vitro methods. This paper illustrates that zebrafish exhibit an immune system that is remarkably similar to humans, both in terms of types of immune cells and signaling pathways. Moreover, the developmental state of the larval immune system is highly similar to human neonates. We provide examples of zebrafish larvae being used to study infections with pathogens commonly causing neonatal sepsis and discuss known limitations. We believe this species could expedite research into immune regulation during neonatal sepsis and may hold keys for the discovery of new biomarkers and novel treatment targets as well as for screening of targeted drug therapies.
Collapse
|
22
|
Modifying TIMER to generate a slow-folding DsRed derivative for optimal use in quickly-dividing bacteria. PLoS Pathog 2021; 17:e1009284. [PMID: 34214139 PMCID: PMC8291646 DOI: 10.1371/journal.ppat.1009284] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 07/20/2021] [Accepted: 06/14/2021] [Indexed: 11/29/2022] Open
Abstract
It is now well appreciated that members of pathogenic bacterial populations exhibit heterogeneity in growth rates and metabolic activity, and it is known this can impact the ability to eliminate all members of the bacterial population during antibiotic treatment. It remains unclear which pathways promote slowed bacterial growth within host tissues, primarily because it has been difficult to identify and isolate slow growing bacteria from host tissues for downstream analyses. To overcome this limitation, we have developed a novel variant of TIMER, a slow-folding fluorescent protein, named DsRed42, to identify subsets of slowly dividing bacteria within host tissues. The original TIMER folds too slowly for fluorescence accumulation in quickly replicating bacterial species (Escherichia coli, Yersinia pseudotuberculosis), however DsRed42 accumulates red fluorescence in late stationary phase cultures of E. coli and Y. pseudotuberculosis. We show DsRed42 signal also accumulates during exposure to sources of nitric oxide (NO), suggesting DsRed42 signal detects growth-arrested bacterial cells. In a mouse model of Y. pseudotuberculosis deep tissue infection, DsRed42 signal was detected, and primarily accumulates in bacteria expressing markers of stationary phase growth. There was no significant overlap between DsRed42 signal and NO-exposed subpopulations of bacteria within host tissues, suggesting NO stress was transient, allowing bacteria to recover from this stress and resume replication. This novel DsRed42 variant represents a tool that will enable additional studies of slow-growing subpopulations of bacteria, specifically within bacterial species that quickly divide. We have generated a variant of TIMER that can be used to mark slow-growing subsets of Yersinia pseudotuberculosis, which has a relatively short division time, similar to E. coli. We used a combination of site-directed and random mutagenesis to generate DsRed42, which has red fluorescent signal accumulation in post-exponential or stationary phase cells. Since this variant accumulates only red fluorescence, it is no longer a TIMER protein, and is more appropriately termed DsRed42. We found that nitric oxide (NO) stress is sufficient to promote DsRed42 signal accumulation in culture, however within host tissues, DsRed42 signal correlates with a stationary phase reporter (dps). These results suggest NO may cause an immediate arrest in bacterial cell division, but during growth in host tissues exposure to NO is transient, allowing bacteria to recover from this stress and resume cell division. Thus instead of indicating a response to host stressors, DsRed42 signal accumulation within host tissues appears to identify slow-growing cells that are experiencing nutrient limitation.
Collapse
|
23
|
Rasheed S, Fries F, Müller R, Herrmann J. Zebrafish: An Attractive Model to Study Staphylococcus aureus Infection and Its Use as a Drug Discovery Tool. Pharmaceuticals (Basel) 2021; 14:594. [PMID: 34205723 PMCID: PMC8235121 DOI: 10.3390/ph14060594] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 11/16/2022] Open
Abstract
Non-mammalian in vivo disease models are particularly popular in early drug discovery. Zebrafish (Danio rerio) is an attractive vertebrate model, the success of which is driven by several advantages, such as the optical transparency of larvae, the small and completely sequenced genome, the small size of embryos and larvae enabling high-throughput screening, and low costs. In this review, we highlight zebrafish models of Staphyloccoccus aureus infection, which are used in drug discovery and for studying disease pathogenesis and virulence. Further, these infection models are discussed in the context of other relevant zebrafish models for pharmacological and toxicological studies as part of early drug profiling. In addition, we examine key differences to commonly applied models of S.aureus infection based on invertebrate organisms, and we compare their frequency of use in academic research covering the period of January 2011 to January 2021.
Collapse
Affiliation(s)
- Sari Rasheed
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, 66123 Saarbrücken, Germany; (S.R.); (F.F.); (R.M.)
- German Centre for Infection Research (DZIF), Partner Site Hannover–Braunschweig, 38124 Braunschweig, Germany
| | - Franziska Fries
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, 66123 Saarbrücken, Germany; (S.R.); (F.F.); (R.M.)
- German Centre for Infection Research (DZIF), Partner Site Hannover–Braunschweig, 38124 Braunschweig, Germany
- Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, 66123 Saarbrücken, Germany; (S.R.); (F.F.); (R.M.)
- German Centre for Infection Research (DZIF), Partner Site Hannover–Braunschweig, 38124 Braunschweig, Germany
- Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Jennifer Herrmann
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, 66123 Saarbrücken, Germany; (S.R.); (F.F.); (R.M.)
- German Centre for Infection Research (DZIF), Partner Site Hannover–Braunschweig, 38124 Braunschweig, Germany
| |
Collapse
|
24
|
Barbieri L, Sorzabal Bellido I, Beckett AJ, Prior IA, Fothergill J, Diaz Fernandez YA, Raval R. One-step preparation of antimicrobial silicone materials based on PDMS and salicylic acid: insights from spatially and temporally resolved techniques. NPJ Biofilms Microbiomes 2021; 7:51. [PMID: 34155220 PMCID: PMC8217222 DOI: 10.1038/s41522-021-00223-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 05/12/2021] [Indexed: 11/09/2022] Open
Abstract
In this work, we introduce a one-step strategy that is suitable for continuous flow manufacturing of antimicrobial PDMS materials. The process is based on the intrinsic capacity of PDMS to react to certain organic solvents, which enables the incorporation of antimicrobial actives such as salicylic acid (SA), which has been approved for use in humans within pharmaceutical products. By combining different spectroscopic and imaging techniques, we show that the surface properties of PDMS remain unaffected while high doses of the SA are loaded inside the PDMS matrix. The SA can be subsequently released under physiological conditions, delivering a strong antibacterial activity. Furthermore, encapsulation of SA inside the PDMS matrix ensured a diffusion-controlled release that was tracked by spatially resolved Raman spectroscopy, Attenuated Total Reflectance IR (ATR-IR), and UV-Vis spectroscopy. The biological activity of the new material was evaluated directly at the surface and in the planktonic state against model pathogenic bacteria, combining confocal laser scanning microscopy, electron microscopy, and cell viability assays. The results showed complete planktonic inhibition for clinically relevant strains of Staphylococcus aureus and Escherichia coli, and a reduction of up to 4 orders of magnitude for viable sessile cells, demonstrating the efficacy of these surfaces in preventing the initial stages of biofilm formation. Our approach adds a new option to existing strategies for the antimicrobial functionalisation of a wide range of products such as catheters, wound dressings and in-dwelling medical devices based on PDMS.
Collapse
Affiliation(s)
- Luca Barbieri
- Open Innovation Hub for Antimicrobial Surfaces, Department of Chemistry, University of Liverpool & National Biofilm Innovation Centre, Liverpool, UK
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - Ioritz Sorzabal Bellido
- Open Innovation Hub for Antimicrobial Surfaces, Department of Chemistry, University of Liverpool & National Biofilm Innovation Centre, Liverpool, UK
| | - Alison J Beckett
- Biomedical Electron Microscopy Unit, University of Liverpool, Liverpool, UK
| | - Ian A Prior
- Biomedical Electron Microscopy Unit, University of Liverpool, Liverpool, UK
| | - Jo Fothergill
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - Yuri A Diaz Fernandez
- Open Innovation Hub for Antimicrobial Surfaces, Department of Chemistry, University of Liverpool & National Biofilm Innovation Centre, Liverpool, UK.
| | - Rasmita Raval
- Open Innovation Hub for Antimicrobial Surfaces, Department of Chemistry, University of Liverpool & National Biofilm Innovation Centre, Liverpool, UK.
| |
Collapse
|
25
|
Staphylococcus aureus cell wall structure and dynamics during host-pathogen interaction. PLoS Pathog 2021; 17:e1009468. [PMID: 33788901 PMCID: PMC8041196 DOI: 10.1371/journal.ppat.1009468] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/12/2021] [Accepted: 03/12/2021] [Indexed: 01/09/2023] Open
Abstract
Peptidoglycan is the major structural component of the Staphylococcus aureus cell wall, in which it maintains cellular integrity, is the interface with the host, and its synthesis is targeted by some of the most crucial antibiotics developed. Despite this importance, and the wealth of data from in vitro studies, we do not understand the structure and dynamics of peptidoglycan during infection. In this study we have developed methods to harvest bacteria from an active infection in order to purify cell walls for biochemical analysis ex vivo. Isolated ex vivo bacterial cells are smaller than those actively growing in vitro, with thickened cell walls and reduced peptidoglycan crosslinking, similar to that of stationary phase cells. These features suggested a role for specific peptidoglycan homeostatic mechanisms in disease. As S. aureus missing penicillin binding protein 4 (PBP4) has reduced peptidoglycan crosslinking in vitro its role during infection was established. Loss of PBP4 resulted in an increased recovery of S. aureus from the livers of infected mice, which coincided with enhanced fitness within murine and human macrophages. Thicker cell walls correlate with reduced activity of peptidoglycan hydrolases. S. aureus has a family of 4 putative glucosaminidases, that are collectively crucial for growth. Loss of the major enzyme SagB, led to attenuation during murine infection and reduced survival in human macrophages. However, loss of the other three enzymes Atl, SagA and ScaH resulted in clustering dependent attenuation, in a zebrafish embryo, but not a murine, model of infection. A combination of pbp4 and sagB deficiencies resulted in a restoration of parental virulence. Our results, demonstrate the importance of appropriate cell wall structure and dynamics during pathogenesis, providing new insight to the mechanisms of disease. The prevalence of methicillin resistant Staphylococcus aureus (MRSA) in both hospitals and the wider community places a huge weight on healthcare providers. To discover new control regimes, it is therefore important to understand how the pathogen behaves within the relevant environment of the host. This is often hampered by the ability to obtain sufficient ex vivo pathogen samples for study. We have developed a method to isolate S. aureus from the infected host to be able to analyse cellular morphology and structure. S. aureus, isolated from an infected kidney abscess are smaller in size, with thicker cell walls than exponentially growing cells in vitro. Their cell wall peptidoglycan also is less crosslinked. These features suggested the role of components controlling cell wall homeostasis as being important for infections. We tested the role of PBP4, known to increase cell wall crosslinking and found a pbp4 mutant to have increased survival in macrophages and fitness within the murine host. Conversely the peptidoglycan hydrolase SagB, whose loss results in thinner cell walls was attenuated in the murine systemic model of infection, with concomitant loss of fitness within macrophages. Our study reveals an important adaptation to the host environment and the role of those components involved in cell wall homeostasis in vivo.
Collapse
|
26
|
Selection of Resistant Bacteria in Mallards Exposed to Subinhibitory Concentrations of Ciprofloxacin in Their Water Environment. Antimicrob Agents Chemother 2021; 65:AAC.01858-20. [PMID: 33318021 DOI: 10.1128/aac.01858-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/10/2020] [Indexed: 12/22/2022] Open
Abstract
Emergence and selection of antibiotic resistance following exposure to high antibiotic concentrations have been repeatedly shown in clinical and agricultural settings, whereas the role of the weak selective pressures exerted by antibiotic levels below the MIC (sub-MIC) in aquatic environments due to anthropogenic contamination remains unclear. Here, we studied how exposure to sub-MIC levels of ciprofloxacin enriches for Escherichia coli with reduced susceptibility to ciprofloxacin using a mallard colonization model. Mallards were inoculated with two isogenic extended-spectrum-β-lactamase (ESBL)-encoding E. coli strains, differing only by a gyrA mutation that results in increased MICs of ciprofloxacin, and exposed to different levels of ciprofloxacin in their swimming water. Changes in the ratios of mutant to parental strains excreted in feces over time and ESBL plasmid spread within the gut microbiota from individual birds were investigated. Results show that in vivo selection of gyrA mutants occurred in mallards during exposure to ciprofloxacin at concentrations previously found in aquatic environments. During colonization, resistance plasmids were readily transferred between strains in the intestines of the mallards, but conjugation frequencies were not affected by ciprofloxacin exposure. Our results highlight the potential for enrichment of resistant bacteria in wildlife and underline the importance of reducing antibiotic pollution in the environment.
Collapse
|
27
|
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: 180] [Impact Index Per Article: 45.0] [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
|
28
|
Mahmutovic A, Gillman AN, Lauksund S, Robson Moe NA, Manzi A, Storflor M, Abel Zur Wiesch P, Abel S. RESTAMP - Rate estimates by sequence-tag analysis of microbial populations. Comput Struct Biotechnol J 2021; 19:1035-1051. [PMID: 33613869 PMCID: PMC7878984 DOI: 10.1016/j.csbj.2021.01.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 12/31/2022] Open
Abstract
Microbial division rates determine the speed of mutation accumulation and thus the emergence of antimicrobial resistance. Microbial death rates are affected by antibiotic action and the immune system. Therefore, measuring these rates has advanced our understanding of host-pathogen interactions and antibiotic action. Several methods based on marker-loss or few inheritable neutral markers exist that allow estimating microbial division and death rates, each of which has advantages and limitations. Technical bottlenecks, i.e., experimental sampling events, during the experiment can distort the rate estimates and are typically unaccounted for or require additional calibration experiments. In this work, we introduce RESTAMP (Rate Estimates by Sequence Tag Analysis of Microbial Populations) as a method for determining bacterial division and death rates. This method uses hundreds of fitness neutral sequence barcodes to measure the rates and account for experimental bottlenecks at the same time. We experimentally validate RESTAMP and compare it to established plasmid loss methods. We find that RESTAMP has a number of advantages over plasmid loss or previous marker based techniques. (i) It enables to correct the distortion of rate estimates by technical bottlenecks. (ii) Rate estimates are independent of the sequence tag distribution in the starting culture allowing the use of an arbitrary number of tags. (iii) It introduces a bottleneck sensitivity measure that can be used to maximize the accuracy of the experiment. RESTAMP allows studying microbial population dynamics with great resolution over a wide dynamic range and can thus advance our understanding of host-pathogen interactions or the mechanisms of antibiotic action.
Collapse
Affiliation(s)
- Anel Mahmutovic
- Department of Pharmacy, Faculty of Health Sciences, UiT - The Arctic University of Norway, 9037 Tromsø, Norway
| | - Aaron Nicholas Gillman
- Department of Pharmacy, Faculty of Health Sciences, UiT - The Arctic University of Norway, 9037 Tromsø, Norway.,Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, PA 16802, USA
| | - Silje Lauksund
- Department of Pharmacy, Faculty of Health Sciences, UiT - The Arctic University of Norway, 9037 Tromsø, Norway
| | - Natasha-Anne Robson Moe
- Department of Pharmacy, Faculty of Health Sciences, UiT - The Arctic University of Norway, 9037 Tromsø, Norway
| | - Aime Manzi
- Department of Pharmacy, Faculty of Health Sciences, UiT - The Arctic University of Norway, 9037 Tromsø, Norway
| | - Merete Storflor
- Department of Pharmacy, Faculty of Health Sciences, UiT - The Arctic University of Norway, 9037 Tromsø, Norway.,Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, PA 16802, USA
| | - Pia Abel Zur Wiesch
- Department of Pharmacy, Faculty of Health Sciences, UiT - The Arctic University of Norway, 9037 Tromsø, Norway.,Centre for Molecular Medicine Norway, Nordic EMBL Partnership, 0318 Oslo, Norway.,Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA.,Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Sören Abel
- Department of Pharmacy, Faculty of Health Sciences, UiT - The Arctic University of Norway, 9037 Tromsø, Norway.,Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, PA 16802, USA.,Centre for Molecular Medicine Norway, Nordic EMBL Partnership, 0318 Oslo, Norway.,Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
29
|
Molina-Mora JA, García F. Molecular Determinants of Antibiotic Resistance in the Costa Rican Pseudomonas aeruginosa AG1 by a Multi-omics Approach: A Review of 10 Years of Study. PHENOMICS 2021; 1:129-142. [PMID: 35233560 PMCID: PMC8210740 DOI: 10.1007/s43657-021-00016-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 01/13/2023]
Abstract
Pseudomonas aeruginosa AG1 (PaeAG1) is a Costa Rican strain that was isolated in 2010 in a major Hospital. This strain has resistance to multiple antibiotics such as β-lactams (including carbapenems), aminoglycosides, and fluoroquinolones. PaeAG1 is considered critical (Priority 1) due to its resistance to carbapenems, and it was the first report of a P. aeruginosa isolate carrying both VIM-2 and IMP-18 genes encoding for metallo-β-lactamases (MBL) enzymes (both with carbapenemase activity). Owing to these traits, we have studied this model for 10 years using diverse approaches including multi-omics. In this review, we summarize the main points of the different steps that we have studied in PaeAG1: preliminary analyses of this strain at the genomic and phenomic levels revealed that this microorganism has particular features of antibiotic resistance. In the multi-omics approach, the genome assembly was the initial step to identify the genomic determinants of this strain, including virulence factors, antibiotic resistance genes, as well as a complex accessory genome. Second, a comparative genomic approach was implemented to define and update the phylogenetic relationship among complete P. aeruginosa genomes, the genomic island content in other strains, and the architecture of the two MBL-carrying integrons. Third, the proteomic profile of PaeAG1 was studied after exposure to antibiotics using 2-dimensional gel electrophoresis (2D-GE). Fourth, to study the central response to multiple perturbations in P. aeruginosa, i.e., the core perturbome, a machine learning approach was used. The analysis revealed biological functions and determinants that are shared by different disturbances. Finally, to evaluate the effects of ciprofloxacin (CIP) on PaeAG1, a growth curve comparison, differential expression analysis (RNA-Seq), and network analysis were performed. Using the results of the core perturbome (pathways that also were found in this perturbation with CIP), it was possible to identify the “exclusive” response and determinants of PaeAG1 after exposure to CIP. Altogether, after a decade of study using a multi-omics approach (at genomics, comparative genomics, perturbomics, transcriptomics, proteomics, and phenomics levels), we have provided new insights about the genomic and transcriptomic determinants associated with antibiotic resistance in PaeAG1. These results not only partially explain the high-risk condition of this strain that enables it to conquer nosocomial environments and its multi-resistance profile, but also this information may eventually be used as part of the strategies to fight this pathogen.
Collapse
Affiliation(s)
- Jose Arturo Molina-Mora
- Centro de Investigación en Enfermedades Tropicales (CIET) & Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Fernando García
- Centro de Investigación en Enfermedades Tropicales (CIET) & Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| |
Collapse
|
30
|
Muñoz-Sánchez S, van der Vaart M, Meijer AH. Autophagy and Lc3-Associated Phagocytosis in Zebrafish Models of Bacterial Infections. Cells 2020; 9:cells9112372. [PMID: 33138004 PMCID: PMC7694021 DOI: 10.3390/cells9112372] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/24/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023] Open
Abstract
Modeling human infectious diseases using the early life stages of zebrafish provides unprecedented opportunities for visualizing and studying the interaction between pathogens and phagocytic cells of the innate immune system. Intracellular pathogens use phagocytes or other host cells, like gut epithelial cells, as a replication niche. The intracellular growth of these pathogens can be counteracted by host defense mechanisms that rely on the autophagy machinery. In recent years, zebrafish embryo infection models have provided in vivo evidence for the significance of the autophagic defenses and these models are now being used to explore autophagy as a therapeutic target. In line with studies in mammalian models, research in zebrafish has shown that selective autophagy mediated by ubiquitin receptors, such as p62, is important for host resistance against several bacterial pathogens, including Shigella flexneri, Mycobacterium marinum, and Staphylococcus aureus. Furthermore, an autophagy related process, Lc3-associated phagocytosis (LAP), proved host beneficial in the case of Salmonella Typhimurium infection but host detrimental in the case of S. aureus infection, where LAP delivers the pathogen to a replication niche. These studies provide valuable information for developing novel therapeutic strategies aimed at directing the autophagy machinery towards bacterial degradation.
Collapse
|
31
|
Park YM, Meyer MR, Müller R, Herrmann J. Drug Administration Routes Impact the Metabolism of a Synthetic Cannabinoid in the Zebrafish Larvae Model. Molecules 2020; 25:E4474. [PMID: 33003405 PMCID: PMC7582563 DOI: 10.3390/molecules25194474] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 09/24/2020] [Accepted: 09/27/2020] [Indexed: 12/18/2022] Open
Abstract
Zebrafish (Danio rerio) larvae have gained attention as a valid model to study in vivo drug metabolism and to predict human metabolism. The microinjection of compounds, oligonucleotides, or pathogens into zebrafish embryos at an early developmental stage is a well-established technique. Here, we investigated the metabolism of zebrafish larvae after microinjection of methyl 2-(1-(5-fluoropentyl)-1H-pyrrolo[2,3-b]pyridine-3-carboxamido)-3,3-dimethylbutanoate (7'N-5F-ADB) as a representative of recently introduced synthetic cannabinoids. Results were compared to human urine data and data from the in vitro HepaRG model and the metabolic pathway of 7'N-5F-ADB were reconstructed. Out of 27 metabolites detected in human urine samples, 19 and 15 metabolites were present in zebrafish larvae and HepaRG cells, respectively. The route of administration to zebrafish larvae had a major impact and we found a high number of metabolites when 7'N-5F-ADB was microinjected into the caudal vein, heart ventricle, or hindbrain. We further studied the spatial distribution of the parent compound and its metabolites by mass spectrometry imaging (MSI) of treated zebrafish larvae to demonstrate the discrepancy in metabolite profiles among larvae exposed through different administration routes. In conclusion, zebrafish larvae represent a superb model for studying drug metabolism, and when combined with MSI, the optimal administration route can be determined based on in vivo drug distribution.
Collapse
Affiliation(s)
- Yu Mi Park
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy, Saarland University, Campus E8 1, 66123 Saarbrücken, Germany;
- Environmental Safety Group, Korea Institute of Science and Technology (KIST) Europe, 66123 Saarbrücken, Germany
| | - Markus R. Meyer
- Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, 66421 Homburg, Germany;
| | - Rolf Müller
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy, Saarland University, Campus E8 1, 66123 Saarbrücken, Germany;
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig Germany, 38124 Braunschweig, Germany
| | - Jennifer Herrmann
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy, Saarland University, Campus E8 1, 66123 Saarbrücken, Germany;
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig Germany, 38124 Braunschweig, Germany
| |
Collapse
|
32
|
Transcriptomic determinants of the response of ST-111 Pseudomonas aeruginosa AG1 to ciprofloxacin identified by a top-down systems biology approach. Sci Rep 2020; 10:13717. [PMID: 32792590 PMCID: PMC7427096 DOI: 10.1038/s41598-020-70581-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/25/2020] [Indexed: 12/13/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that thrives in diverse environments and causes a variety of human infections. Pseudomonas aeruginosa AG1 (PaeAG1) is a high-risk sequence type 111 (ST-111) strain isolated from a Costa Rican hospital in 2010. PaeAG1 has both blaVIM-2 and blaIMP-18 genes encoding for metallo-β-lactamases, and it is resistant to β-lactams (including carbapenems), aminoglycosides, and fluoroquinolones. Ciprofloxacin (CIP) is an antibiotic commonly used to treat P. aeruginosa infections, and it is known to produce DNA damage, triggering a complex molecular response. In order to evaluate the effects of a sub-inhibitory CIP concentration on PaeAG1, growth curves using increasing CIP concentrations were compared. We then measured gene expression using RNA-Seq at three time points (0, 2.5 and 5 h) after CIP exposure to identify the transcriptomic determinants of the response (i.e. hub genes, gene clusters and enriched pathways). Changes in expression were determined using differential expression analysis and network analysis using a top–down systems biology approach. A hybrid model using database-based and co-expression analysis approaches was implemented to predict gene–gene interactions. We observed a reduction of the growth curve rate as the sub-inhibitory CIP concentrations were increased. In the transcriptomic analysis, we detected that over time CIP treatment resulted in the differential expression of 518 genes, showing a complex impact at the molecular level. The transcriptomic determinants were 14 hub genes, multiple gene clusters at different levels (associated to hub genes or as co-expression modules) and 15 enriched pathways. Down-regulation of genes implicated in several metabolism pathways, virulence elements and ribosomal activity was observed. In contrast, amino acid catabolism, RpoS factor, proteases, and phenazines genes were up-regulated. Remarkably, > 80 resident-phage genes were up-regulated after CIP treatment, which was validated at phenomic level using a phage plaque assay. Thus, reduction of the growth curve rate and increasing phage induction was evidenced as the CIP concentrations were increased. In summary, transcriptomic and network analyses, as well as the growth curves and phage plaque assays provide evidence that PaeAG1 presents a complex, concentration-dependent response to sub-inhibitory CIP exposure, showing pleiotropic effects at the systems level. Manipulation of these determinants, such as phage genes, could be used to gain more insights about the regulation of responses in PaeAG1 as well as the identification of possible therapeutic targets. To our knowledge, this is the first report of the transcriptomic analysis of CIP response in a ST-111 high-risk P. aeruginosa strain, in particular using a top-down systems biology approach.
Collapse
|
33
|
Panchal VV, Griffiths C, Mosaei H, Bilyk B, Sutton JAF, Carnell OT, Hornby DP, Green J, Hobbs JK, Kelley WL, Zenkin N, Foster SJ. Evolving MRSA: High-level β-lactam resistance in Staphylococcus aureus is associated with RNA Polymerase alterations and fine tuning of gene expression. PLoS Pathog 2020; 16:e1008672. [PMID: 32706832 PMCID: PMC7380596 DOI: 10.1371/journal.ppat.1008672] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 06/02/2020] [Indexed: 12/21/2022] Open
Abstract
Most clinical MRSA (methicillin-resistant S. aureus) isolates exhibit low-level β-lactam resistance (oxacillin MIC 2-4 μg/ml) due to the acquisition of a novel penicillin binding protein (PBP2A), encoded by mecA. However, strains can evolve high-level resistance (oxacillin MIC ≥256 μg/ml) by an unknown mechanism. Here we have developed a robust system to explore the basis of the evolution of high-level resistance by inserting mecA into the chromosome of the methicillin-sensitive S. aureus SH1000. Low-level mecA-dependent oxacillin resistance was associated with increased expression of anaerobic respiratory and fermentative genes. High-level resistant derivatives had acquired mutations in either rpoB (RNA polymerase subunit β) or rpoC (RNA polymerase subunit β') and these mutations were shown to be responsible for the observed resistance phenotype. Analysis of rpoB and rpoC mutants revealed decreased growth rates in the absence of antibiotic, and alterations to, transcription elongation. The rpoB and rpoC mutations resulted in decreased expression to parental levels, of anaerobic respiratory and fermentative genes and specific upregulation of 11 genes including mecA. There was however no direct correlation between resistance and the amount of PBP2A. A mutational analysis of the differentially expressed genes revealed that a member of the S. aureus Type VII secretion system is required for high level resistance. Interestingly, the genomes of two of the high level resistant evolved strains also contained missense mutations in this same locus. Finally, the set of genetically matched strains revealed that high level antibiotic resistance does not incur a significant fitness cost during pathogenesis. Our analysis demonstrates the complex interplay between antibiotic resistance mechanisms and core cell physiology, providing new insight into how such important resistance properties evolve.
Collapse
Affiliation(s)
- Viralkumar V. Panchal
- Department of Molecular Biology and Biotechnology, University of Sheffield, Western Bank, Sheffield, United Kingdom
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, United Kingdom
| | - Caitlin Griffiths
- Centre for Bacterial Cell Biology, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Hamed Mosaei
- Centre for Bacterial Cell Biology, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Bohdan Bilyk
- Department of Molecular Biology and Biotechnology, University of Sheffield, Western Bank, Sheffield, United Kingdom
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, United Kingdom
| | - Joshua A. F. Sutton
- Department of Molecular Biology and Biotechnology, University of Sheffield, Western Bank, Sheffield, United Kingdom
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, United Kingdom
| | - Oliver T. Carnell
- Department of Molecular Biology and Biotechnology, University of Sheffield, Western Bank, Sheffield, United Kingdom
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, United Kingdom
| | - David P. Hornby
- Department of Molecular Biology and Biotechnology, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | - Jeffrey Green
- Department of Molecular Biology and Biotechnology, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | - Jamie K. Hobbs
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, United Kingdom
- Department of Physics and Astronomy, University of Sheffield, Sheffield, United Kingdom
| | - William L. Kelley
- Department of Microbiology and Molecular Medicine, University Hospital and Medical School of Geneva, Geneva, Switzerland
| | - Nikolay Zenkin
- Centre for Bacterial Cell Biology, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Simon J. Foster
- Department of Molecular Biology and Biotechnology, University of Sheffield, Western Bank, Sheffield, United Kingdom
- The Florey Institute for Host-Pathogen Interactions, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
34
|
Candida albicans Genetic Background Influences Mean and Heterogeneity of Drug Responses and Genome Stability during Evolution in Fluconazole. mSphere 2020; 5:5/3/e00480-20. [PMID: 32581072 PMCID: PMC7316494 DOI: 10.1128/msphere.00480-20] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Antimicrobial resistance is an evolutionary phenomenon with clinical implications. We tested how replicates from diverse strains of Candida albicans, a prevalent human fungal pathogen, evolve in the commonly prescribed antifungal drug fluconazole. Replicates on average increased in fitness in the level of drug they were evolved to, with the least fit parental strains improving the most. Very few replicates increased resistance above the drug level they were evolved in. Notably, many replicates increased in genome size and changed in drug tolerance (a drug response where a subpopulation of cells grow slowly in high levels of drug), and variability among replicates in fitness, tolerance, and genome size was higher in strains that initially were more sensitive to the drug. Genetic background influenced the average degree of adaptation and the evolved variability of many phenotypes, highlighting that different strains from the same species may respond and adapt very differently during adaptation. The importance of within-species diversity in determining the evolutionary potential of a population to evolve drug resistance or tolerance is not well understood, including in eukaryotic pathogens. To examine the influence of genetic background, we evolved replicates of 20 different clinical isolates of Candida albicans, a human fungal pathogen, in fluconazole, the commonly used antifungal drug. The isolates hailed from the major C. albicans clades and had different initial levels of drug resistance and tolerance to the drug. The majority of replicates rapidly increased in fitness in the evolutionary environment, with the degree of improvement inversely correlated with parental strain fitness in the drug. Improvement was largely restricted to up to the evolutionary level of drug: only 4% of the evolved replicates increased resistance (MIC) above the evolutionary level of drug. Prevalent changes were altered levels of drug tolerance (slow growth of a subpopulation of cells at drug concentrations above the MIC) and increased diversity of genome size. The prevalence and predominant direction of these changes differed in a strain-specific manner, but neither correlated directly with parental fitness or improvement in fitness. Rather, low parental strain fitness was correlated with high levels of heterogeneity in fitness, tolerance, and genome size among evolved replicates. Thus, parental strain background is an important determinant in mean improvement to the evolutionary environment as well as the diversity of evolved phenotypes, and the range of possible responses of a pathogen to an antimicrobial drug cannot be captured by in-depth study of a single strain background. IMPORTANCE Antimicrobial resistance is an evolutionary phenomenon with clinical implications. We tested how replicates from diverse strains of Candida albicans, a prevalent human fungal pathogen, evolve in the commonly prescribed antifungal drug fluconazole. Replicates on average increased in fitness in the level of drug they were evolved to, with the least fit parental strains improving the most. Very few replicates increased resistance above the drug level they were evolved in. Notably, many replicates increased in genome size and changed in drug tolerance (a drug response where a subpopulation of cells grow slowly in high levels of drug), and variability among replicates in fitness, tolerance, and genome size was higher in strains that initially were more sensitive to the drug. Genetic background influenced the average degree of adaptation and the evolved variability of many phenotypes, highlighting that different strains from the same species may respond and adapt very differently during adaptation.
Collapse
|
35
|
Vlazaki M, Huber J, Restif O. Integrating mathematical models with experimental data to investigate the within-host dynamics of bacterial infections. Pathog Dis 2020; 77:5704399. [PMID: 31942996 PMCID: PMC6986552 DOI: 10.1093/femspd/ftaa001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/13/2020] [Indexed: 12/23/2022] Open
Abstract
Bacterial infections still constitute a major cause of mortality and morbidity worldwide. The unavailability of therapeutics, antimicrobial resistance and the chronicity of infections due to incomplete clearance contribute to this phenomenon. Despite the progress in antimicrobial and vaccine development, knowledge about the effect that therapeutics have on the host–bacteria interactions remains incomplete. Insights into the characteristics of bacterial colonization and migration between tissues and the relationship between replication and host- or therapeutically induced killing can enable efficient design of treatment approaches. Recently, innovative experimental techniques have generated data enabling the qualitative characterization of aspects of bacterial dynamics. Here, we argue that mathematical modeling as an adjunct to experimental data can enrich the biological insight that these data provide. However, due to limited interdisciplinary training, efforts to combine the two remain limited. To promote this dialogue, we provide a categorization of modeling approaches highlighting their relationship to data generated by a range of experimental techniques in the area of in vivo bacterial dynamics. We outline common biological themes explored using mathematical models with case studies across all pathogen classes. Finally, this review advocates multidisciplinary integration to improve our mechanistic understanding of bacterial infections and guide the use of existing or new therapies.
Collapse
Affiliation(s)
- Myrto Vlazaki
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, CB3 0ES, Cambridge, UK
| | - John Huber
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, CB3 0ES, Cambridge, UK
| | - Olivier Restif
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, CB3 0ES, Cambridge, UK
| |
Collapse
|
36
|
Linnerz T, Hall CJ. The Diverse Roles of Phagocytes During Bacterial and Fungal Infections and Sterile Inflammation: Lessons From Zebrafish. Front Immunol 2020; 11:1094. [PMID: 32582182 PMCID: PMC7289964 DOI: 10.3389/fimmu.2020.01094] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 05/06/2020] [Indexed: 12/23/2022] Open
Abstract
The immediate and natural reaction to both infectious challenges and sterile insults (wounds, tissue trauma or crystal deposition) is an acute inflammatory response. This inflammatory response is mediated by activation of the innate immune system largely comprising professional phagocytes (neutrophils and macrophages). Zebrafish (danio rerio) larvae possess many advantages as a model organism, including their genetic tractability and highly conserved innate immune system. Exploiting these attributes and the live imaging potential of optically transparent zebrafish larvae has greatly contributed to our understanding of how neutrophils and macrophages orchestrate the initiation and resolution phases of inflammatory responses. Numerous bacterial and fungal infection models have been successfully established using zebrafish as an animal model and studies investigating neutrophil and macrophage behavior to sterile insults have also provided unique insights. In this review we highlight how examining the larval zebrafish response to specific bacterial and fungal pathogens has uncovered cellular and molecular mechanisms behind a variety of phagocyte responses, from those that protect the host to those that are detrimental. We also describe how modeling sterile inflammation in larval zebrafish has provided an opportunity to dissect signaling pathways that control the recruitment, and fate, of phagocytes at inflammatory sites. Finally, we briefly discuss some current limitations, and opportunities to improve, the zebrafish model system for studying phagocyte biology.
Collapse
Affiliation(s)
- Tanja Linnerz
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Christopher J Hall
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
37
|
Hou X, Zhang X, Zhao W, Zeng C, Deng B, McComb DW, Du S, Zhang C, Li W, Dong Y. Vitamin lipid nanoparticles enable adoptive macrophage transfer for the treatment of multidrug-resistant bacterial sepsis. NATURE NANOTECHNOLOGY 2020; 15:41-46. [PMID: 31907443 PMCID: PMC7181370 DOI: 10.1038/s41565-019-0600-1] [Citation(s) in RCA: 173] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 11/20/2019] [Indexed: 05/05/2023]
Abstract
Sepsis, a condition caused by severe infections, affects more than 30 million people worldwide every year and remains the leading cause of death in hospitals1,2. Moreover, antimicrobial resistance has become an additional challenge in the treatment of sepsis3, and thus, alternative therapeutic approaches are urgently needed2,3. Here, we show that adoptive transfer of macrophages containing antimicrobial peptides linked to cathepsin B in the lysosomes (MACs) can be applied for the treatment of multidrug-resistant bacteria-induced sepsis in mice with immunosuppression. The MACs are constructed by transfection of vitamin C lipid nanoparticles that deliver antimicrobial peptide and cathepsin B (AMP-CatB) mRNA. The vitamin C lipid nanoparticles allow the specific accumulation of AMP-CatB in macrophage lysosomes, which is the key location for bactericidal activities. Our results demonstrate that adoptive MAC transfer leads to the elimination of multidrug-resistant bacteria, including Staphylococcus aureus and Escherichia coli, leading to the complete recovery of immunocompromised septic mice. Our work provides an alternative strategy for overcoming multidrug-resistant bacteria-induced sepsis and opens up possibilities for the development of nanoparticle-enabled cell therapy for infectious diseases.
Collapse
Affiliation(s)
- Xucheng Hou
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Xinfu Zhang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, China
| | - Weiyu Zhao
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Chunxi Zeng
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Binbin Deng
- Center for Electron Microscopy and Analysis, The Ohio State University, Columbus, OH, USA
| | - David W McComb
- Center for Electron Microscopy and Analysis, The Ohio State University, Columbus, OH, USA
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, USA
| | - Shi Du
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Chengxiang Zhang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Wenqing Li
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA.
- Department of Biomedical Engineering, Center for Clinical and Translational Science, Comprehensive Cancer Center, Dorothy M. Davis Heart & Lung Research Institute, Department of Radiation Oncology, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
38
|
Epinecidin-1 Protects against Methicillin Resistant Staphylococcus aureus Infection and Sepsis in Pyemia Pigs. Mar Drugs 2019; 17:md17120693. [PMID: 31835381 PMCID: PMC6950563 DOI: 10.3390/md17120693] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/03/2019] [Accepted: 12/05/2019] [Indexed: 12/16/2022] Open
Abstract
Methicillin resistant Staphylococcus aureus (MRSA) may be found on the skin, nose, and throats of long-term hospitalized patients. While MRSA infections are usually minor, serious infections and death may occur in immunocompromised or diabetic patients, or after exposure of MRSA to blood. This report demonstrates that the antimicrobial peptide (AMP) epinecidin-1 (Epi-1) efficiently protects against MRSA infection in a pyemia pig model. We first found that Epi-1 exhibits bactericidal activity against MRSA. Next, pharmacokinetic analysis revealed that Epi-1 was stable in serum for 4 h after injection, followed by a gradual decrease. This pharmacokinetic profile suggested Epi-1 may bind serum albumin, which was confirmed in vitro. Harmful effects were not observed for doses up to 100 mg/kg body weight in pigs. When Epi-1 was supplied as a curative agent 30 min post-infection, MRSA-induced abnormalities in blood uric acid (UA), blood urea nitrogen (BUN), creatine (CRE), GOT, and GPT levels were restored to normal levels. We further showed that the bactericidal activity of Epi-1 was higher than that of the antibiotic drug vancomycin. Epi-1 significantly decreased MRSA counts in the blood, liver, kidney, heart, and lungs of infected pigs. Elevated levels of serum C reactive protein (CRP), proinflammatory cytokine IL6, IL1β, and TNFα were also attenuated by Epi-1 treatment. Moreover, the MRSA genes, enterotoxin (et)-A, et-B, intrinsic methicillin resistance A (mecA), and methicillin resistance factor A (femA), were significantly reduced or abolished in MRSA-infected pigs after treatment with Epi-1. Hematoxylin and eosin staining of heart, liver, lung, and kidney sections indicated that Epi-1 attenuated MRSA toxicity in infected pigs. A survival study showed that the pyemia pigs infected with MRSA alone died within a week, whereas the pigs post-treated with 2.5 mg/kg Epi-1 were completely protected against death. The present investigation, thus, demonstrates that Epi-1 effectively protects pyemia pigs against pathogenic MRSA without major toxic side effects.
Collapse
|
39
|
Bacigalupe R, Tormo-Mas MÁ, Penadés JR, Fitzgerald JR. A multihost bacterial pathogen overcomes continuous population bottlenecks to adapt to new host species. SCIENCE ADVANCES 2019; 5:eaax0063. [PMID: 31807698 PMCID: PMC6881152 DOI: 10.1126/sciadv.aax0063] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 10/01/2019] [Indexed: 06/10/2023]
Abstract
While many bacterial pathogens are restricted to single host species, some have the capacity to undergo host switches, leading to the emergence of new clones that are a threat to human and animal health. However, the bacterial traits that underpin a multihost ecology are not well understood. Following transmission to a new host, bacterial populations are influenced by powerful forces such as genetic drift that reduce the fixation rate of beneficial mutations, limiting the capacity for host adaptation. Here, we implement a novel experimental model of bacterial host switching to investigate the ability of the multihost pathogen Staphylococcus aureus to adapt to new species under continuous population bottlenecks. We demonstrate that beneficial mutations accumulated during infection can overcome genetic drift and sweep through the population, leading to host adaptation. Our findings highlight the remarkable capacity of some bacteria to adapt to distinct host niches in the face of powerful antagonistic population forces.
Collapse
Affiliation(s)
- Rodrigo Bacigalupe
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh EH25 9RG, UK
| | - María Ángeles Tormo-Mas
- Centro de Investigación y Tecnología Animal, Instituto Valenciano de Investigaciones Agrarias, Segorbe 12400, Spain
- Severe Infection Group of Instituto de Investigación Sanitaria La Fe, 106 Avenida Fernando Abril Martorell, Valencia 46026, Spain
| | - José R. Penadés
- Departamento de Ciencias Biomédicas, Universidad CEU Cardenal Herrera, Moncada 46113, Spain
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK
| | - J. Ross Fitzgerald
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh EH25 9RG, UK
| |
Collapse
|
40
|
The Case for Modeling Human Infection in Zebrafish. Trends Microbiol 2019; 28:10-18. [PMID: 31604611 DOI: 10.1016/j.tim.2019.08.005] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/19/2019] [Accepted: 08/16/2019] [Indexed: 12/25/2022]
Abstract
Zebrafish (Danio rerio) larvae are widely recognized for studying host-pathogen interactions in vivo because of their optical transparency, genetic manipulability, and translational potential. The development of the zebrafish immune system is well understood, thereby use of larvae enables investigation solely in the context of innate immunity. As a result, infection of zebrafish with natural fish pathogens including Mycobacterium marinum has significantly advanced our understanding of bacterial pathogenesis and vertebrate host defense. However, new work using a variety of human pathogens (bacterial, viral, and fungal) has illuminated the versatility of the zebrafish infection model, revealing unexpected and important concepts underlying infectious disease. We propose that this knowledge can inform studies in higher animal models and help to develop treatments to combat human infection.
Collapse
|
41
|
Adaptation Through Lifestyle Switching Sculpts the Fitness Landscape of Evolving Populations: Implications for the Selection of Drug-Resistant Bacteria at Low Drug Pressures. Genetics 2019; 211:1029-1044. [PMID: 30670539 DOI: 10.1534/genetics.119.301834] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 01/16/2019] [Indexed: 12/21/2022] Open
Abstract
Novel genotypes evolve under selection through mutations in pre-existing genes. However, mutations have pleiotropic phenotypic effects that influence the fitness of emerging genotypes in complex ways. The evolution of antimicrobial resistance is mediated by selection of mutations in genes coding for antibiotic-target proteins. Drug-resistance is commonly associated with a fitness cost due to the impact of resistance-conferring mutations on protein function and/or stability. These costs are expected to prohibit the selection of drug-resistant mutations at low drug pressures. Using laboratory evolution of rifampicin resistance in Escherichia coli, we show that when exposed intermittently to low concentration (0.1 × minimal inhibitory concentration) of rifampicin, the evolution of canonical drug resistance was indeed unfavorable. Instead, these bacterial populations adapted by evolving into small-colony variants that displayed enhanced pellicle-forming ability. This shift in lifestyle from planktonic to pellicle-like was necessary for enhanced fitness at low drug pressures, and was mediated by the genetic activation of the fim operon promoter, which allowed expression of type I fimbriae. Upon continued low drug exposure, these bacteria evolved exclusively into high-level drug-resistant strains through mutations at a limited set of loci within the rifampicin-resistance determining region of the rpoB gene. We show that our results are explained by mutation-specific epistasis, resulting in differential impact of lifestyle switching on the competitive fitness of different rpoB mutations. Thus, lifestyle-alterations that are selected at low selection pressures have the potential to modify the fitness effects of mutations, change the genetic structure, and affect the ultimate fate of evolving populations.
Collapse
|
42
|
Buchan KD, Foster SJ, Renshaw SA. Staphylococcus aureus: setting its sights on the human innate immune system. MICROBIOLOGY-SGM 2019; 165:367-385. [PMID: 30625113 DOI: 10.1099/mic.0.000759] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Staphylococcus aureus has colonized humans for at least 10 000 years, and today inhabits roughly a third of the population. In addition, S. aureus is a major pathogen that is responsible for a significant disease burden, ranging in severity from mild skin and soft-tissue infections to life-threatening endocarditis and necrotizing pneumonia, with treatment often hampered by resistance to commonly available antibiotics. Underpinning its versatility as a pathogen is its ability to evade the innate immune system. S. aureus specifically targets innate immunity to establish and sustain infection, utilizing a large repertoire of virulence factors to do so. Using these factors, S. aureus can resist phagosomal killing, impair complement activity, disrupt cytokine signalling and target phagocytes directly using proteolytic enzymes and cytolytic toxins. Although most of these virulence factors are well characterized, their importance during infection is less clear, as many display species-specific activity against humans or against animal hosts, including cows, horses and chickens. Several staphylococcal virulence factors display species specificity for components of the human innate immune system, with as few as two amino acid changes reducing binding affinity by as much as 100-fold. This represents a major issue for studying their roles during infection, which cannot be examined without the use of humanized infection models. This review summarizes the major factors S. aureus uses to impair the innate immune system, and provides an in-depth look into the host specificity of S. aureus and how this problem is being approached.
Collapse
Affiliation(s)
- Kyle D Buchan
- 1The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Simon J Foster
- 2Department of Molecular Biology and Biotechnology, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Stephen A Renshaw
- 1The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| |
Collapse
|
43
|
Guérillot R, Li L, Baines S, Howden B, Schultz MB, Seemann T, Monk I, Pidot SJ, Gao W, Giulieri S, Gonçalves da Silva A, D’Agata A, Tomita T, Peleg AY, Stinear TP, Howden BP. Comprehensive antibiotic-linked mutation assessment by resistance mutation sequencing (RM-seq). Genome Med 2018; 10:63. [PMID: 30165908 PMCID: PMC6117896 DOI: 10.1186/s13073-018-0572-z] [Citation(s) in RCA: 23] [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: 04/14/2018] [Accepted: 07/24/2018] [Indexed: 12/15/2022] Open
Abstract
Mutation acquisition is a major mechanism of bacterial antibiotic resistance that remains insufficiently characterised. Here we present RM-seq, a new amplicon-based deep sequencing workflow based on a molecular barcoding technique adapted from Low Error Amplicon sequencing (LEA-seq). RM-seq allows detection and functional assessment of mutational resistance at high throughput from mixed bacterial populations. The sensitive detection of very low-frequency resistant sub-populations permits characterisation of antibiotic-linked mutational repertoires in vitro and detection of rare resistant populations during infections. Accurate quantification of resistance mutations enables phenotypic screening of mutations conferring pleiotropic phenotypes such as in vivo persistence, collateral sensitivity or cross-resistance. RM-seq will facilitate comprehensive detection, characterisation and surveillance of resistant bacterial populations ( https://github.com/rguerillot/RM-seq ).
Collapse
Affiliation(s)
- Romain Guérillot
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection & Immunity, Melbourne, Victoria Australia
| | - Lucy Li
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection & Immunity, Melbourne, Victoria Australia
| | - Sarah Baines
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection & Immunity, Melbourne, Victoria Australia
| | - Brian Howden
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection & Immunity, Melbourne, Victoria Australia
| | - Mark B. Schultz
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection & Immunity, Melbourne, Victoria Australia
- Doherty Applied Microbial Genomics, The University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, Victoria Australia
- Microbiological Diagnostic Unit Public Health Laboratory, The University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, Victoria Australia
| | - Torsten Seemann
- Doherty Applied Microbial Genomics, The University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, Victoria Australia
- Melbourne Bioinformatics, The University of Melbourne, Melbourne, Victoria Australia
| | - Ian Monk
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection & Immunity, Melbourne, Victoria Australia
| | - Sacha J. Pidot
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection & Immunity, Melbourne, Victoria Australia
| | - Wei Gao
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection & Immunity, Melbourne, Victoria Australia
| | - Stefano Giulieri
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection & Immunity, Melbourne, Victoria Australia
| | - Anders Gonçalves da Silva
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection & Immunity, Melbourne, Victoria Australia
- Doherty Applied Microbial Genomics, The University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, Victoria Australia
- Microbiological Diagnostic Unit Public Health Laboratory, The University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, Victoria Australia
| | - Anthony D’Agata
- Microbiological Diagnostic Unit Public Health Laboratory, The University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, Victoria Australia
| | - Takehiro Tomita
- Microbiological Diagnostic Unit Public Health Laboratory, The University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, Victoria Australia
| | - Anton Y. Peleg
- Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria Australia
- Infection and Immunity Theme, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria Australia
| | - Timothy P. Stinear
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection & Immunity, Melbourne, Victoria Australia
- Doherty Applied Microbial Genomics, The University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, Victoria Australia
| | - Benjamin P. Howden
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection & Immunity, Melbourne, Victoria Australia
- Doherty Applied Microbial Genomics, The University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, Victoria Australia
- Microbiological Diagnostic Unit Public Health Laboratory, The University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, Victoria Australia
- Infectious Diseases Department, Austin Health, Heidelberg, Victoria Australia
| |
Collapse
|
44
|
Giulieri SG, Baines SL, Guerillot R, Seemann T, Gonçalves da Silva A, Schultz M, Massey RC, Holmes NE, Stinear TP, Howden BP. Genomic exploration of sequential clinical isolates reveals a distinctive molecular signature of persistent Staphylococcus aureus bacteraemia. Genome Med 2018; 10:65. [PMID: 30103826 PMCID: PMC6090636 DOI: 10.1186/s13073-018-0574-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/27/2018] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Large-scale genomic studies of within-host diversity in Staphylococcus aureus bacteraemia (SAB) are needed to understanding bacterial adaptation underlying persistence and thus refining the role of genomics in management of SAB. However, available comparative genomic studies of sequential SAB isolates have tended to focus on selected cases of unusually prolonged bacteraemia, where secondary antimicrobial resistance has developed. METHODS To understand bacterial genetic diversity during SAB more broadly, we applied whole genome sequencing to a large collection of sequential isolates obtained from patients with persistent or relapsing bacteraemia. After excluding genetically unrelated isolates, we performed an in-depth genomic analysis of point mutations and chromosome structural variants arising within individual SAB episodes. RESULTS We show that, while adaptation pathways are heterogenous and episode-specific, isolates from persistent bacteraemia have a distinctive molecular signature, characterised by a low mutation frequency and high proportion of non-silent mutations. Analysis of structural genomic variants revealed that these often overlooked genetic events are commonly acquired during SAB. We discovered that IS256 insertion may represent the most effective driver of within-host microevolution in selected lineages, with up to three new insertion events per isolate even in the absence of other mutations. Genetic mechanisms resulting in significant phenotypic changes, such as increases in vancomycin resistance, development of small colony phenotypes, and decreases in cytotoxicity, included mutations in key genes (rpoB, stp, agrA) and an IS256 insertion upstream of the walKR operon. CONCLUSIONS This study provides for the first time a large-scale analysis of within-host genomic changes during invasive S. aureus infection and describes specific patterns of adaptation that will be informative for both understanding S. aureus pathoadaptation and utilising genomics for management of complicated S. aureus infections.
Collapse
Affiliation(s)
- Stefano G Giulieri
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection & Immunity, Melbourne, Australia.,Infectious Disease Department, Austin Health, Melbourne, Australia.,Infectious Diseases Service, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Sarah L Baines
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection & Immunity, Melbourne, Australia
| | - Romain Guerillot
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection & Immunity, Melbourne, Australia
| | - Torsten Seemann
- Microbiological Diagnostic Unit Public Health Laboratory, The University of Melbourne at The Doherty Institute of Infection and Immunity, Melbourne, Australia.,Melbourne Bioinformatics, The University of Melbourne, Melbourne, Australia
| | - Anders Gonçalves da Silva
- Microbiological Diagnostic Unit Public Health Laboratory, The University of Melbourne at The Doherty Institute of Infection and Immunity, Melbourne, Australia
| | - Mark Schultz
- Microbiological Diagnostic Unit Public Health Laboratory, The University of Melbourne at The Doherty Institute of Infection and Immunity, Melbourne, Australia
| | - Ruth C Massey
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Natasha E Holmes
- Infectious Disease Department, Austin Health, Melbourne, Australia
| | - Timothy P Stinear
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection & Immunity, Melbourne, Australia
| | - Benjamin P Howden
- Department of Microbiology and Immunology, The University of Melbourne at the Doherty Institute for Infection & Immunity, Melbourne, Australia. .,Infectious Disease Department, Austin Health, Melbourne, Australia. .,Microbiological Diagnostic Unit Public Health Laboratory, The University of Melbourne at The Doherty Institute of Infection and Immunity, Melbourne, Australia.
| |
Collapse
|
45
|
Human skin commensals augment Staphylococcus aureus pathogenesis. Nat Microbiol 2018; 3:881-890. [PMID: 30013237 PMCID: PMC6207346 DOI: 10.1038/s41564-018-0198-3] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 06/15/2018] [Indexed: 02/07/2023]
Abstract
All bacterial infections occur within a polymicrobial environment, from which a pathogen population emerges to establish disease within a host. Emphasis has been placed on prevention of pathogen dominance by competing microflora acting as probiotics1. Here we show that virulence of the human pathogen, Staphylococcus aureus is augmented by native, polymicrobial, commensal skin flora and individual species acting as “proinfectious agents”. The outcome is pathogen proliferation but not commensal. Pathogenesis augmentation can be mediated by particulate cell wall peptidoglycan (PGN), reducing the S. aureus infectious dose by over 1000-fold. This phenomenon occurs using a range of S. aureus strains, infection models and is not mediated by established receptor-mediated pathways including Nod1, Nod2, Myd88 and the NLPR3 inflammasome. During mouse sepsis, augmentation depends on liver resident macrophages (Kupffer cells, KC), that capture and internalise both pathogen and ‘proinfectious agent’, leading to reduced production of reactive oxygen species, pathogen survival and subsequent multiple liver abscess formation. The augmented infection model more closely resembles the natural situation and establishes the role of resident environmental microflora in initiation of disease by an invading pathogen. As human microflora is ubiquitous2 its role in increasing susceptibility to infection S. aureus highlights potential strategies for disease prevention.
Collapse
|
46
|
Garner E, Chen C, Xia K, Bowers J, Engelthaler DM, McLain J, Edwards MA, Pruden A. Metagenomic Characterization of Antibiotic Resistance Genes in Full-Scale Reclaimed Water Distribution Systems and Corresponding Potable Systems. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2018; 52:6113-6125. [PMID: 29741366 DOI: 10.1021/acs.est.7b05419] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Water reclamation provides a valuable resource for meeting nonpotable water demands. However, little is known about the potential for wastewater reuse to disseminate antibiotic resistance genes (ARGs). Here, samples were collected seasonally in 2014-2015 from four U.S. utilities' reclaimed and potable water distribution systems before treatment, after treatment, and at five points of use (POU). Shotgun metagenomic sequencing was used to profile the resistome (i.e., full contingent of ARGs) of a subset ( n = 38) of samples. Four ARGs ( qnrA, blaTEM, vanA, sul1) were quantified by quantitative polymerase chain reaction. Bacterial community composition (via 16S rRNA gene amplicon sequencing), horizontal gene transfer (via quantification of intI1 integrase and plasmid genes), and selection pressure (via detection of metals and antibiotics) were investigated as potential factors governing the presence of ARGs. Certain ARGs were elevated in all ( sul1; p ≤ 0.0011) or some ( blaTEM, qnrA; p ≤ 0.0145) reclaimed POU samples compared to corresponding potable samples. Bacterial community composition was weakly correlated with ARGs (Adonis, R2 = 0.1424-0.1734) and associations were noted between 193 ARGs and plasmid-associated genes. This study establishes that reclaimed water could convey greater abundances of certain ARGs than potable waters and provides observations regarding factors that likely control ARG occurrence in reclaimed water systems.
Collapse
Affiliation(s)
- Emily Garner
- Via Department of Civil and Environmental Engineering , Virginia Tech , Blacksburg , Virginia 24061 , United States
| | - Chaoqi Chen
- Department of Crop and Soil Environmental Sciences , Virginia Tech , Blacksburg , Virginia 24061 , United States
| | - Kang Xia
- Department of Crop and Soil Environmental Sciences , Virginia Tech , Blacksburg , Virginia 24061 , United States
| | - Jolene Bowers
- Translational Genomics Research Institute , Flagstaff , Arizona 86005 , United States
| | - David M Engelthaler
- Translational Genomics Research Institute , Flagstaff , Arizona 86005 , United States
| | - Jean McLain
- Water Resources Research Center , University of Arizona , Tucson , Arizona 85719 , United States
| | - Marc A Edwards
- Via Department of Civil and Environmental Engineering , Virginia Tech , Blacksburg , Virginia 24061 , United States
| | - Amy Pruden
- Via Department of Civil and Environmental Engineering , Virginia Tech , Blacksburg , Virginia 24061 , United States
| |
Collapse
|
47
|
Pollitt EJG, Szkuta PT, Burns N, Foster SJ. Staphylococcus aureus infection dynamics. PLoS Pathog 2018; 14:e1007112. [PMID: 29902272 PMCID: PMC6019756 DOI: 10.1371/journal.ppat.1007112] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 06/26/2018] [Accepted: 05/21/2018] [Indexed: 01/22/2023] Open
Abstract
Staphylococcus aureus is a human commensal that can also cause systemic infections. This transition requires evasion of the immune response and the ability to exploit different niches within the host. However, the disease mechanisms and the dominant immune mediators against infection are poorly understood. Previously it has been shown that the infecting S. aureus population goes through a population bottleneck, from which very few bacteria escape to establish the abscesses that are characteristic of many infections. Here we examine the host factors underlying the population bottleneck and subsequent clonal expansion in S. aureus infection models, to identify underpinning principles of infection. The bottleneck is a common feature between models and is independent of S. aureus strain. Interestingly, the high doses of S. aureus required for the widely used "survival" model results in a reduced population bottleneck, suggesting that host defences have been simply overloaded. This brings into question the applicability of the survival model. Depletion of immune mediators revealed key breakpoints and the dynamics of systemic infection. Loss of macrophages, including the liver Kupffer cells, led to increased sensitivity to infection as expected but also loss of the population bottleneck and the spread to other organs still occurred. Conversely, neutrophil depletion led to greater susceptibility to disease but with a concomitant maintenance of the bottleneck and lack of systemic spread. We also used a novel microscopy approach to examine abscess architecture and distribution within organs. From these observations we developed a conceptual model for S. aureus disease from initial infection to mature abscess. This work highlights the need to understand the complexities of the infectious process to be able to assign functions for host and bacterial components, and why S. aureus disease requires a seemingly high infectious dose and how interventions such as a vaccine may be more rationally developed.
Collapse
Affiliation(s)
- Eric J. G. Pollitt
- Department of Molecular Biology and Biotechnology, Firth Court, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | - Piotr T. Szkuta
- Department of Molecular Biology and Biotechnology, Firth Court, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | - Nicola Burns
- Department of Molecular Biology and Biotechnology, Firth Court, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | - Simon J. Foster
- Department of Molecular Biology and Biotechnology, Firth Court, University of Sheffield, Western Bank, Sheffield, United Kingdom
| |
Collapse
|
48
|
Kumar S, Saini V, Maurya IK, Sindhu J, Kumari M, Kataria R, Kumar V. Design, synthesis, DFT, docking studies and ADME prediction of some new coumarinyl linked pyrazolylthiazoles: Potential standalone or adjuvant antimicrobial agents. PLoS One 2018; 13:e0196016. [PMID: 29672633 PMCID: PMC5908142 DOI: 10.1371/journal.pone.0196016] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 04/04/2018] [Indexed: 12/26/2022] Open
Abstract
The control of antimicrobial resistance (AMR) seems to have come to a dead end. The major consequences of the use and abuse of antibacterial drugs are the development of resistant strains due to genetic mutability of both pathogenic and nonpathogenic microorganisms. We, herein, report the synthesis, characterization and biological activities of coumarin-thiazole-pyrazole (CTP) molecular hybrids with an effort to explore and overcome the increasing antimicrobial resistance. The compounds were characterized by analyzing their IR, Mass, 1H and13C NMR spectral data and elemental analysis. The in vitro antimicrobial activity of the synthesized compounds was investigated against various pathogenic strains; the results obtained were further explained with the help of DFT and molecular orbital calculations. Compound 1b and 1f displayed good antimicrobial activity and synergistic effects when used with kanamycin and amphotericin B. Furthermore, in vitro cytotoxicity of compounds 1b and 1f were studied against HeLa cells (cervical cancer cell) and Hek-293 cells. The results of molecular docking study were used to better rationalize the action and prediction of the binding modes of these compounds.
Collapse
Affiliation(s)
- Sunil Kumar
- Department of Chemistry & Centre of Advance Studies in Chemistry, Panjab University, Chandigarhs, India
| | - Vikram Saini
- Department of Biotechnology, AIIMS- New Delhi, Delhi, India
| | - Indresh K. Maurya
- Department of Microbial Biotechnology, Panjab University, Chandigarh, India
| | | | - Mukesh Kumari
- Department of Chemistry, Kurukshetra University, Kurukshetra, India
| | - Ramesh Kataria
- Department of Chemistry & Centre of Advance Studies in Chemistry, Panjab University, Chandigarhs, India
- * E-mail: (RK); (VK)
| | - Vinod Kumar
- Department of Chemistry, M. M. University, Mullana-Ambala, India
- * E-mail: (RK); (VK)
| |
Collapse
|
49
|
Torraca V, Mostowy S. Zebrafish Infection: From Pathogenesis to Cell Biology. Trends Cell Biol 2018; 28:143-156. [PMID: 29173800 PMCID: PMC5777827 DOI: 10.1016/j.tcb.2017.10.002] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 10/06/2017] [Accepted: 10/09/2017] [Indexed: 12/11/2022]
Abstract
The study of host-pathogen interactions has illuminated fundamental research avenues in both infection and cell biology. Zebrafish (Danio rerio) larvae are genetically tractable, optically accessible, and present a fully functional innate immune system with macrophages and neutrophils that mimic their mammalian counterparts. A wide variety of pathogenic bacteria have been investigated using zebrafish models, providing unprecedented resolution of the cellular response to infection in vivo. In this review, we illustrate how zebrafish models have contributed to our understanding of cellular microbiology by providing an in vivo platform to study host-pathogen interactions from the single cell to whole animal level. We also highlight discoveries made from zebrafish infection that hold great promise for translation into novel therapies for humans.
Collapse
Affiliation(s)
- Vincenzo Torraca
- Section of Microbiology, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Serge Mostowy
- Section of Microbiology, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK.
| |
Collapse
|
50
|
McVicker G, Prajsnar TK, Foster SJ. Construction and Use of Staphylococcus aureus Strains to Study Within-Host Infection Dynamics. Methods Mol Biol 2018; 1736:17-27. [PMID: 29322455 DOI: 10.1007/978-1-4939-7638-6_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The study of the dynamics that occur during the course of a bacterial infection has been attempted using several methods. Here we discuss the construction of a set of antibiotic-resistant, otherwise-isogenic Staphylococcus aureus strains that can be used to observe the progress of systemic disease in a mouse model at various time-points postinfection. The strains can likewise be used to study the progression of infection in other animal infection models, such as the zebrafish embryo. Furthermore, the use of antibiotic resistance tags provides a convenient system with which to investigate the effect of antimicrobial chemotherapy during disease.
Collapse
Affiliation(s)
- Gareth McVicker
- Krebs Institute, University of Sheffield, Sheffield, UK
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, UK
- Bateson Centre, University of Sheffield, Sheffield, UK
| | - Tomasz K Prajsnar
- Krebs Institute, University of Sheffield, Sheffield, UK
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, UK
- Bateson Centre, University of Sheffield, Sheffield, UK
| | - Simon J Foster
- Krebs Institute, University of Sheffield, Sheffield, UK.
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, UK.
- Bateson Centre, University of Sheffield, Sheffield, UK.
| |
Collapse
|