301
|
Role of CgTpo4 in Polyamine and Antimicrobial Peptide Resistance: Determining Virulence in Candida glabrata. Int J Mol Sci 2021; 22:ijms22031376. [PMID: 33573089 PMCID: PMC7866538 DOI: 10.3390/ijms22031376] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/18/2021] [Accepted: 01/23/2021] [Indexed: 01/28/2023] Open
Abstract
Candida glabrata is an emerging fungal pathogen whose success depends on its ability to resist antifungal drugs but also to thrive against host defenses. In this study, the predicted multidrug transporter CgTpo4 (encoded by ORF CAGL0L10912g) is described as a new determinant of virulence in C. glabrata, using the infection model Galleria mellonella. The CgTPO4 gene was found to be required for the C. glabrata ability to kill G. mellonella. The transporter encoded by this gene is also necessary for antimicrobial peptide (AMP) resistance, specifically against histatin-5. Interestingly, G. mellonella’s AMP expression was found to be strongly activated in response to C. glabrata infection, suggesting AMPs are a key antifungal defense. CgTpo4 was also found to be a plasma membrane exporter of polyamines, especially spermidine, suggesting that CgTpo4 is able to export polyamines and AMPs, thus conferring resistance to both stress agents. Altogether, this study presents the polyamine exporter CgTpo4 as a determinant of C. glabrata virulence, which acts by protecting the yeast cells from the overexpression of AMPs, deployed as a host defense mechanism.
Collapse
|
302
|
Burkholderia pseudomallei OMVs derived from infection mimicking conditions elicit similar protection to a live-attenuated vaccine. NPJ Vaccines 2021; 6:18. [PMID: 33514749 PMCID: PMC7846723 DOI: 10.1038/s41541-021-00281-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 01/07/2021] [Indexed: 02/07/2023] Open
Abstract
Burkholderia pseudomallei is a Gram-negative, facultative intracellular bacillus that causes the disease melioidosis. B. pseudomallei expresses a number of proteins that contribute to its intracellular survival in the mammalian host. We previously demonstrated that immunization with OMVs derived from B. pseudomallei grown in nutrient-rich media protects mice against lethal disease. Here, we evaluated if OMVs derived from B. pseudomallei grown under macrophage-mimicking growth conditions could be enriched with intracellular-stage proteins in order to improve the vaccine. We show that OMVs produced in this manner (M9 OMVs) contain proteins associated with intracellular survival yet are non-toxic to living cells. Immunization of mice provides significant protection against pulmonary infection similar to that achieved with a live attenuated vaccine and is associated with increased IgG, CD4+, and CD8+ T cells. OMVs possess inherent adjuvanticity and drive DC activation and maturation. These results indicate that M9 OMVs constitute a new promising vaccine against melioidosis.
Collapse
|
303
|
Hesketh-Best PJ, Mouritzen MV, Shandley-Edwards K, Billington RA, Upton M. Galleria mellonella larvae exhibit a weight-dependent lethal median dose when infected with methicillin-resistant Staphylococcus aureus. Pathog Dis 2021; 79:6121426. [PMID: 33503238 DOI: 10.1093/femspd/ftab003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
Galleria mellonella is a recognised model to study antimicrobial efficacy; however, standardisation across the scientific field and investigations of methodological components are needed. Here, we investigate the impact of weight on mortality following infection with Methicillin-resistant Staphylococcus aureus (MRSA). Larvae were separated into six weight groups (180-300 mg at 20 mg intervals) and infected with a range of doses of MRSA to determine the 50% lethal dose (LD50), and the 'lipid weight' of larvae post-infection was quantified. A model of LD50 values correlated with weight was developed. The LD50 values, as estimated by our model, were further tested in vivo to prove our model. We establish a weight-dependent LD50 in larvae against MRSA and demonstrate that G. mellonella is a stable model within 180-260 mg. We present multiple linear models correlating weight with: LD50, lipid weight, and larval length. We demonstrate that the lipid weight is reduced as a result of MRSA infection, identifying a potentially new measure in which to understand the immune response. Finally, we demonstrate that larval length can be a reasonable proxy for weight. Refining the methodologies in which to handle and design experiments involving G. mellonella, we can improve the reliability of this powerful model.
Collapse
Affiliation(s)
- Poppy J Hesketh-Best
- School of Biological and Marine Sciences, University of Plymouth, Drake Circus, Plymouth, PL4 8AA, UK
| | - Michelle V Mouritzen
- School of Biomedical Sciences, University of Plymouth, Derriford Research Facility, Plymouth Science Park, Plymouth, PL6 8BT, UK
| | - Kayleigh Shandley-Edwards
- School of Biological and Marine Sciences, University of Plymouth, Drake Circus, Plymouth, PL4 8AA, UK
| | - Richard A Billington
- School of Biological and Marine Sciences, University of Plymouth, Drake Circus, Plymouth, PL4 8AA, UK
| | - Mathew Upton
- School of Biomedical Sciences, University of Plymouth, Derriford Research Facility, Plymouth Science Park, Plymouth, PL6 8BT, UK
| |
Collapse
|
304
|
Frei A, King AP, Lowe GJ, Cain AK, Short FL, Dinh H, Elliott AG, Zuegg J, Wilson JJ, Blaskovich MAT. Nontoxic Cobalt(III) Schiff Base Complexes with Broad-Spectrum Antifungal Activity. Chemistry 2021; 27:2021-2029. [PMID: 33231906 PMCID: PMC7855930 DOI: 10.1002/chem.202003545] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/03/2020] [Indexed: 12/21/2022]
Abstract
Resistance to currently available antifungal drugs has quietly been on the rise but overshadowed by the alarming spread of antibacterial resistance. There is a striking lack of attention to the threat of drug-resistant fungal infections, with only a handful of new drugs currently in development. Given that metal complexes have proven to be useful new chemotypes in the fight against diseases such as cancer, malaria, and bacterial infections, it is reasonable to explore their possible utility in treating fungal infections. Herein we report a series of cobalt(III) Schiff base complexes with broad-spectrum antifungal activity. Some of these complexes show minimum inhibitory concentrations (MIC) in the low micro- to nanomolar range against a series of Candida and Cryptococcus yeasts. Additionally, we demonstrate that these compounds show no cytotoxicity against both bacterial and human cells. Finally, we report the first in vivo toxicity data on these compounds in Galleria mellonella, showing that doses as high as 266 mg kg-1 are tolerated without adverse effects, paving the way for further in vivo studies of these complexes.
Collapse
Affiliation(s)
- Angelo Frei
- Centre for Superbug SolutionsInstitute for Molecular BioscienceThe University of QueenslandSt. LuciaQLD4072Australia
| | - A. Paden King
- Department of Chemistry and Chemical BiologyCornell UniversityIthacaNY14853USA
| | - Gabrielle J. Lowe
- Centre for Superbug SolutionsInstitute for Molecular BioscienceThe University of QueenslandSt. LuciaQLD4072Australia
| | - Amy K. Cain
- Department of Molecular SciencesMacquarie UniversitySydneyNSW2109Australia
| | - Francesca L. Short
- Department of Molecular SciencesMacquarie UniversitySydneyNSW2109Australia
| | - Hue Dinh
- Department of Molecular SciencesMacquarie UniversitySydneyNSW2109Australia
- Department of Biological SciencesMacquarie UniversitySydneyNSW2109Australia
| | - Alysha G. Elliott
- Centre for Superbug SolutionsInstitute for Molecular BioscienceThe University of QueenslandSt. LuciaQLD4072Australia
| | - Johannes Zuegg
- Centre for Superbug SolutionsInstitute for Molecular BioscienceThe University of QueenslandSt. LuciaQLD4072Australia
| | - Justin J. Wilson
- Department of Chemistry and Chemical BiologyCornell UniversityIthacaNY14853USA
| | - Mark A. T. Blaskovich
- Centre for Superbug SolutionsInstitute for Molecular BioscienceThe University of QueenslandSt. LuciaQLD4072Australia
| |
Collapse
|
305
|
Javed M, Jentzsch B, Heinrich M, Ueltzhoeffer V, Peter S, Schoppmeier U, Angelov A, Schwarz S, Willmann M. Transcriptomic Basis of Serum Resistance and Virulence Related Traits in XDR P. aeruginosa Evolved Under Antibiotic Pressure in a Morbidostat Device. Front Microbiol 2021; 11:619542. [PMID: 33569046 PMCID: PMC7868568 DOI: 10.3389/fmicb.2020.619542] [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: 10/20/2020] [Accepted: 12/30/2020] [Indexed: 01/11/2023] Open
Abstract
Colistin is a last resort antibiotic against the critical status pathogen Pseudomonas aeruginosa. Virulence and related traits such as biofilm formation and serum resistance after exposure to sub-inhibitory levels of colistin have been underexplored. We cultivated P. aeruginosa in a semi-automated morbidostat device with colistin, metronidazole and a combination of the two antibiotics for 21 days, and completed RNA-Seq to uncover the transcriptional changes over time. Strains became resistant to colistin within this time period. Colistin-resistant strains show significantly increased biofilm formation: the cell density in biofilm increases under exposure to colistin, while the addition of metronidazole can remove this effect. After 7 days of colistin exposure, strains develop an ability to grow in serum, suggesting that colistin drives bacterial modifications conferring a protective effect from serum complement factors. Of note, strains exposed to colistin showed a decrease in virulence, when measured using the Galleria mellonella infection model. These phenotypic changes were characterized by a series of differential gene expression changes, particularly those related to LPS modifications, spermidine synthesis (via speH and speE) and the major stress response regulator rpoS. Our results suggest a clinically important bacterial evolution under sub-lethal antibiotic concentration leading to potential for significant changes in the clinical course of infection.
Collapse
Affiliation(s)
- Mumina Javed
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Institute of Medical Microbiology and Hygiene, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | - Benedikt Jentzsch
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Institute of Medical Microbiology and Hygiene, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | - Maximilian Heinrich
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Institute of Medical Microbiology and Hygiene, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | - Viola Ueltzhoeffer
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Institute of Medical Microbiology and Hygiene, Tübingen, Germany
| | - Silke Peter
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Institute of Medical Microbiology and Hygiene, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | - Ulrich Schoppmeier
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Institute of Medical Microbiology and Hygiene, Tübingen, Germany
| | - Angel Angelov
- NGS Competence Center Tübingen (NCCT), Tübingen, Germany
| | - Sandra Schwarz
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Institute of Medical Microbiology and Hygiene, Tübingen, Germany
| | - Matthias Willmann
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Institute of Medical Microbiology and Hygiene, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.,Eurofins MVZ Medizinisches Labor Gelsenkirchen, Gelsenkirchen, Germany
| |
Collapse
|
306
|
Cieślik M, Bagińska N, Górski A, Jończyk-Matysiak E. Animal Models in the Evaluation of the Effectiveness of Phage Therapy for Infections Caused by Gram-Negative Bacteria from the ESKAPE Group and the Reliability of Its Use in Humans. Microorganisms 2021; 9:206. [PMID: 33498243 PMCID: PMC7909267 DOI: 10.3390/microorganisms9020206] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/12/2021] [Accepted: 01/16/2021] [Indexed: 12/12/2022] Open
Abstract
The authors emphasize how extremely important it is to highlight the role played by animal models in an attempt to determine possible phage interactions with the organism into which it was introduced as well as to determine the safety and effectiveness of phage therapy in vivo taking into account the individual conditions of a given organism and its physiology. Animal models in which phages are used make it possible, among other things, to evaluate the effective therapeutic dose and to choose the possible route of phage administration depending on the type of infection developed. These results cannot be applied in detail to the human body, but the knowledge gained from animal experiments is invaluable and very helpful. We would like to highlight how useful animal models may be for the possible effectiveness evaluation of phage therapy in the case of infections caused by gram-negative bacteria from the ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacter species) group of pathogens. In this review, we focus specifically on the data from the last few years.
Collapse
Affiliation(s)
- Martyna Cieślik
- Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.C.); (N.B.); (A.G.)
| | - Natalia Bagińska
- Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.C.); (N.B.); (A.G.)
| | - Andrzej Górski
- Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.C.); (N.B.); (A.G.)
- Phage Therapy Unit, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| | - Ewa Jończyk-Matysiak
- Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.C.); (N.B.); (A.G.)
| |
Collapse
|
307
|
Paulson AR, O’Callaghan M, Zhang XX, Rainey PB, Hurst MRH. In vivo transcriptome analysis provides insights into host-dependent expression of virulence factors by Yersinia entomophaga MH96, during infection of Galleria mellonella. G3 (BETHESDA, MD.) 2021; 11:jkaa024. [PMID: 33561230 PMCID: PMC7849909 DOI: 10.1093/g3journal/jkaa024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/19/2020] [Indexed: 12/31/2022]
Abstract
The function of microbes can be inferred from knowledge of genes specifically expressed in natural environments. Here, we report the in vivo transcriptome of the entomopathogenic bacterium Yersinia entomophaga MH96, captured during initial, septicemic, and pre-cadaveric stages of intrahemocoelic infection in Galleria mellonella. A total of 1285 genes were significantly upregulated by MH96 during infection; 829 genes responded to in vivo conditions during at least one stage of infection, 289 responded during two stages of infection, and 167 transcripts responded throughout all three stages of infection compared to in vitro conditions at equivalent cell densities. Genes upregulated during the earliest infection stage included components of the insecticidal toxin complex Yen-TC (chi1, chi2, and yenC1), genes for rearrangement hotspot element containing protein yenC3, cytolethal distending toxin cdtAB, and vegetative insecticidal toxin vip2. Genes more highly expressed throughout the infection cycle included the putative heat-stable enterotoxin yenT and three adhesins (usher-chaperone fimbria, filamentous hemagglutinin, and an AidA-like secreted adhesin). Clustering and functional enrichment of gene expression data also revealed expression of genes encoding type III and VI secretion system-associated effectors. Together these data provide insight into the pathobiology of MH96 and serve as an important resource supporting efforts to identify novel insecticidal agents.
Collapse
Affiliation(s)
- Amber R Paulson
- Forage Science, AgResearch Ltd., Lincoln 8140, New Zealand
- New Zealand Institute for Advanced Study, Massey University, Auckland 0745, New Zealand
- Department of Biology, Queen’s University, Kingston, ON K7L 3N6, Canada
| | | | - Xue-Xian Zhang
- School of Natural and Computational Sciences, Massey University, Auckland 0745, New Zealand
| | - Paul B Rainey
- New Zealand Institute for Advanced Study, Massey University, Auckland 0745, New Zealand
- Laboratoire de Génétique de l’Evolution CBI, ESPCI Paris, Université PSL, CNRS, Paris 75005, France
- Department of Microbial Population Biology, Max Planck Institute for Evolutionary Biology, Plön 24306, Germany
| | - Mark R H Hurst
- Forage Science, AgResearch Ltd., Lincoln 8140, New Zealand
| |
Collapse
|
308
|
Li C, Hou S, Ma X, Li J, Huo L, Zhang P, Hao X, Zhu X. Epigenetic regulation of virulence and the transcription of ribosomal protein genes involves a YEATS family protein in Cryptococcus deneoformans. FEMS Yeast Res 2021; 21:6095727. [PMID: 33440003 DOI: 10.1093/femsyr/foab001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 01/11/2021] [Indexed: 01/18/2023] Open
Abstract
Epigenetic marks or post-translational modifications on histones have important regulatory roles in gene expression in eukaryotic organisms. The epigenetic regulation of gene expression in the pathogenic yeast Cryptococcus deneoformans remains largely undetermined. The YEATS domain proteins are readers of crotonylated lysine residues in histones. Here, we reported the identification of a single-copy gene putatively coding for a YEATS domain protein (Yst1) in C. deneoformans. To define its function, we created a mutant strain, yst1Δ, using CRISPR-Cas9 editing. yst1Δ exhibited defects in phenotype, for instance, it was hypersensitive to osmotic stress in the presence of 1.3 M NaCl or KCl. Furthermore, it was hypersensitive to 1% Congo red, suggesting defects in the cell wall. Interestingly, RNA-seq data revealed that Yst1p was critical for the expression of genes encoding the ribosomal proteins, that is, most were expressed with significantly lower levels of mRNA in yst1Δ than in the wild-type strain. The mutant strain was hypersensitive to low temperature and anti-ribosomal drugs, which we putatively attribute to the impairment in ribosomal function. In addition, the yst1Δ strain was less virulent to Galleria mellonella. These results generally suggest that Yst1, as a histone modification reader, might be a key coordinator of the transcriptome of this human pathogen. Yst1 could be a potential target for novel antifungal drugs, which might lead to significant developments in the clinical treatment of cryptococcosis.
Collapse
Affiliation(s)
- Chenxi Li
- Beijing Key Laboratory of Genetic Engineering Drug and Biotechnology, Institute of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University (CLS-BNU), Beijing 100875, PR China
| | - Shaonan Hou
- Beijing Key Laboratory of Genetic Engineering Drug and Biotechnology, Institute of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University (CLS-BNU), Beijing 100875, PR China
| | - Xiaoyu Ma
- Beijing Key Laboratory of Genetic Engineering Drug and Biotechnology, Institute of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University (CLS-BNU), Beijing 100875, PR China
| | - Jiajia Li
- Beijing Key Laboratory of Genetic Engineering Drug and Biotechnology, Institute of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University (CLS-BNU), Beijing 100875, PR China
| | - Liang Huo
- Beijing Key Laboratory of Genetic Engineering Drug and Biotechnology, Institute of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University (CLS-BNU), Beijing 100875, PR China
| | - Ping Zhang
- Beijing Key Laboratory of Genetic Engineering Drug and Biotechnology, Institute of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University (CLS-BNU), Beijing 100875, PR China
| | - Xiaoran Hao
- Beijing Key Laboratory of Genetic Engineering Drug and Biotechnology, Institute of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University (CLS-BNU), Beijing 100875, PR China
| | - Xudong Zhu
- Beijing Key Laboratory of Genetic Engineering Drug and Biotechnology, Institute of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University (CLS-BNU), Beijing 100875, PR China
| |
Collapse
|
309
|
Transcription Inhibitors with XRE DNA-Binding and Cupin Signal-Sensing Domains Drive Metabolic Diversification in Pseudomonas. mSystems 2021; 6:6/1/e00753-20. [PMID: 33436508 PMCID: PMC7901475 DOI: 10.1128/msystems.00753-20] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Bacteria of the Pseudomonas genus, including the major human pathogen Pseudomonas aeruginosa, are known for their complex regulatory networks and high number of transcription factors, which contribute to their impressive adaptive ability. However, even in the most studied species, most of the regulators are still uncharacterized. Transcription factors (TFs) are instrumental in the bacterial response to new environmental conditions. They can act as direct signal sensors and subsequently induce changes in gene expression leading to physiological adaptation. Here, by combining transcriptome sequencing (RNA-seq) and cistrome determination (DAP-seq), we studied a family of eight TFs in Pseudomonas aeruginosa. This family, encompassing TFs with XRE-like DNA-binding and cupin signal-sensing domains, includes the metabolic regulators ErfA, PsdR, and PauR and five so-far-unstudied TFs. The genome-wide delineation of their regulons identified 39 regulatory interactions with genes mostly involved in metabolism. We found that the XRE-cupin TFs are inhibitors of their neighboring genes, forming local, functional units encoding proteins with functions in condition-specific metabolic pathways. Growth phenotypes of isogenic mutants highlighted new roles for PauR and PA0535 in polyamines and arginine metabolism. The phylogenetic analysis of this family of regulators across the bacterial kingdom revealed a wide diversity of such metabolic regulatory modules and identified species with potentially higher metabolic versatility. Numerous genes encoding uncharacterized XRE-cupin TFs were found near metabolism-related genes, illustrating the need of further systematic characterization of transcriptional regulatory networks in order to better understand the mechanisms of bacterial adaptation to new environments. IMPORTANCE Bacteria of the Pseudomonas genus, including the major human pathogen Pseudomonas aeruginosa, are known for their complex regulatory networks and high number of transcription factors, which contribute to their impressive adaptive ability. However, even in the most studied species, most of the regulators are still uncharacterized. With the recent advances in high-throughput sequencing methods, it is now possible to fill this knowledge gap and help the understanding of how bacteria adapt and thrive in new environments. By leveraging these methods, we provide an example of a comprehensive analysis of an entire family of transcription factors and bring new insights into metabolic and regulatory adaptation in the Pseudomonas genus.
Collapse
|
310
|
Luiz de Freitas L, Pereira da Silva F, Fernandes KM, Carneiro DG, Licursi de Oliveira L, Martins GF, Dantas Vanetti MC. The virulence of Salmonella Enteritidis in Galleria mellonella is improved by N-dodecanoyl-homoserine lactone. Microb Pathog 2021; 152:104730. [PMID: 33444697 DOI: 10.1016/j.micpath.2021.104730] [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: 08/07/2020] [Revised: 01/01/2021] [Accepted: 01/03/2021] [Indexed: 01/18/2023]
Abstract
Salmonella is a food and waterborne pathogen responsible for outbreaks worldwide, and it can survive during passage through the stomach and inside host phagocytic cells. Virulence genes are required for infection and survival in macrophages, and some are under the regulation of the quorum sensing (QS) system. This study investigated the influence of the autoinducer 1 (AI-1), N-dodecanoyl-homoserine lactone (C12-HSL), on the virulence of Salmonella PT4 using Galleria mellonella as an infection model. Salmonella PT4 was grown in the presence and absence of C12-HSL under anaerobic conditions for 7 h, and the expression of rpoS, arcA, arcB, and invA genes was evaluated. After the inoculation of G. mellonella with the median lethal dose (LD50) of Salmonella PT4, the survival of bacteria inside the larvae and their health status (health index scoring) were monitored, as well as the pigment, nitric oxide (NO), superoxide dismutase (SOD), and catalase (CAT) production. Also, the hemocyte viability, the induction of caspase-3, and microtubule-associated light chain 3 (LC3) protein in hemocytes were evaluated. Salmonella PT4 growing in the presence of C12-HSL showed increased rpoS, arcA, arcB, and invA expression and promoted higher larvae mortality and worse state of health after 24 h of infection. The C12-HSL also increased the persistence of Salmonella PT4 in the hemolymph and in the hemocytes. The highest pigmentation, NO production, and antioxidant enzymes were verified in the larva hemolymph infected with Salmonella PT4 grown with C12-HSL. Hemocytes from larvae infected with Salmonella PT4 grown with C12-HSL showed lower viability and higher production of caspase-3 and LC3. Taken together, these findings suggest that C12-HSL could be involved in the virulence of Salmonella PT4.
Collapse
Affiliation(s)
- Leonardo Luiz de Freitas
- Departmento de Microbiologia, Universidade Federal de Viçosa (UFV), Viçosa, Minas Gerais, Brazil
| | | | - Kenner Morais Fernandes
- Departamento de Biologia Geral, Universidade Federal de Viçosa (UFV), Viçosa, Minas Gerais, Brazil
| | - Deisy Guimarães Carneiro
- Departmento de Microbiologia, Universidade Federal de Viçosa (UFV), Viçosa, Minas Gerais, Brazil
| | | | - Gustavo Ferreira Martins
- Departamento de Biologia Geral, Universidade Federal de Viçosa (UFV), Viçosa, Minas Gerais, Brazil
| | | |
Collapse
|
311
|
Bao P, Li C, Ou H, Ji S, Chen Y, Gao J, Yue X, Shen J, Ding D. A peptide-based aggregation-induced emission bioprobe for selective detection and photodynamic killing of Gram-negative bacteria. Biomater Sci 2021; 9:437-442. [PMID: 33146160 DOI: 10.1039/d0bm01330g] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A bioprobe with aggregation-induced emission characteristics is developed, which can visualize and photodynamically ablate Gram-negative bacteria in a selective manner.
Collapse
Affiliation(s)
- Pingping Bao
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction
- Tianjin Stomatological Hospital
- The Affiliated Stomatological Hospital of Nankai University
- Tianjin 300041
- China
| | - Cong Li
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction
- Tianjin Stomatological Hospital
- The Affiliated Stomatological Hospital of Nankai University
- Tianjin 300041
- China
| | - Hanlin Ou
- Key Laboratory of Bioactive Materials
- Ministry of Education
- and College of Life Sciences
- Nankai University
- Tianjin 300071
| | - Shenglu Ji
- Key Laboratory of Bioactive Materials
- Ministry of Education
- and College of Life Sciences
- Nankai University
- Tianjin 300071
| | - Yao Chen
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction
- Tianjin Stomatological Hospital
- The Affiliated Stomatological Hospital of Nankai University
- Tianjin 300041
- China
| | - Jing Gao
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction
- Tianjin Stomatological Hospital
- The Affiliated Stomatological Hospital of Nankai University
- Tianjin 300041
- China
| | - Xin Yue
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction
- Tianjin Stomatological Hospital
- The Affiliated Stomatological Hospital of Nankai University
- Tianjin 300041
- China
| | - Jing Shen
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction
- Tianjin Stomatological Hospital
- The Affiliated Stomatological Hospital of Nankai University
- Tianjin 300041
- China
| | - Dan Ding
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction
- Tianjin Stomatological Hospital
- The Affiliated Stomatological Hospital of Nankai University
- Tianjin 300041
- China
| |
Collapse
|
312
|
Zhang Y, Fan J, Ye J, Lu L. The fungal-specific histone acetyltransferase Rtt109 regulates development, DNA damage response, and virulence in Aspergillus fumigatus. Mol Microbiol 2020; 115:1191-1206. [PMID: 33300219 DOI: 10.1111/mmi.14665] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/03/2020] [Accepted: 12/03/2020] [Indexed: 12/16/2022]
Abstract
In eukaryotes, histone acetylation catalyzed by histone acetyltransferase (HAT) has been demonstrated to be critical for various physiological processes. However, the biological functions of HAT and the underlying mechanism by which HAT-regulated processes are involved in fungal development and virulence in the human opportunistic pathogen Aspergillus fumigatus remain largely unexplored. Here, we functionally characterized the roles of Rtt109 in A. fumigatus, an ortholog of Saccharomyces cerevisiae histone acetyltransferase Rtt109. In vivo and in vitro HAT assays revealed that AfRtt109 functions as a canonical histone acetyltransferase, acetylating lysines 9 and 56 of histone H3. Deletion of Afrtt109 leads to severe defects in vegetative growth, conidiation, and causes reduced virulence in the Galleria mellonella model, as well as hypersensitivity to genotoxic agents. Moreover, site-directed mutagenesis revealed that the conserved arginine residues R265 and R306 of Rtt109 are required for the H3K9 and H3K56 acetylation and virulence of A. fumigatus. Unexpectedly, R265E and R306E mutants did not exhibit any detectable phenotypic defects, implying that A. fumigatus Rtt109 regulates fungal development via histone acetylation-independent mechanisms. Together, our results revealed the critical role of fungal-specific HAT Rtt109 in regulating fungal development and virulence, and suggested that it may serve as a unique target for antifungal therapies.
Collapse
Affiliation(s)
- Yuanwei Zhang
- Jiangsu Key Laboratory for Microbes and Functional Genomics, Jiangsu Engineering and Technology Research Centre for Microbiology; College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Jialu Fan
- Jiangsu Key Laboratory for Microbes and Functional Genomics, Jiangsu Engineering and Technology Research Centre for Microbiology; College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Jing Ye
- Jiangsu Key Laboratory for Microbes and Functional Genomics, Jiangsu Engineering and Technology Research Centre for Microbiology; College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Ling Lu
- Jiangsu Key Laboratory for Microbes and Functional Genomics, Jiangsu Engineering and Technology Research Centre for Microbiology; College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
313
|
González J, Salvador M, Özkaya Ö, Spick M, Reid K, Costa C, Bailey MJ, Avignone Rossa C, Kümmerli R, Jiménez JI. Loss of a pyoverdine secondary receptor in Pseudomonas aeruginosa results in a fitter strain suitable for population invasion. ISME JOURNAL 2020; 15:1330-1343. [PMID: 33323977 DOI: 10.1038/s41396-020-00853-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 11/03/2020] [Accepted: 11/20/2020] [Indexed: 01/27/2023]
Abstract
The rapid emergence of antibiotic resistant bacterial pathogens constitutes a critical problem in healthcare and requires the development of novel treatments. Potential strategies include the exploitation of microbial social interactions based on public goods, which are produced at a fitness cost by cooperative microorganisms, but can be exploited by cheaters that do not produce these goods. Cheater invasion has been proposed as a 'Trojan horse' approach to infiltrate pathogen populations with strains deploying built-in weaknesses (e.g., sensitiveness to antibiotics). However, previous attempts have been often unsuccessful because population invasion by cheaters was prevented by various mechanisms including the presence of spatial structure (e.g., growth in biofilms), which limits the diffusion and exploitation of public goods. Here we followed an alternative approach and examined whether the manipulation of public good uptake and not its production could result in potential 'Trojan horses' suitable for population invasion. We focused on the siderophore pyoverdine produced by the human pathogen Pseudomonas aeruginosa MPAO1 and manipulated its uptake by deleting and/or overexpressing the pyoverdine primary (FpvA) and secondary (FpvB) receptors. We found that receptor synthesis feeds back on pyoverdine production and uptake rates, which led to strains with altered pyoverdine-associated costs and benefits. Moreover, we found that the receptor FpvB was advantageous under iron-limited conditions but revealed hidden costs in the presence of an antibiotic stressor (gentamicin). As a consequence, FpvB mutants became the fittest strain under gentamicin exposure, displacing the wildtype in liquid cultures, and in biofilms and during infections of the wax moth larvae Galleria mellonella, which both represent structured environments. Our findings reveal that an evolutionary trade-off associated with the costs and benefits of a versatile pyoverdine uptake strategy can be harnessed for devising a Trojan-horse candidate for medical interventions.
Collapse
Affiliation(s)
- Jaime González
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Manuel Salvador
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Özhan Özkaya
- Department of Quantitative Medicine, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Matt Spick
- Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Kate Reid
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Catia Costa
- Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Melanie J Bailey
- Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | | | - Rolf Kümmerli
- Department of Quantitative Medicine, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - José I Jiménez
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK. .,Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
314
|
Senior NJ, Titball RW. Isolation and primary culture of Galleria mellonella hemocytes for infection studies. F1000Res 2020; 9:1392. [PMID: 33520196 PMCID: PMC7818094 DOI: 10.12688/f1000research.27504.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2021] [Indexed: 10/10/2023] Open
Abstract
Galleria mellonella larvae are increasingly used to study the mechanisms of virulence of microbial pathogens and to assess the efficacy of antimicrobials. The G. mellonella model can faithfully reproduce many aspects of microbial disease which are seen in mammals, and therefore allows a reduction in the use of mammals. The model is now being widely used by researchers in universities, research institutes and industry. An attraction of the model is the interaction between pathogen and host. Hemocytes are specialised phagocytic cells which resemble neutrophils in mammals and play a major role in the response of the larvae to infection. However, the detailed interactions of hemocytes with pathogens is poorly understood, and is complicated by the presence of different sub-populations of cells. We report here a method for the isolation of hemocytes from Galleria mellonella. A needle-stick injury of larvae, before harvesting, markedly increased the recovery of hemocytes in the hemolymph. The majority of the hemocytes recovered were granulocyte-like cells. The hemocytes survived for at least 7 days in culture at either 28°C or 37°C. Pre-treatment of larvae with antibiotics did not enhance the survival of the cultured hemocytes. Our studies highlight the importance of including sham injected, rather than un-injected, controls when the G. mellonella model is used to test antimicrobial compounds. Our method will now allow investigations of the interactions of microbial pathogens with insect hemocytes enhancing the value of G. mellonella as an alternative model to replace the use of mammals, and for studies on hemocyte biology.
Collapse
Affiliation(s)
- Nicola J. Senior
- College of Life and Environmental Sciences - Biosciences, University of Exeter, Exeter, Devon, EX4 4QD, UK
| | - Richard W. Titball
- College of Life and Environmental Sciences - Biosciences, University of Exeter, Exeter, Devon, EX4 4QD, UK
| |
Collapse
|
315
|
Alves LA, Ganguly T, Harth-Chú ÉN, Kajfasz J, Lemos JA, Abranches J, Mattos-Graner RO. PepO is a target of the two-component systems VicRK and CovR required for systemic virulence of Streptococcus mutans. Virulence 2020; 11:521-536. [PMID: 32427040 PMCID: PMC7239026 DOI: 10.1080/21505594.2020.1767377] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/10/2020] [Accepted: 03/29/2020] [Indexed: 12/14/2022] Open
Abstract
Streptococcus mutans, a cariogenic species, is often associated with cardiovascular infections. Systemic virulence of specific S. mutans serotypes has been associated with the expression of the collagen- and laminin-binding protein Cnm, which is transcriptionally regulated by VicRK and CovR. In this study, we characterized a VicRK- and CovR-regulated gene, pepO, coding for a conserved endopeptidase. Transcriptional and protein analyses revealed that pepO is highly expressed in S. mutans strains resistant to complement immunity (blood isolates) compared to oral isolates. Gel mobility assay, transcriptional, and Western blot analyses revealed that pepO is repressed by VicR and induced by CovR. Deletion of pepO in the Cnm+ strain OMZ175 (OMZpepO) or in the Cnm- UA159 (UApepO) led to an increased susceptibility to C3b deposition, and to low binding to complement proteins C1q and C4BP. Additionally, pepO mutants showed diminished ex vivo survival in human blood and impaired capacity to kill G. mellonella larvae. Inactivation of cnm in OMZ175 (OMZcnm) resulted in increased resistance to C3b deposition and unaltered blood survival, although both pepO and cnm mutants displayed attenuated virulence in G. mellonella. Unlike OMZcnm, OMZpepO could invade HCAEC endothelial cells. Supporting these phenotypes, recombinant proteins rPepO and rCnmA showed specific profiles of binding to C1q, C4BP, and to other plasma (plasminogen, fibronectin) and extracellular matrix proteins (type I collagen, laminin). Therefore this study identifies a novel VicRK/CovR-target required for immune evasion and host persistence, pepO, expanding the roles of VicRK and CovR in regulating S. mutans virulence.
Collapse
Affiliation(s)
- Lívia A. Alves
- Department of Oral Diagnosis, Piracicaba Dental School – State University of Campinas, Piracicaba, SP, Brazil
| | - Tridib Ganguly
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Érika N. Harth-Chú
- Department of Oral Diagnosis, Piracicaba Dental School – State University of Campinas, Piracicaba, SP, Brazil
| | - Jessica Kajfasz
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - José A. Lemos
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Jacqueline Abranches
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Renata O. Mattos-Graner
- Department of Oral Diagnosis, Piracicaba Dental School – State University of Campinas, Piracicaba, SP, Brazil
| |
Collapse
|
316
|
Alonso B, Fernández-Barat L, Di Domenico EG, Marín M, Cercenado E, Merino I, de Pablos M, Muñoz P, Guembe M. Characterization of the virulence of Pseudomonas aeruginosa strains causing ventilator-associated pneumonia. BMC Infect Dis 2020; 20:909. [PMID: 33261585 PMCID: PMC7706020 DOI: 10.1186/s12879-020-05534-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 10/21/2020] [Indexed: 12/14/2022] Open
Abstract
Background The objective of this study was to evaluate the virulence of P. aeruginosa ventilator-associated pneumonia (VAP) strains (cases) in terms of biofilm production and other phenotypic and genotypic virulence factors compared to P. aeruginosa strains isolated from other infections (controls). Methods Biofilm production was tested to assess biomass production and metabolic activity using crystal violet binding assay and XTT assay, respectively. Pigment production (pyocyanin and pyoverdine) was evaluated using cetrimide agar. Virulence genes were detected by conventional multiplex PCR and virulence was tested in an in vivo model in Galleria mellonella larvae. Results We did not find statistically significant differences between VAP and no-VAP strains (p > 0.05) regarding biofilm production. VAP strains had no production of pyocyanin after 24 h of incubation (p = 0.023). The distribution of virulence genes between both groups were similar (p > 0.05). VAP strains were less virulent than non-VAP strains in an in vivo model of G. mellonella (p < 0.001). Conclusion The virulence of VAP-Pseudomonas aeruginosa does not depend on biofilm formation, production of pyoverdine or the presence of some virulence genes compared to P. aeruginosa isolated from non-invasive locations. However, VAP strains showed attenuated virulence compared to non-VAP strains in an in vivo model of G. mellonella. Supplementary Information Supplementary information accompanies this paper at 10.1186/s12879-020-05534-1.
Collapse
Affiliation(s)
- Beatriz Alonso
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain. .,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.
| | - Laia Fernández-Barat
- Centro de Investigación Biomedica En Red-Enfermedades Respiratorias (CibeRes, CB06/06/0028) and Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Center for Biomedical Research CELLEX, School of Medicine, University of Barcelona, Barcelona, Spain
| | | | - Mercedes Marín
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Emilia Cercenado
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Irene Merino
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Red Española de Investigación en Patología Infecciosa (REIPI), Madrid, Spain.,Group For Biomedical Research in Sepsis (BioSepsis) Hospital Clínico Universitario de Valladolid, Valladolid, Spain.,Centro de Investigación Biomedica En Red - Enfermedades Respiratorias (CibeRes, CB06/06/0028), Barcelona, Spain.,National Health System, SACYL/IECSCYL, Valladolid, Spain
| | - Manuela de Pablos
- Servicio de Microbiología y Parasitología Hospital Universitario La Paz, Madrid, Spain
| | - Patricia Muñoz
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,CIBER Enfermedades Respiratorias-CIBERES (CB06/06/0058), Madrid, Spain.,Medicine Department, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - María Guembe
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| |
Collapse
|
317
|
Phenotypic and Transcriptomic Analyses of Seven Clinical Stenotrophomonas maltophilia Isolates Identify a Small Set of Shared and Commonly Regulated Genes Involved in the Biofilm Lifestyle. Appl Environ Microbiol 2020; 86:AEM.02038-20. [PMID: 33097507 DOI: 10.1128/aem.02038-20] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/04/2020] [Indexed: 12/19/2022] Open
Abstract
Stenotrophomonas maltophilia is one of the most frequently isolated multidrug-resistant nosocomial opportunistic pathogens. It contributes to disease progression in cystic fibrosis (CF) patients and is frequently isolated from wounds, infected tissues, and catheter surfaces. On these diverse surfaces S. maltophilia lives in single-species or multispecies biofilms. Since very little is known about common processes in biofilms of different S. maltophilia isolates, we analyzed the biofilm profiles of 300 clinical and environmental isolates from Europe of the recently identified main lineages Sgn3, Sgn4, and Sm2 to Sm18. The analysis of the biofilm architecture of 40 clinical isolates revealed the presence of multicellular structures and high phenotypic variability at a strain-specific level. Further, transcriptome analyses of biofilm cells of seven clinical isolates identified a set of 106 shared strongly expressed genes and 33 strain-specifically expressed genes. Surprisingly, the transcriptome profiles of biofilm versus planktonic cells revealed that just 9.43% ± 1.36% of all genes were differentially regulated. This implies that just a small set of shared and commonly regulated genes is involved in the biofilm lifestyle. Strikingly, iron uptake appears to be a key factor involved in this metabolic shift. Further, metabolic analyses implied that S. maltophilia employs a mostly fermentative growth mode under biofilm conditions. The transcriptome data of this study together with the phenotypic and metabolic analyses represent so far the largest data set on S. maltophilia biofilm versus planktonic cells. This study will lay the foundation for the identification of strategies for fighting S. maltophilia biofilms in clinical and industrial settings.IMPORTANCE Microorganisms living in a biofilm are much more tolerant to antibiotics and antimicrobial substances than planktonic cells are. Thus, the treatment of infections caused by microorganisms living in biofilms is extremely difficult. Nosocomial infections (among others) caused by S. maltophilia, particularly lung infection among CF patients, have increased in prevalence in recent years. The intrinsic multidrug resistance of S. maltophilia and the increased tolerance to antimicrobial agents of its biofilm cells make the treatment of S. maltophilia infection difficult. The significance of our research is based on understanding the common mechanisms involved in biofilm formation of different S. maltophilia isolates, understanding the diversity of biofilm architectures among strains of this species, and identifying the differently regulated processes in biofilm versus planktonic cells. These results will lay the foundation for the treatment of S. maltophilia biofilms.
Collapse
|
318
|
Wojda I, Staniec B, Sułek M, Kordaczuk J. The greater wax moth Galleria mellonella: biology and use in immune studies. Pathog Dis 2020; 78:ftaa057. [PMID: 32970818 PMCID: PMC7683414 DOI: 10.1093/femspd/ftaa057] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/28/2020] [Indexed: 01/04/2023] Open
Abstract
The greater wax moth Galleria mellonella is an invertebrate that is increasingly being used in scientific research. Its ease of reproduction, numerous offspring, short development cycle, and finally, its known genome and immune-related transcriptome provide a convenient research model for investigation of insect immunity at biochemical and molecular levels. Galleria immunity, consisting of only innate mechanisms, shows adaptive plasticity, which has recently become the subject of intensive scientific research. This insect serves as a mini host in studies of the pathogenicity of microorganisms and in vivo tests of the effectiveness of single virulence factors as well as new antimicrobial compounds. Certainly, the Galleria mellonella species deserves our attention and appreciation for its contribution to the development of research on innate immune mechanisms. In this review article, we describe the biology of the greater wax moth, summarise the main advantages of using it as a model organism and present some of the main techniques facilitating work with this insect.
Collapse
Affiliation(s)
- Iwona Wojda
- Maria Curie Sklodowska University, Institute of Biological Sciences, Department of Immunobiology, Akademicka 19, 20-033 Lublin, Poland
| | - Bernard Staniec
- Maria Curie Sklodowska University, Institute of Biological Sciences, Department of Zoology and Nature Protection, Akademicka 19, 20-033 Lublin, Poland
| | - Michał Sułek
- Maria Curie Sklodowska University, Institute of Biological Sciences, Department of Immunobiology, Akademicka 19, 20-033 Lublin, Poland
| | - Jakub Kordaczuk
- Maria Curie Sklodowska University, Institute of Biological Sciences, Department of Immunobiology, Akademicka 19, 20-033 Lublin, Poland
| |
Collapse
|
319
|
Romera D, Aguilera-Correa JJ, García-Coca M, Mahillo-Fernández I, Viñuela-Sandoval L, García-Rodríguez J, Esteban J. The Galleria mellonella infection model as a system to investigate the virulence of Candida auris strains. Pathog Dis 2020; 78:5937422. [PMID: 33098293 DOI: 10.1093/femspd/ftaa067] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 10/22/2020] [Indexed: 02/06/2023] Open
Abstract
Candida auris is a multiresistant pathogenic yeast commonly isolated from bloodstream infections in immunocompromised patients. In this work, we infected Galleria mellonella larvae with 105 CFU of a reference strains and two clinical isolates of C. albicans and C. auris and we compared the outcomes of infection between both species. Larvae were evaluated every 24 h for a total of 120 h following the G. mellonella Health Index Scoring System, and survival, activity, melanization and cocoon formation were monitored. Our results showed that clinical isolates were significantly more pathogenic than reference strains independently of the tested species, producing lower survival and activity scores and higher melanization scores and being C. albicans strains more virulent than C. auris strains. We did not find differences in mortality between aggregative and non-aggregative C. auris strains, although non-aggregative strains produced significantly lower activity scores and higher melanization scores than aggregative ones. Survival assays using Galleria mellonella have been previously employed to examine and classify strains of this and other microbial species based on their virulence before scaling the experiments to a mammal model. Taken together, these results show how a more complete evaluation of the model can improve the study of C. auris isolates.
Collapse
Affiliation(s)
- David Romera
- Department of Clinical Microbiology, IIS Fundación Jiménez Díaz, UAM. Avda. Reyes Católicos 2, 28040 Madrid, Spain
| | - John-Jairo Aguilera-Correa
- Department of Clinical Microbiology, IIS Fundación Jiménez Díaz, UAM. Avda. Reyes Católicos 2, 28040 Madrid, Spain
| | - Marta García-Coca
- Department of Clinical Microbiology, IIS Fundación Jiménez Díaz, UAM. Avda. Reyes Católicos 2, 28040 Madrid, Spain
| | - Ignacio Mahillo-Fernández
- Epidemiology and Biostatistics Service, Fundación Jiménez Díaz University Hospital, Av. Reyes Católicos, 2. 28040 Madrid, Spain
| | | | - Julio García-Rodríguez
- Department of Microbiology, La Paz University Hospital, Paseo de la Castellana, 261, 28046 Madrid, Spain
| | - Jaime Esteban
- Department of Clinical Microbiology, IIS Fundación Jiménez Díaz, UAM. Avda. Reyes Católicos 2, 28040 Madrid, Spain
| |
Collapse
|
320
|
Bispo M, Anaya-Sanchez A, Suhani S, Raineri EJM, López-Álvarez M, Heuker M, Szymański W, Romero Pastrana F, Buist G, Horswill AR, Francis KP, van Dam GM, van Oosten M, van Dijl JM. Fighting Staphylococcus aureus infections with light and photoimmunoconjugates. JCI Insight 2020; 5:139512. [PMID: 33048846 PMCID: PMC7710284 DOI: 10.1172/jci.insight.139512] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 10/07/2020] [Indexed: 12/19/2022] Open
Abstract
Infections caused by multidrug-resistant Staphylococcus aureus, especially methicillin-resistant S. aureus (MRSA), are responsible for high mortality and morbidity worldwide. Resistant lineages were previously confined to hospitals but are now also causing infections among healthy individuals in the community. It is therefore imperative to explore therapeutic avenues that are less prone to raise drug resistance compared with today’s antibiotics. An opportunity to achieve this ambitious goal could be provided by targeted antimicrobial photodynamic therapy (aPDT), which relies on the combination of a bacteria-specific targeting agent and light-induced generation of ROS by an appropriate photosensitizer. Here, we conjugated the near-infrared photosensitizer IRDye700DX to a fully human mAb, specific for the invariantly expressed staphylococcal antigen immunodominant staphylococcal antigen A (IsaA). The resulting immunoconjugate 1D9-700DX was characterized biochemically and in preclinical infection models. As demonstrated in vitro, in vivo, and in a human postmortem orthopedic implant infection model, targeted aPDT with 1D9-700DX is highly effective. Importantly, combined with the nontoxic aPDT-enhancing agent potassium iodide, 1D9-700DX overcomes the antioxidant properties of human plasma and fully eradicates high titers of MRSA. We show that the developed immunoconjugate 1D9-700DX targets MRSA and kills it upon illumination with red light, without causing collateral damage to human cells. An immunoconjugate for targeted photodynamic therapy of Staphylococcus aureus infections kills MRSA with high efficacy upon illumination with red light.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wiktor Szymański
- Department of Radiology, Medical Imaging Center, University of Groningen, University Medical Center Groningen, Groningen, Netherlands.,Stratingh Institute for Chemistry, University of Groningen, Groningen, Netherlands
| | | | | | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | | | - Gooitzen M van Dam
- Department of Surgery, Division of Surgical Oncology, Nuclear Medicine and Molecular Imaging, Intensive Care, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | | | | |
Collapse
|
321
|
Monitoring Gene Expression during a Galleria mellonella Bacterial Infection. Microorganisms 2020; 8:microorganisms8111798. [PMID: 33207842 PMCID: PMC7697238 DOI: 10.3390/microorganisms8111798] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 12/05/2022] Open
Abstract
Galleria mellonella larvae are an alternative in vivo model that has been extensively used to study the virulence and pathogenicity of different bacteria due to its practicality and lack of ethical constraints. However, the larvae possess intrinsic autofluorescence that obstructs the use of fluorescent proteins to study bacterial infections, hence better methodologies are needed. Here, we report the construction of a promoter probe vector with bioluminescence expression as well as the optimization of a total bacterial RNA extraction protocol to enhance the monitoring of in vivo infections. By employing the vector to construct different gene promoter fusions, variable gene expression levels were efficiently measured in G. mellonella larvae at various time points during the course of infection and without much manipulation of the larvae. Additionally, our optimized RNA extraction protocol facilitates the study of transcriptional gene levels during an in vivo infection. The proposed methodologies will greatly benefit bacterial infection studies as they can contribute to a better understanding of the in vivo infection processes and pathogen–mammalian host interactions.
Collapse
|
322
|
CsrA Supports both Environmental Persistence and Host-Associated Growth of Acinetobacter baumannii. Infect Immun 2020; 88:IAI.00259-20. [PMID: 32989034 DOI: 10.1128/iai.00259-20] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 09/18/2020] [Indexed: 11/20/2022] Open
Abstract
Acinetobacter baumannii is an opportunistic and frequently multidrug-resistant Gram-negative bacterial pathogen that primarily infects critically ill individuals. Indirect transmission from patient to patient in hospitals can drive infections, supported by this organism's abilities to persist on dry surfaces and rapidly colonize susceptible individuals. To investigate how A. baumannii survives on surfaces, we cultured A. baumannii in liquid media for several days and then analyzed isolates that lost the ability to survive drying. One of these isolates carried a mutation that affected the gene encoding the carbon storage regulator CsrA. As we began to examine the role of CsrA in A. baumannii, we observed that the growth of ΔcsrA mutant strains was inhibited in the presence of amino acids. The ΔcsrA mutant strains had a reduced ability to survive drying and to form biofilms but an improved ability to tolerate increased osmolarity compared with the wild type. We also examined the importance of CsrA for A. baumannii virulence. The ΔcsrA mutant strains had a greatly reduced ability to kill Galleria mellonella larvae, could not replicate in G. mellonella hemolymph, and also had a growth defect in human serum. Together, these results show that CsrA is essential for the growth of A. baumannii on host-derived substrates and is involved in desiccation tolerance, implying that CsrA controls key functions involved in the transmission of A. baumannii in hospitals.
Collapse
|
323
|
Westmeier D, Siemer S, Vallet C, Steinmann J, Docter D, Buer J, Knauer SK, Stauber RH. Boosting nanotoxicity to combat multidrug-resistant bacteria in pathophysiological environments. NANOSCALE ADVANCES 2020; 2:5428-5440. [PMID: 36132026 PMCID: PMC9419095 DOI: 10.1039/d0na00644k] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/21/2020] [Indexed: 06/15/2023]
Abstract
Nanomaterials are promising novel antibiotics, but often ineffective. We found that nanomaterial-bacteria complex formation occurred with various nanomaterials. The bactericidal activity of NMs strongly depends on their physical binding to (multidrug-resistant) bacteria. Nanomaterials' binding and antibiotic effect was reduced by various pathophysiological biomolecule coronas strongly inhibiting their antibiotic effects. We show from analytical to in vitro to in vivo that nanomaterial-based killing could be restored by acidic pH treatments. Here, complex formation of negatively-charged, plasma corona-covered, nanomaterials with bacteria was electrostatically enhanced by reducing bacteria's negative surface charge. Employing in vivo skin infection models, acidic pH-induced complex formation was critical to counteract Staphylococcus aureus infections by silver nanomaterials. We explain why nano-antibiotics show reduced activity and provide a clinically practical solution.
Collapse
Affiliation(s)
- Dana Westmeier
- ENT Department, University Medical Center Mainz Langenbeckstrasse 1 55131 Mainz Germany
| | - Svenja Siemer
- ENT Department, University Medical Center Mainz Langenbeckstrasse 1 55131 Mainz Germany
| | - Cecilia Vallet
- Department of Molecular Biology II, Center for Medical Biotechnology/Nanointegration (ZMB/CENIDE), University Duisburg-Essen, Universitätsstrasse 5 45117 Essen Germany
| | - Jörg Steinmann
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen Hufelandstrasse 55 45112 Essen Germany
| | - Dominic Docter
- ENT Department, University Medical Center Mainz Langenbeckstrasse 1 55131 Mainz Germany
| | - Jan Buer
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen Hufelandstrasse 55 45112 Essen Germany
| | - Shirley K Knauer
- Department of Molecular Biology II, Center for Medical Biotechnology/Nanointegration (ZMB/CENIDE), University Duisburg-Essen, Universitätsstrasse 5 45117 Essen Germany
| | - Roland H Stauber
- ENT Department, University Medical Center Mainz Langenbeckstrasse 1 55131 Mainz Germany
| |
Collapse
|
324
|
Functional redundancy of Burkholderia pseudomallei phospholipase C enzymes and their role in virulence. Sci Rep 2020; 10:19242. [PMID: 33159122 PMCID: PMC7648637 DOI: 10.1038/s41598-020-76186-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 10/21/2020] [Indexed: 11/12/2022] Open
Abstract
Phospholipase C (PLC) enzymes are key virulence factors in several pathogenic bacteria. Burkholderia pseudomallei, the causative agent of melioidosis, possesses at least three plc genes (plc1, plc2 and plc3). We found that in culture medium plc1 gene expression increased with increasing pH, whilst expression of the plc3 gene was pH (4.5 to 9.0) independent. Expression of the plc2 gene was not detected in culture medium. All three plc genes were expressed during macrophage infection by B. pseudomallei K96243. Comparing B. pseudomallei wild-type with plc mutants revealed that plc2, plc12 or plc123 mutants showed reduced intracellular survival in macrophages and reduced plaque formation in HeLa cells. However, plc1 or plc3 mutants showed no significant differences in plaque formation compared to wild-type bacteria. These findings suggest that Plc2, but not Plc1 or Plc3 are required for infection of host cells. In Galleria mellonella, plc1, plc2 or plc3 mutants were not attenuated compared to the wild-type strain, but multiple plc mutants showed reduced virulence. These findings indicate functional redundancy of the B. pseudomallei phospholipases in virulence.
Collapse
|
325
|
Santos TA, Scorzoni L, Santos ADC, Junqueira JC, Anbinder AL. Galleria mellonella as an experimental model for studying periodontopathogens. J Indian Soc Periodontol 2020; 24:593-596. [PMID: 33424181 PMCID: PMC7781244 DOI: 10.4103/jisp.jisp_631_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 05/29/2020] [Accepted: 06/11/2020] [Indexed: 11/04/2022] Open
Abstract
In the present study, Galleria mellonella was evaluated as a potential infection model for periodontal bacteria, more specifically, Porphyromonas gingivalis, Fusobacterium nucleatum, and Aggregatibacter actinomycetemcomitans. All the bacteria evaluated were pathogenic to G. mellonella, causing their death in a concentration-dependent manner, and a decrease in their hemocyte count. Moreover, it was possible to recover the bacteria from the larvae hemolymph and determine the colony-forming units per larvae. G. mellonella is an effective model that may help to better understand the host-microbe interactions in periodontics.
Collapse
Affiliation(s)
- Thaís Aguiar Santos
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (Unesp), São José dos Campos, SP, Brazil
| | - Liliana Scorzoni
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (Unesp), São José dos Campos, SP, Brazil
| | - Aline de Castro Santos
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (Unesp), São José dos Campos, SP, Brazil
| | - Juliana Campos Junqueira
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (Unesp), São José dos Campos, SP, Brazil
| | - Ana Lia Anbinder
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (Unesp), São José dos Campos, SP, Brazil
| |
Collapse
|
326
|
Pereira MF, Rossi CC. Overview of rearing and testing conditions and a guide for optimizing Galleria mellonella breeding and use in the laboratory for scientific purposes. APMIS 2020; 128:607-620. [PMID: 32970339 DOI: 10.1111/apm.13082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The greater wax moth Galleria mellonella is an increasingly popular and consolidated alternative infection model to assess microbial virulence and the effectiveness of antimicrobial compounds. The lack of G. mellonella suppliers aiming at scientific purposes and a lack of well-established protocols for raising and testing these animals may impact results and reproducibility between different laboratories. In this review, we discuss the state of the art of rearing the larvae in situ, providing an overview of breeding and testing conditions commonly used and their influence on larval health and experiments results, from setting up the environment, providing the ideal diet, understanding the effects of pretreatments, choosing the best testing conditions, to exploring the most from the results obtained. Meanwhile, we guide the reader through the most practical ways of dealing with G. mellonella to achieve successful experiments.
Collapse
Affiliation(s)
- Monalessa Fábia Pereira
- Laboratório de Bioquímica e Microbiologia, Departamento de Ciências Biológicas, Universidade do Estado de Minas Gerais, Carangola, MG, Brazil
| | - Ciro César Rossi
- Laboratório de Microbiologia Molecular, Departamento de Microbiologia Médica, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
327
|
Piatek M, Sheehan G, Kavanagh K. UtilisingGalleria mellonella larvae for studying in vivo activity of conventional and novel antimicrobial agents. Pathog Dis 2020; 78:5917982. [DOI: 10.1093/femspd/ftaa059] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 10/02/2020] [Indexed: 12/15/2022] Open
Abstract
ABSTRACTThe immune response of insects displays many structural and functional similarities to the innate immune response of mammals. As a result of these conserved features, insects may be used for evaluating microbial virulence or for testing the in vivo efficacy and toxicity of antimicrobial compounds and results show strong similarities to those from mammals. Galleria mellonella larvae are widely used in this capacity and have the advantage of being easy to use, inexpensive to purchase and house, and being free from the ethical and legal restrictions that relate to the use of mammals in these tests. Galleria mellonella larvae may be used to assess the in vivo toxicity and efficacy of novel antimicrobial compounds. A wide range of antibacterial and antifungal therapies have been evaluated in G. mellonella larvae and results have informed subsequent experiments in mammals. While insect larvae are a convenient and reproducible model to use, care must be taken in their use to ensure accuracy of results. The objective of this review is to provide a comprehensive account of the use of G. mellonella larvae for assessing the in vivo toxicity and efficacy of a wide range of antibacterial and antifungal agents.
Collapse
Affiliation(s)
- Magdalena Piatek
- SSPC Pharma Research Centre, Department of Biology, Maynooth University, Co. Kildare W23 F2H6, Ireland
| | - Gerard Sheehan
- Institute of Microbiology and Infection, University of Birmingham, Birmingham B15 2TT, UK
| | - Kevin Kavanagh
- SSPC Pharma Research Centre, Department of Biology, Maynooth University, Co. Kildare W23 F2H6, Ireland
| |
Collapse
|
328
|
Tavares RDS, Tacão M, Figueiredo AS, Duarte AS, Esposito F, Lincopan N, Manaia CM, Henriques I. Genotypic and phenotypic traits of bla CTX-M-carrying Escherichia coli strains from an UV-C-treated wastewater effluent. WATER RESEARCH 2020; 184:116079. [PMID: 32717492 DOI: 10.1016/j.watres.2020.116079] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 06/11/2023]
Abstract
Wastewater treatment plants (WWTPs) are relevant sources of antibiotic resistance into aquatic environments. Disinfection of WWTPs' effluents (e.g. by UV-C irradiation) may attenuate this problem, though some clinically relevant bacteria have been shown to survive disinfection. In this study we characterized 25 CTX-M-producing Escherichia coli strains isolated from a WWTP's UV-C-irradiated effluent, aiming to identify putative human health hazards associated with such effluents. Molecular typing indicated that the strains belong to the phylogroups A, B2 and C and clustered into 9 multilocus sequence types (STs), namely B2:ST131 (n = 7), A:ST58 (n = 1), A:ST155 (n = 4), C:ST410 (n = 2), A:ST453 (n = 2), A:ST617 (n = 2), A:ST744 (n = 1), A:ST1284 (n = 3) and a putative novel ST (n = 3). PCR-screening identified 9 of the 20 antibiotic resistance genes investigated [i.e. sul1, sul2, sul3, tet(A), tet(B), blaOXA-1-like, aacA4, aacA4-cr and qnrS1]. The more prevalent were sul1, sul2 (n = 15 isolates) and tet(A) (n = 14 isolates). Plasmid restriction analysis indicated diverse plasmid content among strains (14 distinct profiles) and mating assays yielded cefotaxime-resistant transconjugants for 8 strains. Two of the transconjugants displayed a multi-drug resistance (MDR) phenotype. All strains were classified as cytotoxic to Vero cells (9 significantly more cytotoxic than the positive control) and 10 of 21 strains were invasive towards this cell line (including all B2:ST131 strains). The 10 strains tested against G. mellonella larvae exhibited a virulent behaviour. Twenty-four and 7 of the 25 strains produced siderophores and haemolysins, respectively. Approximately 66% of the strains formed biofilms. Genome analysis of 6 selected strains identified several virulence genes encoding toxins, siderophores, and colonizing, adhesion and invasion factors. Freshwater microcosms assays showed that after 28 days of incubation 3 out of 6 strains were still detected by cultivation and 4 strains by qPCR. Resistance phenotypes of these strains remained unaltered. Overall, we confirmed WWTP's UV-C-treated outflow as a source of MDR and/or virulent E. coli strains, some probably capable of persisting in freshwater, and that carry conjugative antibiotic resistance plasmids. Hence, disinfected wastewater may still represent a risk for human health. More detailed evaluation of strains isolated from wastewater effluents is urgent, to design treatments that can mitigate the release of such bacteria.
Collapse
Affiliation(s)
- Rafael D S Tavares
- Centre for Environmental and Marine Studies (CESAM), University of Aveiro, Campus Universitário Santiago, 3810-193, Aveiro, Portugal; Department of Biology, University of Aveiro, Campus Universitário Santiago, 3810-193, Aveiro, Portugal
| | - Marta Tacão
- Centre for Environmental and Marine Studies (CESAM), University of Aveiro, Campus Universitário Santiago, 3810-193, Aveiro, Portugal; Department of Biology, University of Aveiro, Campus Universitário Santiago, 3810-193, Aveiro, Portugal.
| | - Ana S Figueiredo
- Department of Biology, University of Aveiro, Campus Universitário Santiago, 3810-193, Aveiro, Portugal
| | - Ana S Duarte
- Universidade Católica Portuguesa, Faculdade de Medicina Dentária, Centro de Investigação Interdisciplinar Em Saúde (CIIS), Estrada da Circunvalação, 3504-505, Viseu, Portugal
| | - Fernanda Esposito
- Department of Clinical Analysis, Faculty of Pharmacy, University of São Paulo, São Paulo, Brazil
| | - Nilton Lincopan
- Department of Clinical Analysis, Faculty of Pharmacy, University of São Paulo, São Paulo, Brazil; Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Célia M Manaia
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina, Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho 1327, 4169-005, Porto, Portugal
| | - Isabel Henriques
- Centre for Environmental and Marine Studies (CESAM), University of Aveiro, Campus Universitário Santiago, 3810-193, Aveiro, Portugal; University of Coimbra, Department of Life Sciences, Faculty of Sciences and Technology, Calçada Martins de Freitas, 3000-456, Coimbra, Portugal
| |
Collapse
|
329
|
Abstract
Porphyromonas gingivalis is a key pathogen of periodontitis, a polymicrobial disease characterized by a chronic inflammation that destroys the tissues supporting the teeth. Thus, understanding the virulence potential of P. gingivalis is essential to maintaining a healthy oral microbiome. In nonoral organisms, CRISPR-Cas systems have been shown to modulate a variety of microbial processes, including protection from exogenous nucleic acids, and, more recently, have been implicated in bacterial virulence. Previously, our clinical findings identified activation of the CRISPR-Cas system in patient samples at the transition to disease; however, the mechanism of contribution to disease remained unknown. The importance of the present study resides in that it is becoming increasingly clear that CRISPR-associated proteins have broader functions than initially thought and that those functions now include their role in the virulence of periodontal pathogens. Studying a P. gingivaliscas3 mutant, we demonstrate that at least one of the CRISPR-Cas systems is involved in the regulation of virulence during infection. The CRISPR (clustered regularly interspaced short palindromic repeat)-Cas system is a unique genomic entity that provides prokaryotic cells with adaptive and heritable immunity. Initial studies identified CRISPRs as central elements used by bacteria to protect against foreign nucleic acids; however, emerging evidence points to CRISPR involvement in bacterial virulence. The present study aimed to identify the participation of one CRISPR-Cas protein, Cas3, in the virulence of the oral pathogen Porphyromonas gingivalis, an organism highly associated with periodontitis. Our results show that compared to the wild type, a mutant with a deletion of the Cas3 gene, an essential nuclease part of the class 1 type I CRISPR-Cas system, increased the virulence of P. gingivalis. In vitro infection modeling revealed only mildly enhanced production of proinflammatory cytokines by THP-1 cells when infected with the mutant strain. Dual transcriptome sequencing (RNA-seq) analysis of infected THP-1 cells showed an increase in expression of genes associated with pathogenesis in response to Δcas3 mutant infection, with the target of Cas3 activities in neutrophil chemotaxis and gene silencing. The importance of cas3 in controlling virulence was corroborated in a Galleria mellonella infection model, where the presence of the Δcas3 mutant resulted in a statistically significant increase in mortality of G. mellonella. A time-series analysis of transcription patterning during infection showed that G. mellonella elicited very different immune responses to the wild-type and the Δcas3 mutant strains and revealed a rearrangement of association in coexpression networks. Together, these observations show for the first time that Cas3 plays a significant role in regulating the virulence of P. gingivalis. IMPORTANCEPorphyromonas gingivalis is a key pathogen of periodontitis, a polymicrobial disease characterized by a chronic inflammation that destroys the tissues supporting the teeth. Thus, understanding the virulence potential of P. gingivalis is essential to maintaining a healthy oral microbiome. In nonoral organisms, CRISPR-Cas systems have been shown to modulate a variety of microbial processes, including protection from exogenous nucleic acids, and, more recently, have been implicated in bacterial virulence. Previously, our clinical findings identified activation of the CRISPR-Cas system in patient samples at the transition to disease; however, the mechanism of contribution to disease remained unknown. The importance of the present study resides in that it is becoming increasingly clear that CRISPR-associated proteins have broader functions than initially thought and that those functions now include their role in the virulence of periodontal pathogens. Studying a P. gingivaliscas3 mutant, we demonstrate that at least one of the CRISPR-Cas systems is involved in the regulation of virulence during infection.
Collapse
|
330
|
Pfefferle K, Lopalco P, Breisch J, Siemund A, Corcelli A, Averhoff B. In vivo synthesis of monolysocardiolipin and cardiolipin by Acinetobacter baumannii phospholipase D and effect on cationic antimicrobial peptide resistance. Environ Microbiol 2020; 22:5300-5308. [PMID: 32929857 DOI: 10.1111/1462-2920.15231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/02/2020] [Accepted: 09/10/2020] [Indexed: 12/29/2022]
Abstract
Acinetobacter baumannii is an opportunistic pathogen, which has become a rising threat in healthcare facilities worldwide due to increasing antibiotic resistances and optimal adaptation to clinical environments and the human host. We reported in a former publication on the identification of three phopholipases of the phospholipase D (PLD) superfamily in A. baumannii ATCC 19606T acting in concerted manner as virulence factors in Galleria mellonella infection and lung epithelial cell invasion. This study focussed on the function of the three PLDs. A Δpld1-3 mutant was defect in biosynthesis of the phospholipids cardiolipin (CL) and monolysocardiolipin (MLCL), whereas the deletion of pld2 and pld3 abolished the production of MLCL. Complementation of the Δpld1-3 mutant with pld1 restored CL biosynthesis demonstrating that the PLD1 is implicated in CL biosynthesis. Complementation of the Δpld1-3 mutant with either pld2 or pld3 restored MLCL and CL production leading to the conclusion that PLD2 and PLD3 are implicated in CL and MLCL production. Mutant studies revealed that two catalytic motifs are essential for the PLD3-mediated biosynthesis of CL and MLCL. The Δpld1-3 mutant exhibited a decreased colistin and polymyxin B resistance indicating a role of CL in cationic antimicrobial peptides (CAMPs) resistance.
Collapse
Affiliation(s)
- Katharina Pfefferle
- Department of Molecular Microbiology and Bioenergetics, Institute of Molecular Biosciences, Goethe-University Frankfurt am Main, Germany
| | - Patrizia Lopalco
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Jennifer Breisch
- Department of Molecular Microbiology and Bioenergetics, Institute of Molecular Biosciences, Goethe-University Frankfurt am Main, Germany
| | - Anna Siemund
- Department of Molecular Microbiology and Bioenergetics, Institute of Molecular Biosciences, Goethe-University Frankfurt am Main, Germany
| | - Angela Corcelli
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Beate Averhoff
- Department of Molecular Microbiology and Bioenergetics, Institute of Molecular Biosciences, Goethe-University Frankfurt am Main, Germany
| |
Collapse
|
331
|
Moya-Andérico L, Admella J, Torrents E. A clearing protocol for Galleria mellonella larvae: Visualization of internalized fluorescent nanoparticles. N Biotechnol 2020; 60:20-26. [PMID: 32866670 DOI: 10.1016/j.nbt.2020.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/01/2020] [Accepted: 08/06/2020] [Indexed: 01/25/2023]
Abstract
Light scattering is a challenge for imaging three-dimensional organisms. A number of new tissue clearing methodologies have been described in recent years, increasing the utilities of clearing techniques to obtain transparent samples. Here, we describe the optimization of a suitable and novel protocol for clearing Galleria mellonella larvae, an alternative infection animal model with a promising potential for the toxicological evaluation of different molecules and materials. This has allowed the visualization of internalised fluorescent nanoparticles using confocal microscopy, opening the door to a wide range of different applications.
Collapse
Affiliation(s)
- Laura Moya-Andérico
- Bacterial Infections and Antimicrobial Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 15-21, 08028, Barcelona, Spain
| | - Joana Admella
- Bacterial Infections and Antimicrobial Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 15-21, 08028, Barcelona, Spain
| | - Eduard Torrents
- Bacterial Infections and Antimicrobial Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 15-21, 08028, Barcelona, Spain; Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, 643 Diagonal Ave., 08028, Barcelona, Spain.
| |
Collapse
|
332
|
Bojanić K, Acke E, Roe WD, Marshall JC, Cornelius AJ, Biggs PJ, Midwinter AC. Comparison of the Pathogenic Potential of Campylobacter jejuni, C. upsaliensis and C. helveticus and Limitations of Using Larvae of Galleria mellonella as an Infection Model. Pathogens 2020; 9:pathogens9090713. [PMID: 32872505 PMCID: PMC7560178 DOI: 10.3390/pathogens9090713] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 11/16/2022] Open
Abstract
Campylobacter enteritis in humans is primarily associated with C. jejuni/coli infection. Other species cause campylobacteriosis relatively infrequently; while this could be attributed to bias in diagnostic methods, the pathogenicity of non-jejuni/coli Campylobacter spp. such as C. upsaliensis and C. helveticus (isolated from dogs and cats) is uncertain. Galleria mellonella larvae are suitable models of the mammalian innate immune system and have been applied to C. jejuni studies. This study compared the pathogenicity of C. jejuni, C. upsaliensis, and C. helveticus isolates. Larvae inoculated with either C. upsaliensis or C. helveticus showed significantly higher survival than those inoculated with C. jejuni. All three Campylobacter species induced indistinguishable histopathological changes in the larvae. C. jejuni could be isolated from inoculated larvae up to eight days post-inoculation whereas C. upsaliensis and C. helveticus could only be isolated in the first two days. There was a significant variation in the hazard rate between batches of larvae, in Campylobacter strains, and in biological replicates as random effects, and in species and bacterial dose as fixed effects. The Galleria model is applicable to other Campylobacter spp. as well as C. jejuni, but may be subject to significant variation with all Campylobacter species. While C. upsaliensis and C. helveticus cannot be considered non-pathogenic, they are significantly less pathogenic than C. jejuni.
Collapse
Affiliation(s)
- Krunoslav Bojanić
- EpiLab, Infectious Disease Research Centre, School of Veterinary Science, Massey University, Palmerston North 4410, New Zealand; (J.C.M.); (P.J.B.); (A.C.M.)
- Correspondence: ; Tel.: +38-514571391
| | - Els Acke
- Klinik für Kleintiere, Veterinärmedizinische Fakultät der Universität Leipzig, 04103 Leipzig, Germany;
| | - Wendi D. Roe
- Department of Pathology, School of Veterinary Science, Massey University, Palmerston North 4410, New Zealand;
| | - Jonathan C. Marshall
- EpiLab, Infectious Disease Research Centre, School of Veterinary Science, Massey University, Palmerston North 4410, New Zealand; (J.C.M.); (P.J.B.); (A.C.M.)
| | - Angela J. Cornelius
- Institute of Environmental Science and Research Limited, Christchurch 8540, New Zealand;
| | - Patrick J. Biggs
- EpiLab, Infectious Disease Research Centre, School of Veterinary Science, Massey University, Palmerston North 4410, New Zealand; (J.C.M.); (P.J.B.); (A.C.M.)
| | - Anne C. Midwinter
- EpiLab, Infectious Disease Research Centre, School of Veterinary Science, Massey University, Palmerston North 4410, New Zealand; (J.C.M.); (P.J.B.); (A.C.M.)
| |
Collapse
|
333
|
Santos TA, Scorzoni L, Correia R, Junqueira JC, Anbinder AL. Interaction between Lactobacillus reuteri and periodontopathogenic bacteria using in vitro and in vivo (G. mellonella) approaches. Pathog Dis 2020; 78:5897357. [DOI: 10.1093/femspd/ftaa044] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 08/24/2020] [Indexed: 12/12/2022] Open
Abstract
ABSTRACT
Periodontitis is a multifactorial inflammatory disease, and the major cause of tooth loss in adults. New therapies have been proposed for its treatment, including the use of probiotics such as Lactobacillus reuteri. The objective of this study was to evaluate the antimicrobial effects of L. reuteri: live, heat-killed and culture filtrate (cell-free supernatant), on periodontopathogenic bacteria (Fusobacterium nucleatum and Aggregatibacter actinomycetemcomitans) in vitro, as well as the in vivo survival curve, hemocyte density and microbial recovery using Galleria mellonella. For in vitro assays, all preparations reduced colony forming units of F. nucleatum, while only live L. reuteri reduced the growth of A. actinomycetemcomitans. All treatments reduced periodontopathogenic bacteria growth in vivo. The treatment with the supernatant increased the survival of larvae infected with F. nucleatum more than the treatment with live L. reuteri, and none of the treatments altered the survival of A. actinomycetemcomitans-infected larvae. In addition, the treatment with L. reuteri preparations did not alter the hemocyte count of F. nucleatum- and A. actinomycetemcomitans-infected larvae. This study demonstrated that L. reuteri preparations exerted antimicrobial effects and increased the survival of G. mellonella infected by F. nucleatum, although only live L. reuteri was able to reduce the growth of A. actinomycetemcomitans in vitro.
Collapse
Affiliation(s)
- Thaís Aguiar Santos
- São Paulo State University (Unesp), Institute of Science and Technology, São José dos Campos, SP. Address: Av Engenheiro Francisco José Longo, 777, Jardim São Dimas, São José dos Campos, SP, Brazil. CEP: 12245-000
| | - Liliana Scorzoni
- São Paulo State University (Unesp), Institute of Science and Technology, São José dos Campos, SP. Address: Av Engenheiro Francisco José Longo, 777, Jardim São Dimas, São José dos Campos, SP, Brazil. CEP: 12245-000
| | - Raquel Correia
- São Paulo State University (Unesp), Institute of Science and Technology, São José dos Campos, SP. Address: Av Engenheiro Francisco José Longo, 777, Jardim São Dimas, São José dos Campos, SP, Brazil. CEP: 12245-000
| | - Juliana Campos Junqueira
- São Paulo State University (Unesp), Institute of Science and Technology, São José dos Campos, SP. Address: Av Engenheiro Francisco José Longo, 777, Jardim São Dimas, São José dos Campos, SP, Brazil. CEP: 12245-000
| | - Ana Lia Anbinder
- São Paulo State University (Unesp), Institute of Science and Technology, São José dos Campos, SP. Address: Av Engenheiro Francisco José Longo, 777, Jardim São Dimas, São José dos Campos, SP, Brazil. CEP: 12245-000
| |
Collapse
|
334
|
Stauber RH, Westmeier D, Wandrey M, Becker S, Docter D, Ding GB, Thines E, Knauer SK, Siemer S. Mechanisms of nanotoxicity - biomolecule coronas protect pathological fungi against nanoparticle-based eradication. Nanotoxicology 2020; 14:1157-1174. [PMID: 32835557 DOI: 10.1080/17435390.2020.1808251] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Whereas nanotoxicity is intensely studied in mammalian systems, our knowledge of desired or unwanted nano-based effects for microbes is still limited. Fungal infections are global socio-economic health and agricultural problems, and current chemical antifungals may induce adverse side-effects in humans and ecosystems. Thus, nanoparticles are discussed as potential novel and sustainable antifungals via the desired nanotoxicity but often fail in practical applications. In our study, we found that nanoparticles' toxicity strongly depends on their binding to fungal spores, including the clinically relevant pathogen Aspergillus fumigatus as well as common plant pests, such as Botrytis cinerea or Penicillum expansum. Employing a selection of the model and antimicrobial nanoparticles, we found that nanoparticle-spore complex formation is influenced by the NM's physicochemical properties, such as size, identified as a key determinant for our silica model particles. Biomolecule coronas acquired in pathophysiologically and ecologically relevant environments, protected fungi against nanoparticle-induced toxicity as shown by employing antimicrobial ZnO, Ag, or CuO nanoparticles as well as dissolution-resistant quantum dots. Mechanistically, dose-dependent corona-mediated resistance was conferred via reducing the physical adsorption of nanoparticles to fungi. The inhibitory effect of biomolecules on nano-based toxicity of Ag NPs was further verified in vivo, using the invertebrate Galleria mellonella as an alternative non-mammalian infection model. We provide the first evidence that biomolecule coronas are not only relevant in mammalian systems but also for nanomaterial designs as future antifungals for human health, biotechnology, and agriculture.
Collapse
Affiliation(s)
| | - Dana Westmeier
- ENT Department, University Medical Center Mainz, Mainz, Germany
| | - Madita Wandrey
- ENT Department, University Medical Center Mainz, Mainz, Germany
| | - Sven Becker
- ENT Department, University Medical Center Mainz, Mainz, Germany
| | - Dominic Docter
- ENT Department, University Medical Center Mainz, Mainz, Germany
| | - Guo-Bin Ding
- Institute for Biotechnology, Shanxi University, Shanxi, China
| | - Eckhard Thines
- Institute for Microbiology, Johannes Gutenberg University, Mainz, Germany
| | - Shirley K Knauer
- Department of Molecular Biology II, Centre for Medical Biotechnology (ZMB)/Center for Nanointegration (CENIDE), University Duisburg-Essen, Essen, Germany
| | - Svenja Siemer
- ENT Department, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
335
|
Seribelli AA, Cruz MF, Vilela FP, Frazão MR, Paziani MH, Almeida F, Medeiros MIC, Rodrigues DDP, Kress MRVZ, Allard MW, Falcão JP. Phenotypic and genotypic characterization of Salmonella Typhimurium isolates from humans and foods in Brazil. PLoS One 2020; 15:e0237886. [PMID: 32810191 PMCID: PMC7437471 DOI: 10.1371/journal.pone.0237886] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/04/2020] [Indexed: 01/25/2023] Open
Abstract
Salmonella enterica subsp. enterica serovar Typhimurium (S. Typhimurium) causes gastroenteritis in many countries. However, in Brazil there are few studies that have conducted a virulence characterization of this serovar. The aim of this study was to evaluate the virulence potential of S. Typhimurium strains isolated in Brazil. Forty S. Typhimurium strains isolated from humans (n = 20) and food (n = 20) from Brazil were studied regarding their invasion and survival in human epithelial cells (Caco-2) and macrophages (U937). Their virulence potential was determined using the Galleria mellonella larvae model combined with the analysis of virulence genes by whole genome sequencing (WGS). A total of 67.5% of the S. Typhimurium studied (32.5% isolated from humans and 35% isolated from food) invaded Caco-2 epithelial cells at levels similar to or greater than the S. Typhimurium SL1344 prototype strain. In addition, 37.5% of the studied strains (25% isolated from humans and 12.5% isolated from food) survived in U937 human macrophages at levels similar to or greater than SL1344. S. Typhimurium strains isolated from humans (40%) and food (25%) showed high or intermediate virulence in G. mellonella larvae after seven days exposure. Approximately, 153 virulence genes of chromosomal and plasmidial origin were detected in the strains studied. In conclusion, the ability of the S. Typhimurium to invade Caco-2 epithelial cells was strain dependent and was not related to the source or the year of isolation. However, S. Typhimurium strains isolated from humans showed greater survival rates in U937 human macrophages, and presented higher proportion of isolates with a virulent profile in G. mellonella in comparison to strains isolated from food suggesting that this difference may be related to the higher frequency of human isolates which contained plasmid genes, such as spvABCDR operon, pefABCD operon, rck and mig-5.
Collapse
Affiliation(s)
- Amanda Aparecida Seribelli
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto–Universidade de São Paulo—USP, Brazil
| | - Marcelo Ferreira Cruz
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto–Universidade de São Paulo—USP, Brazil
| | - Felipe Pinheiro Vilela
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto–Universidade de São Paulo—USP, Brazil
| | - Miliane Rodrigues Frazão
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto–Universidade de São Paulo—USP, Brazil
| | - Mario H. Paziani
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto–Universidade de São Paulo—USP, Brazil
| | - Fernanda Almeida
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto–Universidade de São Paulo—USP, Brazil
| | | | | | - Marcia R. von Zeska Kress
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto–Universidade de São Paulo—USP, Brazil
| | - Marc W. Allard
- Food and Drug Administration—FDA, College Park, Maryland, United States of America
- * E-mail: (JPF); (MWA)
| | - Juliana Pfrimer Falcão
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto–Universidade de São Paulo—USP, Brazil
- * E-mail: (JPF); (MWA)
| |
Collapse
|
336
|
Abstract
The antibiotic resistance crisis has led to renewed interest in phage therapy as an alternative means of treating infection. However, conventional methods for isolating pathogen-specific phage are slow, labor-intensive, and frequently unsuccessful. We have demonstrated that computationally identified prophages carried by near-neighbor bacteria can serve as starting material for production of engineered phages that kill the target pathogen. Our approach and technology platform offer new opportunity for rapid development of phage therapies against most, if not all, bacterial pathogens, a foundational advance for use of phage in treating infectious disease. New therapies are necessary to combat increasingly antibiotic-resistant bacterial pathogens. We have developed a technology platform of computational, molecular biology, and microbiology tools which together enable on-demand production of phages that target virtually any given bacterial isolate. Two complementary computational tools that identify and precisely map prophages and other integrative genetic elements in bacterial genomes are used to identify prophage-laden bacteria that are close relatives of the target strain. Phage genomes are engineered to disable lysogeny, through use of long amplicon PCR and Gibson assembly. Finally, the engineered phage genomes are introduced into host bacteria for phage production. As an initial demonstration, we used this approach to produce a phage cocktail against the opportunistic pathogen Pseudomonas aeruginosa PAO1. Two prophage-laden P. aeruginosa strains closely related to PAO1 were identified, ATCC 39324 and ATCC 27853. Deep sequencing revealed that mitomycin C treatment of these strains induced seven phages that grow on P. aeruginosa PAO1. The most diverse five phages were engineered for nonlysogeny by deleting the integrase gene (int), which is readily identifiable and typically conveniently located at one end of the prophage. The Δint phages, individually and in cocktails, killed P. aeruginosa PAO1 in liquid culture as well as in a waxworm (Galleria mellonella) model of infection. IMPORTANCE The antibiotic resistance crisis has led to renewed interest in phage therapy as an alternative means of treating infection. However, conventional methods for isolating pathogen-specific phage are slow, labor-intensive, and frequently unsuccessful. We have demonstrated that computationally identified prophages carried by near-neighbor bacteria can serve as starting material for production of engineered phages that kill the target pathogen. Our approach and technology platform offer new opportunity for rapid development of phage therapies against most, if not all, bacterial pathogens, a foundational advance for use of phage in treating infectious disease.
Collapse
|
337
|
Functional Analysis of a Fibronectin Binding Protein of Streptococcus parasanguinis FW213. Curr Microbiol 2020; 77:3430-3440. [PMID: 32761388 DOI: 10.1007/s00284-020-02152-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 07/28/2020] [Indexed: 10/23/2022]
Abstract
Streptococcus parasanguinis is a primary colonizer of dental plaque and an opportunistic pathogen for subacute endocarditis. A putative fibronectin binding protein (Spaf_1409) that lacks both an N-terminal signal peptide and a C-terminal cell wall-anchoring motif was identified from the S. parasanguinis FW213 genome. Spaf_1409 was abundantly present in the cytoplasm and also was found in the cell wall preparation and culture supernatant. By using an isogenic mutant strain, MPH4, Spaf_1409 was found to mediate the binding of S. parasanguinis FW213 to fibronectin. Inactivation of Spaf_1409 did not significantly alter the mass of static biofilm, but reduced the resistance of S. parasanguinis against the shearing force in a flow cell biofilm system, resulting in scattered biofilm. The mortality in Galleria mellonella larvae infected with MPH4 was higher than in those infected with wild-type S. parasanguinis. However, fewer viable bacterial cells were recovered from larvae infected with MPH4, compared to those infected with wild-type S. parasanguinis, up to 42 h post infection, suggesting that the infection by MPH4, but not the growth, was responsible for the elevated mortality. The phagocytic analysis using flow cytometry indicated that Spaf_1409 participates in the recognition of S. parasanguinis FW213 by RAW264.7 macrophages, suggesting that inactivation of Spaf_1409 intensified the immune responses in larvae, leading to larval death. Taken together, the data indicate that Spaf_1409 plays different roles in the development of dental biofilm and in systemic infections.
Collapse
|
338
|
Rossoni RD, de Barros PP, Mendonça IDC, Medina RP, Silva DHS, Fuchs BB, Junqueira JC, Mylonakis E. The Postbiotic Activity of Lactobacillus paracasei 28.4 Against Candida auris. Front Cell Infect Microbiol 2020; 10:397. [PMID: 32850495 PMCID: PMC7417517 DOI: 10.3389/fcimb.2020.00397] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/29/2020] [Indexed: 12/11/2022] Open
Abstract
Candida auris has emerged as a medically important pathogen with considerable resistance to antifungal agents. The ability to produce biofilms is an important pathogenicity feature of this species that aids escape of host immune responses and antimicrobial agents. The objective of this study was to verify antifungal action using in vitro and in vivo models of the Lactobacillus paracasei 28.4 probiotic cells and postbiotic activity of crude extract (LPCE) and fraction 1 (LPF1), derived from L. paracasei 28.4 supernatant. Both live cells and cells free supernatant of L. paracasei 28.4 inhibited C. auris suggesting probiotic and postbiotic effects. The minimum inhibitory concentration (MIC) for LPCE was 15 mg/mL and ranges from 3.75 to 7.5 mg/mL for LPF1. Killing kinetics determined that after 24 h treatment with LPCE or LPF1 there was a complete reduction of viable C. auris cells compared to fluconazole, which decreased the initial inoculum by 1-logCFU during the same time period. LPCE and LPF1 significantly reduced the biomass (p = 0.0001) and the metabolic activity (p = 0.0001) of C. auris biofilm. There was also a total reduction (~108 CFU/mL) in viability of persister C. auris cells after treatment with postbiotic elements (p < 0.0001). In an in vivo study, injection of LPCE and LPF1 into G. mellonella larvae infected with C. auris prolonged survival of these insects compared to a control group (p < 0.05) and elicited immune responses by increasing the number of circulating hemocytes and gene expression of antimicrobial peptide galiomicin. We concluded that the L. paracasei 28.4 cells and postbiotic elements (LPCE and LPF1) have antifungal activity against planktonic cells, biofilms, and persister cells of C. auris. Postbiotic supplementation derived from L. paracasei 28.4 protected G. mellonella infected with C. auris and enhanced its immune status indicating a dual function in modulating a host immune response.
Collapse
Affiliation(s)
- Rodnei Dennis Rossoni
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, São José dos Campos, Brazil.,Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School at Brown University, Providence, RI, United States
| | - Patrícia Pimentel de Barros
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, São José dos Campos, Brazil.,Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School at Brown University, Providence, RI, United States
| | - Iatã do Carmo Mendonça
- Department of Organic Chemistry, Center for Bioassays, Biosynthesis and Ecophysiology of Natural Products, Institute of Chemistry, São Paulo State University, UNESP, Araraquara, Brazil
| | - Rebeca Previate Medina
- Department of Organic Chemistry, Center for Bioassays, Biosynthesis and Ecophysiology of Natural Products, Institute of Chemistry, São Paulo State University, UNESP, Araraquara, Brazil
| | - Dulce Helena Siqueira Silva
- Department of Organic Chemistry, Center for Bioassays, Biosynthesis and Ecophysiology of Natural Products, Institute of Chemistry, São Paulo State University, UNESP, Araraquara, Brazil
| | - Beth Burgwyn Fuchs
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School at Brown University, Providence, RI, United States
| | - Juliana Campos Junqueira
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, São José dos Campos, Brazil
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School at Brown University, Providence, RI, United States
| |
Collapse
|
339
|
Weidensdorfer M, Ishikawa M, Hori K, Linke D, Djahanschiri B, Iruegas R, Ebersberger I, Riedel-Christ S, Enders G, Leukert L, Kraiczy P, Rothweiler F, Cinatl J, Berger J, Hipp K, Kempf VAJ, Göttig S. The Acinetobacter trimeric autotransporter adhesin Ata controls key virulence traits of Acinetobacter baumannii. Virulence 2020; 10:68-81. [PMID: 31874074 PMCID: PMC6363060 DOI: 10.1080/21505594.2018.1558693] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Acinetobacter baumannii is a Gram-negative pathogen that causes a multitude of nosocomial infections. The Acinetobacter trimeric autotransporter adhesin (Ata) belongs to the superfamily of trimeric autotransporter adhesins which are important virulence factors in many Gram-negative species. Phylogenetic profiling revealed that ata is present in 78% of all sequenced A. baumannii isolates but only in 2% of the closely related species A. calcoaceticus and A. pittii. Employing a markerless ata deletion mutant of A. baumannii ATCC 19606 we show that adhesion to and invasion into human endothelial and epithelial cells depend on Ata. Infection of primary human umbilical cord vein endothelial cells (HUVECs) with A. baumannii led to the secretion of interleukin (IL)-6 and IL-8 in a time- and Ata-dependent manner. Furthermore, infection of HUVECs by WT A. baumannii was associated with higher rates of apoptosis via activation of caspases-3 and caspase-7, but not necrosis, in comparison to ∆ata. Ata deletion mutants were furthermore attenuated in their ability to kill larvae of Galleria mellonella and to survive in larvae when injected at sublethal doses. This indicates that Ata is an important multifunctional virulence factor in A. baumannii that mediates adhesion and invasion, induces apoptosis and contributes to pathogenicity in vivo.
Collapse
Affiliation(s)
- Marko Weidensdorfer
- Institute for Medical Microbiology and Infection Control, University Hospital, Goethe University, Frankfurt, Germany
| | - Masahito Ishikawa
- Department of Biotechnology, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Katsutoshi Hori
- Department of Biotechnology, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Dirk Linke
- Department of Biosciences, Section for Genetics and Evolutionary Biology, University of Oslo, Oslo, Norway
| | - Bardya Djahanschiri
- Department for Applied Bioinformatics, Institute of Cell Biology and Neuroscience, Goethe University, Frankfurt, Germany
| | - Ruben Iruegas
- Department for Applied Bioinformatics, Institute of Cell Biology and Neuroscience, Goethe University, Frankfurt, Germany
| | - Ingo Ebersberger
- Department for Applied Bioinformatics, Institute of Cell Biology and Neuroscience, Goethe University, Frankfurt, Germany.,Senckenberg Biodiversity and Climate Research Centre Frankfurt (BIK-F), Frankfurt, Germany
| | - Sara Riedel-Christ
- Institute for Medical Microbiology and Infection Control, University Hospital, Goethe University, Frankfurt, Germany
| | - Giulia Enders
- Institute for Medical Microbiology and Infection Control, University Hospital, Goethe University, Frankfurt, Germany
| | - Laura Leukert
- Institute for Medical Microbiology and Infection Control, University Hospital, Goethe University, Frankfurt, Germany
| | - Peter Kraiczy
- Institute for Medical Microbiology and Infection Control, University Hospital, Goethe University, Frankfurt, Germany
| | - Florian Rothweiler
- Institute of Medical Virology, University Hospital, Goethe University, Frankfurt, Germany
| | - Jindrich Cinatl
- Institute of Medical Virology, University Hospital, Goethe University, Frankfurt, Germany
| | - Jürgen Berger
- Electron Microscopy Facility, Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Katharina Hipp
- Electron Microscopy Facility, Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Volkhard A J Kempf
- Institute for Medical Microbiology and Infection Control, University Hospital, Goethe University, Frankfurt, Germany
| | - Stephan Göttig
- Institute for Medical Microbiology and Infection Control, University Hospital, Goethe University, Frankfurt, Germany
| |
Collapse
|
340
|
Thieme L, Hartung A, Makarewicz O, Pletz MW. In vivo synergism of ampicillin, gentamicin, ceftaroline and ceftriaxone against Enterococcus faecalis assessed in the Galleria mellonella infection model. J Antimicrob Chemother 2020; 75:2173-2181. [PMID: 32357212 DOI: 10.1093/jac/dkaa129] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/25/2020] [Accepted: 03/11/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The unfavourable safety profile of aminoglycosides and the synergistic effects observed in vitro have prompted the development of novel dual β-lactam therapies, e.g. ampicillin/ceftriaxone or ampicillin/ceftaroline, for the treatment of Enterococcus faecalis endocarditis. OBJECTIVES For comparison with in vitro chequerboard assay results, a partial chequerboard setup of ampicillin/gentamicin, ampicillin/ceftriaxone and ampicillin/ceftaroline against E. faecalis was established in the Galleria mellonella larval infection model. METHODS Discrimination of synergistic and additive interactions was based on the evaluation of larval survival, bacterial quantity in the haemolymph and a pathology score index (internal to the workgroup). Single and multiple dosing schemes based on the half-life of ampicillin were applied. Pharmacokinetic data of the antibiotics in the larvae were determined via agar plate diffusion assays. RESULTS Ampicillin and ceftriaxone exhibited strain-specific synergistic interactions in the larvae under both dosing regimens, while the other two combinations showed additive effects. Ampicillin/ceftaroline was inferior to ampicillin/ ceftriaxone. Not all synergistic effects observed in vitro could be replicated in the larvae. CONCLUSIONS Our results suggest superior efficacy of ampicillin/ceftriaxone for the treatment of high-inoculum enterococcal infections, for at least some strains, but question the benefit of the current standard of adding the nephrotoxic gentamicin compared with the safer ceftriaxone. This is the first study to develop a scheme for differentiation between additive and synergistic effects in larvae and apply a multiple-antibiotic dosing scheme based on the pharmacokinetics of ampicillin. The model allows the analysis of synergistic effects of antimicrobials in an in vivo setting, but the clinical correlation warrants further study.
Collapse
Affiliation(s)
- Lara Thieme
- Institute for Infectious Diseases and Infection Control, Jena University Hospital, Jena, Germany
| | - Anita Hartung
- Institute for Infectious Diseases and Infection Control, Jena University Hospital, Jena, Germany
| | - Oliwia Makarewicz
- Institute for Infectious Diseases and Infection Control, Jena University Hospital, Jena, Germany
| | - Mathias W Pletz
- Institute for Infectious Diseases and Infection Control, Jena University Hospital, Jena, Germany
| |
Collapse
|
341
|
Wintachai P, Naknaen A, Thammaphet J, Pomwised R, Phaonakrop N, Roytrakul S, Smith DR. Characterization of extended-spectrum-β-lactamase producing Klebsiella pneumoniae phage KP1801 and evaluation of therapeutic efficacy in vitro and in vivo. Sci Rep 2020; 10:11803. [PMID: 32678251 PMCID: PMC7367294 DOI: 10.1038/s41598-020-68702-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 06/30/2020] [Indexed: 12/13/2022] Open
Abstract
Extended spectrum β lactamase-producing Klebsiella pneumoniae (ESBL-KP) is being reported with high morbidity and mortality rates and is considered as the highest priority for new antimicrobial strategies. To develop an alternative antimicrobial agent, phage KP1801 with broad lytic activity was isolated. The genome of phage KP1801 was double stranded DNA of 49,835 base pairs, with a GC content of 50.26%. There were 75 putative open reading frames. Phage KP1801 was classified as being in the order Caudovirales, belonging to the Siphoviridae family. About 323 proteins were detected by shotgun proteome analysis. The phage inhibited biofilm formation and reduced pre-formed biofilm in a dose dependent manner. Scanning electron microscopic studies demonstrated a membrane damage of bacterial cells treated with phage, resulting in cell death. Prophylactic and therapeutic efficacies of the phage were evaluated in Galleria mellonella. Administration of ESBL-KP infection with phage significantly improved the survival of G. mellonella. The number of intracellular bacteria in larvae showed a significant decrease compared with untreated control while the number of phage increased. These studies suggested that phage KP1801 has the potential for development as an alternative for antibiotics and biocontrol agents against ESBL-KP infection.
Collapse
Affiliation(s)
| | - Ampapan Naknaen
- Department of Microbiology, Prince of Songkla University, Songkhla, 90112, Thailand
| | - Jirapath Thammaphet
- School of Science, Walailak University, Nakhon Si Thammarat, 80161, Thailand
| | - Rattanaruji Pomwised
- Department of Microbiology, Prince of Songkla University, Songkhla, 90112, Thailand
| | - Narumon Phaonakrop
- Proteomics Research Laboratory, Thailand Science Park, Pathum Thani, 12120, Thailand
| | - Sittiruk Roytrakul
- Proteomics Research Laboratory, Thailand Science Park, Pathum Thani, 12120, Thailand
| | - Duncan R Smith
- Institute of Molecular Biosciences, Mahidol University, Bangkok, 73170, Thailand
| |
Collapse
|
342
|
Cools F, Triki D, Geerts N, Delputte P, Fourches D, Cos P. In vitro and in vivo Evaluation of in silico Predicted Pneumococcal UDPG:PP Inhibitors. Front Microbiol 2020; 11:1596. [PMID: 32760374 PMCID: PMC7373766 DOI: 10.3389/fmicb.2020.01596] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/18/2020] [Indexed: 11/25/2022] Open
Abstract
Pneumonia, of which Streptococcus pneumoniae is the most common causative agent, is considered one of the three top leading causes of death worldwide. As seen in other bacterial species, antimicrobial resistance is on the rise for this pathogen. Therefore, there is a pressing need for novel antimicrobial strategies to combat these infections. Recently, uridine diphosphate glucose pyrophosphorylase (UDPG:PP) has been put forward as a potential drug target worth investigating. Moreover, earlier research demonstrated that streptococci lacking a functional galU gene (encoding for UDPG:PP) were characterized by significantly reduced in vitro and in vivo virulence. Therefore, in this study we evaluated the anti-virulence activity of potential UDPG:PP inhibitors. They were selected in silico using a tailor-made streptococcal homology model, based on earlier listerial research. While the compounds didn’t affect bacterial growth, nor affected in vitro adhesion to and phagocytosis in macrophages, the amount of polysaccharide capsule was significantly reduced after co-incubation with these inhibitors. Moreover, co-incubation proved to have a positive effect on survival in an in vivo Galleria mellonella larval infection model. Therefore, rather than targeting bacterial survival directly, these compounds proved to have an effect on streptococcal virulence by lowering the amount of polysaccharide and thereby probably boosting recognition of this pathogen by the innate immune system. While the compounds need adaptation to broaden their activity to more streptococcal strains rather than being strain-specific, this study consolidates UDPG:PP as a potential novel drug target.
Collapse
Affiliation(s)
- Freya Cools
- Department of Pharmaceutical Sciences, Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Dhoha Triki
- Department of Chemistry, Bioinformatics Research Center, North Carolina State University, Raleigh, NC, United States
| | - Nele Geerts
- Department of Pharmaceutical Sciences, Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Peter Delputte
- Department of Pharmaceutical Sciences, Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Denis Fourches
- Department of Chemistry, Bioinformatics Research Center, North Carolina State University, Raleigh, NC, United States
| | - Paul Cos
- Department of Pharmaceutical Sciences, Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
343
|
Kathayat D, Antony L, Deblais L, Helmy YA, Scaria J, Rajashekara G. Small Molecule Adjuvants Potentiate Colistin Activity and Attenuate Resistance Development in Escherichia coli by Affecting pmrAB System. Infect Drug Resist 2020; 13:2205-2222. [PMID: 32764996 PMCID: PMC7360418 DOI: 10.2147/idr.s260766] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/16/2020] [Indexed: 12/22/2022] Open
Abstract
Background Colistin is one of the last-resort antibiotics to treat multi-drug resistant (MDR) Gram-negative bacterial infections in humans. Further, colistin has been also used to prevent and treat Enterobacteriaceae infections in food animals. However, chromosomal mutations and mobile colistin resistance (mcr) genes, which confer resistance to colistin, have been detected in bacterial isolates from food animals and humans worldwide; thus, limiting the use of colistin. Therefore, strategies that could aid in ameliorating colistin resistance are critically needed. Objective Investigate the adjuvant potential of novel small molecules (SMs) on colistin. Materials and Methods Previously, we identified 11 membrane-affecting SMs with bactericidal activity against avian pathogenic Escherichia coli (APEC). Here, we investigated the potentiation effect of those SMs on colistin using checkerboard assays and wax moth (Galleria mellonella) larval model. The impact of the SM combination on colistin resistance evolution was also investigated by analyzing whole genome sequences of APEC isolates passaged with colistin alone or in combination with SMs followed by quantitating pmrCAB and pmrH expression in those isolates. Results The SM combination synergistically reduced the minimum bactericidal concentration of colistin by at least 10-fold. In larvae, the SM combination increased the efficacy of colistin by two-fold with enhanced (>50%) survival and reduced (>4 logs) APEC load. Further, the SM combination decreased the frequency (5/6 to 1/6) of colistin resistance evolution and downregulated the pmrCAB and pmrH expression. Previously unknown mutations in pmrB (L14Q, T92P) and pmrA (A80V), which were predicted deleterious, were identified in the colistin-resistant (ColR) APEC isolates when passaged with colistin alone but not in combination with SMs. Our study also identified mutations in hypothetical and several phage-related proteins in ColR APEC isolates in concurrent with pmrAB mutations. Conclusion Our study identified two SMs (SM2 and SM3) that potentiated the colistin activity and attenuated the development of colistin resistance in APEC. These SMs can be developed as anti-evolution drugs that can slow down colistin resistance development.
Collapse
Affiliation(s)
- Dipak Kathayat
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH 44691, USA
| | - Linto Antony
- Animal Disease Research and Diagnostic Laboratory, Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD 57007, USA
| | - Loic Deblais
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH 44691, USA
| | - Yosra A Helmy
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH 44691, USA
| | - Joy Scaria
- Animal Disease Research and Diagnostic Laboratory, Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD 57007, USA
| | - Gireesh Rajashekara
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH 44691, USA
| |
Collapse
|
344
|
Fuentes-Castillo D, Navas-Suárez PE, Gondim MF, Esposito F, Sacristán C, Fontana H, Fuga B, Piovani C, Kooij R, Lincopan N, Catão-Dias JL. Genomic characterization of multidrug-resistant ESBL-producing Escherichia coli ST58 causing fatal colibacillosis in critically endangered Brazilian merganser (Mergus octosetaceus). Transbound Emerg Dis 2020; 68:258-266. [PMID: 32544292 PMCID: PMC8246901 DOI: 10.1111/tbed.13686] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/03/2020] [Accepted: 06/08/2020] [Indexed: 12/12/2022]
Abstract
Even though antimicrobial‐resistant bacteria have begun to be detected in wildlife, raising important issues related to their transmission and persistence of clinically important pathogens in the environment, little is known about the role of these bacteria on wildlife health, especially on endangered species. The Brazilian merganser (Mergus octosetaceus) is one of the most threatened waterfowl in the world, classified as Critically Endangered by the International Union for Conservation of Nature. In 2019, a fatal case of sepsis was diagnosed in an 8‐day‐old Brazilian merganser inhabiting a zoological park. At necropsy, major gross lesions were pulmonary and hepatic congestion. Using microbiologic and genomic methods, we identified a multidrug‐resistant (MDR) extended‐spectrum β‐lactamase (ESBL) CTX‐M‐8‐producing Escherichia coli (designed as PMPU strain) belonging to the international clone ST58, in coelomic cavity, oesophagus, lungs, small intestine and cloaca samples. PMPU strain harboured a broad resistome against antibiotics (cephalosporins, tetracyclines, aminoglycosides, sulphonamides, trimethoprim and quinolones), domestic/hospital disinfectants and heavy metals (arsenic, mercury, lead, copper and silver). Additionally, the virulence of E. coli PMPU strain was confirmed using a wax moth (Galleria mellonella) infection model, and it was supported by the presence of virulence genes encoding toxins, adherence factors, invasins and iron acquisition systems. Broad resistome and virulome of PMPU contributed to therapeutic failure and death of the animal. In brief, we report for the first time a fatal colibacillosis by MDR ESBL‐producing E. coli in critically endangered Brazilian merganser, highlighting that besides colonization, critical priority pathogens are threatening wildlife. E. coli ST58 clone has been previously reported in humans, food‐producing animals, wildlife and environment, supporting broad adaptation and persistence at human–animal–environment interface.
Collapse
Affiliation(s)
- Danny Fuentes-Castillo
- Department of Pathology, School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, Brazil.,One Health Brazilian Resistance Project (OneBR), São Paulo, Brazil
| | - Pedro Enrique Navas-Suárez
- Department of Pathology, School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Fernanda Esposito
- One Health Brazilian Resistance Project (OneBR), São Paulo, Brazil.,Department of Clinical Analysis, Faculty of Pharmacy, University of São Paulo, São Paulo, Brazil
| | - Carlos Sacristán
- Department of Pathology, School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, Brazil
| | - Herrison Fontana
- One Health Brazilian Resistance Project (OneBR), São Paulo, Brazil.,Department of Clinical Analysis, Faculty of Pharmacy, University of São Paulo, São Paulo, Brazil
| | - Bruna Fuga
- One Health Brazilian Resistance Project (OneBR), São Paulo, Brazil.,Department of Clinical Analysis, Faculty of Pharmacy, University of São Paulo, São Paulo, Brazil.,Department of Microbiology, Instituto de Ciências Biomédicas, University of São Paulo, São Paulo, Brazil
| | | | | | - Nilton Lincopan
- One Health Brazilian Resistance Project (OneBR), São Paulo, Brazil.,Department of Clinical Analysis, Faculty of Pharmacy, University of São Paulo, São Paulo, Brazil.,Department of Microbiology, Instituto de Ciências Biomédicas, University of São Paulo, São Paulo, Brazil
| | - José Luiz Catão-Dias
- Department of Pathology, School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
345
|
Yu X, Zheng B, Xiao F, Jin Y, Guo L, Xu H, Luo Q, Xiao Y. Effect of Short-Term Antimicrobial Therapy on the Tolerance and Antibiotic Resistance of Multidrug-Resistant Staphylococcus capitis. Infect Drug Resist 2020; 13:2017-2026. [PMID: 32636655 PMCID: PMC7335296 DOI: 10.2147/idr.s254141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/26/2020] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Bacteria undergo adaptive mutation in the host. However, the specific effect of antimicrobial use on bacterial evolution and genome mutations related to bacterial survival within a patient is unclear. MATERIALS AND METHODS Three S. capitis strains were sequentially isolated from cerebrospinal fluid of a clinical inpatient. Antimicrobial susceptibility, growth rate, biofilm formation and whole blood survival of these strains were measured. Relative fitness was calculated. The virulence was examined in the Galleria mellonella model. Whole-genome sequencing and in silico analysis were performed to explore the genetic mechanisms of the changes in antimicrobial resistance phenotype. Hypothetical proteins are cloned, expressed and characterized by detection the susceptibility to gentamycin. RESULTS The first isolate was susceptible to rifampin (MIC=0.25 μg/mL), resistant to gentamicin (MIC=16 μg/mL), while the later two isolates were resistant to rifampin (MIC >64 μg/mL), susceptible to gentamicin (MIC=4 μg/mL). For the latter two strains, compared to the first, frameshift mutation in a hypothetical protein encoding gene and base substitutions (in genes saeR, moaA and rpoB) were discovered. The mutation of rpoB gene caused rifampicin resistance. Mutations in saeR, moaA and hypothetical gene are associated with changes in other biological traits. Amino acid sequence-based structure and function identification of the hypothetical protein indicated that a mutation in the encoding gene might be associated with altered aminoglycoside susceptibility. Growth curve showed that the later two isolates grew faster than the first isolate with a positive fitness advantage of 13.5%, and 14.8%, accordingly. Biofilm form ability and whole blood survival of the derivative mutants were also enhanced. No significant differences of virulence in the G. mellonella model were observed. CONCLUSION We report here for the first time that short-term clinical antibiotic use was associated with resistance mutations, collateral sensitivity, and positive in vivo fitness advantages to S. capitis during infection.
Collapse
Affiliation(s)
- Xiao Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Beiwen Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Feng Xiao
- Neurosurgery Department, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Ye Jin
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Lihua Guo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Hao Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Qixia Luo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Yonghong Xiao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| |
Collapse
|
346
|
Fang R, Sun Y, Dai W, Zheng X, Tian X, Zhang X, Wang C, Cao J, Zhou T. Mutations in the MepRAB efflux system contribute to the in vitro development of tigecycline resistance in Staphylococcus aureus. J Glob Antimicrob Resist 2020; 22:631-636. [PMID: 32590185 DOI: 10.1016/j.jgar.2020.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 03/24/2020] [Accepted: 06/01/2020] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE To characterize the evolutionary pathways of tigecycline (TGC) resistance and alterations in the biological characteristics of hospital-derived Staphylococcus aureus isolates under selective pressure. METHODS Three clinical S. aureus strains and one standard S. aureus strain, ATCC 29213, were used for the in vitro selection of TGC-resistant S. aureus variants with gradient concentrations of TGC. Changes in drug resistance and genetic alterations in resistance-related genes (operon mepRAB and rpsJ) in mutant strains were determined. The efflux inhibitor assay for MepA and the fitness cost, determined by comparing the growth and virulence of parental and mutant strains, were also investigated. RESULTS Mutants induced in vitro showed a 64- to 128-fold increase in the minimum inhibitory concentration (MIC) of TGC. Substitution mutations were detected in the transcriptional repressor mepR and the efflux pump gene mepA. A K57M amino acid substitution occurred in the ribosomal S10 protein-encoding gene rpsJ. The MICs of TGC in the final mutants were significantly decreased in the presence of efflux pump inhibitors. It was worth noting that growth was unaffected by TGC resistance selection in vitro, with the exception of one strain, and the MICs of other antibiotics and virulence were also unaffected. CONCLUSIONS The evolution of TGC resistance in S. aureus in vitro is associated with a loss-of-function mutation in the efflux pump transcriptional repressor mepR and a missense mutation in the efflux pump-encoding gene mepA. Our work further validated the resistance mechanisms of S. aureus to TGC and reported previously undiscovered mutations.
Collapse
Affiliation(s)
- Renchi Fang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Yao Sun
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Weisi Dai
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Xiangkuo Zheng
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xuebin Tian
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiucai Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Chong Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Jianming Cao
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Tieli Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
347
|
Peters DL, McCutcheon JG, Dennis JJ. Characterization of Novel Broad-Host-Range Bacteriophage DLP3 Specific to Stenotrophomonas maltophilia as a Potential Therapeutic Agent. Front Microbiol 2020; 11:1358. [PMID: 32670234 PMCID: PMC7326821 DOI: 10.3389/fmicb.2020.01358] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 05/27/2020] [Indexed: 01/04/2023] Open
Abstract
A novel Siphoviridae phage specific to the bacterial species Stenotrophomonas maltophilia was isolated from a pristine soil sample and characterized as a second member of the newly established Delepquintavirus genus. Phage DLP3 possesses one of the broadest host ranges of any S. maltophilia phage yet characterized, infecting 22 of 29 S. maltophilia strains. DLP3 has a genome size of 96,852 bp and a G+C content of 58.4%, which is significantly lower than S. maltophilia host strain D1571 (G+C content of 66.9%). The DLP3 genome encodes 153 coding domain sequences covering 95% of the genome, including five tRNA genes with different specificities. The DLP3 lysogen exhibits a growth rate increase during the exponential phase of growth as compared to the wild type strain. DLP3 also encodes a functional erythromycin resistance protein, causing lysogenic conversion of the host D1571 strain. Although a temperate phage, DLP3 demonstrates excellent therapeutic potential because it exhibits a broad host range, infects host cells through the S. maltophilia type IV pilus, and exhibits lytic activity in vivo. Undesirable traits, such as its temperate lifecycle, can be eliminated using genetic techniques to produce a modified phage useful in the treatment of S. maltophilia bacterial infections.
Collapse
Affiliation(s)
- Danielle L Peters
- Department of Biological Sciences, Faculty of Science, University of Alberta, Edmonton, AB, Canada
| | - Jaclyn G McCutcheon
- Department of Biological Sciences, Faculty of Science, University of Alberta, Edmonton, AB, Canada
| | - Jonathan J Dennis
- Department of Biological Sciences, Faculty of Science, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
348
|
Antibiotics Act with vB_AbaP_AGC01 Phage against Acinetobacter baumannii in Human Heat-Inactivated Plasma Blood and Galleria mellonella Models. Int J Mol Sci 2020; 21:ijms21124390. [PMID: 32575645 PMCID: PMC7352404 DOI: 10.3390/ijms21124390] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/11/2020] [Accepted: 06/19/2020] [Indexed: 12/16/2022] Open
Abstract
Increasing multidrug resistance has led to renewed interest in phage-based therapy. A combination of the bacteriophages and antibiotics presents a promising approach enhancing the phage therapy effectiveness. First, phage candidates for therapy should be deeply characterized. Here we characterize the bacteriophage vB_AbaP_AGC01 that poses antibacterial activity against clinical Acinetobacter baumannii strains. Moreover, besides genomic and phenotypic analysis our study aims to analyze phage–antibiotic combination effectiveness with the use of ex vivo and in vivo models. The phage AGC01 efficiently adsorbs to A. baumannii cells and possesses a bacteriolytic lifecycle resulting in high production of progeny phages (317 ± 20 PFU × cell−1). The broad host range (50.27%, 93 out of 185 strains) against A. baumannii isolates and the inability of AGC01 to infect other bacterial species show its high specificity. Genomic analysis revealed a high similarity of the AGC01 genome sequence with that of the Friunavirus genus from a subfamily of Autographivirinae. The AGC01 is able to significantly reduce the A. baumannii cell count in a human heat-inactivated plasma blood model (HIP-B), both alone and in combination with antibiotics (gentamicin (GEN), ciprofloxacin (CIP), and meropenem (MER)). The synergistic action was observed when a combination of phage treatment with CIP or MER was used. The antimicrobial activity of AGC01 and phage-antibiotic combinations was confirmed using an in vivo larva model. This study shows the greatest increase in survival of G. mellonella larvae when the combination of phage (MOI = 1) and MER was used, which increased larval survival from 35% to 77%. Hence, AGC01 represents a novel candidate for phage therapy. Additionally, our study suggests that phages and antibiotics can act synergistically for greater antimicrobial effect when used as combination therapy.
Collapse
|
349
|
Moman R, O'Neill CA, Ledder RG, Cheesapcharoen T, McBain AJ. Mitigation of the Toxic Effects of Periodontal Pathogens by Candidate Probiotics in Oral Keratinocytes, and in an Invertebrate Model. Front Microbiol 2020; 11:999. [PMID: 32612578 PMCID: PMC7308727 DOI: 10.3389/fmicb.2020.00999] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/24/2020] [Indexed: 12/23/2022] Open
Abstract
The larvae of the wax moth Galleria mellonella and human oral keratinocytes were used to investigate the protective activity of the candidate oral probiotics Lactobacillus rhamnosus GG (LHR), Lactobacillus reuteri (LR), and Streptococcus salivarius K-12 (SS) against the periodontal pathogens Fusobacterium nucleatum (FN), Porphyromonas gingivalis (PG), and Aggregatibacter actinomycetemcomitans (AA). Probiotics were delivered to the larvae (i) concomitantly with the pathogen in the same larval pro-leg; (ii) concomitantly with the pathogen in different pro-legs, and (iii) before inoculation with the pathogen in different pro-legs. Probiotics were delivered as viable cells, cell lysates or cell supernatants to the oral keratinocytes concomitantly with the pathogen. The periodontal pathogens killed at least 50% of larvae within 24 h although PG and FN were significantly more virulent than AA in the order FN > PG > AA and were also significantly lethal to mammalian cells. The candidate probiotics, however, were not lethal to the larvae or human oral keratinocytes at doses up to 107 cells/larvae. Wax worm survival rates increased up to 60% for some probiotic/pathogen combinations compared with control larvae inoculated with pathogens only. SS was the most effective probiotic against FN challenge and LHR the least, in simultaneous administration and pre-treatment, SS and LR were generally the most protective against all pathogens (up to 60% survival). For P. gingivalis, LR > LHR > SS, and for A. actinomycetemcomitans SS > LHR and LR. Administering the candidate probiotics to human oral keratinocytes significantly decreased the toxic effects of the periodontal pathogens. In summary, the periodontal pathogens were variably lethal to G. mellonella and human oral keratinocytes and the candidate probiotics had measurable protective effects, which were greatest when administrated simultaneously with the periodontal pathogens, suggesting protective effects based on bacterial interaction, and providing a basis for mechanistic studies.
Collapse
Affiliation(s)
- Raja Moman
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Tripoli, Tripoli, Libya
| | - Catherine A O'Neill
- Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
| | - Ruth G Ledder
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Tanaporn Cheesapcharoen
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Andrew J McBain
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
350
|
Vera‐González N, Bailey‐Hytholt CM, Langlois L, Camargo Ribeiro F, Souza Santos EL, Junqueira JC, Shukla A. Anidulafungin liposome nanoparticles exhibit antifungal activity against planktonic and biofilm
Candida albicans. J Biomed Mater Res A 2020; 108:2263-2276. [DOI: 10.1002/jbm.a.36984] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 03/27/2020] [Accepted: 04/04/2020] [Indexed: 01/29/2023]
Affiliation(s)
- Noel Vera‐González
- School of Engineering, Center for Biomedical Engineering, Institute for Molecular and Nanoscale Innovation, Brown University Providence Rhode Island USA
| | - Christina M. Bailey‐Hytholt
- School of Engineering, Center for Biomedical Engineering, Institute for Molecular and Nanoscale Innovation, Brown University Providence Rhode Island USA
| | - Luc Langlois
- Department of Chemistry Brown University Providence Rhode Island USA
| | - Felipe Camargo Ribeiro
- Institute of Science and Technology, São Paulo State University (UNESP) São Paulo Brazil
| | | | | | - Anita Shukla
- School of Engineering, Center for Biomedical Engineering, Institute for Molecular and Nanoscale Innovation, Brown University Providence Rhode Island USA
| |
Collapse
|