201
|
Alves MM, Batista C, Mil-Homens D, Grenho L, Fernandes MH, Santos CF. Enhanced antibacterial activity of Rosehip extract-functionalized Mg(OH) 2 nanoparticles: An in vitro and in vivo study. Colloids Surf B Biointerfaces 2022; 217:112643. [PMID: 35759895 DOI: 10.1016/j.colsurfb.2022.112643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/07/2022] [Accepted: 06/13/2022] [Indexed: 10/18/2022]
Abstract
The development of nanoparticles as antimicrobial agents against pathogenic bacteria has emerged as one of the leading global healthcare challenges. In this study, Mg(OH)2 NPs with controlled morphology and nanometric size, using two distinct counterions, chloride or nitrate, have been synthesized using Rosehip (RH) extract that has privileges beyond conventional chemical and physical methods. Various physicochemical techniques were used to characterize the RH-functionalized Mg-based NPs. They exhibited a spherical shape with a diameter of ~10 nm, low crystallinity compared to non-functionalized NPs, high polyphenol content, and negative zeta potential in three different media (H2O, TSB, and cell medium). The resulting RH-functionalized Mg-based NPs also exhibited an increased antibacterial activity against Gram-positive (S. Epidermis and S. aureus) and Gram-negative (E. Coli) bacteria compared to those prepared in pure water (0 % RH), an effect that was well evident with low NPs contents (250 μg/mL). A preliminary attempt to elucidate their mechanism of action revealed that RH-functionalized Mg-based NPs could disrupt cellular structures (bacterial cell wall and cytoplasmic membrane) and damage the bacterial cell, as confirmed by TEM imaging. Noteworthy is that Mg-based NPs exhibited higher toxicity to bacteria than to eukaryotic cells. More significantly, was their enhanced in vivo efficacy in a Galleria mellonella invertebrate animal model, when infected with S. aureus bacteria. Overall, our findings indicate that well-engineered Rosehip magnesium-based nanoparticles can be used as a green non-cytotoxic polyphenolic source in different antibacterial applications for the biomedical industry.
Collapse
Affiliation(s)
- Marta M Alves
- Centro Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, Lisboa 1049-001, Portugal
| | - Catarina Batista
- EST Setúbal, CDP2T, Instituto Politécnico de Setúbal, Campus IPS, Setúbal 2910, Portugal
| | - Dalila Mil-Homens
- iBB - Institute for Bioengineering and Biosciences and i4HB, Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, Lisboa 1049-001, Portugal
| | - Liliana Grenho
- Faculdade de Medicina Dentária, Laboratory for Bone Metabolism and Regeneration, Universidade do Porto, Porto 4200-393, Portugal; LAQV/REQUIMTE, U. Porto, Porto 4160-007, Portugal
| | - Maria H Fernandes
- Faculdade de Medicina Dentária, Laboratory for Bone Metabolism and Regeneration, Universidade do Porto, Porto 4200-393, Portugal; LAQV/REQUIMTE, U. Porto, Porto 4160-007, Portugal.
| | - Catarina F Santos
- Centro Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, Lisboa 1049-001, Portugal; EST Setúbal, CDP2T, Instituto Politécnico de Setúbal, Campus IPS, Setúbal 2910, Portugal.
| |
Collapse
|
202
|
Insecticidal features displayed by the beneficial rhizobacterium Pseudomonas chlororaphis PCL1606. Int Microbiol 2022; 25:679-689. [PMID: 35670867 PMCID: PMC9526686 DOI: 10.1007/s10123-022-00253-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 05/19/2022] [Accepted: 05/28/2022] [Indexed: 10/28/2022]
Abstract
The biocontrol rhizobacterium Pseudomonas chlororaphis is one of the bacterial species of the P. fluorescens group where insecticide fit genes have been found. Fit toxin, supported with other antimicrobial compounds, gives the bacterial the ability to repel and to fight against eukaryotic organisms, such as nematodes and insect larvae, thus protecting the plant host and itself. Pseudomonas chlororaphis PCL1606 is an antagonistic rhizobacterium isolated from avocado roots and show efficient biocontrol against fungal soil-borne disease. The main antimicrobial compound produced by P. chlororaphis PCL606 is 2-hexyl-5-propyl resorcinol (HPR), which plays a crucial role in effective biocontrol against fungal pathogens. Further analysis of the P. chlororaphis PCL1606 genome showed the presence of hydrogen cyanide (HCN), pyrrolnitrin (PRN), and homologous fit genes. To test the insecticidal activity and to determine the bases for such activity, single and double mutants on the biosynthetic genes of these four compounds were tested in a Galleria mellonella larval model using inoculation by injection. The results revealed that Fit toxin and HPR in combination are involved in the insecticide phenotype of P. chlororaphis PCL1606, and additional compounds such as HCN and PRN could be considered supporting compounds.
Collapse
|
203
|
Methods for Using the Galleria mellonella Invertebrate Model to Probe Enterococcus faecalis Pathogenicity. Methods Mol Biol 2022; 2427:177-183. [PMID: 35619034 DOI: 10.1007/978-1-0716-1971-1_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The Enterococci, mainly Enterococcus faecalis and E. faecium, are ubiquitous members of the human gastrointestinal tract consortia but also a leading cause of opportunistic infections. The global rise in human-associated enterococcal infections, often caused by multidrug resistant strains, highlights an urgent need to identify the bacterial factors contributing to its pathogenicity such that new therapies can be devised. The use of the Galleria mellonella (greater wax moth) larvae, commonly known as wax worm, as a model to study host-pathogen interactions has allowed the identification and characterization of numerous bacterial factors that contribute to disease in humans, serving both as an alternative and complementary approach to mammalian models. Here, we describe the methods for using G. mellonella to characterize the virulence factors of E. faecalis.
Collapse
|
204
|
Calvigioni M, Cara A, Celandroni F, Mazzantini D, Panattoni A, Tirloni E, Bernardi C, Pinotti L, Stella S, Ghelardi E. Characterization of a Bacillus cereus strain associated with a large feed-related outbreak of severe infection in pigs. J Appl Microbiol 2022; 133:1078-1088. [PMID: 35611609 PMCID: PMC9543730 DOI: 10.1111/jam.15636] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/12/2022] [Accepted: 05/19/2022] [Indexed: 12/03/2022]
Abstract
Aims Bacillus cereus is often responsible for foodborne diseases and both local and systemic infections in humans. Cases of infection in other mammals are rather rare. In this study, we report a B. cereus feed‐related outbreak that caused the death of 6234 pigs in Italy. Methods and Results Massive doses of a Gram‐positive, spore‐forming bacterium were recovered from the animal feed, faeces of survived pigs and intestinal content of dead ones. The B. cereus MM1 strain was identified by MALDI‐TOF MS and typified by RAPD‐PCR. The isolate was tested for the production of PC‐PLC, proteases, hemolysins and biofilm, for motility, as well as for the presence of genes encoding tissue‐degrading enzymes and toxins. Antimicrobial resistance and pathogenicity in Galleria mellonella larvae were also investigated. Our results show that the isolated B. cereus strain is swimming‐proficient, produces PC‐PLC, proteases, hemolysins, biofilm and carries many virulence genes. The strain shows high pathogenicity in G. mellonella larvae. Conclusions The isolated B. cereus strain demonstrates an aggressive profile of pathogenicity and virulence, being able to produce a wide range of determinants potentially hazardous to pigs' health. Significance and Impact of Study This study highlights the proficiency of B. cereus to behave as a devastating pathogen in swine if ingested at high doses and underlines that more stringent quality controls are needed for livestock feeds and supplements.
Collapse
Affiliation(s)
- Marco Calvigioni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Alice Cara
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Francesco Celandroni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Diletta Mazzantini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Adelaide Panattoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Erica Tirloni
- Department of Health, Animal Science and Food Safety, University of Milan, Italy
| | - Cristian Bernardi
- Department of Health, Animal Science and Food Safety, University of Milan, Italy
| | - Luciano Pinotti
- Department of Health, Animal Science and Food Safety, University of Milan, Italy
| | - Simone Stella
- Department of Health, Animal Science and Food Safety, University of Milan, Italy
| | - Emilia Ghelardi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy.,Research Center Nutraceuticals and Food for Health-Nutrafood, University of Pisa, Italy
| |
Collapse
|
205
|
Kang M, Kim W, Lee J, Jung HS, Jeon CO, Park W. 6-Bromo-2-naphthol from Silene armeria extract sensitizes Acinetobacter baumannii strains to polymyxin. Sci Rep 2022; 12:8546. [PMID: 35595766 PMCID: PMC9123208 DOI: 10.1038/s41598-022-11995-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 05/04/2022] [Indexed: 12/02/2022] Open
Abstract
The overuse of antibiotics has led to the emergence of multidrug-resistant bacteria, which are resistant to various antibiotics. Combination therapies using natural compounds with antibiotics have been found to have synergistic effects against several pathogens. Synergistic natural compounds can potentiate the effects of polymyxins for the treatment of Acinetobacter baumannii infection. Out of 120 types of plant extracts, only Silene armeria extract (SAE) showed a synergistic effect with polymyxin B (PMB) in our fractional inhibitory concentration and time-kill analyses. The survival rate of G. mellonella infected with A. baumannii ATCC 17978 increased following the synergistic treatment. Interestingly, the addition of osmolytes, such as trehalose, canceled the synergistic effect of SAE with PMB; however, the underlying mechanism remains unclear. Quadrupole time-of-flight liquid chromatography-mass spectrometry revealed 6-bromo-2-naphthol (6B2N) to be a major active compound that exhibited synergistic effects with PMB. Pretreatment with 6B2N made A. baumannii cells more susceptible to PMB exposure in a time- and concentration-dependent manner, indicating that 6B2N exhibits consequential synergistic action with PMB. Moreover, the exposure of 6B2N-treated cells to PMB led to higher membrane leakage and permeability. The present findings provide a promising approach for utilizing plant extracts as adjuvants to reduce the toxicity of PMB in A. baumannii infection.
Collapse
Affiliation(s)
- Mingyeong Kang
- Laboratory of Molecular Environmental Microbiology, Department of Environmental Science and Ecological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Wonjae Kim
- Laboratory of Molecular Environmental Microbiology, Department of Environmental Science and Ecological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Jaebok Lee
- Laboratory of Molecular Environmental Microbiology, Department of Environmental Science and Ecological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hye Su Jung
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Che Ok Jeon
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Woojun Park
- Laboratory of Molecular Environmental Microbiology, Department of Environmental Science and Ecological Engineering, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
206
|
Virulence and DNA sequence analysis of Cronobacter spp. isolated from infant cereals. Int J Food Microbiol 2022; 376:109745. [DOI: 10.1016/j.ijfoodmicro.2022.109745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/21/2022] [Accepted: 05/23/2022] [Indexed: 11/20/2022]
|
207
|
Davis CM, Ruest MK, Cole JH, Dennis JJ. The Isolation and Characterization of a Broad Host Range Bcep22-like Podovirus JC1. Viruses 2022; 14:938. [PMID: 35632679 PMCID: PMC9144972 DOI: 10.3390/v14050938] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 02/04/2023] Open
Abstract
Bacteriophage JC1 is a Podoviridae phage with a C1 morphotype, isolated on host strain Burkholderia cenocepacia Van1. Phage JC1 is capable of infecting an expansive range of Burkholderia cepacia complex (Bcc) species. The JC1 genome exhibits significant similarity and synteny to Bcep22-like phages and to many Ralstonia phages. The genome of JC1 was determined to be 61,182 bp in length with a 65.4% G + C content and is predicted to encode 76 proteins and 1 tRNA gene. Unlike the other Lessieviruses, JC1 encodes a putative helicase gene in its replication module, and it is in a unique organization not found in previously analyzed phages. The JC1 genome also harbours 3 interesting moron genes, that encode a carbon storage regulator (CsrA), an N-acetyltransferase, and a phosphoadenosine phosphosulfate (PAPS) reductase. JC1 can stably lysogenize its host Van1 and integrates into the 5' end of the gene rimO. This is the first account of stable integration identified for Bcep22-like phages. JC1 has a higher global virulence index at 37 °C than at 30 °C (0.8 and 0.21, respectively); however, infection efficiency and lysogen stability are not affected by a change in temperature, and no observable temperature-sensitive switch between lytic and lysogenic lifestyle appears to exist. Although JC1 can stably lysogenize its host, it possesses some desirable characteristics for use in phage therapy. Phage JC1 has a broad host range and requires the inner core of the bacterial LPS for infection. Bacteria that mutate to evade infection by JC1 may develop a fitness disadvantage as seen in previously characterized LPS mutants lacking inner core.
Collapse
Affiliation(s)
| | | | | | - Jonathan J. Dennis
- Department of Biological Sciences, University of Alberta, CW 405 Biological Sciences Building, Edmonton, AB T6G 2E9, Canada; (C.M.D.); (M.K.R.); (J.H.C.)
| |
Collapse
|
208
|
Ribani A, Taurisano V, Utzeri VJ, Fontanesi L. Honey Environmental DNA Can Be Used to Detect and Monitor Honey Bee Pests: Development of Methods Useful to Identify Aethina tumida and Galleria mellonella Infestations. Vet Sci 2022; 9:213. [PMID: 35622741 PMCID: PMC9147136 DOI: 10.3390/vetsci9050213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/09/2022] [Accepted: 04/26/2022] [Indexed: 11/17/2022] Open
Abstract
Environmental DNA (eDNA) contained in honey derives from the organisms that directly and indirectly have been involved in the production process of this matrix and that have played a role in the hive ecosystems where the honey has been produced. In this study we set up PCR-based assays to detect the presence of DNA traces left in the honey by two damaging honey bee pests: the small hive beetle (Aethina tumida) and the greater wax moth (Galleria mellonella). DNA was extracted from 82 honey samples produced in Italy and amplified using two specific primer pairs that target the mitochondrial gene cytochrome oxidase I (COI) of A. tumida and two specific primer pairs that target the same gene in G. mellonella. The limit of detection was tested using sequential dilutions of the pest DNA. Only one honey sample produced in Calabria was positive for A. tumida whereas about 66% of all samples were positively amplified for G. mellonella. The use of honey eDNA could be important to establish early and effective measures to contain at the local (e.g., apiary) or regional scales these two damaging pests and, particularly for the small hive beetle, to prevent its widespread diffusion.
Collapse
Affiliation(s)
- Anisa Ribani
- Department of Agricultural and Food Sciences, University of Bologna, Viale Giuseppe Fanin 46, 40127 Bologna, Italy; (A.R.); (V.T.); (V.J.U.)
- GRIFFA srl, Viale Fanin 48, 40127 Bologna, Italy
| | - Valeria Taurisano
- Department of Agricultural and Food Sciences, University of Bologna, Viale Giuseppe Fanin 46, 40127 Bologna, Italy; (A.R.); (V.T.); (V.J.U.)
| | - Valerio Joe Utzeri
- Department of Agricultural and Food Sciences, University of Bologna, Viale Giuseppe Fanin 46, 40127 Bologna, Italy; (A.R.); (V.T.); (V.J.U.)
- GRIFFA srl, Viale Fanin 48, 40127 Bologna, Italy
| | - Luca Fontanesi
- Department of Agricultural and Food Sciences, University of Bologna, Viale Giuseppe Fanin 46, 40127 Bologna, Italy; (A.R.); (V.T.); (V.J.U.)
| |
Collapse
|
209
|
Paquette AR, Payne SR, McKay GA, Brazeau-Henrie JT, Darnowski MG, Kammili A, Bernal F, Mah TF, Gruenheid S, Nguyen D, Boddy CN. RpoN-Based stapled peptides with improved DNA binding suppress Pseudomonas aeruginosa virulence. RSC Med Chem 2022; 13:445-455. [PMID: 35647551 PMCID: PMC9020619 DOI: 10.1039/d1md00371b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 02/02/2022] [Indexed: 11/21/2022] Open
Abstract
Stapled peptides have the ability to mimic α-helices involved in protein binding and have proved to be effective pharmacological agents for disrupting protein-protein interactions. DNA-binding proteins such as transcription factors bind their cognate DNA sequences via an α-helix interacting with the major groove of DNA. We previously developed a stapled peptide based on the bacterial alternative sigma factor RpoN capable of binding the RpoN DNA promoter sequence and inhibiting RpoN-mediated expression in Escherichia coli. We have elucidated a structure-activity relationship for DNA binding by this stapled peptide, improving DNA binding affinity constants in the high nM range. Lead peptides were shown to have low toxicity as determined by their low hemolytic activity at 100 μM and were shown to have anti-virulence activity in a Galleria mellonella model of Pseudomonas aeruginosa infection. These findings support further preclinical development of stapled peptides as antivirulence agents targeting P. aeruginosa.
Collapse
Affiliation(s)
- André R. Paquette
- Department of Chemistry and Biomolecular Sciences, University of OttawaOttawaONK1N 6N5 Canada
| | - Sterling R. Payne
- Laboratory of Protein Dynamics and Signaling, National Cancer Institute, National Institutes of HealthFrederickMD 21702USA
| | - Geoffrey A. McKay
- Meakins-Christie Laboratories, Research Institute of the McGill University Health CentreMontrealQuebec H4A 3J1Canada
| | | | - Micheal G. Darnowski
- Department of Chemistry and Biomolecular Sciences, University of OttawaOttawaONK1N 6N5 Canada
| | - Anitha Kammili
- Department of Chemistry and Biomolecular Sciences, University of Ottawa Ottawa ON K1N 6N5 Canada
| | - Federico Bernal
- Laboratory of Protein Dynamics and Signaling, National Cancer Institute, National Institutes of HealthFrederickMD 21702USA
| | - Thien-Fah Mah
- Department of Biochemistry, Microbiology, and Immunology, University of OttawaOttawaONK1H 8M5Canada
| | | | - Dao Nguyen
- Meakins-Christie Laboratories, Research Institute of the McGill University Health CentreMontrealQuebec H4A 3J1Canada,Department of Medicine, McGill UniversityMontrealQuebec H4A 3J1Canada
| | - Christopher N. Boddy
- Department of Chemistry and Biomolecular Sciences, University of OttawaOttawaONK1N 6N5 Canada
| |
Collapse
|
210
|
Mariotti M, Lombardini G, Rizzo S, Scarafile D, Modesto M, Truzzi E, Benvenuti S, Elmi A, Bertocchi M, Fiorentini L, Gambi L, Scozzoli M, Mattarelli P. Potential Applications of Essential Oils for Environmental Sanitization and Antimicrobial Treatment of Intensive Livestock Infections. Microorganisms 2022; 10:822. [PMID: 35456873 PMCID: PMC9029798 DOI: 10.3390/microorganisms10040822] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 11/17/2022] Open
Abstract
The extensive use of antibiotics has contributed to the current antibiotic resistance crisis. Livestock infections of Salmonella spp, Clostridium spp. and E. coli antimicrobial-resistant bacteria represent a public threat to human and animal health. To reduce the incidence of these zoonoses, essential oils (EOs) could be effective antibiotic alternatives. This study aims at identifying EOs safe for use, effective both in complementary therapy and in the environmental sanitization of intensive farming. Natural products were chemo-characterized by gas chromatography. Three S. Typhimurium, three C. perfringens and four E. coli strains isolated from poultry and swine farms were used to assess the antimicrobial properties of nine EOs and a modified GR-OLI (mGR-OLI). The toxicity of the most effective ones (Cinnamomum zeylanicum, Cz; Origanum vulgare, Ov) was also evaluated on porcine spermatozoa and Galleria mellonella larvae. Cz, Ov and mGR-OLI showed the strongest antimicrobial activity; their volatile components were also able to significantly inhibit the growth of tested strains. In vitro, Ov toxicity was slightly lower than Cz, while it showed no toxicity on G. mellonella larvae. In conclusion, the study confirms the importance of evaluating natural products to consolidate the idea of safe EO applications in reducing and preventing intensive livestock infections.
Collapse
Affiliation(s)
- Melinda Mariotti
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (G.L.); (S.R.)
| | - Giulia Lombardini
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (G.L.); (S.R.)
| | - Silvia Rizzo
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (G.L.); (S.R.)
| | - Donatella Scarafile
- Dipartimento di Scienze e Tecnologie Agro-Alimentari, Università di Bologna, Viale G. Fanin 42, 40127 Bologna, Italy; (D.S.); (M.M.); (P.M.)
| | - Monica Modesto
- Dipartimento di Scienze e Tecnologie Agro-Alimentari, Università di Bologna, Viale G. Fanin 42, 40127 Bologna, Italy; (D.S.); (M.M.); (P.M.)
| | - Eleonora Truzzi
- Dipartimento di Scienze della Vita, Università di Modena e Reggio Emilia, Via G. Campi 103, 41125 Modena, Italy; (E.T.); (S.B.)
| | - Stefania Benvenuti
- Dipartimento di Scienze della Vita, Università di Modena e Reggio Emilia, Via G. Campi 103, 41125 Modena, Italy; (E.T.); (S.B.)
| | - Alberto Elmi
- Dipartimento di Scienze Mediche Veterinarie, Università di Bologna, Via Tolara di Sopra 50, Ozzano dell’Emilia, 40064 Bologna, Italy; (A.E.); (M.B.)
| | - Martina Bertocchi
- Dipartimento di Scienze Mediche Veterinarie, Università di Bologna, Via Tolara di Sopra 50, Ozzano dell’Emilia, 40064 Bologna, Italy; (A.E.); (M.B.)
| | - Laura Fiorentini
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna (IZSLER)—Sede Territoriale di Forlì, Via Don Eugenio Servadei 3E/3F, 47122 Forlì, Italy; (L.F.); (L.G.)
| | - Lorenzo Gambi
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna (IZSLER)—Sede Territoriale di Forlì, Via Don Eugenio Servadei 3E/3F, 47122 Forlì, Italy; (L.F.); (L.G.)
| | - Maurizio Scozzoli
- Società Italiana per la Ricerca sugli Oli Essenziali (SIROE), Viale Regina Elena 299, 00161 Rome, Italy;
| | - Paola Mattarelli
- Dipartimento di Scienze e Tecnologie Agro-Alimentari, Università di Bologna, Viale G. Fanin 42, 40127 Bologna, Italy; (D.S.); (M.M.); (P.M.)
| |
Collapse
|
211
|
Pegorin Brasil GS, de Barros PP, Miranda MCR, de Barros NR, Junqueira JC, Gomez A, Herculano RD, de Mendonça RJ. Natural latex serum: characterization and biocompatibility assessment using Galleria mellonella as an alternative in vivo model. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2022; 33:705-726. [PMID: 34927570 DOI: 10.1080/09205063.2021.2014027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 06/14/2023]
Abstract
Natural latex serum (NLS) is one of the natural rubber latex fractions from Hevea brasiliensis tree, which is formed by centrifuged serum and is composed of proteins, acids, nucleotides, salts and carbohydrates. The proteins present in NLS have demonstrated several interesting biological properties, including angiogenic, healing, osteogenic, anti-inflammatory, antimicrobial, in addition to inducing neovascularization, bone formation and osseointegration. Thus, we proposed to characterize NLS by physicochemical techniques and to investigate the biocompatibility by toxicological assays and safety test in Galleria mellonella. Infrared spectrum showed vibrational bands characteristic of amide I, II and III that are linked to the protein content, which was confirmed by the High Performance Liquid Chromatography profile and by the Electrophoresis analysis. This material did not exhibit hemolytic (rate <0.5%) and cytotoxic effects (viability >70%) and was able to enhance the proliferation of fibroblasts (>600%) after 3 days. The pronounced proliferative effect observed in fibroblast cells can be explained by the presence of the fibroblast growth factor (FGF) like protein revealed by the Western blot test. Moreover, NLS did not provoke toxic effects (survival ∼ 80%) on the G. mellonella model, indicating that it is a biocompatible and safe material.
Collapse
Affiliation(s)
- Giovana Sant'Ana Pegorin Brasil
- Institute of Chemistry, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
- Department of Biotechnology and Bioprocess Engineering, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Patrícia Pimentel de Barros
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos, São Paulo, Brazil
- Multicampi School of Medical Sciences, Federal University of Rio Grande do Norte (UFRN), Caico, Rio Grande do Norte, Brazil
| | | | | | - Juliana Campos Junqueira
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos, São Paulo, Brazil
| | - Alejandro Gomez
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA
| | - Rondinelli Donizetti Herculano
- Department of Biotechnology and Bioprocess Engineering, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Ricardo José de Mendonça
- Department of Biochemistry, Pharmacology and Physiology, Federal University of Triangulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| |
Collapse
|
212
|
In Vitro & In Vivo Studies on Identifying and Designing Temporin-1CEh from the Skin Secretion of Rana chensinensis as the Optimised Antibacterial Prototype Drug. Pharmaceutics 2022; 14:pharmaceutics14030604. [PMID: 35335979 PMCID: PMC8949600 DOI: 10.3390/pharmaceutics14030604] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/25/2022] [Accepted: 03/04/2022] [Indexed: 11/17/2022] Open
Abstract
Amphibian skin secretion is an ideal source of antimicrobial peptides that are difficult to induce drug resistance to due to their membrane-targeting mechanism as a new treatment scheme. In this study, a natural antimicrobial peptide Temporin-1CEh was identified by molecular cloning and mass spectrometry from the skin secretions of the Chinese forest frog (Rana chensinensis). Through the study of the structure and biological activity, it was found that Temporin-1CEh was a helical peptide from the Temporin family, and possessed good anti-Gram-positive bacteria activity through the mechanism of membrane destruction. Seven analogues were further designed to obtain broad-spectrum antimicrobial activity and higher stability in different physiological conditions. The results showed that T1CEh-KKPWW showed potent antibacterial activity with significantly increasing the activity against Gram-negative bacteria in vitro and in vivo with low haemolysis. In addition, T1CEh-KKPWW2 showed high sensitivity to the pH, serum or salts conditions, which applied a branched structure to allow the active units of the peptide to accumulate. Even though the haemolytic activity was increased, the stable antibacterial activity made this novel analogue meet the conditions to become a potential candidate in future antimicrobial and antibiofilm applications.
Collapse
|
213
|
Alhayek A, Khan ES, Schönauer E, Däinghaus T, Shafiei R, Voos K, Han MK, Ducho C, Posselt G, Wessler S, Brandstetter H, Haupenthal J, del Campo A, Hirsch AK. Inhibition of Collagenase Q1 of Bacillus cereus as a Novel Antivirulence Strategy for the Treatment of Skin-Wound Infections. ADVANCED THERAPEUTICS 2022; 5:2100222. [PMID: 35310821 PMCID: PMC7612511 DOI: 10.1002/adtp.202100222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Indexed: 01/02/2023]
Abstract
Despite the progress in surgical techniques and antibiotic prophylaxis, opportunistic wound infections with Bacillus cereus remain a public health problem. Secreted toxins are one of the main factors contributing to B. cereus pathogenicity. A promising strategy to treat such infections is to target these toxins and not the bacteria. Although the exoenzymes produced by B. cereus are thoroughly investigated, little is known about the role of B. cereus collagenases in wound infections. In this report, the collagenolytic activity of secreted collagenases (Col) is characterized in the B. cereus culture supernatant (csn) and its isolated recombinantly produced ColQ1 is characterized. The data reveals that ColQ1 causes damage on dermal collagen (COL). This results in gaps in the tissue, which might facilitate the spread of bacteria. The importance of B. cereus collagenases is also demonstrated in disease promotion using two inhibitors. Compound 2 shows high efficacy in peptidolytic, gelatinolytic, and COL degradation assays. It also preserves the fibrillar COLs in skin tissue challenged with ColQ1, as well as the viability of skin cells treated with B. cereus csn. A Galleria mellonella model highlights the significance of collagenase inhibition in vivo.
Collapse
Affiliation(s)
- Alaa Alhayek
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Helmholtz Centre for Infection Research (HZI) 38124 Saarbrücken, Germany; Department of Pharmacy Saarland University, Saarbrücken Campus Campus E8.1, 66123 Saarbrücken, Germany
| | - Essak S. Khan
- Leibniz Institute for New Materials (INM) Saarland University Campus D2 2, 66123 Saarbrücken, Germany
| | - Esther Schönauer
- Department of Biosciences and Medical Biology Hellbrunner Str. 34 University of Salzburg Salzburg 5020, Austria
| | - Tobias Däinghaus
- Leibniz Institute for New Materials (INM) Saarland University Campus D2 2, 66123 Saarbrücken, Germany
| | - Roya Shafiei
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Helmholtz Centre for Infection Research (HZI) 38124 Saarbrücken, Germany
| | - Katrin Voos
- Department of Pharmacy Pharmaceutical and Medicinal Chemistry Saarland University Campus C2 3, 66123 Saarbrücken, Germany
| | - Mitchell K.L. Han
- Leibniz Institute for New Materials (INM) Saarl and University Campus D2 2, 66123 Saarbrücken, Germany
| | - Christian Ducho
- Department of Pharmacy Pharmaceutical and Medicinal Chemistry Saarland University Campus C2 3, 66123 Saarbrücken, Germany
| | - Gernot Posselt
- Department of Biosciences and Medical Biology Hellbrunner Str. 34 University of Salzburg Salzburg 5020, Austria
| | - Silja Wessler
- Department of Biosciences and Medical Biology Hellbrunner Str. 34 University of Salzburg Salzburg 5020, Austria
| | - Hans Brandstetter
- Department of Biosciences and Medical Biology Hellbrunner Str. 34 University of Salzburg Salzburg 5020, Austria
| | - Jörg Haupenthal
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Helmholtz Centre for Infection Research (HZI) 38124 Saarbrücken, Germany
| | - Aránzazu del Campo
- Leibniz Institute for New Materials (INM) Saarland University Campus D2 2, 66123 Saarbrücken, Germany; Chemistry Department Saarland University 66123 Saarbrücken, Germany
| | - Anna K.H. Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) Helmholtz Centre for Infection Research (HZI) 38124 Saarbrücken, Germany; Department of Pharmacy Saarland University, Saarbrücken Campus Campus E8.1, 66123 Saarbrücken, Germany
| |
Collapse
|
214
|
Araújo D, Mil-Homens D, Henriques M, Silva S. Anti-EFG1 2′-OMethylRNA oligomer inhibits Candida albicans filamentation and attenuates the candidiasis in Galleria mellonella. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 27:517-523. [PMID: 35036062 PMCID: PMC8728520 DOI: 10.1016/j.omtn.2021.12.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 12/14/2021] [Indexed: 11/20/2022]
Abstract
EFG1 is a central transcriptional regulator of filamentation that is an important virulence factor of Candida albicans. This study serves to assess in vivo the applicability of the anti-EFG1 2′-OMethylRNA oligomer for inhibiting C.albicans filamentation and to attenuate candidiasis, using the Galleria mellonella model. For that, larvae infected with a lethal concentration of C. albicans cells were treated with a single dose and with a double dose of the anti-EFG1 2′OMe oligomer (at 40 and 100 nM). The anti-EFG1 2′OMe oligomer toxicity and effect on larvae survival was evaluated. No evidence of anti-EFG1 2′OMe oligomer toxicity was observed and the treatment with double dose of 2′OMe oligomer empowered larvae survival over 24 h (by 90%–100%) and prolonged its efficacy until 72 h of infection (by 30%). Undoubtedly, this work validates the in vivo therapeutic potential of anti-EFG1 2′OMe oligomer for controlling C. albicans infections.
Collapse
|
215
|
Guan XN, Zhang T, Yang T, Dong Z, Yang S, Lan L, Gan J, Yang CG. Covalent sortase A inhibitor ML346 prevents Staphylococcus aureus infection of Galleria mellonella. RSC Med Chem 2022; 13:138-149. [PMID: 35308030 PMCID: PMC8864484 DOI: 10.1039/d1md00316j] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 10/31/2021] [Indexed: 12/16/2023] Open
Abstract
The housekeeping sortase A (SrtA), a membrane-associated cysteine transpeptidase, is responsible for anchoring surface proteins to the cell wall peptidoglycan in Gram-positive bacteria. This process is essential for the regulation of bacterial virulence and pathogenicity. Therefore, SrtA is considered to be an ideal target for antivirulence therapy. In this study, we report that ML346, a compound with a barbituric acid and cinnamaldehyde scaffold, functions as an irreversible inhibitor of Staphylococcus aureus SrtA (SaSrtA) and Streptococcus pyogenes SrtA (SpSrtA) in vitro at low micromolar concentrations. According to our X-ray crystal structure of the SpSrtAΔN81/ML346 complex (Protein Data Bank ID: 7V6K), ML346 covalently modifies the thiol group of Cys208 in the active site of SpSrtA. Importantly, ML346 significantly attenuated the virulence phenotypes of S. aureus and exhibited inhibitory effects on Galleria mellonella larva infection caused by S. aureus. Collectively, our results indicate that ML346 has potential for development as a covalent antivirulence agent for treating S. aureus infections, including methicillin-resistant S. aureus.
Collapse
Affiliation(s)
- Xiang-Na Guan
- Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- University of the Chinese Academy of Sciences Beijing 100049 China
| | - Tao Zhang
- Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
| | - Teng Yang
- Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals, Guizhou University Guiyang 550025 China
| | - Ze Dong
- Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
| | - Song Yang
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals, Guizhou University Guiyang 550025 China
| | - Lefu Lan
- Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- University of the Chinese Academy of Sciences Beijing 100049 China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences Hangzhou 310024 China
| | - Jianhua Gan
- School of Life Sciences, Fudan University Shanghai 200433 China
| | - Cai-Guang Yang
- Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- University of the Chinese Academy of Sciences Beijing 100049 China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences Hangzhou 310024 China
| |
Collapse
|
216
|
Castledine M, Padfield D, Sierocinski P, Soria Pascual J, Hughes A, Mäkinen L, Friman VP, Pirnay JP, Merabishvili M, de Vos D, Buckling A. Parallel evolution of Pseudomonas aeruginosa phage resistance and virulence loss in response to phage treatment in vivo and in vitro. eLife 2022; 11:73679. [PMID: 35188102 PMCID: PMC8912922 DOI: 10.7554/elife.73679] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 02/20/2022] [Indexed: 12/02/2022] Open
Abstract
With rising antibiotic resistance, there has been increasing interest in treating pathogenic bacteria with bacteriophages (phage therapy). One limitation of phage therapy is the ease at which bacteria can evolve resistance. Negative effects of resistance may be mitigated when resistance results in reduced bacterial growth and virulence, or when phage coevolves to overcome resistance. Resistance evolution and its consequences are contingent on the bacteria-phage combination and their environmental context, making therapeutic outcomes hard to predict. One solution might be to conduct ‘in vitro evolutionary simulations’ using bacteria-phage combinations from the therapeutic context. Overall, our aim was to investigate parallels between in vitro experiments and in vivo dynamics in a human participant. Evolutionary dynamics were similar, with high levels of resistance evolving quickly with limited evidence of phage evolution. Resistant bacteria—evolved in vitro and in vivo—had lower virulence. In vivo, this was linked to lower growth rates of resistant isolates, whereas in vitro phage resistant isolates evolved greater biofilm production. Population sequencing suggests resistance resulted from selection on de novo mutations rather than sorting of existing variants. These results highlight the speed at which phage resistance can evolve in vivo, and how in vitro experiments may give useful insights for clinical evolutionary outcomes.
Collapse
Affiliation(s)
- Meaghan Castledine
- College of Life and Environmental Sciences, University of Exeter, Penryn, United Kingdom
| | - Daniel Padfield
- College of Life and Environmental Sciences, University of Exeter, Penryn, United Kingdom
| | - Pawel Sierocinski
- College of Life and Environmental Sciences, University of Exeter, Penryn, United Kingdom
| | - Jesica Soria Pascual
- College of Life and Environmental Sciences, University of Exeter, Penryn, United Kingdom
| | - Adam Hughes
- College of Life and Environmental Sciences, University of Exeter, Penryn, United Kingdom
| | - Lotta Mäkinen
- College of Life and Environmental Sciences, University of Exeter, Penryn, United Kingdom
| | | | - Jean-Paul Pirnay
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| | - Maya Merabishvili
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| | - Daniel de Vos
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| | - Angus Buckling
- College of Life and Environmental Sciences, University of Exeter, Penryn, United Kingdom
| |
Collapse
|
217
|
Kaya C, Konstantinović J, Kany AM, Andreas A, Kramer JS, Brunst S, Weizel L, Rotter MJ, Frank D, Yahiaoui S, Müller R, Hartmann RW, Haupenthal J, Proschak E, Wichelhaus TA, Hirsch AKH. N-Aryl Mercaptopropionamides as Broad-Spectrum Inhibitors of Metallo-β-Lactamases. J Med Chem 2022; 65:3913-3922. [PMID: 35188771 DOI: 10.1021/acs.jmedchem.1c01755] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Drug-resistant pathogens pose a global challenge to public health as they cause diseases that are extremely difficult to cure. Metallo-β-lactamases (MBLs) are a diverse set of zinc-containing enzymes that catalyze the hydrolysis of β-lactam drugs, including carbapenems, which are considered as the last resort to fight severe infections. To restore the activity of current β-lactam antibiotics and to offer an orthogonal strategy to the discovery of new antibiotics, we have identified a series of polar N-aryl mercaptopropionamide derivatives as potent inhibitors of several class B1 MBLs. We have identified a hit structure with high selectivity restoring the effect of imipenem and reducing minimum inhibitory concentration (MIC) values up to 256-fold in resistant isolates from Escherichia coli. Furthermore, the combination of imipenem with our inhibitor showed in vivo efficacy in a Galleria mellonella model, increasing the survival rate of infected larvae by up to 31%.
Collapse
Affiliation(s)
- Cansu Kaya
- Helmholtz Institute for Pharmaceutical Research Saarland, (HIPS)─Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany.,Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| | - Jelena Konstantinović
- Helmholtz Institute for Pharmaceutical Research Saarland, (HIPS)─Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Andreas M Kany
- Helmholtz Institute for Pharmaceutical Research Saarland, (HIPS)─Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Anastasia Andreas
- Helmholtz Institute for Pharmaceutical Research Saarland, (HIPS)─Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany.,Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| | - Jan S Kramer
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438 Frankfurt, Germany
| | - Steffen Brunst
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438 Frankfurt, Germany
| | - Lilia Weizel
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438 Frankfurt, Germany
| | - Marco J Rotter
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438 Frankfurt, Germany
| | - Denia Frank
- Institute of Medical Microbiology and Infection Control, University Hospital Frankfurt, Paul-Ehrlich-Straße 40, 60596 Frankfurt, Germany
| | - Samir Yahiaoui
- Helmholtz Institute for Pharmaceutical Research Saarland, (HIPS)─Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland, (HIPS)─Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany.,Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany.,Helmholtz International Lab for Anti-infectives, Campus E8.1, 66123 Saarbrücken, Germany
| | - Rolf W Hartmann
- Helmholtz Institute for Pharmaceutical Research Saarland, (HIPS)─Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany.,Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| | - Jörg Haupenthal
- Helmholtz Institute for Pharmaceutical Research Saarland, (HIPS)─Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Ewgenij Proschak
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438 Frankfurt, Germany
| | - Thomas A Wichelhaus
- Institute of Medical Microbiology and Infection Control, University Hospital Frankfurt, Paul-Ehrlich-Straße 40, 60596 Frankfurt, Germany
| | - Anna K H Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland, (HIPS)─Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany.,Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany.,Helmholtz International Lab for Anti-infectives, Campus E8.1, 66123 Saarbrücken, Germany
| |
Collapse
|
218
|
Morales-Ruíz LM, Rodríguez-Cisneros M, Kerber-Díaz JC, Rojas-Rojas FU, Ibarra JA, Estrada-de Los Santos P. Burkholderia orbicola sp. nov., a novel species within the Burkholderia cepacia complex. Arch Microbiol 2022; 204:178. [PMID: 35174425 DOI: 10.1007/s00203-022-02778-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 11/30/2022]
Abstract
Genome analysis of strains placed in the NCBI genome database as Burkholderia cenocepacia defined nine genomic species groups. The largest group (259 strains) corresponds to B. cenocepacia and the second largest group (58 strains) was identified as "Burkholderia servocepacia", a Burkholderia species classification which has not been validly published. The publication of "B. servocepacia" did not comply with Rule 27 and Recommendation 30 from the International Code of Nomenclature of Prokaryotes (ICNP) and have errors in the type strain name and the protologue describing the novel species. Here, we correct the position of this species by showing essential information that meets the criteria defined by ICNP. After additional analysis complying with taxonomic criteria, we propose that the invalid "B. servocepacia" be renamed as Burkholderia orbicola sp. nov. The original study proposing "B. servocepacia" was misleading, because this name derives from the Latin "servo" meaning "to protect/watch over", and the authors proposed this based on the beneficial biocontrol properties of several strains within the group. However, it is clear that "B. servocepacia" isolates are capable of opportunistic infection, and the proposed name Burkholderia orbicola sp. nov. takes into account these diverse phenotypic traits. The type strain is TAtl-371 T (= LMG 30279 T = CM-CNRG 715 T).
Collapse
Affiliation(s)
- Leslie-Mariana Morales-Ruíz
- Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. Plan de Ayala s/n, Col. Santo Tomás, Alcaldía Miguel Hidalgo, C.P. 11340, Mexico City, México
| | - Mariana Rodríguez-Cisneros
- Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. Plan de Ayala s/n, Col. Santo Tomás, Alcaldía Miguel Hidalgo, C.P. 11340, Mexico City, México
| | - Jeniffer-Chris Kerber-Díaz
- Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. Plan de Ayala s/n, Col. Santo Tomás, Alcaldía Miguel Hidalgo, C.P. 11340, Mexico City, México
| | - Fernando-Uriel Rojas-Rojas
- Laboratorio de Ciencias AgroGenómicas, Escuela Nacional de Estudios Superiores Unidad León, Universidad Nacional Autónoma de México (ENES-León UNAM), Blvd. UNAM 2011, 37684, León, Guanajuato, México.,Laboratorio Nacional PlanTECC, Escuela Nacional de Estudios Superiores Unidad León, Universidad Nacional Autónoma de México (ENES-León), Blvd. UNAM 2011, 37684, León, Guanajuato, México
| | - J Antonio Ibarra
- Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. Plan de Ayala s/n, Col. Santo Tomás, Alcaldía Miguel Hidalgo, C.P. 11340, Mexico City, México
| | - Paulina Estrada-de Los Santos
- Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prol. Plan de Ayala s/n, Col. Santo Tomás, Alcaldía Miguel Hidalgo, C.P. 11340, Mexico City, México.
| |
Collapse
|
219
|
Marena GD, Ramos MADS, Lima LC, Chorilli M, Bauab TM. Galleria mellonella for systemic assessment of anti-Candida auris using amphotericin B loaded in nanoemulsion. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 807:151023. [PMID: 34662607 DOI: 10.1016/j.scitotenv.2021.151023] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 06/13/2023]
Abstract
Galleria mellonella is a model that uses adult larvae to assess the prophylactic, therapeutic, and acute toxic potential of substances. Given their benefits, G. mellonella models are being employed in investigations of systemic infections caused by highly resistant microorganisms. Among the multiresistant microorganisms, we highlight Candida auris, a yeast with high mortality potential and resistance. Among the potential drugs, amphotericin B (AmB) stands out; however, microbial resistance episodes and side effects caused by low selectivity have been observed. The incorporation of AmB into a nanoemulsion (NE) can contribute to the control of C. auris infections and resistance as well as decrease the side effects of this drug. This study aimed to develop AmB-loaded NE (NEA) and evaluate its antifungal action against C. auris in G. mellonella. NEs were obtained by using sunflower oil and cholesterol as the oily phase, polyoxyethylene 20 cetyl ether (Brij® 58) and soy phosphatidylcholine as the surfactant system, and PBS buffer as the aqueous phase. An alternative in vivo assay with G. mellonella for acute toxicity and infection was performed using adult stage larvae (200 mg to 400 mg). According to the obtained results, NE and NEA exhibited sizes of 43 and 48 nm, respectively. The PDI was 0.285 and 0.389 for NE and NEA, respectively. The ZP showed electronegativity for both systems, with -3.77 mV and -3.80 mV for NE and NEA, respectively. Acute toxicity showed that free AmB had greater acute toxicity potential than NEA. The survival assay showed high larval viability. NEA had a better antifungal profile against systemic infection in G. mellonella. It is concluded that the alternative model proved to be an efficient in vivo assay to determine the toxicity and evaluate the therapeutic property of free AmB and NEA in systemic infections caused by C. auris.
Collapse
Affiliation(s)
- Gabriel Davi Marena
- Department of Drugs and Medicines, São Paulo State University (UNESP), School of Pharmaceutical Sciences, Campus Araraquara, São Paulo State 14.800-903, Brazil
| | - Matheus Aparecido Dos Santos Ramos
- Department of Drugs and Medicines, São Paulo State University (UNESP), School of Pharmaceutical Sciences, Campus Araraquara, São Paulo State 14.800-903, Brazil
| | - Laura Caminitti Lima
- Department of Biological Sciences, São Paulo State University (UNESP), School of Pharmaceutical Sciences, Campus Araraquara, São Paulo State 14.800-903, Brazil
| | - Marlus Chorilli
- Department of Drugs and Medicines, São Paulo State University (UNESP), School of Pharmaceutical Sciences, Campus Araraquara, São Paulo State 14.800-903, Brazil.
| | - Tais Maria Bauab
- Department of Biological Sciences, São Paulo State University (UNESP), School of Pharmaceutical Sciences, Campus Araraquara, São Paulo State 14.800-903, Brazil.
| |
Collapse
|
220
|
Shen L, Zhang J, Xue J, Du L, Yuan L, Nie H, Dai S, Yu Q, Li Y. Regulation of ECP fimbriae-related genes by the transcriptional regulator RcsAB in Klebsiella pneumoniae NTUH-K2044. J Basic Microbiol 2022; 62:593-603. [PMID: 35132658 DOI: 10.1002/jobm.202100595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 01/06/2022] [Accepted: 01/15/2022] [Indexed: 11/11/2022]
Abstract
Klebsiella pneumoniae is one of the major pathogens causing nosocomial infections. The regulator of capsule synthesis (Rcs) system is a complex signal transduction pathway that is involved in the regulation of virulence factors of K. pneumoniae as an important transcriptional regulator. The RcsAB box-like sequence was found to be present in the promoter-proximal regions of ykgK, one of the ECP fimbriae-related genes, which suggested the expression of ECP fimbriae may be regulated by RcsAB. The ykgK gene in K. pneumoniae has 86% similarity to the ecpR gene in Escherichia coli. Nucleotide sequence alignment revealed a similar ECP fimbriae gene cluster including six genes in K. pneumoniae, which was proved to be on the same operon in this study. The electrophoretic mobility shift assay and DNase I assay, relative fluorescence expression, β-galactosidase activity, and relative gene expression of ykgK in the wild-type and mutant strains were performed to determine the transcriptional regulation mechanism of RcsAB on ECP fimbriae. The mutant ΔykgK and complementary strain ΔykgK/cΔykgK were constructed to complete the Galleria mellonella larvae infection experiment and biofilm formation assay. This study showed that RcsAB binds directly to the promoter region of the ykgK gene to positively regulate ECP fimbriae-related gene clusters, and then positively affect the biofilm formation.
Collapse
Affiliation(s)
- Lifei Shen
- School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Jiaxue Zhang
- Chongqing Jiangbei District Center for Disease Control and Prevention, Chongqing, China
| | - Jian Xue
- Zunyi Medical and Pharmaceutical College, Zunyi, China
| | - Ling Du
- School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Lingyue Yuan
- School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Hao Nie
- School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Sue Dai
- School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Qian Yu
- School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Yingli Li
- School of Public Health and Management, Chongqing Medical University, Chongqing, China
| |
Collapse
|
221
|
Hamed A, Pullinger G, Stevens M, Farveen F, Freestone P. Characterisation of the E. coli and Salmonella qseC and qseE mutants reveals a metabolic rather than adrenergic receptor role. FEMS Microbiol Lett 2022; 369:6524176. [PMID: 35137015 PMCID: PMC8897314 DOI: 10.1093/femsle/fnac012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 12/31/2021] [Accepted: 02/04/2022] [Indexed: 11/14/2022] Open
Abstract
Catecholamine stress hormones (norepinephrine, epinephrine, and dopamine) are signals that have been shown to be used as environmental cues, which affect the growth and virulence of normal microbiota as well as pathogenic bacteria. It has been reported that Escherichia coli and Salmonella use the two-component system proteins QseC and QseE to recognise catecholamines and so act as bacterial adrenergic receptors. In this study, we mutated the E. coli O157:H7 and Salmonella enterica serovar Typhimurium genes encoding QseC and QseE and found that this did not block stress hormone responsiveness in either species. Motility, biofilm formation, and analysis of virulence of the mutants using two infection models were similar to the wild-type strains. The main differences in phenotypes of the qseC and qseE mutants were responses to changes in temperature and growth in different media particularly with respect to salt, carbon, and nitrogen salt sources. In this physiological respect, it was also found that the phenotypes of the qseC and qseE mutants differed between E. coli and Salmonella. These findings collectively suggest that QseC and QseE are not essential for E. coli and Salmonella to respond to stress hormones and that the proteins may be playing a role in regulating metabolism.
Collapse
Affiliation(s)
- Abdalla Hamed
- Department of Microbiology and Immunology, Faculty of Medicine, University of Zawia, Zawiya QP7X+536, Libya
| | - Gillian Pullinger
- Division of Microbiology, Institute for Animal Health, Compton, Newbury RG20 7NN, United Kingdom
| | - Mark Stevens
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian EH25 9RG, United Kingdom
| | - Fathima Farveen
- Department of Respiratory Sciences, University of Leicester, University Road, Leicester LE1 7RH, United Kingdom
| | - Primrose Freestone
- Corresponding author: Department of Respiratory Sciences, University of Leicester, University Road, Leicester LE1 7RH, United Kingdom. Tel: +44 (0)116 2525656; Fax: +44 (0)116 2525030; E-mail:
| |
Collapse
|
222
|
Zhu L, Shuai X, Xu L, Sun Y, Lin Z, Zhou Z, Meng L, Chen H. Mechanisms underlying the effect of chlorination and UV disinfection on VBNC state Escherichia coli isolated from hospital wastewater. JOURNAL OF HAZARDOUS MATERIALS 2022; 423:127228. [PMID: 34547689 DOI: 10.1016/j.jhazmat.2021.127228] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 09/10/2021] [Accepted: 09/11/2021] [Indexed: 06/13/2023]
Abstract
The occurrence of viable but non-culturable (VBNC) bacteria in the wastewater system poses a huge threat to environmental and public health, in particular in hospital wastewater treatment system (HWTS). HWTS-oriented studies have been conducted to assess the effectiveness of chlorination and UV disinfection using indigenous bacteria. Results revealed that the VBNC Escherichia coli and ARGs remained persistent even at high chlorination (12 mg/L for 2.5 h) and UV doses (1000 mJ/cm2). The molecular mechanisms underlying chlorination-/UV-induced VBNC state in E. coli were explored through the transcriptomics and results suggested that most energy-dependent physiological activities (e.g., metabolism) have been suppressed in VBNC E. coli, while the pathogenicity-related genes varied insignificantly compared to the culturable cells, indicating that the VBNC E. coli could potentially display pathogenicity. Further Galleria mellonella model experiment has confirmed that although the disinfection-induced VBNC state made cells less infectious, these cells could regain their pathogenicity after resuscitation. This in vitro study can be used as a reference for studies on infections from VBNC bacteria and highlights the health risk due to VBNC pathogens in hospital effluents. There is a need to develop effluent standards specifically for healthcare facilities, and a stricter downstream disinfection strategy should be considered for the removal of VBNC cells and ARGs in the effluent.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Environmental Engineering, College of Environmental and Resource Sciences; Zhejiang University, Hangzhou 310058, China
| | - Xinyi Shuai
- Department of Environmental Engineering, College of Environmental and Resource Sciences; Zhejiang University, Hangzhou 310058, China
| | - Like Xu
- Department of Chemistry, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Yujie Sun
- Department of Environmental Engineering, College of Environmental and Resource Sciences; Zhejiang University, Hangzhou 310058, China
| | - Zejun Lin
- Department of Environmental Engineering, College of Environmental and Resource Sciences; Zhejiang University, Hangzhou 310058, China
| | - Zhenchao Zhou
- Department of Environmental Engineering, College of Environmental and Resource Sciences; Zhejiang University, Hangzhou 310058, China
| | - Lingxuan Meng
- Department of Environmental Engineering, College of Environmental and Resource Sciences; Zhejiang University, Hangzhou 310058, China
| | - Hong Chen
- Department of Environmental Engineering, College of Environmental and Resource Sciences; Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
223
|
Aghababa H, Ting YT, Pilapitiya D, Loh JM, Young PG, Proft T. Complement evasion factor (CEF), a novel immune evasion factor of Streptococcus pyogenes. Virulence 2022; 13:225-240. [PMID: 35094646 PMCID: PMC8803112 DOI: 10.1080/21505594.2022.2027629] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Streptococcus pyogenes, a leading human pathogen, is responsible for a wide range of diseases, including skin and soft tissue infections and severe invasive diseases. S. pyogenes produces a large arsenal of virulence factors, including several immune evasion factors. We have identified an open reading frame (spy0136) in the S. pyogenes SF370 genome encoding a protein of unknown function. Using recombinant Spy0136 in a pull-down assay with human plasma and ELISA, we have identified four complement proteins (C1r, C1s, C3, and C5) as binding partners. Treatment of the complement proteins with PNGase F abrogated binding to C1s, C3, and C5, indicating glycan-dependent interactions. rSpy0136 inhibited complement-mediated hemolysis and interfered with all three complement pathways in a Wieslab complement assay. Furthermore, rSpy0136 inhibited deposition of the C3b opsonin and the membrane attack complex (MAC) on the surface of S. pyogenes. We therefore named the previously unknown protein ‘complement evasion factor’ (CEF). An S. pyogenes Δspy0136/cef deletion mutant showed decreased virulence in an in-vitro whole blood killing assay and a Galleria mellonella (wax moth) infection model. Furthermore, an L. lactis spy0136/cef gain-of-function mutant showed increased survival during growth in whole human blood. Analysis of serum samples from patients with invasive S. pyogenes revealed Spy0136/CEF sero-conversion indicating expression during disease. In summary, we have identified a novel S. pyogenes immune evasion factor that binds to several complement proteins to interfere with complement function. This is the first example of a S. pyogenes virulence factor binding to several different target proteins via glycan-dependent interactions.
Collapse
Affiliation(s)
- Haniyeh Aghababa
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - Yi Tian Ting
- School of Biological Sciences, the University of Auckland, Auckland, New Zealand
- Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Devaki Pilapitiya
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
- University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Jacelyn M.S. Loh
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Biomolecular Discoveries. The University of Auckland, Auckland, New Zealand
| | - Paul G. Young
- School of Biological Sciences, the University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Biomolecular Discoveries. The University of Auckland, Auckland, New Zealand
| | - Thomas Proft
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Biomolecular Discoveries. The University of Auckland, Auckland, New Zealand
| |
Collapse
|
224
|
RNase R, a New Virulence Determinant of Streptococcus pneumoniae. Microorganisms 2022; 10:microorganisms10020317. [PMID: 35208772 PMCID: PMC8875335 DOI: 10.3390/microorganisms10020317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 11/30/2022] Open
Abstract
Pneumococcal infections have increasingly high mortality rates despite the availability of vaccines and antibiotics. Therefore, the identification of new virulence determinants and the understanding of the molecular mechanisms behind pathogenesis have become of paramount importance in the search of new targets for drug development. The exoribonuclease RNase R has been involved in virulence in a growing number of pathogens. In this work, we used Galleria mellonella as an infection model to demonstrate that the presence of RNase R increases the pneumococcus virulence. Larvae infected with the RNase R mutant show an increased expression level of antimicrobial peptides. Furthermore, they have a lower bacterial load in the hemolymph in the later stages of infection, leading to a higher survival rate of the larvae. Interestingly, pneumococci expressing RNase R show a sudden drop in bacterial numbers immediately after infection, resembling the eclipse phase observed after intravenous inoculation in mice. Concomitantly, we observed a lower number of mutant bacteria inside larval hemocytes and a higher susceptibility to oxidative stress when compared to the wild type. Together, our results indicate that RNase R is involved in the ability of pneumococci to evade the host immune response, probably by interfering with internalization and/or replication inside the larval hemocytes.
Collapse
|
225
|
Comparative Assessment of Bacteriophage and Antibiotic Activity against Multidrug-Resistant Staphylococcus aureus Biofilms. Int J Mol Sci 2022; 23:ijms23031274. [PMID: 35163197 PMCID: PMC8836238 DOI: 10.3390/ijms23031274] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 02/01/2023] Open
Abstract
Problems connected with biofilm-related infections and antibiotic resistance necessitate the investigation and development of novel treatment strategies. Given their unique characteristics, one of the most promising alternatives to conventional antibiotics are bacteriophages. In the in vitro and in vivo larva model study, we demonstrate that phages vB_SauM-A, vB_SauM-C, and vB_SauM-D are effective antibiofilm agents. The exposure of biofilm to phages vB_SauM-A and vB_SauM-D led to 2-3 log reductions in the colony-forming unit number in most of the multidrug-resistant S. aureus strains. It was found that phage application reduced the formed biofilms independently of the used titer. Moreover, the study demonstrated that bacteriophages are more efficient in biofilm biomass removal and reduction in staphylococci count when compared to the antibiotics used. The scanning electron microscopy analysis results are in line with colony forming unit (CFU) counting but not entirely consistent with crystal violet (CV) staining. Additionally, phages vB_SauM-A, vB_SauM-C, and vB_SauM-D can significantly increase the survival rate and extend the survival time of Galleria mellonella larvae.
Collapse
|
226
|
Rocha J, Ferreira C, Mil-Homens D, Busquets A, Fialho AM, Henriques I, Gomila M, Manaia CM. Third generation cephalosporin-resistant Klebsiella pneumoniae thriving in patients and in wastewater: what do they have in common? BMC Genomics 2022; 23:72. [PMID: 35065607 PMCID: PMC8783465 DOI: 10.1186/s12864-021-08279-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/22/2021] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Klebsiella pneumoniae are ubiquitous bacteria and recognized multidrug-resistant opportunistic pathogens that can be released into the environment, mainly through sewage, where they can survive even after wastewater treatment. A major question is if once released into wastewater, the selection of lineages missing clinically-relevant traits may occur. Wastewater (n = 25) and clinical (n = 34) 3rd generation cephalosporin-resistant K. pneumoniae isolates were compared based on phenotypic, genotypic and genomic analyses. RESULTS Clinical and wastewater isolates were indistinguishable based on phenotypic and genotypic characterization. The analysis of whole genome sequences of 22 isolates showed that antibiotic and metal resistance or virulence genes, were associated with mobile genetic elements, mostly transposons, insertion sequences or integrative and conjugative elements. These features were variable among isolates, according to the respective genetic lineage rather than the origin. CONCLUSIONS It is suggested that once acquired, clinically relevant features of K. pneumoniae may be preserved in wastewater, even after treatment. This evidence highlights the high capacity of K. pneumoniae for spreading through wastewater, enhancing the risks of transmission back to humans.
Collapse
Affiliation(s)
- Jaqueline Rocha
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005, Porto, Portugal
| | - Catarina Ferreira
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005, Porto, Portugal
| | - Dalila Mil-Homens
- iBB-Institute of Bioengineering and Biosciences and i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Lisbon, Portugal
- Department of Bioengineering, Instituto Superior Técnico, University of Lisbon, Lisbon, Portugal
| | - Antonio Busquets
- Microbiologia, Departament de Biologia, Universitat de les Illes Balears, Palma de Mallorca, Spain
| | - Arsénio M Fialho
- iBB-Institute of Bioengineering and Biosciences and i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Lisbon, Portugal
- Department of Bioengineering, Instituto Superior Técnico, University of Lisbon, Lisbon, Portugal
| | - Isabel Henriques
- University of Coimbra, Department of Life Sciences, Faculty of Science and Technology, Coimbra, Portugal
- CESAM, University of Aveiro, Aveiro, Portugal
| | - Margarita Gomila
- Microbiologia, Departament de Biologia, Universitat de les Illes Balears, Palma de Mallorca, Spain
| | - Célia M Manaia
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005, Porto, Portugal.
| |
Collapse
|
227
|
O’Shaughnessy M, Piatek M, McCarron P, McCann M, Devereux M, Kavanagh K, Howe O. In Vivo Activity of Metal Complexes Containing 1,10-Phenanthroline and 3,6,9-Trioxaundecanedioate Ligands against Pseudomonas aeruginosa Infection in Galleria mellonella Larvae. Biomedicines 2022; 10:biomedicines10020222. [PMID: 35203432 PMCID: PMC8869450 DOI: 10.3390/biomedicines10020222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 11/16/2022] Open
Abstract
Drug-resistant Pseudomonas aeruginosa is rapidly developing resulting in a serious global threat. Immunocompromised patients are specifically at risk, especially those with cystic fibrosis (CF). Novel metal complexes incorporating 1,10-phenanthroline (phen) ligands have previously demonstrated antibacterial and anti-biofilm effects against resistant P. aeruginosa from CF patients in vitro. Herein, we present the in vivo efficacy of {[Cu(3,6,9-tdda)(phen)2]·3H2O·EtOH}n (Cu-tdda-phen), {[Mn(3,6,9-tdda)(phen)2]·3H2O·EtOH}n (Mn-tdda-phen) and [Ag2(3,6,9-tdda)(phen)4]·EtOH (Ag-tdda-phen) (tddaH2 = 3,6,9-trioxaundecanedioic acid). Individual treatments of these metal-tdda-phen complexes and in combination with the established antibiotic gentamicin were evaluated in vivo in larvae of Galleria mellonella infected with clinical isolates and laboratory strains of P. aeruginosa. G. mellonella were able to tolerate all test complexes up to 10 µg/larva. In addition, the immune response was affected by stimulation of immune cells (hemocytes) and genes that encode for immune-related peptides, specifically transferrin and inducible metallo-proteinase inhibitor. The amalgamation of metal-tdda-phen complexes and gentamicin further intensified this response at lower concentrations, clearing a P. aeruginosa infection that were previously resistant to gentamicin alone. Therefore this work highlights the anti-pseudomonal capabilities of metal-tdda-phen complexes alone and combined with gentamicin in an in vivo model.
Collapse
Affiliation(s)
- Megan O’Shaughnessy
- School of Biological and Health Sciences, Technological University Dublin-City Campus, D07 ADY7 Dublin, Ireland;
- Centre for Biomimetic and Therapeutic Research, FOCAS Research Institute, Technological University Dublin-City Campus, D08 CKP1 Dublin, Ireland; (P.M.); (M.D.)
| | - Magdalena Piatek
- SSPC Pharma Research Centre, Department of Biology, Maynooth University, W23 F2H6 Kildare, Ireland;
| | - Pauraic McCarron
- Centre for Biomimetic and Therapeutic Research, FOCAS Research Institute, Technological University Dublin-City Campus, D08 CKP1 Dublin, Ireland; (P.M.); (M.D.)
| | - Malachy McCann
- Chemistry Department, Maynooth University, W23 F2H6 Kildare, Ireland;
| | - Michael Devereux
- Centre for Biomimetic and Therapeutic Research, FOCAS Research Institute, Technological University Dublin-City Campus, D08 CKP1 Dublin, Ireland; (P.M.); (M.D.)
| | - Kevin Kavanagh
- SSPC Pharma Research Centre, Department of Biology, Maynooth University, W23 F2H6 Kildare, Ireland;
- Correspondence: (K.K.); (O.H.)
| | - Orla Howe
- School of Biological and Health Sciences, Technological University Dublin-City Campus, D07 ADY7 Dublin, Ireland;
- Centre for Biomimetic and Therapeutic Research, FOCAS Research Institute, Technological University Dublin-City Campus, D08 CKP1 Dublin, Ireland; (P.M.); (M.D.)
- Correspondence: (K.K.); (O.H.)
| |
Collapse
|
228
|
Kaskatepe B, Aslan Erdem S, Ozturk S, Safi Oz Z, Subasi E, Koyuncu M, Vlainić J, Kosalec I. Antifungal and Anti-Virulent Activity of Origanum majorana L. Essential Oil on Candida albicans and In Vivo Toxicity in the Galleria mellonella Larval Model. Molecules 2022; 27:molecules27030663. [PMID: 35163928 PMCID: PMC8838586 DOI: 10.3390/molecules27030663] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/12/2022] [Accepted: 01/18/2022] [Indexed: 02/06/2023] Open
Abstract
The aim of this study was to investigate and compare in detail both the antifungal activity in vitro (with planktonic and biofilm-forming cells) and the essential oil composition (EOs) of naturally growing (OMN) and cultivated (OMC) samples of Origanum majorana L. (marjoram). The essential oil composition was analyzed using GC-MS. The major constituent of both EOs was carvacrol: 75.3% and 84%, respectively. Both essential oils showed high antifungal activity against clinically relevant Candida spp. with IC50 and IC90 less than or equal to 0.5 µg mL−1 and inhibition of biofilm with a concentration of 3.5 µg mL−1 or less. Cultivated marjoram oil showed higher anti-biofilm activity against C. albicans. In addition, OMC showed greater inhibition of germ-tube formation (inhibition by 83% in Spider media), the major virulence factor of C. albicans at a concentration of 0.125 µg mL−1. Both EOs modulated cell surface hydrophobicity (CSH), but OMN proved to be more active with a CSH% up to 58.41%. The efficacy of O. majorana EOs was also investigated using Galleria mellonella larvae as a model. It was observed that while the larvae of the control group infected with C. albicans (6.0 × 108 cells) and not receiving treatment died in the controls carried out after 24 h, all larvae in the infected treatment group survived at the end of the 96th hour. When the treatment group and the infected group were evaluated in terms of vital activities, it was found that the difference was statistically significant (p < 0.001). The infection of larvae with C. albicans and the effects of O. majorana EOs on the hemocytes of the model organism and the blastospores of C. albicans were evaluated by light microscopy on slides stained with Giemsa. Cytological examination in the treatment group revealed that C. albicans blastospores were phagocytosed and morphological changes occurred in hemocytes. Our results indicated that the essential oil of both samples showed strong antifungal activities against planktonic and biofilm-forming C. albicans cells and also had an influence on putative virulence factors (germ-tube formation and its length and on CSH).
Collapse
Affiliation(s)
- Banu Kaskatepe
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Ankara University, Ankara 06560, Turkey;
| | - Sinem Aslan Erdem
- Department of Pharmacognosy, Faculty of Pharmacy, Ankara University, Ankara 06560, Turkey;
| | - Sukran Ozturk
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Zonguldak Bulent Ecevit University, Zonguldak 67100, Turkey;
| | - Zehra Safi Oz
- Department of Medical Biology, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak 67100, Turkey;
| | - Eldan Subasi
- Microbiology Laboratory of Application and Research Hospital, Zonguldak Bulent Ecevit University, Zonguldak 67100, Turkey;
| | - Mehmet Koyuncu
- Department of Pharmaceutical Botany, Faculty of Pharmacy, Cyprus International University, Lefkosa 99258, Turkey;
| | | | - Ivan Kosalec
- Faculty of Pharmacy and Biochemistry, Institute for Microbiology, University of Zagreb, 10000 Zagreb, Croatia
- Correspondence: ; Tel.: +385-1639-4492
| |
Collapse
|
229
|
N-acyl-homoserine lactone produced by Rahnella inusitata isolated from the gut of Galleria mellonella influences Salmonella phenotypes. Braz J Microbiol 2022; 53:819-829. [PMID: 35048318 PMCID: PMC9151966 DOI: 10.1007/s42770-022-00681-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/10/2022] [Indexed: 02/01/2023] Open
Abstract
The most studied mechanism of quorum sensing in Gram-negative bacteria is mediated by autoinducer 1 (AI-1), namely, acyl-homoserine lactone (AHL). This system allows communication among different bacterial species and regulates the expression of virulence genes in many pathogens. Although AHL-producing bacteria have been detected in the intestines of humans and other animals, no report was found about AHL-producing bacteria in the insect gut and the possible effects of these autoinducers on enteropathogenic bacteria. Therefore, this study aimed to identify AHL-producing bacteria in the gut of larvae of Galleria mellonella and to evaluate the influence of this quorum sensing signal on the regulation of adhesion and motility phenotypes in the intestinal pathogen Salmonella. Sequencing of the 16S rRNA gene, 16S rRNA gene-based phylogenetic analyses, and phenotypic characterization of gut isolates was performed. The profile of AHLs produced by the isolates was determined using thin-layer chromatography (TLC) and revealed with the biosensor strain Chromobacterium violaceum CV026. Sequencing, phylogenetic analyses and phenotypic characterization of gut isolates showed that the three AHL-producing strains belong to the species Rahnella inusitata, named GM34, GM56, and GM60. The TLC showed that R. inusitata produces a six-carbon AHL. In the presence of cell-free extract of R. inusitata containing AHL and under anaerobic conditions, Salmonella enterica increased the adhesion to stainless steel coupons and presented swarming motility. Extracts from the culture medium of R. inusitata isolates containing AHL increased the adhesion on stainless steel coupons and swarming motility of Salmonella enterica serovar Enteritidis PT4 under anaerobic conditions. The results suggest the possibility of communication between members of the G. mellonella intestinal microbiota with pathogens such as Salmonella.
Collapse
|
230
|
An integrated IncFIB/IncFII plasmid confers hypervirulence and its fitness cost and stability. Eur J Clin Microbiol Infect Dis 2022; 41:681-684. [DOI: 10.1007/s10096-022-04407-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/13/2022] [Indexed: 11/03/2022]
|
231
|
Ménard G, Rouillon A, Cattoir V, Donnio PY. Galleria mellonella as a Suitable Model of Bacterial Infection: Past, Present and Future. Front Cell Infect Microbiol 2022; 11:782733. [PMID: 35004350 PMCID: PMC8727906 DOI: 10.3389/fcimb.2021.782733] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 12/01/2021] [Indexed: 12/16/2022] Open
Abstract
The increasing interest for Galleria mellonella larvae as an infection model is evidenced by the number of papers reporting its use, which increases exponentially since the early 2010s. This popularity was initially linked to limitation of conventional animal models due to financial, technical and ethical aspects. In comparison, alternative models (e.g. models using Caenorhabditis elegans, Drosophila melanogaster or G. mellonella) were cheap, simple to use and not limited by ethical regulation. Since then, similar results have been established with G. mellonella model comparatively to vertebrates, and it is more and more often used as a robust model per se, not only as an alternative to the murine model. This review attempts to summarize the current knowledge supporting the development of this model, both on immunological and microbiological aspects. For that, we focus on investigation of virulence and new therapies for the most important pathogenic bacteria. We also discuss points out directions for standardization, as well as recent advances and new perspectives for monitoring host-pathogen interactions.
Collapse
Affiliation(s)
- Guillaume Ménard
- Univ Rennes, CHU Rennes, INSERM, Bacterial Regulatory RNAs and Medicine (BRM), service de Bactériologie Hygiène-Hospitalière (SB2H), UMR_S 1230, Rennes, France
| | - Astrid Rouillon
- Univ Rennes, INSERM, Bacterial Regulatory RNAs and Medicine (BRM), UMR_S 1230, Rennes, France
| | - Vincent Cattoir
- Univ Rennes, CHU Rennes, INSERM, Bacterial Regulatory RNAs and Medicine (BRM), service de Bactériologie Hygiène-Hospitalière (SB2H), UMR_S 1230, Rennes, France
| | - Pierre-Yves Donnio
- Univ Rennes, CHU Rennes, INSERM, Bacterial Regulatory RNAs and Medicine (BRM), service de Bactériologie Hygiène-Hospitalière (SB2H), UMR_S 1230, Rennes, France
| |
Collapse
|
232
|
Bugs on Drugs: A Drosophila melanogaster Gut Model to Study In Vivo Antibiotic Tolerance of E. coli. Microorganisms 2022; 10:microorganisms10010119. [PMID: 35056568 PMCID: PMC8780219 DOI: 10.3390/microorganisms10010119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/23/2021] [Accepted: 01/05/2022] [Indexed: 02/04/2023] Open
Abstract
With an antibiotic crisis upon us, we need to boost antibiotic development and improve antibiotics’ efficacy. Crucial is knowing how to efficiently kill bacteria, especially in more complex in vivo conditions. Indeed, many bacteria harbor antibiotic-tolerant persisters, variants that survive exposure to our most potent antibiotics and catalyze resistance development. However, persistence is often only studied in vitro as we lack flexible in vivo models. Here, I explored the potential of using Drosophila melanogaster as a model for antimicrobial research, combining methods in Drosophila with microbiology techniques: assessing fly development and feeding, generating germ-free or bacteria-associated Drosophila and in situ microscopy. Adult flies tolerate antibiotics at high doses, although germ-free larvae show impaired development. Orally presented E. coli associates with Drosophila and mostly resides in the crop. E. coli shows an overall high antibiotic tolerance in vivo potentially resulting from heterogeneity in growth rates. The hipA7 high-persistence mutant displays an increased antibiotic survival while the expected low persistence of ΔrelAΔspoT and ΔrpoS mutants cannot be confirmed in vivo. In conclusion, a Drosophila model for in vivo antibiotic tolerance research shows high potential and offers a flexible system to test findings from in vitro assays in a broader, more complex condition.
Collapse
|
233
|
Bhaumik KN, Hetényi A, Olajos G, Martins A, Spohn R, Németh L, Jojart B, Szili P, Dunai A, Jangir PK, Daruka L, Földesi I, Kata D, Pál C, Martinek TA. Rationally designed foldameric adjuvants enhance antibiotic efficacy via promoting membrane hyperpolarization. MOLECULAR SYSTEMS DESIGN & ENGINEERING 2022; 7:21-33. [PMID: 35127141 PMCID: PMC8724909 DOI: 10.1039/d1me00118c] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/06/2021] [Indexed: 05/26/2023]
Abstract
The negative membrane potential of bacterial cells influences crucial cellular processes. Inspired by the molecular scaffold of the antimicrobial peptide PGLa, we have developed antimicrobial foldamers with a computer-guided design strategy. The novel PGLa analogues induce sustained membrane hyperpolarization. When co-administered as an adjuvant, the resulting compounds - PGLb1 and PGLb2 - have substantially reduced the level of antibiotic resistance of multi-drug resistant Escherichia coli, Klebsiella pneumoniae and Shigella flexneri clinical isolates. The observed antibiotic potentiation was mediated by hyperpolarization of the bacterial membrane caused by the alteration of cellular ion transport. Specifically, PGLb1 and PGLb2 are selective ionophores that enhance the Goldman-Hodgkin-Katz potential across the bacterial membrane. These findings indicate that manipulating bacterial membrane electrophysiology could be a valuable tool to overcome antimicrobial resistance.
Collapse
Affiliation(s)
- Kaushik Nath Bhaumik
- Department of Medical Chemistry, University of Szeged Dóm tér 8 Szeged HU-6720 Hungary
| | - Anasztázia Hetényi
- Department of Medical Chemistry, University of Szeged Dóm tér 8 Szeged HU-6720 Hungary
| | - Gábor Olajos
- Department of Medical Chemistry, University of Szeged Dóm tér 8 Szeged HU-6720 Hungary
| | - Ana Martins
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network (ELKH) Szeged Hungary
| | - Réka Spohn
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network (ELKH) Szeged Hungary
| | - Lukács Németh
- Institute of Food Engineering, University of Szeged Szeged Hungary
| | - Balázs Jojart
- Institute of Food Engineering, University of Szeged Szeged Hungary
| | - Petra Szili
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network (ELKH) Szeged Hungary
- Doctoral School of Multidisciplinary Medical Sciences, University of Szeged Szeged Hungary
| | - Anett Dunai
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network (ELKH) Szeged Hungary
| | - Pramod K Jangir
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network (ELKH) Szeged Hungary
| | - Lejla Daruka
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network (ELKH) Szeged Hungary
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged Szeged Hungary
| | - Imre Földesi
- Department of Laboratory Medicine, University of Szeged Szeged Hungary
| | - Diána Kata
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged Szeged Hungary
| | - Csaba Pál
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network (ELKH) Szeged Hungary
| | - Tamás A Martinek
- Department of Medical Chemistry, University of Szeged Dóm tér 8 Szeged HU-6720 Hungary
| |
Collapse
|
234
|
Development of a Phage Cocktail to Target Salmonella Strains Associated with Swine. Pharmaceuticals (Basel) 2022; 15:ph15010058. [PMID: 35056115 PMCID: PMC8777603 DOI: 10.3390/ph15010058] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/20/2021] [Accepted: 12/30/2021] [Indexed: 02/06/2023] Open
Abstract
Infections caused by multidrug resistant Salmonella strains are problematic in swine and are entering human food chains. Bacteriophages (phages) could be used to complement or replace antibiotics to reduce infection within swine. Here, we extensively characterised six broad host range lytic Salmonella phages, with the aim of developing a phage cocktail to prevent or treat infection. Intriguingly, the phages tested differed by one to five single nucleotide polymorphisms. However, there were clear phenotypic differences between them, especially in their heat and pH sensitivity. In vitro killing assays were conducted to determine the efficacy of phages alone and when combined, and three cocktails reduced bacterial numbers by ~2 × 103 CFU/mL within two hours. These cocktails were tested in larvae challenge studies, and prophylactic treatment with phage cocktail SPFM10-SPFM14 was the most efficient. Phage treatment improved larvae survival to 90% after 72 h versus 3% in the infected untreated group. In 65% of the phage-treated larvae, Salmonella counts were below the detection limit, whereas it was isolated from 100% of the infected, untreated larvae group. This study demonstrates that phages effectively reduce Salmonella colonisation in larvae, which supports their ability to similarly protect swine.
Collapse
|
235
|
Prakoso D, Zhu X, Rajeev S. Galleria mellonella infection model to evaluate pathogenic and nonpathogenic Leptospira strains. Vet Microbiol 2022; 264:109295. [PMID: 34875420 DOI: 10.1016/j.vetmic.2021.109295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 01/21/2023]
Abstract
The Galleria mellonella larvae infection model is emerging as a valuable tool for studying various characteristics of infectious agents and host-pathogen interaction. This system has been widely recognized as a high throughput, ethical, and cost-effective invertebrate infection model to study the virulence and pathogenesis of various bacterial pathogens. In this study, we compared the effect of Leptospira infection in G. mellonella larvae infected with Leptospira interrogans serovar Copenhageni (pathogenic) or Leptospira biflexa serovar Patoc (saprophytic) strains. We observed significant pathologic changes such as decreased activity, complete melanization, and lower survival rate in the G. mellonella larvae infected with a pathogenic strain L. interrogans serovar Copenhageni compared to those infected with a nonpathogenic strain L. biflexa serovar Patoc. Our study demonstrates the feasibility and the potential of using G. mellonella larvae as an alternative model to study virulence mechanisms and pathogenesis of Leptospira strains. Once optimized, the G. mellonella infection model can be a potential substitute for hamsters to explore various host and pathogen-related mechanistic events in Leptospira infection.
Collapse
Affiliation(s)
- Dhani Prakoso
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, 37996, Knoxville, TN, United States
| | - Xiaojuan Zhu
- Office of Information Technology, University of Tennessee, Knoxville, TN, United States
| | - Sreekumari Rajeev
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, 37996, Knoxville, TN, United States.
| |
Collapse
|
236
|
Zhang X, Shi S, Yao Z, Zheng X, Li W, Zhang Y, Wang L, Cao J, Zhou T. OUP accepted manuscript. J Antimicrob Chemother 2022; 77:1903-1911. [PMID: 35474013 DOI: 10.1093/jac/dkac128] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/25/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Xiaodong Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Shiyi Shi
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zhuocheng Yao
- Department of Medical Lab Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Xiangkuo Zheng
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Wangyang Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Ying Zhang
- Department of Medical Lab Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Lingbo Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jianming Cao
- Department of Medical Lab Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Tieli Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| |
Collapse
|
237
|
Wang S, Liu H, Mao J, Peng Y, Yan Y, Li Y, Zhang N, Jiang L, Liu Y, Li J, Huang X. Pharmacodynamics of Linezolid Plus Fosfomycin Against Vancomycin-Resistant Enterococcus faecium in a Hollow Fiber Infection Model. Front Microbiol 2022; 12:779885. [PMID: 34970238 PMCID: PMC8714187 DOI: 10.3389/fmicb.2021.779885] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/17/2021] [Indexed: 12/27/2022] Open
Abstract
The optimal therapy for severe infections caused by vancomycin-resistant Enterococcus faecium (VREfm) remains unclear, but the combination of linezolid and fosfomycin may be a good choice. The 24-h static-concentration time-kill study (SCTK) was used to preliminarily explore the pharmacodynamics of linezolid combined with fosfomycin against three clinical isolates. Subsequently, a hollow-fibre infection model (HFIM) was used for the first time to further investigate the pharmacodynamic activity of the co-administration regimen against selected isolates over 72 h. To further quantify the relationship between fosfomycin resistance and bacterial virulence in VREfm, the Galleria mellonella infection model and virulence genes expression experiments were also performed. The results of SCTK showed that the combination of linezolid and fosfomycin had additive effect on all strains. In the HFIM, the dosage regimen of linezolid (12 mg/L, steady-state concentration) combined with fosfomycin (8 g administered intravenously every 8 h as a 1 h infusion) not only produced a sustained bactericidal effect of 3∼4 log10 CFU/mL over 72 h, but also completely eradicated the resistant subpopulations. The expression of virulence genes was down-regulated to at least 0.222-fold in fosfomycin-resistant strains compared with baseline isolate, while survival rates of G. mellonella was increased (G. mellonella survival ≥45% at 72 h). For severe infections caused by VREfm, neither linezolid nor fosfomycin monotherapy regimens inhibited amplification of the resistant subpopulations, and the development of fosfomycin resistance was at the expense of the virulence of VREfm. The combination of linezolid with fosfomycin produced a sustained bactericidal effect and completely eradicated the resistant subpopulations. Linezolid plus Fosfomycin is a promising combination for therapy of severe infections caused by VREfm.
Collapse
Affiliation(s)
- Shuaishuai Wang
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Huiping Liu
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jun Mao
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Yu Peng
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Yisong Yan
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Yaowen Li
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Na Zhang
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Lifang Jiang
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Yanyan Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiabin Li
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaohui Huang
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| |
Collapse
|
238
|
Tiwari V, Panta PR, Billiot CE, Douglass MV, Herrera CM, Trent MS, Doerrler WT. A Klebsiella pneumoniae DedA family membrane protein is required for colistin resistance and for virulence in wax moth larvae. Sci Rep 2021; 11:24365. [PMID: 34934166 PMCID: PMC8692421 DOI: 10.1038/s41598-021-03834-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 12/10/2021] [Indexed: 12/15/2022] Open
Abstract
Ineffectiveness of carbapenems against multidrug resistant pathogens led to the increased use of colistin (polymyxin E) as a last resort antibiotic. A gene belonging to the DedA family encoding conserved membrane proteins was previously identified by screening a transposon library of K. pneumoniae ST258 for sensitivity to colistin. We have renamed this gene dkcA (dedA of Klebsiella required for colistin resistance). DedA family proteins are likely membrane transporters required for viability of Escherichia coli and Burkholderia spp. at alkaline pH and for resistance to colistin in a number of bacterial species. Colistin resistance is often conferred via modification of the lipid A component of bacterial lipopolysaccharide with aminoarabinose (Ara4N) and/or phosphoethanolamine. Mass spectrometry analysis of lipid A of the ∆dkcA mutant shows a near absence of Ara4N in the lipid A, suggesting a requirement for DkcA for lipid A modification with Ara4N. Mutation of K. pneumoniae dkcA resulted in a reduction of the colistin minimal inhibitory concentration to approximately what is found with a ΔarnT strain. We also identify a requirement of DkcA for colistin resistance that is independent of lipid A modification, instead requiring maintenance of optimal membrane potential. K. pneumoniae ΔdkcA displays reduced virulence in Galleria mellonella suggesting colistin sensitivity can cause loss of virulence.
Collapse
Affiliation(s)
- Vijay Tiwari
- grid.64337.350000 0001 0662 7451Department of Biological Sciences, Louisiana State University, Baton Rouge, LA USA
| | - Pradip R. Panta
- grid.64337.350000 0001 0662 7451Department of Biological Sciences, Louisiana State University, Baton Rouge, LA USA
| | - Caitlin E. Billiot
- grid.64337.350000 0001 0662 7451Department of Biological Sciences, Louisiana State University, Baton Rouge, LA USA
| | - Martin V. Douglass
- grid.213876.90000 0004 1936 738XDepartment of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA USA
| | - Carmen M. Herrera
- grid.213876.90000 0004 1936 738XDepartment of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA USA
| | - M. Stephen Trent
- grid.213876.90000 0004 1936 738XDepartment of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA USA
| | - William T. Doerrler
- grid.64337.350000 0001 0662 7451Department of Biological Sciences, Louisiana State University, Baton Rouge, LA USA
| |
Collapse
|
239
|
Gooch HCC, Kiu R, Rudder S, Baker DJ, Hall LJ, Maxwell A. Enterococcus innesii sp. nov., isolated from the wax moth Galleria mellonella. Int J Syst Evol Microbiol 2021; 71:005168. [PMID: 34919037 PMCID: PMC8744253 DOI: 10.1099/ijsem.0.005168] [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] [Indexed: 01/05/2023] Open
Abstract
Four bacterial strains were isolated from two different colony sources of the wax moth Galleria mellonella. They were characterized by a polyphasic approach including 16S rRNA gene sequence analysis, core-genome analysis, average nucleotide identity (ANI) analysis, digital DNA-DNA hybridization (dDDH), determination of G+C content, screening of antibiotic resistance genes, and various phenotypic analyses. Initial analysis of 16S rRNA gene sequence identities indicated that strain GAL7T was potentially very closely related to Enterococcus casseliflavus and Enterococcus gallinarum, having 99.5-99.9 % sequence similarity. However, further analysis of whole genome sequences revealed a genome size of 3.69 Mb, DNA G+C content of 42.35 mol%, and low dDDH and ANI values between the genomes of strain GAL7T and closest phylogenetic relative E. casseliflavus NBRC 100478T of 59.0 and 94.5 %, respectively, indicating identification of a putative new Enterococcus species. In addition, all novel strains encoded the atypical vancomycin-resistance gene vanC-4. Results of phylogenomic, physiological and phenotypic characterization confirmed that strain GAL7T represented a novel species within the genus Enterococcus, for which the name Enterococcus innesii sp. nov. is proposed. The type strain is GAL7T (=DSM 112306T=NCTC 14608T).
Collapse
Affiliation(s)
- Harriet C. C. Gooch
- Dept. Biochemisty & Metabolism, John Innes Centre, Norwich Research Park, Norwich, NR4 7UH, UK
| | - Raymond Kiu
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Steven Rudder
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - David J. Baker
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Lindsay J. Hall
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK,Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK,School of Life Sciences, ZIEL – Institute for Food &Health, Technical University of Munich, Freising, 85354, Germany,*Correspondence: Lindsay J. Hall,
| | - Anthony Maxwell
- Dept. Biochemisty & Metabolism, John Innes Centre, Norwich Research Park, Norwich, NR4 7UH, UK,*Correspondence: Anthony Maxwell,
| |
Collapse
|
240
|
Collins E, Martin C, Blomquist T, Phillips K, Cantlay S, Fisher N, Horzempa J. The utilization of Blaptica dubia cockroaches as an in vivo model to test antibiotic efficacy. Sci Rep 2021; 11:24004. [PMID: 34907348 PMCID: PMC8671488 DOI: 10.1038/s41598-021-03486-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 12/03/2021] [Indexed: 12/03/2022] Open
Abstract
Insects are now well recognized as biologically relevant alternative hosts for dozens of mammalian pathogens and they are routinely used in microbial pathogenesis studies. Unfortunately, these models have yet to be incorporated into the drug development pipeline. The purpose of this work was to begin to evaluate the utility of orange spotted (Blaptica dubia) cockroaches in early antibiotic characterization. To determine whether these model hosts could exhibit mortality when infected with bacteria that are pathogenic to humans, we subjected B. dubia roaches to a range of infectious doses of Klebsiella pneumoniae, Escherichia coli, Staphylococcus aureus, and Acinetobacter baumannii to identify the medial lethal dose. These results showed that lethal disease did not develop following infection of high doses of S. aureus, and A. baumannii. However, cockroaches infected with E. coli and K. pneumoniae succumbed to infection (LD50s of 5.82 × 106 and 2.58 × 106 respectively) suggesting that this model may have limitations based on pathogen specificity. However, because these cockroaches were susceptible to infection from E. coli and K. pneumoniae, we used these bacterial strains for subsequent antibiotic characterization studies. These studies suggested that β-lactam antibiotic persistence and dose was associated with reduction of hemolymph bacterial burden. Moreover, our data indicated that the reduction of bacterial CFU was directly due to the drug activity. Altogether, this work suggests that the orange-spotted cockroach infection model provides an alternative in vivo setting from which antibiotic efficacy can be evaluated.
Collapse
Affiliation(s)
- Elliot Collins
- Department of Biological Sciences, West Liberty University, West Liberty, WV, USA
| | - Caleb Martin
- Department of Biological Sciences, West Liberty University, West Liberty, WV, USA
| | - Tyler Blomquist
- Department of Biological Sciences, West Liberty University, West Liberty, WV, USA
| | - Katherine Phillips
- Department of Biological Sciences, West Liberty University, West Liberty, WV, USA
| | - Stuart Cantlay
- Department of Biological Sciences, West Liberty University, West Liberty, WV, USA
| | | | - Joseph Horzempa
- Department of Biological Sciences, West Liberty University, West Liberty, WV, USA.
| |
Collapse
|
241
|
Zhang X, Zhao Y, Feng L, Xu M, Ge Y, Wang L, Zhang Y, Cao J, Sun Y, Wu Q, Zhou T. Combined With Mefloquine, Resurrect Colistin Active in Colistin-Resistant Pseudomonas aeruginosa in vitro and in vivo. Front Microbiol 2021; 12:790220. [PMID: 34899672 PMCID: PMC8662342 DOI: 10.3389/fmicb.2021.790220] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/08/2021] [Indexed: 11/15/2022] Open
Abstract
Colistin is a polymyxin antibiotic that is widely used for the treatment of multidrug resistant (MDR) Pseudomonas aeruginosa infections, as the last resort. Over the past few years, unreasonable use of antibiotics has resulted in an increase in MDR strains, including colistin-resistant P. aeruginosa. The present study aimed to explore the synergistic effects of mefloquine in combination with colistin for the treatment of colistin-resistant P. aeruginosa in vivo and in vitro. The synergistic effect of the combination of mefloquine and colistin was investigated in vitro using checkerboard method, time-killing assay, biofilm formation inhibition test, and biofilm eradication test. The study also explored the synergistic effects of this combination of drugs in vivo, using a Galleria mellonella infection model. The results for checkerboard method and time killing curve indicated that mefloquine in combination with colistin showed a good antibacterial activity. Furthermore, the combination of these two drugs inhibited biofilm formation and eradicated pre-formed mature biofilms. This synergistic effect was visualized using scanning electron microscopy (SEM), wherein the results showed that the combination of mefloquine and colistin reduced biofilm formation significantly. Further, the application of this combination of drugs to in vivo infection model significantly increased the survival rate of G. mellonella larvae. Altogether, the combination of mefloquine and colistin showed a good synergistic effect in vitro and in vivo, and highlighted its potential to be used as an alternative therapy for the treatment of colistin-resistant P. aeruginosa infection.
Collapse
Affiliation(s)
- Xiaodong Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yining Zhao
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Luozhu Feng
- Department of Medical Lab Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Mengxin Xu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yiru Ge
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lingbo Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ying Zhang
- Department of Medical Lab Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Jianming Cao
- Department of Medical Lab Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Yao Sun
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qing Wu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tieli Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
242
|
Thees AV, Pietrosimone KM, Melchiorre CK, Marden JN, Graf J, Lynes MA, Maltz-Matyschsyk M. PmtA Regulates Pyocyanin Expression and Biofilm Formation in Pseudomonas aeruginosa. Front Microbiol 2021; 12:789765. [PMID: 34867928 PMCID: PMC8636135 DOI: 10.3389/fmicb.2021.789765] [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: 10/05/2021] [Accepted: 10/18/2021] [Indexed: 01/30/2023] Open
Abstract
The opportunistic pathogen Pseudomonas aeruginosa expresses a small molecular weight, cysteine-rich protein (PmtA), identified as a metallothionein (MT) protein family member. The MT family proteins have been well-characterized in eukaryotes as essential for zinc and copper homeostasis, protection against oxidative stress, and the ability to modify a variety of immune activities. Bacterial MTs share sequence homology, antioxidant chemistry, and heavy metal-binding capacity with eukaryotic MTs, however, the impact of bacterial MTs on virulence and infection have not been well-studied. In the present study, we investigated the role of PmtA in P. aeruginosa PAO1 using a PmtA-deficient strain (ΔpmtA). Here we demonstrated the virulence factor, pyocyanin, relies on the expression of PmtA. We showed that PmtA may be protective against oxidative stress, as an alternative antioxidant, glutathione, can rescue pyocyanin expression. Furthermore, the expression of phzM, which encodes a pyocyanin precursor enzyme, was decreased in the ΔpmtA mutant during early stationary phase. Upregulated pmtA expression was previously detected in confluent biofilms, which are essential for chronic infection, and we observed that the ΔpmtA mutant was disrupted for biofilm formation. As biofilms also modulate antibiotic susceptibility, we examined the ΔpmtA mutant susceptibility to antibiotics and found that the ΔpmtA mutant is more susceptible to cefepime and ciprofloxacin than the wild-type strain. Finally, we observed that the deletion of pmtA results in decreased virulence in a waxworm model. Taken together, our results support the conclusion that PmtA is necessary for the full virulence of P. aeruginosa and may represent a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Amy V Thees
- Department of Molecular and Cell Biology, University of Connecticut, Mansfield, CT, United States
| | - Kathryn M Pietrosimone
- Department of Molecular and Cell Biology, University of Connecticut, Mansfield, CT, United States
| | - Clare K Melchiorre
- Department of Molecular and Cell Biology, University of Connecticut, Mansfield, CT, United States
| | - Jeremiah N Marden
- Department of Molecular and Cell Biology, University of Connecticut, Mansfield, CT, United States
| | - Joerg Graf
- Department of Molecular and Cell Biology, University of Connecticut, Mansfield, CT, United States
| | - Michael A Lynes
- Department of Molecular and Cell Biology, University of Connecticut, Mansfield, CT, United States
| | - Michele Maltz-Matyschsyk
- Department of Molecular and Cell Biology, University of Connecticut, Mansfield, CT, United States
| |
Collapse
|
243
|
Cai W, Tang F, Jiang L, Li R, Wang Z, Liu Y. Histone-Like Nucleoid Structuring Protein Modulates the Fitness of tet(X4)-Bearing IncX1 Plasmids in Gram-Negative Bacteria. Front Microbiol 2021; 12:763288. [PMID: 34858374 PMCID: PMC8632487 DOI: 10.3389/fmicb.2021.763288] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 10/14/2021] [Indexed: 11/13/2022] Open
Abstract
The emergence of plasmid-mediated tigecycline resistance gene tet(X4) poses a challenging threat to public health. Based on the analysis of tet(X4)-positive plasmids in the NCBI database, we found that the IncX1-type plasmid is one of the most common vectors for spreading tet(X4) gene, but the mechanisms by which these plasmids adapt to host bacteria and maintain the persistence of antibiotic resistance genes (ARGs) remain unclear. Herein, we investigated the underlying mechanisms of how host bacteria modulate the fitness cost of IncX1 plasmids carrying tet(X4) gene. Interestingly, we found that the tet(X4)-bearing IncX1 plasmids encoding H-NS protein imposed low or no fitness cost in Escherichia coli and Klebsiella pneumoniae; instead, they partially promoted the virulence and biofilm formation in host bacteria. Regression analysis revealed that the expression of hns gene in plasmids was positively linked to the relative fitness of host bacteria. Furthermore, when pCE2::hns was introduced, the fitness of tet(X4)-positive IncX1 plasmid pRF55-1 without hns gene was significantly improved, indicating that hns mediates the improvement of fitness. Finally, we showed that the expression of hns gene is negatively correlated with the expression of tet(X4) gene, suggesting that the regulatory effect of H-NS on adaptability may be attributed to its inhibitory effect on the expression of ARGs. Together, our findings suggest the important role of plasmid-encoded H-NS protein in modulating the fitness of tet(X4)-bearing IncX1 plasmids, which shed new insight into the dissemination of tet(X4) gene in a biological environment.
Collapse
Affiliation(s)
- Wenhui Cai
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Feifei Tang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Lijie Jiang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Ruichao Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China.,Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| | - Zhiqiang Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Yuan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China.,Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| |
Collapse
|
244
|
Krachler AM, Sirisaengtaksin N, Monteith P, Paine CET, Coates CJ, Lim J. Defective phagocyte association during infection of Galleria mellonella with Yersinia pseudotuberculosis is detrimental to both insect host and microbe. Virulence 2021; 12:638-653. [PMID: 33550901 PMCID: PMC7889024 DOI: 10.1080/21505594.2021.1878672] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 11/03/2022] Open
Abstract
Adhesins facilitate bacterial colonization and invasion of host tissues and are considered virulence factors, but their impact on immune-mediated damage as a driver of pathogenesis remains unclear. Yersinia pseudotuberculosis encodes for a multivalent adhesion molecule (MAM), a mammalian cell entry (MCE) family protein and adhesin. MAMs are widespread in Gram-negative bacteria and enable enteric bacteria to colonize epithelial tissues. Their role in bacterial interactions with the host innate immune system and contribution to pathogenicity remains unclear. Here, we investigated howY. pseudotuberculosis MAM contributes to pathogenesis during infection of the Galleria mellonella insect model. We show that Y. pseudotuberculosis MAM is required for efficient bacterial binding and uptake by hemocytes, the host phagocytes. Y. pseudotuberculosis interactions with insect and mammalian phagocytes are determined by bacterial and host factors. Loss of MAM, and deficient microbe-phagocyte interaction, increased pathogenesis in G. mellonella. Diminished phagocyte association also led to increased bacterial clearance. Furthermore, Y. pseudotuberculosis that failed to engage phagocytes hyperactivated humoral immune responses, most notably melanin production. Despite clearing the pathogen, excessive melanization also increased phagocyte death and host mortality. Our findings provide a basis for further studies investigating how microbe- and host-factors integrate to drive pathogenesis in a tractable experimental system.
Collapse
Affiliation(s)
- Anne Marie Krachler
- Department of Microbiology and Molecular Genetics, University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Natalie Sirisaengtaksin
- Department of Microbiology and Molecular Genetics, University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Pauline Monteith
- Biological and Environmental Sciences, University of Stirling, Stirling, UK
| | - C. E. Timothy Paine
- School of Environmental and Rural Sciences, University of New England, Armidale, Australia
| | - Christopher J. Coates
- Department of Biosciences, College of Science, Swansea University, Swansea, Wales UK
| | - Jenson Lim
- Biological and Environmental Sciences, University of Stirling, Stirling, UK
| |
Collapse
|
245
|
Dijokaite A, Humbert MV, Borkowski E, La Ragione RM, Christodoulides M. Establishing an invertebrate Galleria mellonella greater wax moth larval model of Neisseria gonorrhoeae infection. Virulence 2021; 12:1900-1920. [PMID: 34304706 PMCID: PMC8312596 DOI: 10.1080/21505594.2021.1950269] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/21/2021] [Accepted: 06/26/2021] [Indexed: 11/24/2022] Open
Abstract
Neisseria gonorrhoeae (gonococcus) causes the human sexually transmitted disease gonorrhea. Studying gonococcal pathogenesis and developing new vaccines and therapies to combat the increasing prevalence of multi-antibiotic resistant bacteria has made use of many ex vivo models based on human cells and tissues, and in vivo vertebrate models, for example, rodent, pig and human. The focus of the current study was to examine the utility of the invertebrate greater wax moth Galleria mellonella as an in vivo model of gonococcal infection. We observed that a threshold of ~106 - 107 gonococci/larva was required to kill >50% of larvae (P < 0.05), and increased toxicity correlated with reduced health index scores and pronounced histopathological changes such as increases in the total lesion grade, melanized nodules, hemocyte reaction, and multifocal adipose body degeneration. Larval death was independent of the expression of pilus or Opa protein or LOS sialylation within a single gonococcal species studied, but the model could demonstrate relative toxicity of different isolates. N. meningitidis, N. lacatamica and gonococci all killed larvae equally, but were significantly less toxic (P > 0.05) than Pseudomonas aeruginosa. Larvae primed with nontoxic doses of gonococci were more susceptible to subsequent challenge with homologous and heterologous bacteria, and larval survival was significantly reduced (P < 0.05) in infected larvae after depletion of their hemocytes with clodronate-liposomes. The model was used to test the anti-gonococcal properties of antibiotics and novel antimicrobials. Ceftriaxone (P < 0.05) protected larvae from infection with different gonococcal isolates, but not azithromycin or monocaprin or ligand-coated silver nanoclusters (P > 0.05).
Collapse
Affiliation(s)
- Aiste Dijokaite
- Neisseria Research Group, Molecular Microbiology, Academic School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - Maria Victoria Humbert
- Neisseria Research Group, Molecular Microbiology, Academic School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - Emma Borkowski
- Department of Pathology and Infectious Diseases, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Roberto M La Ragione
- Department of Pathology and Infectious Diseases, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Myron Christodoulides
- Neisseria Research Group, Molecular Microbiology, Academic School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| |
Collapse
|
246
|
In Vivo Efficacy of Voriconazole in a Galleria mellonella Model of Invasive Infection Due to Azole-Susceptible or Resistant Aspergillus fumigatus Isolates. J Fungi (Basel) 2021; 7:jof7121012. [PMID: 34946994 PMCID: PMC8708373 DOI: 10.3390/jof7121012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/15/2021] [Accepted: 11/21/2021] [Indexed: 02/05/2023] Open
Abstract
Aspergillus fumigatus is an environmental filamentous fungus responsible for life-threatening infections in humans and animals. Azoles are the first-line treatment for aspergillosis, but in recent years, the emergence of azole resistance in A. fumigatus has changed treatment recommendations. The objective of this study was to evaluate the efficacy of voriconazole (VRZ) in a Galleria mellonella model of invasive infection due to azole-susceptible or azole-resistant A. fumigatus isolates. We also sought to describe the pharmacokinetics of VRZ in the G. mellonella model. G. mellonella larvae were infected with conidial suspensions of azole-susceptible and azole-resistant isolates of A. fumigatus. Mortality curves were used to calculate the lethal dose. Assessment of the efficacy of VRZ or amphotericin B (AMB) treatment was based on mortality in the lethal model and histopathologic lesions. The pharmacokinetics of VRZ were determined in larval hemolymph. Invasive fungal infection was obtained after conidial inoculation. A dose-dependent reduction in mortality was observed after antifungal treatment with AMB and VRZ. VRZ was more effective at treating larvae inoculated with azole-susceptible A. fumigatus isolates than larvae inoculated with azole-resistant isolates. The concentration of VRZ was maximal at the beginning of treatment and gradually decreased in the hemolymph to reach a Cmin (24 h) between 0.11 and 11.30 mg/L, depending on the dose. In conclusion, G. mellonella is a suitable model for testing the efficacy of antifungal agents against A. fumigatus.
Collapse
|
247
|
Wenholz DS, Miller M, Dawson C, Bhadbhade M, Black DS, Griffith R, Dinh H, Cain A, Lewis P, Kumar N. Inhibitors of bacterial RNA polymerase transcription complex. Bioorg Chem 2021; 118:105481. [PMID: 34801947 DOI: 10.1016/j.bioorg.2021.105481] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/02/2021] [Accepted: 11/07/2021] [Indexed: 01/28/2023]
Abstract
A series of hybrid compounds that incorporated anthranilic acid with activated 1H-indoles through a glyoxylamide linker were designed to target bacterial RNA polymerase holoenzyme formation using computational docking. Synthesis, in vitro transcription inhibition assays, and biological testing of the hybrids identified a range of potent anti-transcription inhibitors with activity against a range of pathogenic bacteria with MICs as low as 3.1 μM. A structure activity relationship study identified the key structural components necessary for inhibition of both bacterial growth and transcription. Correlation of in vitro transcription inhibition activity with in vivo mechanism of action was established using fluorescence microscopy and resistance passaging using Gram-positive bacteria showed no resistance development over 30 days. Furthermore, no toxicity was observed from the compounds in a wax moth larvae model, establishing a platform for the development of a series of new antibacterial drugs with an established mode of action.
Collapse
Affiliation(s)
- Daniel S Wenholz
- School of Chemistry, UNSW Sydney, Kensington, NSW 2502, Australia
| | - Michael Miller
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Catherine Dawson
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Mohan Bhadbhade
- Mark Wainwright Analytical Centre, UNSW Sydney, NSW 2052, Australia
| | - David StC Black
- School of Chemistry, UNSW Sydney, Kensington, NSW 2502, Australia
| | - Renate Griffith
- School of Chemistry, UNSW Sydney, Kensington, NSW 2502, Australia
| | - Hue Dinh
- Department of Biological Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Amy Cain
- Department of Biological Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Peter Lewis
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia; School of Chemistry and Molecular Bioscience, University of Wollongong and Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Naresh Kumar
- School of Chemistry, UNSW Sydney, Kensington, NSW 2502, Australia.
| |
Collapse
|
248
|
Li H, Li T, Zhang L, Hu Q, Liao X, Jiang Q, Qiu X, Li L, Draheim RR, Huang Q, Zhou R. Antimicrobial compounds from an FDA-approved drug library with activity against Streptococcus suis. J Appl Microbiol 2021; 132:1877-1886. [PMID: 34800069 DOI: 10.1111/jam.15377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/12/2021] [Accepted: 11/17/2021] [Indexed: 02/03/2023]
Abstract
AIM Antimicrobial resistance (AMR) has become a global concern. Developing novel antimicrobials is one of the most effective approaches in tackling AMR. Considering its relatively low cost and risk, drug repurposing has been proposed as a valuable approach for novel antimicrobial discovery. The aim of this study was to screen for antimicrobial compounds against Streptococcus suis, an important zoonotic bacterial pathogen, from an Food and Drug Administration (FDA)-approved drug library. METHODS AND RESULTS In this study, we tested the antimicrobial activity of 1815 FDA-approved drugs against S. suis. Sixty-seven hits were obtained that showed a growth inhibition of more than 98%. After excluding already known antibiotics and antiseptics, 12 compounds were subjected to minimal inhibition concentration (MIC) assessment against S. suis. This showed that pralatrexate, daunorubicin (hydrochloride), teniposide, aclacinomycin A hydrochloride and floxuridine gave a relatively low MIC, ranging from 0.85 to 5.25 μg/ml. Apart from pralatrexate, the remaining four drugs could also inhibit the growth of antimicrobial-resistant S. suis. It was also demonstrated that these four drugs had better efficacy against Gram-positive bacteria than Gram-negative bacteria. Cytotoxicity assays showed that floxuridine and teniposide had a relatively high 50% cytotoxic concentration (CC50 ). The in vivo efficacy of floxuridine was analysed using a Galleria mellonella larvae infection model, and the results showed that floxuridine was effective in treating S. suis infection in vivo. CONCLUSIONS Five compounds from the FDA-approved drug library showed high antimicrobial activity against S. suis, among which floxuridine displayed potent in vivo efficacy that is worth further development. SIGNIFICANCE AND IMPACT OF STUDY Our study identified several antimicrobial compounds that are effective against S. suis, which provides a valuable starting point for further antimicrobial development.
Collapse
Affiliation(s)
- Haotian Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Tingting Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Liangsheng Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qiao Hu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xia Liao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qinggen Jiang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiuxiu Qiu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Lu Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Cooperative Innovation Center of Sustainable Pig Production, Wuhan, China.,International Research Center for Animal Disease (Ministry of Science & Technology of China), Wuhan, China
| | - Roger R Draheim
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Qi Huang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Cooperative Innovation Center of Sustainable Pig Production, Wuhan, China.,International Research Center for Animal Disease (Ministry of Science & Technology of China), Wuhan, China
| | - Rui Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Cooperative Innovation Center of Sustainable Pig Production, Wuhan, China.,International Research Center for Animal Disease (Ministry of Science & Technology of China), Wuhan, China
| |
Collapse
|
249
|
Xu W, Fang Y, Hu Q, Zhu K. Emerging Risks in Food: Probiotic Enterococci Pose a Threat to Public Health through the Food Chain. Foods 2021; 10:foods10112846. [PMID: 34829127 PMCID: PMC8623795 DOI: 10.3390/foods10112846] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 11/16/2022] Open
Abstract
Probiotics have been associated with clinical infections, toxicity, and antimicrobial resistance transfer, raising public concerns. Probiotic enterococci are emerging food risks as opportunistic pathogens, yet little attention has been paid to them. Herein, we collected 88 enterococcal isolates from probiotic products used for humans, companion animals, livestock, and aquaculture. Results showed that all 88 probiotic enterococcal isolates harbored diverse virulence genes, multiple antimicrobial resistance genes, and mobile genetic elements. Notably, 77 isolates were highly resistant to gentamicin. Representative enterococcal isolates exerted toxic activities in both in vitro and in vivo models. Collectively, our findings suggest that probiotic enterococci may be harmful to hosts and pose a potential threat to public health.
Collapse
Affiliation(s)
| | | | | | - Kui Zhu
- Correspondence: ; Tel.: +86-10-62733695
| |
Collapse
|
250
|
Tao Y, Duma L, Rossez Y. Galleria mellonella as a Good Model to Study Acinetobacter baumannii Pathogenesis. Pathogens 2021; 10:1483. [PMID: 34832638 PMCID: PMC8623143 DOI: 10.3390/pathogens10111483] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 12/29/2022] Open
Abstract
The invertebrate model, Galleria mellonella, has been widely used to study host-pathogen interactions due to its cheapness, ease of handling, and similar mammalian innate immune system. G. mellonella larvae have been proven to be useful and a reliable model for analyzing pathogenesis mechanisms of multidrug resistant Acinetobacter baumannii, an opportunistic pathogen difficult to kill. This review describes the detailed experimental design of G. mellonella/A. baumannii models, and provides a comprehensive comparison of various virulence factors and therapy strategies using the G. mellonella host. These investigations highlight the importance of this host-pathogen model for in vivo pathogen virulence studies. On the long term, further development of the G. mellonella/A. baumannii model will offer promising insights for clinical treatments of A. baumannii infection.
Collapse
Affiliation(s)
- Ye Tao
- Université de Technologie de Compiègne, UPJV, UMR CNRS 7025, Enzyme and Cell Engineering, Centre de Recherche Royallieu–CS 60 319 , 60203 Compiègne, France; (Y.T.); (L.D.)
| | - Luminita Duma
- Université de Technologie de Compiègne, UPJV, UMR CNRS 7025, Enzyme and Cell Engineering, Centre de Recherche Royallieu–CS 60 319 , 60203 Compiègne, France; (Y.T.); (L.D.)
- Université de Reims Champagne-Ardenne, CNRS, ICMR UMR 7312, 51097 Reims, France
| | - Yannick Rossez
- Université Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| |
Collapse
|