151
|
The Cell Wall Deacetylases Spy1094 and Spy1370 Contribute to Streptococcus pyogenes Virulence. Microorganisms 2023; 11:microorganisms11020305. [PMID: 36838272 PMCID: PMC9966966 DOI: 10.3390/microorganisms11020305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/21/2023] [Accepted: 01/22/2023] [Indexed: 01/26/2023] Open
Abstract
Streptococcus pyogenes, or Group A Streptococcus (GAS), is a strictly human pathogen that causes a wide range of diseases, including skin and soft tissue infections, toxic shock syndrome and acute rheumatic fever. We have recently reported that Spy1094 and Spy1370 of S. pyogenes serotype M1 are N-acetylglucosamine (GlcNAc) deacetylases. We have generated spy1094 and spy1370 gene deletion mutants in S. pyogenes and gain-of-function mutants in Lactococcus lactis. Similar to other cell wall deacetylases, our results show that Spy1094 and Spy1370 confer lysozyme-resistance. Furthermore, deletion of the genes decreased S. pyogenes virulence in a human whole blood killing assay and a Galleria mellonella (Greater wax moth) larvae infection model. Expression of the two genes in L. lactis resulted in increased lysozyme resistance and survival in whole human blood, and reduced survival of infected G. mellonella larvae. Deletion of the spy1370, but not the spy1094 gene, decreased resistance to the cationic antimicrobial peptide cecropin B, whereas both enzymes increased biofilm formation, probably resulting from the increase in positive charges due to deacetylation of the cell wall. In conclusion, Spy1094 and Spy1370 are important S. pyogenes virulence factors and might represent attractive targets for the development of antibacterial agents.
Collapse
|
152
|
Tenebrio molitor as a Simple and Cheap Preclinical Pharmacokinetic and Toxicity Model. Int J Mol Sci 2023; 24:ijms24032296. [PMID: 36768618 PMCID: PMC9917132 DOI: 10.3390/ijms24032296] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/20/2023] [Accepted: 01/22/2023] [Indexed: 01/26/2023] Open
Abstract
The progression of drugs into clinical phases requires proper toxicity assessment in animals and the correct identification of possible metabolites. Accordingly, different animal models are used to preliminarily evaluate toxicity and biotransformations. Rodents are the most common models used to preliminarily evaluate the safety of drugs; however, their use is subject to ethical consideration and elevated costs, and strictly regulated by national legislations. Herein, we developed a novel, cheap and convenient toxicity model using Tenebrio molitor coleoptera (TMC). A panel of 15 drugs-including antivirals and antibacterials-with different therapeutic applications was administered to TMC and the LD50 was determined. The values are comparable with those already determined in mice and rats. In addition, a TMC model was used to determine the presence of the main metabolites and in vivo pharmacokinetics (PK), and results were compared with those available from in vitro assays and the literature. Taken together, our results demonstrate that TMC can be used as a novel and convenient preliminary toxicity model to preliminarily evaluate the safety of experimental compounds and the formation of main metabolites, and to reduce the costs and number of rodents, according to 3R principles.
Collapse
|
153
|
Antimicrobial Susceptibility Testing Performed in RPMI 1640 Reveals Azithromycin Efficacy against Carbapenem-Resistant Acinetobacter baumannii and Predicts In Vivo Outcomes in Galleria mellonella. Antimicrob Agents Chemother 2023; 67:e0132022. [PMID: 36468875 PMCID: PMC9872712 DOI: 10.1128/aac.01320-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
Abstract
Antimicrobial susceptibility testing (AST) in RPMI 1640, a more physiologically relevant culture medium, revealed that a substantial proportion of carbapenem-resistant Acinetobacter baumannii isolates were susceptible to azithromycin, a macrolide antibiotic not currently considered effective against A. baumannii. Experiments using Galleria mellonella validated these in vitro data. Our finding that RPMI 1640's predictive accuracy for in vivo outcomes is superior to that of Mueller-Hinton II broth also supports the use of more physiologically relevant AST culturing conditions.
Collapse
|
154
|
Loulou A, Mastore M, Caramella S, Bhat AH, Brivio MF, Machado RAR, Kallel S. Entomopathogenic potential of bacteria associated with soil-borne nematodes and insect immune responses to their infection. PLoS One 2023; 18:e0280675. [PMID: 36689436 PMCID: PMC10045567 DOI: 10.1371/journal.pone.0280675] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/06/2023] [Indexed: 01/24/2023] Open
Abstract
Soil-borne nematodes establish close associations with several bacterial species. Whether they confer benefits to their hosts has been investigated in only a few nematode-bacteria systems. Their ecological function, therefore, remains poorly understood. In this study, we isolated several bacterial species from rhabditid nematodes, molecularly identified them, evaluated their entomopathogenic potential on Galleria mellonella larvae, and measured immune responses of G. mellonella larvae to their infection. Bacteria were isolated from Acrobeloides sp., A. bodenheimeri, Heterorhabditis bacteriophora, Oscheius tipulae, and Pristionchus maupasi nematodes. They were identified as Acinetobacter sp., Alcaligenes sp., Bacillus cereus, Enterobacter sp., Kaistia sp., Lysinibacillus fusiformis, Morganella morganii subsp. morganii, Klebsiella quasipneumoniae subsp. quasipneumoniae, and Pseudomonas aeruginosa. All bacterial strains were found to be highly entomopathogenic as they killed at least 53.33% G. mellonella larvae within 72h post-infection, at a dose of 106 CFU/larvae. Among them, Lysinibacillus fusiformis, Enterobacter sp., Acinetobacter sp., and K. quasipneumoniae subsp. quasipneumoniae were the most entomopathogenic bacteria. Insects strongly responded to bacterial infection. However, their responses were apparently little effective to counteract bacterial infection. Our study, therefore, shows that bacteria associated with soil-borne nematodes have entomopathogenic capacities. From an applied perspective, our study motivates more research to determine the potential of these bacterial strains as biocontrol agents in environmentally friendly and sustainable agriculture.
Collapse
Affiliation(s)
- Ameni Loulou
- Department of Plant Health and Environment, Laboratory of Bio-Aggressor and Integrated Protection in Agriculture, National Agronomic Institute of Tunisia, University of Carthage, Tunis, Tunisia
| | - Maristella Mastore
- Department of Theoretical and Applied Sciences, Laboratory of Comparative Immunology and Parasitology, University of Insubria, Varese, Italy
| | - Sara Caramella
- Department of Theoretical and Applied Sciences, Laboratory of Comparative Immunology and Parasitology, University of Insubria, Varese, Italy
| | - Aashaq Hussain Bhat
- Faculty of Sciences, Experimental Biology Research Group, Institute of Biology, University of Neuchâtel, Neuchâtel, Switzerland
| | - Maurizio Francesco Brivio
- Department of Theoretical and Applied Sciences, Laboratory of Comparative Immunology and Parasitology, University of Insubria, Varese, Italy
| | - Ricardo A. R. Machado
- Faculty of Sciences, Experimental Biology Research Group, Institute of Biology, University of Neuchâtel, Neuchâtel, Switzerland
| | - Sadreddine Kallel
- Department of Plant Health and Environment, Laboratory of Bio-Aggressor and Integrated Protection in Agriculture, National Agronomic Institute of Tunisia, University of Carthage, Tunis, Tunisia
| |
Collapse
|
155
|
Schwenk GR, Glass AM, Ji HF, Ehrlich GD, Navas-Martin S, Król JE, Hall DC. Surfactant-Impregnated MOF-Coated Fabric for Antimicrobial Applications. ACS APPLIED BIO MATERIALS 2023; 6:238-245. [PMID: 36595712 DOI: 10.1021/acsabm.2c00860] [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: 01/05/2023]
Abstract
Since the onset of the SARS-CoV-2 pandemic, the world has witnessed over 617 million confirmed cases and more than 6.54 million confirmed deaths, but the actual totals are likely much higher. The virus has mutated at a significantly faster rate than initially projected, and positive cases continue to surge with the emergence of ever more transmissible variants. According to the CDC, and at the time of this manuscript submission, more than 77% of all current US cases are a result of the B.5 (omicron). The continued emergence of highly transmissible variants makes clear the need for more effective methods of mitigating disease spread. Herein, we have developed an antimicrobial fabric capable of destroying a myriad of microbes including betacoronaviruses. We have demonstrated the capability of this highly porous and nontoxic metal organic framework (MOF), γ-CD-MOF-1, to serve as a host for varied-length benzalkonium chlorides (BACs; active ingredient in Lysol). Molecular docking simulations predicted a binding affinity of up to -4.12 kcal·mol-1, which is comparable to that of other reported guest molecules for this MOF. Similar Raman spectra and powder X-ray diffraction patterns between the unloaded and loaded MOFs, accompanied by a decrease in the Brunauer-Emmett-Teller surface area from 616.20 and 155.55 m2 g-1 respectively, corroborate the suggested potential for pore occupation with BAC. The MOF was grown on polypropylene fabric, exposed to a BAC-loading bath, washed to remove excess BAC from the external surface, and evaluated for its microbicidal activity against various bacterial and viral classes. Significant antimicrobial character was observed against Pseudomonas aeruginosa, Staphylococcus aureus, Escherichia coli, bacteriophage, and betacoronavirus. This study shows that a common mask material (polypropylene) can be coated with BAC-loaded γ-CD-MOF-1 while maintaining the guest molecule's antimicrobial effects.
Collapse
Affiliation(s)
- Gregory R Schwenk
- Department of Chemistry, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Adam M Glass
- Department of Microbiology & Immunology, Philadelphia, Pennsylvania 19102, United States.,Center for Molecular Virology & Translational Neuroscience, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, United States.,Institute for Molecular Medicine & Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, United States
| | - Hai-Feng Ji
- Department of Chemistry, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Garth D Ehrlich
- Center for Advanced Microbial Processing, Center for Surgical Infections and Biofilms, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, United States.,Department of Microbiology & Immunology, Philadelphia, Pennsylvania 19102, United States
| | - Sonia Navas-Martin
- Department of Microbiology & Immunology, Philadelphia, Pennsylvania 19102, United States.,Center for Molecular Virology & Translational Neuroscience, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, United States.,Institute for Molecular Medicine & Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, United States
| | - Jarosław E Król
- Center for Advanced Microbial Processing, Center for Surgical Infections and Biofilms, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, United States.,Department of Microbiology & Immunology, Philadelphia, Pennsylvania 19102, United States.,Institute for Molecular Medicine & Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, United States
| | - Donald C Hall
- Center for Advanced Microbial Processing, Center for Surgical Infections and Biofilms, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, United States.,Department of Microbiology & Immunology, Philadelphia, Pennsylvania 19102, United States.,Institute for Molecular Medicine & Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, United States
| |
Collapse
|
156
|
Potentially Virulent Multi-Drug Resistant Escherichia fergusonii Isolated from Inanimate Surface in a Medical University: Omphisa fuscidentalis as an Alternative for Bacterial Virulence Determination. Diagnostics (Basel) 2023; 13:diagnostics13020279. [PMID: 36673089 PMCID: PMC9858318 DOI: 10.3390/diagnostics13020279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/29/2022] [Accepted: 01/01/2023] [Indexed: 01/14/2023] Open
Abstract
Multi-drug resistant (MDR) bacteria are becoming a worldwide problem due to limited options for treatment. Moreover, patients infected by MDR with highly virulent accessories are worsening the symptoms, even to the point of causing death. In this study, we isolated bacteria from 14 inanimate surfaces that could potentially be reservoirs for the spread of bacterial infections in the medical university. Blood agar media was used for bacterial isolation. The bacterial colony that showed hemolytic activities on each surface was tested for antimicrobial susceptibility against eight different antibiotics. We found that MDR bacterium, namely TB1, which was isolated from a toilet bowl, was non-susceptible to ampicillin, imipenem, chloramphenicol, amoxicillin-clavulanic acid, gentamicin, and tetracycline. Another MDR bacterium isolated from the mobile phone screen of security officers, namely HSO, was resistant to chloramphenicol, gentamicin, tetracycline, and cefixime. An in vivo virulence test of bacterial isolates used Omphisa fuscidentalis larvae as an alternative to Galleria mellonella larvae for the infection model. A virulence test of TB1 in O. fuscidentalis larvae revealed 20% survival in the bacterial density of 104 and 105 CFU/larvae; and 0% survival in the bacterial density of 106 CFU/larvae at 24 h after injection. Bacterial identification was performed for TB1 as a potential virulent isolate. Bacterial identification using partial 16s rRNA gene showed that TB1 exhibited 99.84% identity to Escherichia fergusonii 2611. This study concludes that TB1 is a potentially virulent MDR E. fergusonii isolated from toilet bowls at a medical university.
Collapse
|
157
|
Zhao Y, Feng L, Zhou B, Zhang X, Yao Z, Wang L, Wang Z, Zhou T, Chen L. A newly isolated bacteriophage vB8388 and its synergistic effect with aminoglycosides against multi-drug resistant Klebsiella oxytoca strain FK-8388. Microb Pathog 2023; 174:105906. [PMID: 36494020 DOI: 10.1016/j.micpath.2022.105906] [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: 08/21/2022] [Revised: 11/24/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022]
Abstract
The bacteriophage vB8388 can lyse multi-drug resistant Klebsiella oxytoca strain FK-8388 and maintain stability in a wide range of temperatures (from 4 °C to 80 °C) and pHs (3-11). Bioinformatics analysis showed that vB8388 is a linear double-stranded DNA virus that is 39,750 long with 50.65% G + C content and 44 putative open reading frames (ORFs). Phage vB8388 belongs to the family Autographviridae and possesses a non-contractile tail. The latency period of vB8388 was approximately 20 min. The combination of phage vB8388 and gentamicin, amikacin, or tobramycin could effectively inhibit the growth of K. oxytoca strain FK-8388, with a decrease of more than 4 log units within 12 h in vitro. Phage vB8388 showed a strong synergistic effect with gentamicin that could enhance the anti-biofilm effect of vB8388. The phage + gentamicin combination also showed synergy in vivo in the larval infection model of Galleria mellonella. In conclusion, the findings of this study suggest the potential of phage + antibiotic combination therapy to be used as an alternative therapeutic approach for treating infectious diseases caused by multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Yining Zhao
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang Province, China.
| | - Luozhu Feng
- Department of Medical Lab Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| | - Beibei Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang Province, China.
| | - Xiaodong Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, 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.
| | - Lingbo Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang Province, China.
| | - Zhongyong Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang Province, China.
| | - Tieli Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang Province, China.
| | - Lijiang Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
158
|
Ganbold M, Seo J, Wi YM, Kwon KT, Ko KS. Species identification, antibiotic resistance, and virulence in Enterobacter cloacae complex clinical isolates from South Korea. Front Microbiol 2023; 14:1122691. [PMID: 37032871 PMCID: PMC10076837 DOI: 10.3389/fmicb.2023.1122691] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/21/2023] [Indexed: 04/11/2023] Open
Abstract
This study aimed to identify the species of Enterobacter cloacae complex (ECC) isolates and compare the genotype, antibiotic resistance, and virulence among them. A total of 183 ECC isolates were collected from patients in eight hospitals in South Korea. Based on partial sequences of hsp60 and phylogenetic analysis, all ECC isolates were identified as nine species and six subspecies. Enterobacter hormaechei was the predominant species (47.0%), followed by Enterobacter kobei, Enterobacter asburiae, Enterobacter ludiwigii, and Enterobacter roggenkampii. Multilocus sequence typing analysis revealed that dissemination was not limited to a few clones, but E. hormaechei subsp. xiangfangensis, E. hormaechei subsp. steigerwaltii, and E. ludwigii formed large clonal complexes. Antibiotic resistance rates were different between the ECC species. In particular, E. asburiae, E. kobei, E. roggenkampii, and E. cloacae isolates were highly resistant to colistin, whereas most E. hormaechei and E. ludwigii isolates were susceptible to colistin. Virulence was evaluated through serum bactericidal assay and the Galleria mellonella larvae infection model. Consistency in the results between the serum resistance and the G. mellonella larvae infection assay was observed. Serum bactericidal assay showed that E. hormaechei, E. kobei, and E. ludwigii were significantly more virulent than E. asburiae and E. roggenkampii. In this study, we identified the predominant ECC species in South Korea and observed the differences in antibiotic resistance and virulence between the species. Our findings suggest that correct species identification, as well as continuous monitoring is crucial in clinical settings.
Collapse
Affiliation(s)
- Michidmaral Ganbold
- Department of Microbiology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Jungyu Seo
- Department of Microbiology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Yu Mi Wi
- Division of Infectious Diseases, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Republic of Korea
| | - Ki Tae Kwon
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Kwan Soo Ko
- Department of Microbiology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
- *Correspondence: Kwan Soo Ko,
| |
Collapse
|
159
|
Li X, Chen Z, Jiao J, Wang S, Wang Y, Wu W, Yang H, Lou H. In vitro and in vivo activity of meropenem+avibactam against MBL-producing carbapenem-resistant Klebsiella pneumoniae. Expert Rev Anti Infect Ther 2023; 21:91-98. [PMID: 36433637 DOI: 10.1080/14787210.2022.2153117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/23/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Antibiotic resistance has become a public health problem to be solved worldwide and metallo-β-lactamase (MBL)-producing bacteria make this problem even more challenging. METHODS The interactions of meropenem (MEM) in combination with avibactam (AVI) in growth inhibition on MBL-producing carbapenem-resistant Klebsiella pneumoniae (CRKP) strains were tested. In vitro interactions of MEM+AVI were tested using the microdilution checkerboard assay and time-kill curves. In vivo interactions of MEM+AVI were tested using the Galleria mellonella model. RESULTS All strains were multi-drug resistant strains and six of them were proved to produce MBLs. We show that the combination of MEM+AVI generates profound synergistic effects on growth inhibition of all strains, which was better than that of MEM+vaborbactam or imipenem+relebactam. The time-kill curves further confirmed the potent synergistic antibacterial effects of MEM+AVI against MBL-producing CRKP strains. Galleria mellonella studies were consistent with in vitro analysis. Combining MEM with AVI improved survival rates and mean survival days were obviously prolonged compared to the drug alone and the untreated controls. CONCLUSIONS To our knowledge, this study is the first report of MEM+AVI collaborating against MBL-producing CRKP strains. Our findings showed that the combination of MEM+AVI has the potential for antibiotic drug development to combat MBL-producing pathogens.
Collapse
Affiliation(s)
- Xiuyun Li
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Maternal and Child Health Development Research Center, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| | - Zhaowen Chen
- Medical Department, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| | - Jin Jiao
- Department of Clinical Laboratory, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| | - Shifu Wang
- Microbiology Laboratory, Children's Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Yuehua Wang
- Department of Pharmacy, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Weiwei Wu
- Department of Clinical Laboratory, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| | - Huijun Yang
- Reproductive Medicine Center, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| | - Hongxiang Lou
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
160
|
Updated Review on the Mechanisms of Pathogenicity in Mycobacterium abscessus, a Rapidly Growing Emerging Pathogen. Microorganisms 2022; 11:microorganisms11010090. [PMID: 36677382 PMCID: PMC9866562 DOI: 10.3390/microorganisms11010090] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 01/01/2023] Open
Abstract
In recent years, Mycobacterium abscessus has appeared as an emerging pathogen, with an increasing number of disease cases reported worldwide that mainly occur among patients with chronic lung diseases or impaired immune systems. The treatment of this pathogen represents a challenge due to the multi-drug-resistant nature of this species and its ability to evade most therapeutic approaches. However, although predisposing host factors for disease are well known, intrinsic pathogenicity mechanisms of this mycobacterium are still not elucidated. Like other mycobacteria, intracellular invasiveness and survival inside different cell lines are pathogenic factors related to the ability of M. abscessus to establish infection. Some of the molecular factors involved in this process are well-known and are present in the mycobacterial cell wall, such as trehalose-dimycolate and glycopeptidolipids. The ability to form biofilms is another pathogenic factor that is essential for the development of chronic disease and for promoting mycobacterial survival against the host immune system or different antibacterial treatments. This capability also seems to be related to glycopeptidolipids and other lipid molecules, and some studies have shown an intrinsic relationship between both pathogenic mechanisms. Antimicrobial resistance is also considered a mechanism of pathogenicity because it allows the mycobacterium to resist antimicrobial therapies and represents an advantage in polymicrobial biofilms. The recent description of hyperpathogenic strains with the potential interhuman transmission makes it necessary to increase our knowledge of pathogenic mechanisms of this species to design better therapeutic approaches to the management of these infections.
Collapse
|
161
|
Barkowsky G, Abt C, Pöhner I, Bieda A, Hammerschmidt S, Jacob A, Kreikemeyer B, Patenge N. Antimicrobial Activity of Peptide-Coupled Antisense Peptide Nucleic Acids in Streptococcus pneumoniae. Microbiol Spectr 2022; 10:e0049722. [PMID: 36321914 PMCID: PMC9784828 DOI: 10.1128/spectrum.00497-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 10/19/2022] [Indexed: 12/24/2022] Open
Abstract
Streptococcus pneumoniae is the most common cause of community-acquired pneumonia and is responsible for multiple other infectious diseases, such as meningitis and otitis media, in children. Resistance to penicillins, macrolides, and fluoroquinolones is increasing and, since the introduction of pneumococcal conjugate vaccines (PCVs), vaccine serotypes have been replaced by non-vaccine serotypes. Antisense peptide nucleic acids (PNAs) have been shown to reduce the growth of several pathogenic bacteria in various infection models. PNAs are frequently coupled to cell-penetrating peptides (CPPs) to improve spontaneous cellular PNA uptake. In this study, different CPPs were investigated for their capability to support translocation of antisense PNAs into S. pneumoniae. HIV-1 TAT- and (RXR)4XB-coupled antisense PNAs efficiently reduced the viability of S. pneumoniae strains TIGR4 and D39 in vitro. Two essential genes, gyrA and rpoB, were used as targets for antisense PNAs. Overall, the antimicrobial activity of anti-gyrA PNAs was higher than that of anti-rpoB PNAs. Target gene transcription levels in S. pneumoniae were reduced following antisense PNA treatment. The effect of HIV-1 TAT- and (RXR)4XB-anti-gyrA PNAs on pneumococcal survival was also studied in vivo using an insect infection model. Treatment increased the survival of infected Galleria mellonella larvae. Our results represent a proof of principle and may provide a basis for the development of efficient antisense molecules for treatment of S. pneumoniae infections. IMPORTANCE Streptococcus pneumoniae is the most common cause of community-acquired pneumonia and is responsible for the deaths of up to 2 million children each year. Antibiotic resistance and strain replacement by non-vaccine serotypes are growing problems. For this reason, S. pneumoniae has been added to the WHO "global priority list" of antibiotic-resistant bacteria for which novel antimicrobials are most urgently needed. In this study, we investigated whether CPP-coupled antisense PNAs show antibacterial activity in S. pneumoniae. We demonstrated that HIV-1 TAT- and (RXR)4XB-coupled antisense PNAs were able to kill S. pneumoniae in vitro. The specificity of the antimicrobial effect was verified by reduced target gene transcription levels in S. pneumoniae. Moreover, CPP-antisense PNA treatment increased the survival rate of infected Galleria mellonella larvae in vivo. Based on these results, we believe that efficient antisense PNAs can be developed for the treatment of S. pneumoniae infections.
Collapse
Affiliation(s)
- Gina Barkowsky
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Corina Abt
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Irina Pöhner
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Adam Bieda
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Anette Jacob
- Peps4LS GmbH, Heidelberg, Germany
- Functional Genome Analysis, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Bernd Kreikemeyer
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Nadja Patenge
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| |
Collapse
|
162
|
Dzianach PA, Pérez-Reche FJ, Strachan NJC, Forbes KJ, Dykes GA. The Use of Interdisciplinary Approaches to Understand the Biology of Campylobacter jejuni. Microorganisms 2022; 10:2498. [PMID: 36557751 PMCID: PMC9786101 DOI: 10.3390/microorganisms10122498] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Campylobacter jejuni is a bacterial pathogen recognised as a major cause of foodborne illness worldwide. While Campylobacter jejuni generally does not grow outside its host, it can survive outside of the host long enough to pose a health concern. This review presents an up-to-date description and evaluation of biological, mathematical, and statistical approaches used to understand the behaviour of this foodborne pathogen and suggests future avenues which can be explored. Specifically, the incorporation of mathematical modelling may aid the understanding of C. jejuni biofilm formation both outside and inside the host. Predictive studies may be improved by the introduction of more standardised protocols for assessments of disinfection methods and by assessment of novel physical disinfection strategies as well as assessment of the efficiency of plant extracts on C. jejuni eradication. A full description of the metabolic pathways of C. jejuni, which is needed for the successful application of metabolic models, is yet to be achieved. Finally, a shift from animal models (except for those that are a source of human campylobacteriosis) to human-specific data may be made possible due to recent technological advancements, and this may lead to more accurate predictions of human infections.
Collapse
Affiliation(s)
- Paulina A. Dzianach
- Geospatial Health and Development, Telethon Kids Institute, Perth 6009, Australia
| | | | - Norval J. C. Strachan
- School of Natural and Computing Sciences, University of Aberdeen, Aberdeen AB24 3FX, UK
| | - Ken J. Forbes
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB24 3FX, UK
| | - Gary A. Dykes
- School of Agriculture and Food Sciences, University of Queensland, Brisbane 4072, Australia
| |
Collapse
|
163
|
Nale JY, Thanki AM, Rashid SJ, Shan J, Vinner GK, Dowah ASA, Cheng JKJ, Sicheritz-Pontén T, Clokie MRJ. Diversity, Dynamics and Therapeutic Application of Clostridioides difficile Bacteriophages. Viruses 2022; 14:v14122772. [PMID: 36560776 PMCID: PMC9784644 DOI: 10.3390/v14122772] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/08/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
Clostridioides difficile causes antibiotic-induced diarrhoea and pseudomembranous colitis in humans and animals. Current conventional treatment relies solely on antibiotics, but C. difficile infection (CDI) cases remain persistently high with concomitant increased recurrence often due to the emergence of antibiotic-resistant strains. Antibiotics used in treatment also induce gut microbial imbalance; therefore, novel therapeutics with improved target specificity are being investigated. Bacteriophages (phages) kill bacteria with precision, hence are alternative therapeutics for the targeted eradication of the pathogen. Here, we review current progress in C. difficile phage research. We discuss tested strategies of isolating C. difficile phages directly, and via enrichment methods from various sample types and through antibiotic induction to mediate prophage release. We also summarise phenotypic phage data that reveal their morphological, genetic diversity, and various ways they impact their host physiology and pathogenicity during infection and lysogeny. Furthermore, we describe the therapeutic development of phages through efficacy testing in different in vitro, ex vivo and in vivo infection models. We also discuss genetic modification of phages to prevent horizontal gene transfer and improve lysis efficacy and formulation to enhance stability and delivery of the phages. The goal of this review is to provide a more in-depth understanding of C. difficile phages and theoretical and practical knowledge on pre-clinical, therapeutic evaluation of the safety and effectiveness of phage therapy for CDI.
Collapse
Affiliation(s)
- Janet Y. Nale
- Centre for Epidemiology and Planetary Health, Department of Veterinary and Animal Science, Scotland’s Rural College, Inverness IV2 5NA, UK
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Anisha M. Thanki
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Srwa J. Rashid
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Jinyu Shan
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Gurinder K. Vinner
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
| | - Ahmed S. A. Dowah
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
- School of Pharmacy, University of Lincoln, Lincoln LN6 7TS, UK
| | | | - Thomas Sicheritz-Pontén
- Center for Evolutionary Hologenomics, The Globe Institute, University of Copenhagen, 1353 Copenhagen, Denmark
- Centre of Excellence for Omics-Driven Computational Biodiscovery, AIMST University, Bedong 08100, Kedah, Malaysia
| | - Martha R. J. Clokie
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK
- Correspondence:
| |
Collapse
|
164
|
Guevara MA, Francis JD, Lu J, Manning SD, Doster RS, Moore RE, Gaddy JA. Streptococcus agalactiae cadD Is Critical for Pathogenesis in the Invertebrate Galleria mellonella Model. ACS Infect Dis 2022; 8:2405-2412. [PMID: 36445344 PMCID: PMC10262471 DOI: 10.1021/acsinfecdis.2c00453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Group B Streptococcus (GBS) is a gram-positive bacterium that can cause invasive infections in immunocompromised, elderly, pregnant, or neonatal patients. The invertebrate model, Galleria mellonella, has emerged as an effective tool to study GBS-host interactions; specifically, those conserved within the innate arm of the immune system. We sought to determine the role of metal homeostasis functions in GBS infections of G. mellonella larvae and to validate this model as a tool to study GBS-host interactions. Our results indicate that wild-type GBS infects G. mellonella in a dose-dependent manner, replicates in the invertebrate host, induces larval melanization and larval killing. These results were significantly abrogated in cohorts of larvae infected with the isogenic cadD deletion mutant. Additionally, complementation restored GBS-dependent infection, bacterial burden, larval melanization, and killing to wild-type levels. Together, these results indicate that the G. mellonella model is a useful tool for studying GBS pathogenesis.
Collapse
Affiliation(s)
- Miriam A. Guevara
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, U.S.A
| | - Jamisha D. Francis
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, U.S.A
| | - Jacky Lu
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, U.S.A
| | - Shannon D. Manning
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, 48864, U.S.A
| | - Ryan S. Doster
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, 37232, U.S.A
| | - Rebecca E. Moore
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, U.S.A
| | - Jennifer A. Gaddy
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, U.S.A
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, 37232, U.S.A
- Tennessee Valley Healthcare Systems, Department of Veterans Affairs, Nashville, Tennessee, 37212, U.S.A
| |
Collapse
|
165
|
Starr AM, Zabet-Moghaddam M, San Francisco M. Identification of a novel secreted metabolite cyclo(phenylalanyl-prolyl) from Batrachochytrium dendrobatidis and its effect on Galleria mellonella. BMC Microbiol 2022; 22:293. [PMID: 36482304 PMCID: PMC9730576 DOI: 10.1186/s12866-022-02680-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 10/26/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The fungus, Batrachochytrium dendrobatidis, is the causative agent of chytridiomycosis and a leading cause of global decline in amphibian populations. The first stages of chytridiomycosis include: inflammation, hyperkeratosis, lethargy, loss of righting reflex, and disruption of internal electrolyte levels leading to eventual death of the host. Previous work indicates that B. dendrobatidis can produce immunomodulatory compounds and other secreted molecules that regulate the growth of the fungus. In this study, filtrates of the fungus grown in media and water were subjected to ultra-performance liquid chromatography-mass spectrometry and analyzed using Compound Discoverer 3.0. RESULTS Identification of cyclo(phenylalanyl-prolyl), chitobiose, and S-adenosylmethionine were verified by their retention times and fragmentation patterns from B. dendrobatidis supernatants. Previous studies have analyzed the effects of B. dendrobatidis on amphibian models, in vitro, or in cell culture. We studied the effects of live B. dendrobatidis cells, spent culture filtrates containing secreted metabolites, and cyclo(pheylalanyl-prolyl) on wax moth larvae (Galleria mellonella). Concentrated filtrates caused melanization within 24 h, while live B. dendrobatidis caused melanization within 48 h. CONCLUSIONS Here we show B. dendrobatidis produces secreted metabolites previously unreported. The impacts of these chemicals were tested on an alternate non-amphibian model system that has been used for other fungi to study pathogenicity traits in this fungus.
Collapse
Affiliation(s)
- Amanda M. Starr
- grid.462127.4Bryant & Stratton College, 8141 Hull Street Road, Richmond, VA 23235 USA ,grid.264784.b0000 0001 2186 7496Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409-3131 USA
| | | | - Michael San Francisco
- grid.264784.b0000 0001 2186 7496Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409-3131 USA
| |
Collapse
|
166
|
Lycopene, Mesoporous Silica Nanoparticles and Their Association: A Possible Alternative against Vulvovaginal Candidiasis? Molecules 2022; 27:molecules27238558. [PMID: 36500650 PMCID: PMC9738730 DOI: 10.3390/molecules27238558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/24/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Commonly found colonizing the human microbiota, Candida albicans is a microorganism known for its ability to cause infections, mainly in the vulvovaginal region known as vulvovaginal candidiasis (VVC). This pathology is, in fact, one of the main C. albicans clinical manifestations, changing from a colonizer to a pathogen. The increase in VVC cases and limited antifungal therapy make C. albicans an increasingly frequent risk in women's lives, especially in immunocompromised patients, pregnant women and the elderly. Therefore, it is necessary to develop new therapeutic options, especially those involving natural products associated with nanotechnology, such as lycopene and mesoporous silica nanoparticles. From this perspective, this study sought to assess whether lycopene, mesoporous silica nanoparticles and their combination would be an attractive product for the treatment of this serious disease through microbiological in vitro tests and acute toxicity tests in an alternative in vivo model of Galleria mellonella. Although they did not show desirable antifungal activity for VVC therapy, the present study strongly encourages the use of mesoporous silica nanoparticles impregnated with lycopene for the treatment of other human pathologies, since the products evaluated here did not show toxicity in the in vivo test performed, being therefore, a topic to be further explored.
Collapse
|
167
|
Barton TE, Frost F, Fothergill JL, Neill DR. Challenges and opportunities in the development of novel antimicrobial therapeutics for cystic fibrosis. J Med Microbiol 2022; 71. [PMID: 36748497 DOI: 10.1099/jmm.0.001643] [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: 12/24/2022] Open
Abstract
Chronic respiratory infection is the primary driver of mortality in individuals with cystic fibrosis (CF). Existing drug screening models utilised in preclinical antimicrobial development are unable to mimic the complex CF respiratory environment. Consequently, antimicrobials showing promising activity in preclinical models often fail to translate through to clinical efficacy in people with CF. Model systems used in CF anti-infective drug discovery and development range from antimicrobial susceptibility testing in nutrient broth, through to 2D and 3D in vitro tissue culture systems and in vivo models. No single model fully recapitulates every key aspect of the CF lung. To improve the outcomes of people with CF (PwCF) it is necessary to develop a set of preclinical models that collectively recapitulate the CF respiratory environment to a high degree of accuracy. Models must be validated for their ability to mimic aspects of the CF lung and associated lung infection, through evaluation of biomarkers that can also be assessed following treatment in the clinic. This will give preclinical models greater predictive power for identification of antimicrobials with clinical efficacy. The landscape of CF is changing, with the advent of modulator therapies that correct the function of the CFTR protein, while antivirulence drugs and phage therapy are emerging alternative treatments to chronic infection. This review discusses the challenges faced in current antimicrobial development pipelines, including the advantages and disadvantages of current preclinical models and the impact of emerging treatments.
Collapse
Affiliation(s)
- Thomas E Barton
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Ronald Ross Building, 8 West Derby Street, Liverpool, L69 7BE, UK
| | - Frederick Frost
- Adult Cystic Fibrosis Centre, Liverpool Heart & Chest Hospital NHS Foundation Trust, Liverpool, UK.,Liverpool Centre for Cardiovascular Sciences, University of Liverpool, Liverpool, UK
| | - Joanne L Fothergill
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Ronald Ross Building, 8 West Derby Street, Liverpool, L69 7BE, UK
| | - Daniel R Neill
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Ronald Ross Building, 8 West Derby Street, Liverpool, L69 7BE, UK
| |
Collapse
|
168
|
Moraes RM, Garcia MT, Stossi F, de Barros PP, Junqueira JC, Anbinder AL. Effects of α and β-adrenergic signaling on innate immunity and Porphyromonas gingivalis virulence in an invertebrate model. Virulence 2022; 13:1614-1630. [PMID: 36121102 PMCID: PMC9487758 DOI: 10.1080/21505594.2022.2123302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
To investigate the role of adrenergic signalling (AS) in the host immune response and Porphyromonas gingivalis virulence, we compared norepinephrine (NE) and isoproterenol (ISO) responses in Galleria mellonella. P. gingivalis infection was evaluated by survival; humoral immune responses (i.e. melanization and cecropin and gloverin mRNA expression); cellular immune responses (i.e. haemocyte count, nodulation by histology); and P. gingivalis recovery (CFU/mL). P. gingivalis was cultivated in the presence of ISO (PgISO) or NE and injected into the larvae for survival evaluation. Finally, we co-injected ISO and PgISO to evaluate the concomitant effects on the immune response and bacterial virulence. None of the ligands were toxic to the larvae; ISO increased haemocyte number, even after P. gingivalis infection, by mobilizing sessile haemocytes in a β-adrenergic-specific manner, while NE showed the opposite effect. ISO treatment reduced larval mortality and the number of recovered bacteria, while NE increased mortality and showed no effect on bacterial recovery. ISO and NE had similar effects on melanization and decreased the expression of cecropin. Although co-cultivation with NE and ISO increased the gene expression of bacterial virulence factors in vitro, only the injection of PgISO increased larval death, which was partially reversed by circulating ISO. Therefore, α- and β-adrenergic signalling had opposite effects after P. gingivalis infection. Ultimately, the catecholamine influence on the immune response overcame the effect of more virulent strains. The effect of AS directly on the pathogen found in vitro did not translate to the in vivo setting.
Collapse
Affiliation(s)
- Renata Mendonça Moraes
- Institute of Science and Technology, Biosciences and Diagnosis Department, São Paulo State University (Unesp), São José dos Campos, SP, Brazil
| | - Maíra Terra Garcia
- Institute of Science and Technology, Biosciences and Diagnosis Department, São Paulo State University (Unesp), São José dos Campos, SP, Brazil
| | - Fabio Stossi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA.,GCC Center for Advanced Microscopy and Image Informatics, Houston, Texas, USA
| | - Patrícia Pimentel de Barros
- Institute of Science and Technology, Biosciences and Diagnosis Department, São Paulo State University (Unesp), São José dos Campos, SP, Brazil.,Multicampi School of Medical Sciences, Federal University of Rio Grande do Norte (UFRN), Caicó, RN, Brazil
| | - Juliana Campos Junqueira
- Institute of Science and Technology, Biosciences and Diagnosis Department, São Paulo State University (Unesp), São José dos Campos, SP, Brazil
| | - Ana Lia Anbinder
- Institute of Science and Technology, Biosciences and Diagnosis Department, São Paulo State University (Unesp), São José dos Campos, SP, Brazil
| |
Collapse
|
169
|
Xu M, Qian C, Jia H, Feng L, Shi S, Zhang Y, Wang L, Cao J, Zhou T, Zhou C. Emergence of Ceftazidime-Avibactam Resistance and Decreased Virulence in Carbapenem-Resistant ST11 Klebsiella pneumoniae During Antibiotics Treatment. Infect Drug Resist 2022; 15:6881-6891. [PMID: 36465808 PMCID: PMC9717589 DOI: 10.2147/idr.s387187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/23/2022] [Indexed: 08/27/2023] Open
Abstract
Introduction Carbapenem-resistant Klebsiella pneumoniae (CRKP) poses a serious threat to human public health. Ceftazidime-avibactam (CZA) is currently one of the few effective antibiotics for carbapenem-resistant Enterobacteriaceae (CRE). Methods and Results Here, we analyzed two longitudinal Klebsiella pneumoniae clinical isolates (FK8578, FK8695) that were isolated from an ICU patient during antimicrobial treatment. Broth microdilution method, whole-genome sequencing (WGS) and comparative genomic analysis were used to elucidate the dynamics and mechanisms of antibiotic resistance. String test, quantification of capsule, biofilm inhibition test and Galleria mellonella (G. mellonella) infection model were used to explore the changes in virulence of the two clinical isolates. During antibiotic treatment, CRKP FK8578 underwent a series of drug resistance and virulence changes, including CZA resistance, carbapenem susceptibility and virulence attenuation. The results of WGS showed that mutation of bla KPC-2 to bla KPC-33 was responsible for the change of drug resistance phenotype between FK8578 and FK8695. pLVPK-like virulence plasmid without siderophore synthesis operon was identified in the two strains. On the other hand, the loss of hypermucoviscosity phenotype in the FK8695 strain may be related to a single nucleotide deletion of the rmpA gene, which would further lead to a decrease in virulence. Virulence results showed that compared with FK8578, FK8695 was negative in the string test, with decreased capsular production, smaller amounts of biofilm formation and higher survival rate of G. mellonella. Conclusion This is the first report of CZA resistance and decreased virulence in ST11 CRKP strains during antimicrobial treatment. It is urgent to monitor CZA resistance and timely adjust anti-infective treatment strategies.
Collapse
Affiliation(s)
- Mengxin Xu
- Department of Clinical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, People’s Republic of China
| | - Changrui Qian
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Huaiyu Jia
- Department of Clinical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, People’s Republic of China
| | - Luozhu Feng
- Department of Medical Laboratory Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Shiyi Shi
- Department of Clinical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, People’s Republic of China
| | - Ying Zhang
- Department of Medical Laboratory Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Lingbo Wang
- Department of Clinical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, People’s Republic of China
| | - Jianming Cao
- Department of Medical Laboratory Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Tieli Zhou
- Department of Clinical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, People’s Republic of China
| | - Cui Zhou
- Department of Clinical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Wenzhou, People’s Republic of China
| |
Collapse
|
170
|
Quansah E, Ramoji A, Thieme L, Mirza K, Goering B, Makarewicz O, Heutelbeck A, Meyer-Zedler T, Pletz MW, Schmitt M, Popp J. Label-free multimodal imaging of infected Galleria mellonella larvae. Sci Rep 2022; 12:20416. [PMID: 36437287 PMCID: PMC9701796 DOI: 10.1038/s41598-022-24846-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 11/21/2022] [Indexed: 11/28/2022] Open
Abstract
Non-linear imaging modalities have enabled us to obtain unique morpho-chemical insights into the tissue architecture of various biological model organisms in a label-free manner. However, these imaging techniques have so far not been applied to analyze the Galleria mellonella infection model. This study utilizes for the first time the strength of multimodal imaging techniques to explore infection-related changes in the Galleria mellonella larvae due to massive E. faecalis bacterial infection. Multimodal imaging techniques such as fluorescent lifetime imaging (FLIM), coherent anti-Stokes Raman scattering (CARS), two-photon excited fluorescence (TPEF), and second harmonic generation (SHG) were implemented in conjunction with histological HE images to analyze infection-associated tissue damage. The changes in the larvae in response to the infection, such as melanization, vacuolization, nodule formation, and hemocyte infiltration as a defense mechanism of insects against microbial pathogens, were visualized after Enterococcus faecalis was administered. Furthermore, multimodal imaging served for the analysis of implant-associated biofilm infections by visualizing biofilm adherence on medical stainless steel and ePTFE implants within the larvae. Our results suggest that infection-related changes as well as the integrity of the tissue of G. mellonella larvae can be studied with high morphological and chemical contrast in a label-free manner.
Collapse
Affiliation(s)
- Elsie Quansah
- grid.9613.d0000 0001 1939 2794Institute of Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Friedrich-Schiller-University Jena, Helmholtzweg 4, 07743 Jena, Germany ,grid.418907.30000 0004 0563 7158Leibniz Institute of Photonic Technology, Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745 Jena, Germany
| | - Anuradha Ramoji
- grid.9613.d0000 0001 1939 2794Institute of Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Friedrich-Schiller-University Jena, Helmholtzweg 4, 07743 Jena, Germany ,grid.418907.30000 0004 0563 7158Leibniz Institute of Photonic Technology, Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745 Jena, Germany ,grid.9613.d0000 0001 1939 2794Jena University Hospital, Center for Sepsis Control and Care (CSCC), Friedrich-Schiller-University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Lara Thieme
- grid.9613.d0000 0001 1939 2794Jena University Hospital, Institute of Infectious Diseases and Infection Control, Friedrich-Schiller-University Jena, Am Klinikum 1, 07747 Jena, Germany ,grid.9613.d0000 0001 1939 2794Jena University Hospital, Leibniz Center for Photonics in Infection Research, Friedrich Schiller University Jena, 07747 Jena, Germany
| | - Kamran Mirza
- grid.9613.d0000 0001 1939 2794Jena University Hospital, Institute of Infectious Diseases and Infection Control, Friedrich-Schiller-University Jena, Am Klinikum 1, 07747 Jena, Germany ,grid.9613.d0000 0001 1939 2794Jena University Hospital, Leibniz Center for Photonics in Infection Research, Friedrich Schiller University Jena, 07747 Jena, Germany
| | - Bianca Goering
- grid.9613.d0000 0001 1939 2794ena University Hospital, Institute for Occupational, Social, and Environmental Medicine, J, Friedrich-Schiller-University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Oliwia Makarewicz
- grid.9613.d0000 0001 1939 2794Jena University Hospital, Center for Sepsis Control and Care (CSCC), Friedrich-Schiller-University Jena, Am Klinikum 1, 07747 Jena, Germany ,grid.9613.d0000 0001 1939 2794Jena University Hospital, Institute of Infectious Diseases and Infection Control, Friedrich-Schiller-University Jena, Am Klinikum 1, 07747 Jena, Germany ,grid.9613.d0000 0001 1939 2794Jena University Hospital, Leibniz Center for Photonics in Infection Research, Friedrich Schiller University Jena, 07747 Jena, Germany
| | - Astrid Heutelbeck
- grid.9613.d0000 0001 1939 2794ena University Hospital, Institute for Occupational, Social, and Environmental Medicine, J, Friedrich-Schiller-University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Tobias Meyer-Zedler
- grid.9613.d0000 0001 1939 2794Institute of Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Friedrich-Schiller-University Jena, Helmholtzweg 4, 07743 Jena, Germany ,grid.418907.30000 0004 0563 7158Leibniz Institute of Photonic Technology, Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745 Jena, Germany
| | - Mathias W. Pletz
- grid.9613.d0000 0001 1939 2794Jena University Hospital, Center for Sepsis Control and Care (CSCC), Friedrich-Schiller-University Jena, Am Klinikum 1, 07747 Jena, Germany ,grid.9613.d0000 0001 1939 2794Jena University Hospital, Institute of Infectious Diseases and Infection Control, Friedrich-Schiller-University Jena, Am Klinikum 1, 07747 Jena, Germany ,grid.9613.d0000 0001 1939 2794Jena University Hospital, Leibniz Center for Photonics in Infection Research, Friedrich Schiller University Jena, 07747 Jena, Germany
| | - Michael Schmitt
- grid.9613.d0000 0001 1939 2794Institute of Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Friedrich-Schiller-University Jena, Helmholtzweg 4, 07743 Jena, Germany ,grid.418907.30000 0004 0563 7158Leibniz Institute of Photonic Technology, Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745 Jena, Germany
| | - Jürgen Popp
- grid.9613.d0000 0001 1939 2794Institute of Physical Chemistry (IPC) and Abbe Center of Photonics (ACP), Friedrich-Schiller-University Jena, Helmholtzweg 4, 07743 Jena, Germany ,grid.418907.30000 0004 0563 7158Leibniz Institute of Photonic Technology, Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745 Jena, Germany ,grid.9613.d0000 0001 1939 2794Jena University Hospital, Center for Sepsis Control and Care (CSCC), Friedrich-Schiller-University Jena, Am Klinikum 1, 07747 Jena, Germany
| |
Collapse
|
171
|
Ahlawat S, Sharma KK. Lepidopteran insects: emerging model organisms to study infection by enteropathogens. Folia Microbiol (Praha) 2022; 68:181-196. [PMID: 36417090 DOI: 10.1007/s12223-022-01014-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 11/03/2022] [Indexed: 11/24/2022]
Abstract
The in vivo analysis of a pathogen is a critical step in gaining greater knowledge of pathogen biology and host-pathogen interactions. In the last two decades, there has been a notable rise in the number of studies on developing insects as a model for studying pathogens, which provides various benefits, such as ethical acceptability, relatively short life cycle, and cost-effective care and maintenance relative to routinely used rodent infection models. Furthermore, lepidopteran insects provide many advantages, such as easy handling and tissue extraction due to their large size relative to other invertebrate models, like Caenorhabditis elegans. Additionally, insects have an innate immune system that is highly analogous to vertebrates. In the present review, we discuss the components of the insect's larval immune system, which strengthens its usage as an alternative host, and present an updated overview of the research findings involving lepidopteran insects (Galleria mellonella, Manduca sexta, Bombyx mori, and Helicoverpa armigera) as infection models to study the virulence by enteropathogens due to the homology between insect and vertebrate gut.
Collapse
Affiliation(s)
- Shruti Ahlawat
- Department of Microbiology, Faculty of Allied Health Sciences, SGT University, Gurgaon-Badli Road Chandu, Budhera, Gurugram, 122505, Haryana, India.
| | - Krishna Kant Sharma
- Laboratory of Enzymology and Recombinant DNA Technology, Department of Microbiology, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| |
Collapse
|
172
|
Orekhova A, De Angelis M, Cacciotti A, Reverberi M, Rotili D, Giorgi A, Protto V, Bonincontro G, Fiorentino F, Zgoda V, Mai A, Palamara AT, Simonetti G. Modulation of Virulence-Associated Traits in Aspergillus fumigatus by BET Inhibitor JQ1. Microorganisms 2022; 10:2292. [PMID: 36422362 PMCID: PMC9698166 DOI: 10.3390/microorganisms10112292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/13/2022] [Accepted: 11/15/2022] [Indexed: 08/18/2024] Open
Abstract
Aspergillus fumigatus is a disease-causing, opportunistic fungus that can establish infection due to its capacity to respond to a wide range of environmental conditions. Secreted proteins and metabolites, which play a critical role in fungal-host interactions and pathogenesis, are modulated by epigenetic players, such as bromodomain and extraterminal domain (BET) proteins. In this study, we evaluated the in vitro and in vivo capability of the BET inhibitor JQ1 to modulate the extracellular proteins and virulence of A. fumigatus. The abundance of 25 of the 76 extracellular proteins identified through LC-MS/MS proteomic analysis changed following JQ1 treatment. Among them, a ribonuclease, a chitinase, and a superoxide dismutase were dramatically downregulated. Moreover, the proteomic analysis of A. fumigatus intracellular proteins indicated that Abr2, an intracellular laccase involved in the last step of melanin synthesis, was absent in the JQ1-treated group. To investigate at which level this downregulation occurred and considering the ability of JQ1 to modulate gene expression we checked the level of ABR2, Chitinase, and Superoxide dismutase mRNA expression by qRT-PCR. Finally, the capacity of JQ1 to reduce the virulence of A. fumigatus has been proved using Galleria mellonella larvae, which are an in vivo model to evaluate fungal virulence. Overall, the promising activity exhibited by JQ1 suggests that A. fumigatus is sensitive to BET inhibition and BET proteins may be a viable target for developing antifungal agents.
Collapse
Affiliation(s)
- Anastasia Orekhova
- Department of Public Health and Infectious Diseases, “Sapienza” University of Rome, 00185 Rome, Italy
| | - Marta De Angelis
- Department of Public Health and Infectious Diseases, “Sapienza” University of Rome, 00185 Rome, Italy
| | - Andrea Cacciotti
- Department of Environmental Biology, “Sapienza” University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy
| | - Massimo Reverberi
- Department of Environmental Biology, “Sapienza” University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, “Sapienza” University of Rome, 00185 Rome, Italy
| | - Alessandra Giorgi
- Department of Biochemical Sciences, “Sapienza” University of Rome, 00185 Rome, Italy
| | - Virginia Protto
- Department of Public Health and Infectious Diseases, “Sapienza” University of Rome, 00185 Rome, Italy
| | - Graziana Bonincontro
- Department of Environmental Biology, “Sapienza” University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy
| | - Francesco Fiorentino
- Department of Drug Chemistry and Technologies, “Sapienza” University of Rome, 00185 Rome, Italy
| | - Victor Zgoda
- Institute of Biomedical Chemistry, Moscow, 10 Pogodinskaya Street, Moscow 119121, Russia
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, “Sapienza” University of Rome, 00185 Rome, Italy
| | - Anna Teresa Palamara
- Department of Public Health and Infectious Diseases, “Sapienza” University of Rome, 00185 Rome, Italy
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Giovanna Simonetti
- Department of Environmental Biology, “Sapienza” University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy
| |
Collapse
|
173
|
Sänger PA, Wagner S, Liebler-Tenorio EM, Fuchs TM. Dissecting the invasion of Galleria mellonella by Yersinia enterocolitica reveals metabolic adaptations and a role of a phage lysis cassette in insect killing. PLoS Pathog 2022; 18:e1010991. [PMID: 36399504 PMCID: PMC9718411 DOI: 10.1371/journal.ppat.1010991] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/02/2022] [Accepted: 11/08/2022] [Indexed: 11/19/2022] Open
Abstract
The human pathogen Yersinia enterocolitica strain W22703 is characterized by its toxicity towards invertebrates that requires the insecticidal toxin complex (Tc) proteins encoded by the pathogenicity island Tc-PAIYe. Molecular and pathophysiological details of insect larvae infection and killing by this pathogen, however, have not been dissected. Here, we applied oral infection of Galleria mellonella (Greater wax moth) larvae to study the colonisation, proliferation, tissue invasion, and killing activity of W22703. We demonstrated that this strain is strongly toxic towards the larvae, in which they proliferate by more than three orders of magnitude within six days post infection. Deletion mutants of the genes tcaA and tccC were atoxic for the insect. W22703 ΔtccC, in contrast to W22703 ΔtcaA, initially proliferated before being eliminated from the host, thus confirming TcaA as membrane-binding Tc subunit and TccC as cell toxin. Time course experiments revealed a Tc-dependent infection process starting with midgut colonisation that is followed by invasion of the hemolymph where the pathogen elicits morphological changes of hemocytes and strongly proliferates. The in vivo transcriptome of strain W22703 shows that the pathogen undergoes a drastic reprogramming of central cell functions and gains access to numerous carbohydrate and amino acid resources within the insect. Strikingly, a mutant lacking a phage-related holin/endolysin (HE) cassette, which is located within Tc-PAIYe, resembled the phenotypes of W22703 ΔtcaA, suggesting that this dual lysis cassette may be an example of a phage-related function that has been adapted for the release of a bacterial toxin.
Collapse
Affiliation(s)
| | - Stefanie Wagner
- Friedrich-Loeffler-Institut, Institut für Molekulare Pathogenese, Jena, Germany
| | | | - Thilo M. Fuchs
- Friedrich-Loeffler-Institut, Institut für Molekulare Pathogenese, Jena, Germany
- * E-mail:
| |
Collapse
|
174
|
Yang YS, Huang TW, Huang YC, Huang WC, Hsu SY, Wu HC, Chen FJ, Shang HS, Sytwu HK, Kuo SC. In vitro and in vivo efficacy of minocycline-based therapy for Elizabethkingia anophelis and the impact of reduced minocycline susceptibility. Int J Antimicrob Agents 2022; 60:106678. [PMID: 36184015 DOI: 10.1016/j.ijantimicag.2022.106678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/23/2022] [Accepted: 09/25/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Elizabethkingia anophelis is inherently resistant to multiple antibiotics, except minocycline. This study aimed to determine the in vitro and in vivo efficacy of minocycline monotherapy and combination therapy against susceptible strains and the impact of reduced minocycline susceptibility. METHODS Three clinical isolates and one laboratory-induced mutant with reduced minocycline susceptibility were included. Time-kill and checkerboard assays were used to assess in vitro efficacy and synergy, respectively. Galleria mellonella infection and mouse pneumonia models were used to assess in vivo efficacy, and a mouse thigh infection model was used to determine the bacterial load. RESULTS Minocycline monotherapy exerted a modest inhibitory effect on three clinical minocycline-susceptible E. anophelis isolates in vitro, but delayed G. mellonella death and improved infected mouse survival; it also significantly reduced the in vivo bacterial load. Minocycline had decreased efficacy on G. mellonella and mice infected by the mutant with reduced minocycline susceptibility. Genome comparison revealed several spontaneous mutations associated with reduced minocycline susceptibility. Among eight antibiotics tested in combination with minocycline, rifampin consistently showed in vitro synergy. The addition of rifampin (1 mg/L) reduced the mutant prevention concentration of minocycline from 2-4 mg/L to < 0.5 mg/L. However, compared with monotherapy, the combination of rifampin and minocycline did not further reduce the bacterial load or improve the survival of G. mellonella or mice. CONCLUSION Minocycline monotherapy was in vivo effective against susceptible E. anophelis. Reduced minocycline susceptibility due to spontaneous mutation decreased its therapeutic efficacy. In combination with rifampin, it prevented the in vitro emergence of reduced susceptibility but did not provide additional in vivo survival benefit.
Collapse
Affiliation(s)
- Ya-Sung Yang
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Tzu-Wen Huang
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ying-Chi Huang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
| | - Wei-Cheng Huang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
| | - Shu-Yuan Hsu
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli County, Taiwan
| | - Han-Chieh Wu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
| | - Feng-Jui Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan; National Yang Ming Chiao Tung University, Department of Biological Science & Technology
| | - Hung-Sheng Shang
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Huey-Kang Sytwu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
| | - Shu-Chen Kuo
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan; National Yang Ming Chiao Tung University, Department of Biological Science & Technology.
| |
Collapse
|
175
|
Zou W, Zhang Y, Zhou M, Chen X, Ma C, Wang T, Jiang Y, Chen T, Shaw C, Wang L. Exploring the active core of a novel antimicrobial peptide, palustrin-2LTb, from the Kuatun frog, Hylarana latouchii, using a bioinformatics-directed approach. Comput Struct Biotechnol J 2022; 20:6192-6205. [DOI: 10.1016/j.csbj.2022.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022] Open
|
176
|
Zhang B, Xu X, Song X, Wen Y, Zhu Z, Lv J, Xie X, Chen L, Tang YW, Du H. Emerging and re-emerging KPC-producing hypervirulent Pseudomonas aeruginosa ST697 and ST463 between 2010 and 2021. Emerg Microbes Infect 2022; 11:2735-2745. [DOI: 10.1080/22221751.2022.2140609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Biying Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004 PR China
| | - Xun Xu
- Institute of Active Polymers and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Hereon, Kantstr.55, 14513 Teltow, Germany
| | - Xiaomei Song
- Department of Nursing, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004 PR China
| | - Yicheng Wen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004 PR China
| | - Zhichen Zhu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004 PR China
| | - Jingnan Lv
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004 PR China
| | - Xiaofang Xie
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004 PR China
| | - Liang Chen
- Hackensack Meridian Health Center for Discovery and Innovation, Nutley, NJ, USA
- Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA
| | - Yi-Wei Tang
- Department of Medical Affairs, Danaher Diagnostic Platform/Cepheid (China), New York, NY, USA
| | - Hong Du
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004 PR China
| |
Collapse
|
177
|
Villanueva JA, Crooks AL, Nagy TA, Quintana JLJ, Dalebroux ZD, Detweiler CS. Salmonella enterica Infections Are Disrupted by Two Small Molecules That Accumulate within Phagosomes and Differentially Damage Bacterial Inner Membranes. mBio 2022; 13:e0179022. [PMID: 36135367 PMCID: PMC9601186 DOI: 10.1128/mbio.01790-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/06/2022] [Indexed: 11/23/2022] Open
Abstract
Gram-negative bacteria have a robust cell envelope that excludes or expels many antimicrobial agents. However, during infection, host soluble innate immune factors permeabilize the bacterial outer membrane. We identified two small molecules that exploit outer membrane damage to access the bacterial cell. In standard microbiological media, neither compound inhibited bacterial growth nor permeabilized bacterial outer membranes. In contrast, at micromolar concentrations, JAV1 and JAV2 enabled the killing of an intracellular human pathogen, Salmonella enterica serovar Typhimurium. S. Typhimurium is a Gram-negative bacterium that resides within phagosomes of cells from the monocyte lineage. Under broth conditions that destabilized the lipopolysaccharide layer, JAV2 permeabilized the bacterial inner membrane and was rapidly bactericidal. In contrast, JAV1 activity was more subtle: JAV1 increased membrane fluidity, altered reduction potential, and required more time than JAV2 to disrupt the inner membrane barrier and kill bacteria. Both compounds interacted with glycerophospholipids from Escherichia coli total lipid extract-based liposomes. JAV1 preferentially interacted with cardiolipin and partially relied on cardiolipin production for activity, whereas JAV2 generally interacted with lipids and had modest affinity for phosphatidylglycerol. In mammalian cells, neither compound significantly altered mitochondrial membrane potential at concentrations that killed S. Typhimurium. Instead, JAV1 and JAV2 became trapped within acidic compartments, including macrophage phagosomes. Both compounds improved survival of S. Typhimurium-infected Galleria mellonella larvae. Together, these data demonstrate that JAV1 and JAV2 disrupt bacterial inner membranes by distinct mechanisms and highlight how small, lipophilic, amine-substituted molecules can exploit host soluble innate immunity to facilitate the killing of intravesicular pathogens. IMPORTANCE Innovative strategies for developing new antimicrobials are needed. Combining our knowledge of host-pathogen interactions and relevant drug characteristics has the potential to reveal new approaches to treating infection. We identified two compounds with antibacterial activity specific to infection and with limited host cell toxicity. These compounds appeared to exploit host innate immunity to access the bacterium and differentially damage the bacterial inner membrane. Further, both compounds accumulated within Salmonella-containing and other acidic vesicles, a process known as lysosomal trapping, which protects the host and harms the pathogen. The compounds also increased host survival in an insect infection model. This work highlights the ability of host innate immunity to enable small molecules to act as antibiotics and demonstrates the feasibility of antimicrobial targeting of the inner membrane. Additionally, this study features the potential use of lysosomal trapping to enhance the activities of compounds against intravesicular pathogens.
Collapse
Affiliation(s)
- Joseph A. Villanueva
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Amy L. Crooks
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Toni A. Nagy
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Joaquin L. J. Quintana
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Zachary D. Dalebroux
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Corrella S. Detweiler
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| |
Collapse
|
178
|
Stempinski PR, Goughenour KD, du Plooy LM, Alspaugh JA, Olszewski MA, Kozubowski L. The Cryptococcus neoformans Flc1 Homologue Controls Calcium Homeostasis and Confers Fungal Pathogenicity in the Infected Hosts. mBio 2022; 13:e0225322. [PMID: 36169198 PMCID: PMC9600462 DOI: 10.1128/mbio.02253-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/12/2022] [Indexed: 01/30/2023] Open
Abstract
Cryptococcus neoformans, an opportunistic yeast pathogen, relies on a complex network of stress response pathways that allow for proliferation in the host. In Saccharomyces cerevisiae, stress responses are regulated by integral membrane proteins containing a transient receptor potential (TRP) domain, including the flavin carrier protein 1 (Flc1), which regulates calcium homeostasis and flavin transport. Here, we report that deletion of C. neoformans FLC1 results in cytosolic calcium elevation and increased nuclear content of calcineurin-dependent transcription factor Crz1, which is associated with an aberrant cell wall chitin overaccumulation observed in the flc1Δ mutant. Absence of Flc1 or inhibition of calcineurin with cyclosporine A prevents vacuolar fusion under conditions of combined osmotic and temperature stress, which is reversed in the flc1Δ mutant by the inhibition of TORC1 kinase with rapamycin. Flc1-deficient yeasts exhibit compromised vacuolar fusion under starvation conditions, including conditions that stimulate formation of carbohydrate capsule. Consequently, the flc1Δ mutant fails to proliferate under low nutrient conditions and displays a defect in capsule formation. Consistent with the previously uncharacterized role of Flc1 in vacuolar biogenesis, we find that Flc1 localizes to the vacuole. The flc1Δ mutant presents a survival defect in J774A.1 macrophage cell-line and profound virulence attenuation in both the Galleria mellonella and mouse pulmonary infection models, demonstrating that Flc1 is essential for pathogenicity. Thus, cryptococcal Flc1 functions in calcium homeostasis and links calcineurin and TOR signaling with vacuolar biogenesis to promote survival under conditions associated with vacuolar fusion required for this pathogen's fitness and virulence. IMPORTANCE Cryptococcosis is a highly lethal infection with limited drug choices, most of which are highly toxic or complicated by emerging antifungal resistance. There is a great need for new drug targets that are unique to the fungus. Here, we identify such a potential target, the Flc1 protein, which we show is crucial for C. neoformans stress response and virulence. Importantly, homologues of Flc1 exist in other fungal pathogens, such as Candida albicans and Aspergillus fumigatus, and are poorly conserved in humans, which could translate into wider spectrum therapy associated with minimal toxicity. Thus, Flc1 could be an "Achille's heel" of C. neoformans to be leveraged therapeutically in cryptococcosis and possibly other fungal infections.
Collapse
Affiliation(s)
- Piotr R. Stempinski
- Department of Genetics and Biochemistry, Eukaryotic Pathogens Innovation Center (EPIC), Clemson University, Clemson, South Carolina, USA
| | - Kristie D. Goughenour
- LTC Charles S. Kettles VA Medical Center, Ann Arbor, Michigan, USA
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School
| | - Lukas M. du Plooy
- Departments of Medicine and Molecular Genetics/Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| | - J. Andrew Alspaugh
- Departments of Medicine and Molecular Genetics/Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Michal A. Olszewski
- LTC Charles S. Kettles VA Medical Center, Ann Arbor, Michigan, USA
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School
| | - Lukasz Kozubowski
- Department of Genetics and Biochemistry, Eukaryotic Pathogens Innovation Center (EPIC), Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
179
|
Frei A, Elliott AG, Kan A, Dinh H, Bräse S, Bruce AE, Bruce MR, Chen F, Humaidy D, Jung N, King AP, Lye PG, Maliszewska HK, Mansour AM, Matiadis D, Muñoz MP, Pai TY, Pokhrel S, Sadler PJ, Sagnou M, Taylor M, Wilson JJ, Woods D, Zuegg J, Meyer W, Cain AK, Cooper MA, Blaskovich MAT. Metal Complexes as Antifungals? From a Crowd-Sourced Compound Library to the First In Vivo Experiments. JACS AU 2022; 2:2277-2294. [PMID: 36311838 PMCID: PMC9597602 DOI: 10.1021/jacsau.2c00308] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/01/2022] [Accepted: 07/27/2022] [Indexed: 06/16/2023]
Abstract
There are currently fewer than 10 antifungal drugs in clinical development, but new fungal strains that are resistant to most current antifungals are spreading rapidly across the world. To prevent a second resistance crisis, new classes of antifungal drugs are urgently needed. Metal complexes have proven to be promising candidates for novel antibiotics, but so far, few compounds have been explored for their potential application as antifungal agents. In this work, we report the evaluation of 1039 metal-containing compounds that were screened by the Community for Open Antimicrobial Drug Discovery (CO-ADD). We show that 20.9% of all metal compounds tested have antimicrobial activity against two representative Candida and Cryptococcus strains compared with only 1.1% of the >300,000 purely organic molecules tested through CO-ADD. We identified 90 metal compounds (8.7%) that show antifungal activity while not displaying any cytotoxicity against mammalian cell lines or hemolytic properties at similar concentrations. The structures of 21 metal complexes that display high antifungal activity (MIC ≤1.25 μM) are discussed and evaluated further against a broad panel of yeasts. Most of these have not been previously tested for antifungal activity. Eleven of these metal complexes were tested for toxicity in the Galleria mellonella moth larva model, revealing that only one compound showed signs of toxicity at the highest injected concentration. Lastly, we demonstrated that the organo-Pt(II) cyclooctadiene complex Pt1 significantly reduces fungal load in an in vivo G. mellonella infection model. These findings showcase that the structural and chemical diversity of metal-based compounds can be an invaluable tool in the development of new drugs against infectious diseases.
Collapse
Affiliation(s)
- Angelo Frei
- Centre
for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland4072, Australia
- Department
of Chemistry, Biochemistry & Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012Bern, Switzerland
| | - Alysha G. Elliott
- Centre
for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland4072, Australia
| | - Alex Kan
- Molecular
Mycology Research Laboratory, Centre for Infectious Diseases and Microbiology,
Faculty of Medicine and Health, Sydney Medical School, Westmead Clinical
School, Sydney Institute for Infectious Diseases, Westmead Hospital-Research
and Education Network, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW2145, Australia
| | - Hue Dinh
- School
of Natural Sciences, ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW2109, Australia
| | - Stefan Bräse
- Institute
of Organic Chemistry, Karlsruhe Institute
of Technology, Fritz-Haber-Weg 6, 76131Karlsruhe, Germany
- Institute
of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, 76344Eggenstein-Leopoldshafen, Germany
| | - Alice E. Bruce
- Department
of Chemistry, University of Maine, Orono, Maine04469, United States
| | - Mitchell R. Bruce
- Department
of Chemistry, University of Maine, Orono, Maine04469, United States
| | - Feng Chen
- Department
of Chemistry, University of Warwick, Gibbet Hill Road, CoventryCV4 7AL, U.K.
| | - Dhirgam Humaidy
- Department
of Chemistry, University of Maine, Orono, Maine04469, United States
| | - Nicole Jung
- Karlsruhe
Nano Micro Facility (KNMF), Karlsruhe Institute
of Technology, Hermann-von-Helmholtz-Platz 1, 76344Eggenstein-Leopoldshafen, Germany
- Institute
of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, 76344Eggenstein-Leopoldshafen, Germany
| | - A. Paden King
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York14853, United States
| | - Peter G. Lye
- School
of Science and Technology, University of
New England, Armidale, NSW2351, Australia
| | - Hanna K. Maliszewska
- School
of Chemistry, University of East Anglia, Norwich Research Park, NorwichNR4 7TJ, U.K.
| | - Ahmed M. Mansour
- Chemistry
Department, Faculty of Science, Cairo University, Giza12613, Egypt
| | - Dimitris Matiadis
- Institute
of Biosciences & Applications, National
Centre for Scientific Research “Demokritos”, 15310Athens, Greece
| | - María Paz Muñoz
- School
of Chemistry, University of East Anglia, Norwich Research Park, NorwichNR4 7TJ, U.K.
| | - Tsung-Yu Pai
- Molecular
Mycology Research Laboratory, Centre for Infectious Diseases and Microbiology,
Faculty of Medicine and Health, Sydney Medical School, Westmead Clinical
School, Sydney Institute for Infectious Diseases, Westmead Hospital-Research
and Education Network, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW2145, Australia
| | - Shyam Pokhrel
- Department
of Chemistry, University of Maine, Orono, Maine04469, United States
| | - Peter J. Sadler
- Department
of Chemistry, University of Warwick, Gibbet Hill Road, CoventryCV4 7AL, U.K.
| | - Marina Sagnou
- Institute
of Biosciences & Applications, National
Centre for Scientific Research “Demokritos”, 15310Athens, Greece
| | - Michelle Taylor
- School
of Science and Technology, University of
New England, Armidale, NSW2351, Australia
| | - Justin J. Wilson
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York14853, United States
| | - Dean Woods
- School
of Science and Technology, University of
New England, Armidale, NSW2351, Australia
| | - Johannes Zuegg
- Centre
for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland4072, Australia
| | - Wieland Meyer
- Molecular
Mycology Research Laboratory, Centre for Infectious Diseases and Microbiology,
Faculty of Medicine and Health, Sydney Medical School, Westmead Clinical
School, Sydney Institute for Infectious Diseases, Westmead Hospital-Research
and Education Network, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW2145, Australia
| | - Amy K. Cain
- School
of Natural Sciences, ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW2109, Australia
| | - Matthew A. Cooper
- Centre
for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland4072, Australia
| | - Mark A. T. Blaskovich
- Centre
for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland4072, Australia
| |
Collapse
|
180
|
Engineering Antibacterial Activities and Biocompatibility of Hyperbranched Lysine-based Random Copolymers. CHINESE JOURNAL OF POLYMER SCIENCE 2022. [DOI: 10.1007/s10118-022-2859-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
181
|
Zhang L, Wang X, Hua X, Yu Y, Leptihn S, Loh B. Therapeutic evaluation of the Acinetobacter baumannii phage Phab24 for clinical use. Virus Res 2022; 320:198889. [PMID: 35970267 DOI: 10.1016/j.virusres.2022.198889] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/03/2022] [Accepted: 08/11/2022] [Indexed: 11/23/2022]
Abstract
Phages have shown to be effective in treating bacterial infections. However, when evaluating the therapeutic potential of novel phage isolates which have the ability to infect and kill a pathogen, it is important to include parameters such as stability (crucial for storage and delivery), infection dynamics in vitro and in vivo (for efficacy and dosing), and an in-depth genome analysis (to exclude the presence of virulence or lysogeny genes), among others. In this study, we characterized bacteriophage Phab24, which infects a colistin-resistant strain of the notorious nosocomial pathogen Acinetobacter baumannii. Our study is crucial for the use of Phab24 in therapy, while also advancing our understanding of phage predation.
Collapse
Affiliation(s)
- Liwei Zhang
- Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, Zhejiang University, International Campus, Haining, Zhejiang, China
| | - Xiaoqing Wang
- School of Medicine, Lishui University, Lishui, China
| | - Xiaoting Hua
- Department of Infectious Diseases, Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yunsong Yu
- Department of Infectious Diseases, Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sebastian Leptihn
- Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, Zhejiang University, International Campus, Haining, Zhejiang, China; Department of Infectious Diseases, Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; University of Edinburgh Medical School, Biomedical Sciences, College of Medicine & Veterinary Medicine, The University of Edinburgh, 1 George Square, Edinburgh EH8 9JZ, United Kingdom.
| | - Belinda Loh
- Department of Vaccines and Infection Models, Fraunhofer Institute for Cell Therapy and Immunology, Perlickstrasse 1, Leipzig 04103, Germany.
| |
Collapse
|
182
|
Virulence Potential of Biofilm-Producing Staphylococcus pseudintermedius, Staphylococcus aureus and Staphylococcus coagulans Causing Skin Infections in Companion Animals. Antibiotics (Basel) 2022; 11:antibiotics11101339. [PMID: 36289997 PMCID: PMC9598800 DOI: 10.3390/antibiotics11101339] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/21/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022] Open
Abstract
Coagulase-positive staphylococci (CoPS) account for most bacteria-related pyoderma in companion animals. Emergence of methicillin-resistant strains of Staphylococcus pseudintermedius (MRSP), Staphylococcus aureus (MRSA) or Staphylococcus coagulans (MRSC), often with multidrug-resistant (MDR) phenotypes, is a public health concern. The study collection comprised 237 staphylococci (S. pseudintermedius (n = 155), S. aureus (n = 55) and S. coagulans (n = 27)) collected from companion animals, previously characterized regarding resistance patterns and clonal lineages. Biofilm production was detected for 51.0% (79/155), 94.6% (52/55) and 88.9% (24/27) of the S. pseudintermedius, S. aureus and S. coagulans, respectively, and was a frequent trait of the predominant S. pseudintermedius and S. aureus clonal lineages. The production of biofilm varied with NaCl supplementation of the growth media. All S. pseudintermedius and S. aureus strains carried icaADB. Kaplan–Meier survival analysis of Galleria mellonella infected with different CoPS revealed a higher virulence potential of S. aureus when compared with other CoPS. Our study highlights a high frequency of biofilm production by prevalent antimicrobial-resistant clonal lineages of CoPS associated with animal pyoderma, potentially related with a higher virulence potential and persistent or recurrent infections.
Collapse
|
183
|
Wang Y, Kong J, Zhang X, Liu Y, Huang Z, Yuan L, Zhang Y, Cao J, Chen L, Liu Y, Zhou T. Plumbagin resurrect colistin susceptible against colistin-resistant Pseudomonas aeruginosa in vitro and in vivo. Front Microbiol 2022; 13:1020652. [PMID: 36274701 PMCID: PMC9579824 DOI: 10.3389/fmicb.2022.1020652] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/08/2022] [Indexed: 11/22/2022] Open
Abstract
The global emergence and spread of multi-drug resistant (MDR) strains is becoming increasingly worrisome due to the overuse of broad-spectrum antibiotics. Colistin, the last resort for treating MDR strains infections, has once again returned to the clinician’s choice. However, with the widespread use of colistin, colistin-resistant gram-negative bacteria (GNB) have subsequently emerged, including colistin-resistant Pseudomonas aeruginosa (COL-R PA). Therefore, available solutions are urgently needed to respond to this situation. Here, we inspiringly found that the combination of plumbagin and colistin had an efficiently inhibitory effect for colistin-resistant P. aeruginosa in vitro through checkerboard assay and time-kill assay. The combinatorial inhibition of biofilm formation was clearly demonstrated by crystal violet staining and scanning electron microscopy (SEM), and this combination can not only inhibited biofilm formation but also eradicated the mature biofilm. Erythrocytes hemolysis test showed that plumbagin has negligible hemolysis ability. In addition, the increased survival rate of Galleria mellonella (G. mellonella) larva confirmed this combination as same as effective in vivo. As for the mechanism of this combination, propidium iodide (PI) staining showed colistin combined with plumbagin could significantly change the membrane permeability, thus exerting synergistic antibacterial activity. In conclusion, the combination of plumbagin and colistin shows a prominently synergistic antibacterial effect in vitro and in vivo, providing a promising option for the therapy of COL-R PA infection.
Collapse
Affiliation(s)
- Yue Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingchun Kong
- Department of Medical Lab Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaodong Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yan Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zeyu Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lu Yuan
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, China
| | - Ying Zhang
- Department of Medical Lab Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianming Cao
- Department of Medical Lab Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lijiang Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yong Liu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, China
- Yong Liu,
| | - Tieli Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- *Correspondence: Tieli Zhou,
| |
Collapse
|
184
|
Bernabè G, Marzaro G, Di Pietra G, Otero A, Bellato M, Pauletto A, Scarpa M, Sut S, Chilin A, Dall’Acqua S, Brun P, Castagliuolo I. A novel phenolic derivative inhibits AHL-dependent quorum sensing signaling in Pseudomonas aeruginosa. Front Pharmacol 2022; 13:996871. [PMID: 36204236 PMCID: PMC9531014 DOI: 10.3389/fphar.2022.996871] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Increasing antibiotic resistance and the decline in the pharmaceutical industry’s investments have amplified the need for novel treatments for multidrug-resistant bacteria. Quorum sensing (QS) inhibitors reduce pathogens’ virulence without selective pressure on bacteria and provide an alternative to conventional antibiotic-based therapies. P. aeruginosa uses complex QS signaling to control virulence and biofilm formation. We aimed to identify inhibitors of P. aeruginosa QS acting on acyl-homoserine lactones (AHL)-mediated circuits. Bioluminescence and qRT-PCR assays were employed to screen a library of 81 small phenolic derivatives to reduce AHL-dependent signaling. We identified GM-50 as the most active compound inhibiting the expression of AHL-regulated genes but devoid of cytotoxic activity in human epithelial cells and biocidal effects on bacteria. GM-50 reduces virulence factors such as rhamnolipids, pyocyanin, elastase secretion, and swarming motility in P. aeruginosa PAO1 laboratory strain. By molecular docking, we provide evidence that GM-50 highly interacts with RhlR. GM-50 significantly improved aztreonam-mediated biofilm disruption. Moreover, GM-50 prevents adhesion of PAO1 and inflammatory damage in the human A549 cell line and protects Galleria mellonella from PAO1-mediated killing. GM-50 significantly reduces virulence factors in 20 P. aeruginosa clinical isolates from patients with respiratory tract infections. In conclusion, GM-50 inhibits AHL-signaling, reduces virulence factors, enhances the anti-biofilm activity of aztreonam, and protects G. mellonella larvae from damage induced by P. aeruginosa. Since GM-50 is active on clinical strains, it represents a starting point for identifying and developing new phenolic derivatives acting as QS-inhibitors in P. aeruginosa infections.
Collapse
Affiliation(s)
- Giulia Bernabè
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Giovanni Marzaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | | | - Ana Otero
- Departamento de Microbioloxía e Parasitoloxía, Facultade de Bioloxía-CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Massimo Bellato
- Department of Information Engineering, University of Padua, Padua, Italy
| | - Anthony Pauletto
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Melania Scarpa
- Laboratory of Advanced Translational Research, Veneto Institute of Oncology IOV—IRCCS, Padua, Italy
| | - Stefania Sut
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Adriana Chilin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Stefano Dall’Acqua
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Paola Brun
- Department of Molecular Medicine, University of Padua, Padua, Italy
- *Correspondence: Paola Brun,
| | | |
Collapse
|
185
|
Meccatti VM, Figueiredo-Godoi LMA, Pereira TC, de Lima PMN, Abu Hasna A, Senna LB, Marcucci MC, Junqueira JC, de Oliveira LD. The biocompatibility and antifungal effect of Rosmarinus officinalis against Candida albicans in Galleria mellonella model. Sci Rep 2022; 12:15611. [PMID: 36114199 PMCID: PMC9481548 DOI: 10.1038/s41598-022-19425-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/29/2022] [Indexed: 11/25/2022] Open
Abstract
This study was performed to evaluate the biocompatibility and antifungal effect of Rosmarinus officinalis against Candida albicans in Galleria mellonella model. Five different concentrations of R. officinalis glycolic extract (50; 25; 12.5 e 6.25 mg/mL) were used to evaluate its biocompatibility in G. mellonella model, in which the nystatin suspension (100; 50; 25; 12.5 e 6.25%) was used as a control group. The antifungal action of R. officinalis glycolic extract was evaluated on C. albicans for 72, 48 and 12 h at two different phases: (1) using the extract as therapeutic agent; and (2) using the extract as prophylactic agent. PBS was used as a negative control group. G. mellonella survival curves were plotted using the Kaplan-Meier method and statistical analysis was performed using the log-rank test (Mantel-Cox) and the significance level was set at (α ≤ 0.05). There was no significant difference among the groups in which all were biocompatible except of a significant death rate of 26.6% with nystatin 100%. In phase 1, it was found that after 7 days, there was no statistically significant difference among the prophylactic treatment groups. In phase 2, the groups of R. officinalis 6.25 mg/mL for 72 h and R. officinalis of 12.5 mg/mL for 24 h promoted the survival rate of the larvae in comparison with the control group with a significant difference (p = 0.017) and (p = 0.032) respectively. Therefore, R. officinalis extract is biocompatible in different concentrations and can be used as a prophylactic agent against fungal infection.
Collapse
Affiliation(s)
- Vanessa Marques Meccatti
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (ICT-UNESP), São José dos Campos, SP, Brazil
| | - Lívia Mara Alves Figueiredo-Godoi
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (ICT-UNESP), São José dos Campos, SP, Brazil
| | - Thaís Cristine Pereira
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (ICT-UNESP), São José dos Campos, SP, Brazil
| | - Patrícia Michelle Nagai de Lima
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (ICT-UNESP), São José dos Campos, SP, Brazil
| | - Amjad Abu Hasna
- Department of Restorative Dentistry, Endodontics Division, Institute of Science and Technology, São Paulo State University (ICT-UNESP), Av. Eng. Francisco José Longo Avenue 777, São José dos Campos, SP, CEP 12245-000, Brazil.
| | - Lavinia Barbosa Senna
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (ICT-UNESP), São José dos Campos, SP, Brazil
| | - Maria Cristina Marcucci
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (ICT-UNESP), São José dos Campos, SP, Brazil
| | - Juliana Campos Junqueira
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (ICT-UNESP), São José dos Campos, SP, Brazil
| | - Luciane Dias de Oliveira
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (ICT-UNESP), São José dos Campos, SP, Brazil
| |
Collapse
|
186
|
Natural Substrates and Culture Conditions to Produce Pigments from Potential Microbes in Submerged Fermentation. FERMENTATION-BASEL 2022. [DOI: 10.3390/fermentation8090460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Pigments from bacteria, fungi, yeast, cyanobacteria, and microalgae have been gaining more demand in the food, leather, and textile industries due to their natural origin and effective bioactive functions. Mass production of microbial pigments using inexpensive and ecofriendly agro-industrial residues is gaining more demand in the current research due to their low cost, natural origin, waste utilization, and high pigment stimulating characteristics. A wide range of natural substrates has been employed in submerged fermentation as carbon and nitrogen sources to enhance the pigment production from these microorganisms to obtain the required quantity of pigments. Submerged fermentation is proven to yield more pigment when added with agro-waste residues. Hence, in this review, aspects of potential pigmented microbes such as diversity, natural substrates that stimulate more pigment production from bacteria, fungi, yeast, and a few microalgae under submerged culture conditions, pigment identification, and ecological functions are detailed for the benefit of industrial personnel, researchers, and other entrepreneurs to explore pigmented microbes for multifaceted applications. In addition, some important aspects of microbial pigments are covered herein to disseminate the knowledge.
Collapse
|
187
|
Kaul L, Abdo AI, Coenye T, Krom BP, Hoogenkamp MA, Zannettino ACW, Süss R, Richter K. The combination of diethyldithiocarbamate and copper ions is active against Staphylococcus aureus and Staphylococcus epidermidis biofilms in vitro and in vivo. Front Microbiol 2022; 13:999893. [PMID: 36160243 PMCID: PMC9500474 DOI: 10.3389/fmicb.2022.999893] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/18/2022] [Indexed: 12/13/2022] Open
Abstract
Staphylococcus aureus and Staphylococcus epidermidis are associated with life-threatening infections. Despite the best medical care, these infections frequently occur due to antibiotic resistance and the formation of biofilms of these two bacteria (i.e., clusters of bacteria embedded in a matrix). As a consequence, there is an urgent need for effective anti-biofilm treatments. Here, we describe the antibacterial properties of a combination treatment of diethyldithiocarbamate (DDC) and copper ions (Cu2+) and their low toxicity in vitro and in vivo. The antibacterial activity of DDC and Cu2+ was assessed in vitro against both planktonic and biofilm cultures of S. aureus and S. epidermidis using viability assays, microscopy, and attachment assays. Cytotoxicity of DDC and Cu2+ (DDC-Cu2+) was determined using a human fibroblast cell line. In vivo antimicrobial activity and toxicity were monitored in Galleria mellonella larvae. DDC-Cu2+ concentrations of 8 μg/ml DDC and 32 μg/ml Cu2+ resulted in over 80% MRSA and S. epidermidis biofilm killing, showed synergistic and additive effects in both planktonic and biofilm cultures of S. aureus and S. epidermidis, and synergized multiple antibiotics. DDC-Cu2+ inhibited MRSA and S. epidermidis attachment and biofilm formation in the xCELLigence and Bioflux systems. In vitro and in vivo toxicity of DDC, Cu2+ and DDC-Cu2+ resulted in > 70% fibroblast viability and > 90% G. mellonella survival. Treatment with DDC-Cu2+ significantly increased the survival of infected larvae (87% survival of infected, treated larvae vs. 47% survival of infected, untreated larvae, p < 0.001). Therefore, DDC-Cu2+ is a promising new antimicrobial with activity against planktonic and biofilm cultures of S. epidermidis and S. aureus and low cytotoxicity in vitro. This gives us high confidence to progress to mammalian animal studies, testing the antimicrobial efficacy and safety of DDC-Cu2+.
Collapse
Affiliation(s)
- Laurine Kaul
- Richter Lab, Basil Hetzel Institute for Translational Health Research, Department of Surgery, University of Adelaide, Adelaide, SA, Australia
- Department of Pharmaceutical Technology and Biopharmacy, Institute of Pharmaceutical Sciences, University of Freiburg, Freiburg, Germany
- Faculty of Health and Medical Sciences, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Adrian I. Abdo
- Richter Lab, Basil Hetzel Institute for Translational Health Research, Department of Surgery, University of Adelaide, Adelaide, SA, Australia
- Faculty of Health and Medical Sciences, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Gent, Belgium
| | - Bastiaan P. Krom
- Department of Preventive Dentistry, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Michel A. Hoogenkamp
- Department of Preventive Dentistry, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Andrew C. W. Zannettino
- Faculty of Health and Medical Sciences, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Precision Medicine Theme, South Australian Health & Medical Research Institute, Adelaide, SA, Australia
- Central Adelaide Local Health Network, Adelaide, SA, Australia
| | - Regine Süss
- Department of Pharmaceutical Technology and Biopharmacy, Institute of Pharmaceutical Sciences, University of Freiburg, Freiburg, Germany
| | - Katharina Richter
- Richter Lab, Basil Hetzel Institute for Translational Health Research, Department of Surgery, University of Adelaide, Adelaide, SA, Australia
- Faculty of Health and Medical Sciences, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Institute for Photonics and Advanced Sensing, University of Adelaide, Adelaide, SA, Australia
- *Correspondence: Katharina Richter,
| |
Collapse
|
188
|
Larval gut microbiome of Pelidnota luridipes (Coleoptera: Scarabaeidae): high bacterial diversity, different metabolic profiles on gut chambers and species with probiotic potential. World J Microbiol Biotechnol 2022; 38:210. [PMID: 36050590 DOI: 10.1007/s11274-022-03387-1] [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: 08/05/2021] [Accepted: 08/11/2022] [Indexed: 10/14/2022]
Abstract
Pelidnota luridipes Blanchard (1850) is a tropical beetle of the family Scarabaeidae, whose larvae live on wood without parental care. Microbiota of mid- and hindgut of larvae was evaluated by culture-dependent and independent methods, and the results show a diverse microbiota, with most species of bacteria and fungi shared between midgut and hindgut. We isolated 272 bacterial and 29 yeast isolates, identified in 57 and 7 species, respectively, while using metabarcoding, we accessed 1,481 and 267 OTUs of bacteria and fungi, respectively. The composition and abundance of bacteria and fungi differed between mid- and hindgut, with a tendency for higher richness and diversity of yeasts in the midgut, and bacteria on the hindgut. Some taxa are abundant in the intestine of P. luridipes larvae, such as Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria; as well as Saccharomycetales and Trichosporonales yeasts. Mid- and hindgut metabolic profiles differ (e.g. biosynthesis of amino acids, cofactors, and lipopolysaccharides) with higher functional diversity in the hindgut. Isolates have different functional traits such as secretion of hydrolytic enzymes and antibiosis against pathogens. Apiotrichum siamense L29A and Bacillus sp. BL17B protected larvae of the moth Galleria mellonella, against infection by the pathogens Listeria monocytogenes ATCC19111 and Pseudomonas aeruginosa ATCC 9027. This is the first work with the larval microbiome of a Rutelini beetle, demonstrating its diversity and potential in prospecting microbial products as probiotics. The functional role of microbiota for the nutrition and adaptability of P. luridipes larvae needs to be evaluated in the future.
Collapse
|
189
|
Asai M, Li Y, Spiropoulos J, Cooley W, Everest DJ, Kendall SL, Martín C, Robertson BD, Langford PR, Newton SM. Galleria mellonella as an infection model for the virulent Mycobacterium tuberculosis H37Rv. Virulence 2022; 13:1543-1557. [PMID: 36052440 PMCID: PMC9481108 DOI: 10.1080/21505594.2022.2119657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (MTB), is a leading cause of infectious disease mortality. Animal infection models have contributed substantially to our understanding of TB, yet their biological and non-biological limitations are a research bottleneck. There is a need for more ethically acceptable, economical, and reproducible TB infection models capable of mimicking key aspects of disease. Here, we demonstrate and present a basic description of how Galleria mellonella (the greater wax moth, Gm) larvae can be used as a low cost, rapid, and ethically more acceptable model for TB research. This is the first study to infect Gm with the fully virulent MTB H37Rv, the most widely used strain in research. Infection of Gm with MTB resulted in a symptomatic lethal infection, the virulence of which differed from both attenuated Mycobacterium bovis BCG and auxotrophic MTB strains. The Gm-MTB model can also be used for anti-TB drug screening, although CFU enumeration from Gm is necessary for confirmation of mycobacterial load reducing activity of the tested compound. Furthermore, comparative virulence of MTB isogenic mutants can be determined in Gm. However, comparison of mutant phenotypes in Gm against conventional models must consider the limitations of innate immunity. Our findings indicate that Gm will be a practical, valuable, and advantageous additional model to be used alongside existing models to advance tuberculosis research.
Collapse
Affiliation(s)
- Masanori Asai
- Section of Paediatric Infectious Diseases, Department of Infectious Disease, Imperial College London, London, UK
| | - Yanwen Li
- Section of Paediatric Infectious Diseases, Department of Infectious Disease, Imperial College London, London, UK
| | - John Spiropoulos
- Department of Pathology, Animal and Plant Health Agency, Addlestone, UK
| | - William Cooley
- Department of Pathology, Animal and Plant Health Agency, Addlestone, UK
| | - David J Everest
- Department of Pathology, Animal and Plant Health Agency, Addlestone, UK
| | - Sharon L Kendall
- Centre for Emerging, Endemic and Exotic Diseases, Pathobiology and Population Sciences, Royal Veterinary College, Hartfield, UK
| | - Carlos Martín
- Department of Microbiology, Facultad de Medicina Universidad de Zaragoza, CIBERES, (ISCIII), Spain
| | - Brian D Robertson
- MRC Centre for Molecular Bacteriology and Infection, Department of Infectious Disease, Imperial College London, UK
| | - Paul R Langford
- Section of Paediatric Infectious Diseases, Department of Infectious Disease, Imperial College London, London, UK
| | - Sandra M Newton
- Section of Paediatric Infectious Diseases, Department of Infectious Disease, Imperial College London, London, UK
| |
Collapse
|
190
|
Zhang J, Sun R, Chen Z, Zhou C, Ma C, Zhou M, Chen X, Chen T, Shaw C, Wang L. Evaluation of the Antimicrobial Properties of a Natural Peptide from Vespa mandarinia Venom and Its Synthetic Analogues as a Possible Route to Defeat Drug-Resistant Microbes. BIOLOGY 2022; 11:1263. [PMID: 36138742 PMCID: PMC9495676 DOI: 10.3390/biology11091263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/12/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022]
Abstract
Antimicrobial peptides (AMPs) from wasp venom have a good track record and potential for drug development as tools against development of antimicrobial resistance. Herein, the biological function and activity profile of peptide VM, which was discovered in the venom of the wasp, Vespamandarinia, and several of its third-position substituted analogues, were investigated. VM had potent antimicrobial activity against Gram-positive bacteria and biofilm, and all modified peptides achieved the significant enhancement of these capacities. The various physicochemical properties of amino acids substituted in analogues, generated the different mechanisms of action of bacterial membrane disruption. VM-3K showed a maximum 8-fold enhancement of antibacterial activity against Gram-positive bacteria and also presented microbicidal properties against Gram-negative bacteria and fungi. This peptide also exhibited a high killing efficiency at low concentration and had a comparable selectivity index to VM. Furthermore, VM-3K produced a 90% survival of S. aureus-infected waxworms at a concentration of 5.656 mg/kg, at which concentration the natural template peptide only achieved 50% survival. This peptide also lacked short-term resistance generation. Thus, peptide VM-3K could be a promising broad-spectrum antimicrobial candidate for addressing the current antibiotic-resistant infection crisis. It is worth mentioning that this investigation on the relationship between peptide structure and mechanism of action could become an important aspect of drug research on short peptides.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiaoling Chen
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | | | | | - Lei Wang
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| |
Collapse
|
191
|
Achromobacter spp. Adaptation in Cystic Fibrosis Infection and Candidate Biomarkers of Antimicrobial Resistance. Int J Mol Sci 2022; 23:ijms23169265. [PMID: 36012535 PMCID: PMC9409262 DOI: 10.3390/ijms23169265] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/29/2022] [Accepted: 08/12/2022] [Indexed: 11/24/2022] Open
Abstract
Achromobacter spp. can establish occasional or chronic lung infections in patients with cystic fibrosis (CF). Chronic colonization has been associated with worse prognosis highlighting the need to identify markers of bacterial persistence. To this purpose, we analyzed phenotypic features of 95 Achromobacter spp. isolates from 38 patients presenting chronic or occasional infection. Virulence was tested in Galleria mellonella larvae, cytotoxicity was tested in human bronchial epithelial cells, biofilm production in static conditions was measured by crystal violet staining and susceptibility to selected antibiotics was tested by the disk diffusion method. The presence of genetic loci associated to the analyzed phenotypic features was evaluated by a genome-wide association study. Isolates from occasional infection induced significantly higher mortality of G. mellonella larvae and showed a trend for lower cytotoxicity than chronic infection isolates. No significant difference was observed in biofilm production among the two groups. Additionally, antibiotic susceptibility testing showed that isolates from chronically-infected patients were significantly more resistant to sulfonamides and meropenem than occasional isolates. Candidate genetic biomarkers associated with antibiotic resistance or sensitivity were identified. Achromobacter spp. strains isolated from people with chronic and occasional lung infection exhibit different virulence and antibiotic susceptibility features, which could be linked to persistence in CF lungs. This underlines the possibility of identifying predictive biomarkers of persistence that could be useful for clinical purposes.
Collapse
|
192
|
Sohrabi M, Alizade Naini M, Rasekhi A, Oloomi M, Moradhaseli F, Ayoub A, Bazargani A, Hashemizadeh Z, Shahcheraghi F, Badmasti F. Emergence of K1 ST23 and K2 ST65 hypervirulent klebsiella pneumoniae as true pathogens with specific virulence genes in cryptogenic pyogenic liver abscesses Shiraz Iran. Front Cell Infect Microbiol 2022; 12:964290. [PMID: 36017366 PMCID: PMC9396702 DOI: 10.3389/fcimb.2022.964290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
Hypervirulent Klebsiella pneumoniae (hvKp) pathotype is emerging worldwide in pyogenic liver abscesses (PLAs). However, the role of virulence factors in pathogenicity remains unclear. On the other hand, the epidemiology of PLAs in Iran is unknown. From July 2020 to April 2022, bacterial species were isolated and identified from the drainage samples of 54 patients with PLAs. K. pneumoniae as the most common pathogen of pyogenic liver abscesses was identified in 20 (37%) of the 54 patients. We analyzed the clinical and microbiological characteristics of K. pneumoniae-related pyogenic liver abscesses. Antibiotic susceptibility testes and string test were performed. 16S rRNA, antibiotic resistance, and virulence genes were determined by polymerase chain reaction amplification. Clonal relatedness of isolates was identified by multilocus sequence typing. Virulence levels were assessed in the Galleria mellonella larval infection model. Four hvKp isolates (K1/K2) were found to be responsible for cryptogenic PLAs, and 16 classical K. pneumoniae isolates (non-K1/K2) were associated with non-cryptogenic PLAs. Three capsular serotype K1 strains belonged to sequence type 23 (ST23) and one K2 strain to ST65. Meanwhile, the non-K1/K2 strains belonged to other STs. ST231 was the most common strain among the classical K. pneumoniae strains. Compared with the non-K1/K2 strains, capsular serotypes K1/K2 strains were less resistant to antibiotics, had positive string test results, and had more virulence genes. In Galleria mellonella, a concentration of 106 colony-forming units of the K1 hvKp strain resulted in 100% death at 24 hours, confirming the higher virulence of the hvKp strain compared with cKp. K. pneumoniae isolates represented that the acquisition of any plasmid or chromosomal virulence genes contributes to pathogenicity and high prevalence in PLAs. Meanwhile, hvKp isolates with a specific genetic background were detected in cryptogenic PLAs.
Collapse
Affiliation(s)
- Maryam Sohrabi
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| | - Mahvash Alizade Naini
- Department of Internal Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Rasekhi
- Department of Radiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mana Oloomi
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Farzad Moradhaseli
- Department of Radiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Ayoub
- Department of Radiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abdollah Bazargani
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Hashemizadeh
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fereshteh Shahcheraghi
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
- *Correspondence: Farzad Badmasti, ; Fereshteh Shahcheraghi,
| | - Farzad Badmasti
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
- *Correspondence: Farzad Badmasti, ; Fereshteh Shahcheraghi,
| |
Collapse
|
193
|
Polunin KE, Fedotkina OS, Polunina IA, Buryak AK. Optimizing the Chromatographic Separation of Antibacterial Peptides of Galleria mellonella. RUSSIAN JOURNAL OF PHYSICAL CHEMISTRY A 2022. [DOI: 10.1134/s0036024422080209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
194
|
El Haddad L, Angelidakis G, Clark JR, Mendoza JF, Terwilliger AL, Chaftari CP, Duna M, Yusuf ST, Harb CP, Stibich M, Maresso A, Chemaly RF. Genomic and Functional Characterization of Vancomycin-Resistant Enterococci-Specific Bacteriophages in the Galleria mellonella Wax Moth Larvae Model. Pharmaceutics 2022; 14:1591. [PMID: 36015218 PMCID: PMC9414631 DOI: 10.3390/pharmaceutics14081591] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022] Open
Abstract
Phages are naturally occurring viruses that selectively kill bacterial species without disturbing the individual's normal flora, averting the collateral damage of antimicrobial usage. The safety and the effectiveness of phages have been mainly confirmed in the food industry as well as in animal models. In this study, we report on the successful isolation of phages specific to Vancomycin-resistant Enterococci, including Enterococcus faecium (VREfm) and Enterococcus faecalis from sewage samples, and demonstrate their efficacy and safety for VREfm infection in the greater wax moth Galleria mellonella model. No virulence-associated genes, antibiotic resistance genes or integrases were detected in the phages' genomes, rendering them safe to be used in an in vivo model. Phages may be considered as potential agents for therapy for bacterial infections secondary to multidrug-resistant organisms such as VREfm.
Collapse
Affiliation(s)
- Lynn El Haddad
- Department of Medicine, University of Florida, Gainesville, FL 32611, USA; (L.E.H.); (J.F.M.)
| | - Georgios Angelidakis
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (G.A.); (C.P.C.); (M.D.); (S.T.Y.); (C.P.H.); (M.S.)
| | - Justin R. Clark
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (J.R.C.); (A.L.T.); (A.M.)
| | - Jesus F. Mendoza
- Department of Medicine, University of Florida, Gainesville, FL 32611, USA; (L.E.H.); (J.F.M.)
| | - Austen L. Terwilliger
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (J.R.C.); (A.L.T.); (A.M.)
| | - Christopher P. Chaftari
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (G.A.); (C.P.C.); (M.D.); (S.T.Y.); (C.P.H.); (M.S.)
| | - Mark Duna
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (G.A.); (C.P.C.); (M.D.); (S.T.Y.); (C.P.H.); (M.S.)
| | - Serena T. Yusuf
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (G.A.); (C.P.C.); (M.D.); (S.T.Y.); (C.P.H.); (M.S.)
| | - Cynthia P. Harb
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (G.A.); (C.P.C.); (M.D.); (S.T.Y.); (C.P.H.); (M.S.)
| | - Mark Stibich
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (G.A.); (C.P.C.); (M.D.); (S.T.Y.); (C.P.H.); (M.S.)
- Xenex Disinfection Services, San Antonio, TX 78216, USA
| | - Anthony Maresso
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (J.R.C.); (A.L.T.); (A.M.)
| | - Roy F. Chemaly
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (G.A.); (C.P.C.); (M.D.); (S.T.Y.); (C.P.H.); (M.S.)
| |
Collapse
|
195
|
Tao S, Chen H, Li N, Wang T, Liang W. The Spread of Antibiotic Resistance Genes In Vivo Model. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2022; 2022:3348695. [PMID: 35898691 PMCID: PMC9314185 DOI: 10.1155/2022/3348695] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/26/2022] [Accepted: 06/30/2022] [Indexed: 12/20/2022]
Abstract
Infections caused by antibiotic-resistant bacteria are a major public health threat. The emergence and spread of antibiotic resistance genes (ARGs) in the environment or clinical setting pose a serious threat to human and animal health worldwide. Horizontal gene transfer (HGT) of ARGs is one of the main reasons for the dissemination of antibiotic resistance in vitro and in vivo environments. There is a consensus on the role of mobile genetic elements (MGEs) in the spread of bacterial resistance. Most drug resistance genes are located on plasmids, and the spread of drug resistance genes among microorganisms through plasmid-mediated conjugation transfer is the most common and effective way for the spread of multidrug resistance. Experimental studies of the processes driving the spread of antibiotic resistance have focused on simple in vitro model systems, but the current in vitro protocols might not correctly reflect the HGT of antibiotic resistance genes in realistic conditions. This calls for better models of how resistance genes transfer and disseminate in vivo. The in vivo model can better mimic the situation that occurs in patients, helping study the situation in more detail. This is crucial to develop innovative strategies to curtail the spread of antibiotic resistance genes in the future. This review aims to give an overview of the mechanisms of the spread of antibiotic resistance genes and then demonstrate the spread of antibiotic resistance genes in the in vivo model. Finally, we discuss the challenges in controlling the spread of antibiotic resistance genes and their potential solutions.
Collapse
Affiliation(s)
- Shuan Tao
- School of Medical, Jiangsu University, Zhenjiang, Jiangsu Province, China
- Lianyungang Clinical College of Jiangsu University, Lianyungang, Jiangsu Province, China
| | - Huimin Chen
- School of Medical, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Na Li
- Bengbu Medical College, Bengbu, Anhui Province, China
| | - Tong Wang
- Nanjing Brain Hospital Affiliated Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Wei Liang
- Lianyungang Clinical College of Jiangsu University, Lianyungang, Jiangsu Province, China
| |
Collapse
|
196
|
Galleria mellonella as a Novel In Vivo Model to Screen Natural Product-Derived Modulators of Innate Immunity. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12136587] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Immunomodulators are drugs that either stimulate or suppress the immune system in response to an immunopathological disease or cancer. The majority of clinically approved immunomodulators are either chemically synthesised (e.g., dexamethasone) or protein-based (e.g., monoclonal antibodies), whose uses are limited due to toxicity issues, poor bioavailability, or prohibitive cost. Nature is an excellent source of novel compounds, as it is estimated that almost half of all licenced medicines are derived from nature or inspired by natural product (NP) structures. The clinical success of the fungal-derived immunosuppressant cyclosporin A demonstrates the potential of natural products as immunomodulators. Conventionally, the screening of NP molecules for immunomodulation is performed in small animal models; however, there is a growing impetus to replace animal models with more ethical alternatives. One novel approach is the use of Galleria melonella larvae as an in vivo model of immunity. Despite lacking adaptive antigen-specific immunity, this insect possesses an innate immune system comparable to mammals. In this review, we will describe studies that have used this alternative in vivo model to assess the immunomodulating activity of synthetic and NP-derived compounds, outline the array of bioassays employed, and suggest strategies to enhance the use of this model in future research.
Collapse
|
197
|
Connections between Exoproteome Heterogeneity and Virulence in the Oral Pathogen Aggregatibacter actinomycetemcomitans. mSystems 2022; 7:e0025422. [PMID: 35695491 PMCID: PMC9239275 DOI: 10.1128/msystems.00254-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aggregatibacter actinomycetemcomitans is a Gram-negative bacterial pathogen associated with severe periodontitis and nonoral diseases. Clinical isolates of A. actinomycetemcomitans display a rough (R) colony phenotype with strong adherent properties. Upon prolonged culturing, nonadherent strains with a smooth (S) colony phenotype emerge. To date, most virulence studies on A. actinomycetemcomitans have been performed with S strains of A. actinomycetemcomitans, whereas the virulence of clinical R isolates has received relatively little attention. Since the extracellular proteome is the main bacterial reservoir of virulence factors, the present study was aimed at a comparative analysis of this subproteome fraction for a collection of R isolates and derivative S strains, in order to link particular proteins to the virulence of A. actinomycetemcomitans with serotype b. To assess the bacterial virulence, we applied different infection models based on larvae of the greater wax moth Galleria mellonella, a human salivary gland-derived epithelial cell line, and freshly isolated neutrophils from healthy human volunteers. A total number of 351 extracellular A. actinomycetemcomitans proteins was identified by mass spectrometry, with the S strains consistently showing more extracellular proteins than their parental R isolates. A total of 50 known extracellular virulence factors was identified, of which 15 were expressed by all investigated bacteria. Importantly, the comparison of differences in exoproteome composition and virulence highlights critical roles of 10 extracellular proteins in the different infection models. Together, our findings provide novel clues for understanding the virulence of A. actinomycetemcomitans and for development of potential preventive or therapeutic avenues to neutralize this important oral pathogen. IMPORTANCE Periodontitis is one of the most common inflammatory diseases worldwide, causing high morbidity and decreasing the quality of life of millions of people. The bacterial pathogen Aggregatibacter actinomycetemcomitans is strongly associated with aggressive forms of periodontitis. Moreover, it has been implicated in serious nonoral infections, including endocarditis and brain abscesses. Therefore, it is important to investigate how A. actinomycetemcomitans can cause disease. In the present study, we applied a mass spectrometry approach to make an inventory of the virulence factors secreted by different clinical A. actinomycetemcomitans isolates and derivative strains that emerged upon culturing. We subsequently correlated the secreted virulence factors to the pathogenicity of the investigated bacteria in different infection models. The results show that a limited number of extracellular virulence factors of A. actinomycetemcomitans have central roles in pathogenesis, indicating that they could be druggable targets to prevent or treat oral disease.
Collapse
|
198
|
Sellera FP, Cardoso B, Fuentes-Castillo D, Esposito F, Sano E, Fontana H, Fuga B, Goldberg DW, Seabra LAV, Antonelli M, Sandri S, Kolesnikovas CKM, Lincopan N. Genomic Analysis of a Highly Virulent NDM-1-Producing Escherichia coli ST162 Infecting a Pygmy Sperm Whale ( Kogia breviceps) in South America. Front Microbiol 2022; 13:915375. [PMID: 35755998 PMCID: PMC9231830 DOI: 10.3389/fmicb.2022.915375] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/18/2022] [Indexed: 11/29/2022] Open
Abstract
Carbapenemase-producing Enterobacterales are rapidly spreading and adapting to different environments beyond hospital settings. During COVID-19 lockdown, a carbapenem-resistant NDM-1-positive Escherichia coli isolate (BA01 strain) was recovered from a pygmy sperm whale (Kogia breviceps), which was found stranded on the southern coast of Brazil. BA01 strain belonged to the global sequence type (ST) 162 and carried the blaNDM–1, besides other medically important antimicrobial resistance genes. Additionally, genes associated with resistance to heavy metals, biocides, and glyphosate were also detected. Halophilic behavior (tolerance to > 10% NaCl) of BA01 strain was confirmed by tolerance tests of NaCl minimal inhibitory concentration, whereas halotolerance associated genes katE and nhaA, which encodes for catalase and Na+/H+ antiporter cytoplasmic membrane, respectively, were in silico confirmed. Phylogenomics clustered BA01 with poultry- and human-associated ST162 lineages circulating in European and Asian countries. Important virulence genes, including the astA (a gene encoding an enterotoxin associated with human and animal infections) were detected, whereas in vivo experiments using the Galleria mellonella infection model confirmed the virulent behavior of the BA01 strain. WHO critical priority carbapenemase-producing pathogens in coastal water are an emerging threat that deserves the urgent need to assess the role of the aquatic environment in its global epidemiology.
Collapse
Affiliation(s)
- Fábio P Sellera
- Department of Internal Medicine, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil.,One Health Brazilian Resistance Project (OneBR), São Paulo, Brazil.,School of Veterinary Medicine, Metropolitan University of Santos, Santos, Brazil
| | - Brenda Cardoso
- One Health Brazilian Resistance Project (OneBR), São Paulo, Brazil.,Department of Microbiology, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Danny Fuentes-Castillo
- One Health Brazilian Resistance Project (OneBR), São Paulo, Brazil.,Departamento de Patología y Medicina Preventiva, Facultad de Ciencias Veterinarias, Universidad de Concepción, Chillán, Chile
| | - Fernanda Esposito
- One Health Brazilian Resistance Project (OneBR), São Paulo, Brazil.,Department of Clinical Analysis, School of Pharmacy, University of São Paulo, São Paulo, Brazil
| | - Elder Sano
- One Health Brazilian Resistance Project (OneBR), São Paulo, Brazil.,Department of Microbiology, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Herrison Fontana
- One Health Brazilian Resistance Project (OneBR), São Paulo, Brazil.,Department of Clinical Analysis, School of Pharmacy, University of São Paulo, São Paulo, Brazil
| | - Bruna Fuga
- One Health Brazilian Resistance Project (OneBR), São Paulo, Brazil.,Department of Microbiology, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil.,Department of Clinical Analysis, School of Pharmacy, University of São Paulo, São Paulo, Brazil
| | | | - Lourdes A V Seabra
- School of Veterinary Medicine, Metropolitan University of Santos, Santos, Brazil
| | | | | | | | - Nilton Lincopan
- One Health Brazilian Resistance Project (OneBR), São Paulo, Brazil.,Department of Microbiology, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil.,Department of Clinical Analysis, School of Pharmacy, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
199
|
Duman Erbaş E, Gwokyalya R, Altuntaş H, Kutrup B. Screening the immunotoxicity of different food preservative agents on the model organism Galleria mellonella L. (Lepidoptera: Pyralidae) larvae. Drug Chem Toxicol 2022:1-11. [PMID: 35758106 DOI: 10.1080/01480545.2022.2091589] [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: 11/03/2022]
Abstract
Immunotoxic effects of sodium benzoate (SB, E211), sodium nitrate (SNa, E251), and sodium nitrite (SNi, E250), a few of the most common food preservatives, on the model organism Galleria mellonella L. (Lepidoptera: Pyralidae) larvae were investigated in this study. The last instar larvae were used for all experimental analyses. For this purpose, median lethal doses of SB, SNa, and SNi were applied to the larvae by the force-feeding method. We found that force-feeding G. mellonella larvae with SB, SNa, and SNi significantly reduced the larval total hemocyte counts, prohemocyte, and granulocyte ratios but increased plasmatocyte, spherulocyte, and oenocyte ratios, as well as the hemocyte mitotic indices and micronucleus frequency. The spreading ability of hemocytes and hemocyte-mediated immune responses were lower in the SB, SNa-, and SNi-treated larval groups compared to controls. Apoptotic indices were higher in all larval groups treated with food preservatives, but increments in necrotic indices were only significantly higher in SNi-treated larvae compared to controls. Our research shows that SB, SNa, and SNi have immunotoxic and cytotoxic potential on G. mellonella larvae. Thus, we suggest that G. mellonella larvae can be used as preliminary in vivo models to screen the immunotoxic effects of food preservative agents.
Collapse
Affiliation(s)
- Emine Duman Erbaş
- Department of Biology, Faculty of Science, Karadeniz Technical University, Trabzon, Turkey
| | - Rehemah Gwokyalya
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
| | - Hülya Altuntaş
- Department of Biology, Faculty of Science, Eskisehir Technical University, Eskişehir, Turkey
| | - Bilal Kutrup
- Department of Biology, Faculty of Science, Karadeniz Technical University, Trabzon, Turkey
| |
Collapse
|
200
|
Flagellotropic Bacteriophages: Opportunities and Challenges for Antimicrobial Applications. Int J Mol Sci 2022; 23:ijms23137084. [PMID: 35806089 PMCID: PMC9266447 DOI: 10.3390/ijms23137084] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/18/2022] [Accepted: 06/21/2022] [Indexed: 12/22/2022] Open
Abstract
Bacteriophages (phages) are the most abundant biological entities in the biosphere. As viruses that solely infect bacteria, phages have myriad healthcare and agricultural applications including phage therapy and antibacterial treatments in the foodservice industry. Phage therapy has been explored since the turn of the twentieth century but was no longer prioritized following the invention of antibiotics. As we approach a post-antibiotic society, phage therapy research has experienced a significant resurgence for the use of phages against antibiotic-resistant bacteria, a growing concern in modern medicine. Phages are extraordinarily diverse, as are their host receptor targets. Flagellotropic (flagellum-dependent) phages begin their infection cycle by attaching to the flagellum of their motile host, although the later stages of the infection process of most of these phages remain elusive. Flagella are helical appendages required for swimming and swarming motility and are also of great importance for virulence in many pathogenic bacteria of clinical relevance. Not only is bacterial motility itself frequently important for virulence, as it allows pathogenic bacteria to move toward their host and find nutrients more effectively, but flagella can also serve additional functions including mediating bacterial adhesion to surfaces. Flagella are also a potent antigen recognized by the human immune system. Phages utilizing the flagellum for infections are of particular interest due to the unique evolutionary tradeoff they force upon their hosts: by downregulating or abolishing motility to escape infection by a flagellotropic phage, a pathogenic bacterium would also likely attenuate its virulence. This factor may lead to flagellotropic phages becoming especially potent antibacterial agents. This review outlines past, present, and future research of flagellotropic phages, including their molecular mechanisms of infection and potential future applications.
Collapse
|