1
|
Eichberg J, Oberpaul M, Hartwig C, Geißler AH, Culmsee C, Vilcinskas A, Böttcher-Friebertshäuser E, Brückner H, Degenkolb T, Hardes K. Structural characterization and bioactivity profiling of the fungal peptaibiotic tolypin reveal protective effects against influenza viruses. Arch Pharm (Weinheim) 2024; 357:e2400384. [PMID: 39031917 DOI: 10.1002/ardp.202400384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/22/2024]
Abstract
In a bioprospection for new antivirals, we tested nonribosomally biosynthesized polypeptide antibiotics in MDCK II cells for their actions on influenza A and B viruses (IAV/IBV). Only tolypin, a mixture of closely related 16-residue peptaibiotics from the fungus Tolypocladium inflatum IE 1897, showed promising activity. It was selected for further investigation and structural characterization by ultrahigh performance liquid chromatography coupled to high-resolution mass spectrometry (UHPLC-HR-MS/MS) and ultrahigh performance liquid chromatography coupled to in-source collision-induced dissociation tandem mass spectrometry (UHPLC-isCID-HR-MS/MS), revealing 12 partially co-eluting individual peptides that were fully sequenced. Since tolypin-related efrapeptins are potent inhibitors of F1/Fo-ATPase, we screened tolypin for its toxicity against MDCK II cells and larvae of the greater wax moth Galleria mellonella. We found that a nontoxic concentration of tolypin (1 µg/mL) reduced the titer of two IBV strains by 4-5 log values, and that of an H3N2 strain by 1-2 log values, but the H1N1pdm strain was not affected. The higher concentrations of tolypin were cytostatic to MDCK II cells, shifted their metabolism from oxidative phosphorylation to glycolysis, and induced paralysis in G. mellonella, supporting the inhibition of F1/Fo-ATPase as the mode of action. Our results lay the foundations for future work to investigate the interplay between viral replication and cellular energy metabolism, as well as the development of drugs that target host factors.
Collapse
Affiliation(s)
- Johanna Eichberg
- Branch for Bioresources of the Fraunhofer IME, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Giessen, Germany
- BMBF Junior Research Group in Infection Research "ASCRIBE", Giessen, Germany
| | - Markus Oberpaul
- Branch for Bioresources of the Fraunhofer IME, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Giessen, Germany
- BMBF Junior Research Group in Infection Research "ASCRIBE", Giessen, Germany
| | - Christoph Hartwig
- Branch for Bioresources of the Fraunhofer IME, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Giessen, Germany
| | - Andrea Helga Geißler
- Department of Insect Biotechnology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Carsten Culmsee
- Institute for Pharmacology and Clinical Pharmacy, Biochemical-Pharmacological Center Marburg, University of Marburg, Marburg, Germany
| | - Andreas Vilcinskas
- Branch for Bioresources of the Fraunhofer IME, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Giessen, Germany
- Department of Insect Biotechnology, Justus-Liebig-University of Giessen, Giessen, Germany
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE-TBG), Frankfurt, Germany
| | | | - Hans Brückner
- Department of Food Sciences, Interdisciplinary Research Centre for Biosystems, Land Use and Nutrition, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Thomas Degenkolb
- Department of Insect Biotechnology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Kornelia Hardes
- Branch for Bioresources of the Fraunhofer IME, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Giessen, Germany
- BMBF Junior Research Group in Infection Research "ASCRIBE", Giessen, Germany
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE-TBG), Frankfurt, Germany
| |
Collapse
|
2
|
Noakes F, Smitten KL, Maple LEC, Bernardino de la Serna J, Robertson CC, Pritchard D, Fairbanks SD, Weinstein JA, Smythe CGW, Thomas JA. Phenazine Cations as Anticancer Theranostics †. J Am Chem Soc 2024; 146:12836-12849. [PMID: 38683943 PMCID: PMC11082890 DOI: 10.1021/jacs.4c03491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 05/02/2024]
Abstract
The biological properties of two water-soluble organic cations based on polypyridyl structures commonly used as ligands for photoactive transition metal complexes designed to interact with biomolecules are investigated. A cytotoxicity screen employing a small panel of cell lines reveals that both cations show cytotoxicity toward cancer cells but show reduced cytotoxicity to noncancerous HEK293 cells with the more extended system being notably more active. Although it is not a singlet oxygen sensitizer, the more active cation also displayed enhanced potency on irradiation with visible light, making it active at nanomolar concentrations. Using the intrinsic luminescence of the cations, their cellular uptake was investigated in more detail, revealing that the active compound is more readily internalized than its less lipophilic analogue. Colocalization studies with established cell probes reveal that the active cation predominantly localizes within lysosomes and that irradiation leads to the disruption of mitochondrial structure and function. Stimulated emission depletion (STED) nanoscopy and transmission electron microscopy (TEM) imaging reveal that treatment results in distinct lysosomal swelling and extensive cellular vacuolization. Further imaging-based studies confirm that treatment with the active cation induces lysosomal membrane permeabilization, which triggers lysosome-dependent cell-death due to both necrosis and caspase-dependent apoptosis. A preliminary toxicity screen in the Galleria melonella animal model was carried out on both cations and revealed no detectable toxicity up to concentrations of 80 mg/kg. Taken together, these studies indicate that this class of synthetically easy-to-access photoactive compounds offers potential as novel therapeutic leads.
Collapse
Affiliation(s)
- Felicity
F. Noakes
- Department
of Chemistry, The University of Sheffield, Western Bank, Sheffield S3 7HF, U.K.
- Department
of Biomedical Science, The University of
Sheffield, Western Bank, Sheffield S10 2TN, U.K.
| | - Kirsty L. Smitten
- Department
of Chemistry, The University of Sheffield, Western Bank, Sheffield S3 7HF, U.K.
- Department
of Molecular Biology and Biotechnology, The University of Sheffield, Western Bank, Sheffield S10 2TN, U.K.
| | - Laura E. C. Maple
- Department
of Biomedical Science, The University of
Sheffield, Western Bank, Sheffield S10 2TN, U.K.
| | - Jorge Bernardino de la Serna
- National
Heart and Lung Institute, Imperial College
London, London SW7 2AZ, U.K.
- Central
Laser
Facility, Rutherford Appleton Laboratory, Research Complex at Harwell, Science and Technology Facilities Council, Harwell-Oxford, Didcot OX11 0QX, U.K.
| | - Craig C. Robertson
- Department
of Chemistry, The University of Sheffield, Western Bank, Sheffield S3 7HF, U.K.
| | - Dylan Pritchard
- Department
of Chemistry, The University of Sheffield, Western Bank, Sheffield S3 7HF, U.K.
| | - Simon D. Fairbanks
- Department
of Chemistry, The University of Sheffield, Western Bank, Sheffield S3 7HF, U.K.
| | - Julia A. Weinstein
- Department
of Chemistry, The University of Sheffield, Western Bank, Sheffield S3 7HF, U.K.
| | - Carl G. W. Smythe
- Department
of Biomedical Science, The University of
Sheffield, Western Bank, Sheffield S10 2TN, U.K.
| | - Jim A. Thomas
- Department
of Chemistry, The University of Sheffield, Western Bank, Sheffield S3 7HF, U.K.
| |
Collapse
|
3
|
Liu H, Xu T, Xue Z, Huang M, Wang T, Zhang M, Yang R, Guo Y. Current Development of Thiazole-Containing Compounds as Potential Antibacterials against Methicillin-Resistant Staphylococcus aureus. ACS Infect Dis 2024; 10:350-370. [PMID: 38232301 DOI: 10.1021/acsinfecdis.3c00647] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
The emergence of multi-drug-resistant bacteria is threatening to human health and life around the world. In particular, methicillin-resistant Staphylococcus aureus (MRSA) causes fatal injuries to human beings and serious economic losses to animal husbandry due to its easy transmission and difficult treatment. Currently, the development of novel, highly effective, and low-toxicity antimicrobials is important to combat MRSA infections. Thiazole-containing compounds with good biological activity are widely used in clinical practice, and appropriate structural modifications make it possible to develop new antimicrobials. Here, we review thiazole-containing compounds and their antibacterial effects against MRSA reported in the past two decades and discuss their structure-activity relationships as well as the corresponding antimicrobial mechanisms. Some thiazole-containing compounds exhibit potent antibacterial efficacy in vitro and in vivo after appropriate structural modifications and could be used as antibacterial candidates. This Review provides insights into the development of thiazole-containing compounds as antimicrobials to combat MRSA infections.
Collapse
Affiliation(s)
- Hang Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan Province, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Ting Xu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan Province, China
| | - Zihan Xue
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Meijuan Huang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Tingting Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Miaomiao Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Ruige Yang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan Province, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| | - Yong Guo
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, Hunan Province, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, Henan Province, China
| |
Collapse
|
4
|
Xiao Y, Wan C, Wu X, Xu Y, Chen Y, Rao L, Wang B, Shen L, Han W, Zhao H, Shi J, Zhang J, Song Z, Yu F. Novel small-molecule compound YH7 inhibits the biofilm formation of Staphylococcus aureus in a sarX-dependent manner. mSphere 2024; 9:e0056423. [PMID: 38170984 PMCID: PMC10826350 DOI: 10.1128/msphere.00564-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 11/20/2023] [Indexed: 01/05/2024] Open
Abstract
The emergence of antibiotic-resistant and biofilm-producing Staphylococcus aureus isolates presents major challenges for treating staphylococcal infections. Biofilm inhibition is an important anti-virulence strategy. In this study, a novel maleimide-diselenide hybrid compound (YH7) was synthesized and demonstrated remarkable antimicrobial activity against methicillin-resistant S. aureus (MRSA) and methicillin-susceptible S. aureus (MSSA) in both planktonic cultures and biofilms. The minimum inhibitory concentration (MIC) of YH7 for S. aureus isolates was 16 µg/mL. Quantification of biofilms demonstrated that the sub-MIC (4 µg/mL) of YH7 significantly inhibits biofilm formation in both MSSA and MRSA. Confocal laser scanning microscopy analysis further confirmed the biofilm inhibitory potential of YH7. YH7 also significantly suppressed bacterial adherence to A549 cells. Moreover, YH7 treatment significantly inhibited S. aureus colonization in nasal tissue of mice. Preliminary mechanistic studies revealed that YH7 exerted potent biofilm-suppressing effects by inhibiting polysaccharide intercellular adhesin (PIA) synthesis, rather than suppressing bacterial autolysis. Real-time quantitative PCR data indicated that YH7 downregulated biofilm formation-related genes (clfA, fnbA, icaA, and icaD) and the global regulatory gene sarX, which promotes PIA synthesis. The sarX-dependent antibiofilm potential of YH7 was validated by constructing S. aureus NCTC8325 sarX knockout and complementation strains. Importantly, YH7 demonstrated a low potential to induce drug resistance in S. aureus and exhibited non-toxic to rabbit erythrocytes, A549, and BEAS-2B cells at antibacterial concentrations. In vivo toxicity assays conducted on Galleria mellonella further confirmed that YH7 is biocompatible. Overall, YH7 demonstrated potent antibiofilm activity supports its potential as an antimicrobial agent against S. aureus biofilm-related infections. IMPORTANCE Biofilm-associated infections, characterized by antibiotic resistance and persistence, present a formidable challenge in healthcare. Traditional antibacterial agents prove inadequate against biofilms. In this study, the novel compound YH7 demonstrates potent antibiofilm properties by impeding the adhesion and the polysaccharide intercellular adhesin production of Staphylococcus aureus. Notably, its exceptional efficacy against both methicillin-resistant and methicillin-susceptible strains highlights its broad applicability. This study highlights the potential of YH7 as a novel therapeutic agent to address the pressing issue of biofilm-driven infections.
Collapse
Affiliation(s)
- Yanghua Xiao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Public Health, Nanchang University, Nanchang, China
| | - Cailing Wan
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Public Health, Nanchang University, Nanchang, China
| | - Xiaocui Wu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yanlei Xu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yao Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Lulin Rao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bingjie Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Li Shen
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Weihua Han
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huilin Zhao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Junhong Shi
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiao Zhang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zengqiang Song
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Fangyou Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
5
|
Zhang J, Shen L, Zhou P, Chen S, Wang B, Wan C, Han W, Rao L, Zhao H, Wang X, Wu C, Shi J, Xiao Y, Song Z, Yu F, Lin C. A novel small-molecule compound S-342-3 effectively inhibits the biofilm formation of Staphylococcus aureus. Microbiol Spectr 2023; 11:e0159623. [PMID: 37819121 PMCID: PMC10714762 DOI: 10.1128/spectrum.01596-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 09/06/2023] [Indexed: 10/13/2023] Open
Abstract
IMPORTANCE Biofilms are an important virulence factor in Staphylococcus aureus and are characterized by a structured microbial community consisting of bacterial cells and a secreted extracellular polymeric matrix. Inhibition of biofilm formation is an effective measure to control S. aureus infection. Here, we have synthesized a small molecule compound S-342-3, which exhibits potent inhibition of biofilm formation in both MRSA and MSSA. Further investigations revealed that S-342-3 exerts inhibitory effects on biofilm formation by reducing the production of polysaccharide intercellular adhesin and preventing bacterial adhesion. Our study has confirmed that the inhibitory effect of S-342-3 on biofilm is achieved by downregulating the expression of genes responsible for biofilm formation. In addition, S-342-3 is non-toxic to Galleria mellonella larvae and A549 cells. Consequently, this study demonstrates the efficacy of a biologically safe compound S-342-3 in inhibiting biofilm formation in S. aureus, thereby providing a promising antibiofilm agent for further research.
Collapse
Affiliation(s)
- Jiao Zhang
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Li Shen
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Peiyao Zhou
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shuying Chen
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bingjie Wang
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Cailin Wan
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, China
| | - Weihua Han
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lulin Rao
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huilin Zhao
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xinyi Wang
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chunyang Wu
- Department of Respiratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Junhong Shi
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yanghua Xiao
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, China
| | - Zengqiang Song
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Fangyou Yu
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chunchan Lin
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
6
|
Wang S, Yin Y, Zai X, Gu Y, Guo F, Shao F, Zhang Y, Li Y, Li R, Zhang J, Xu J, Chen W. A novel Galleria mellonella experimental model for zoonotic pathogen Brucella. Virulence 2023; 14:2268496. [PMID: 37817444 PMCID: PMC10599192 DOI: 10.1080/21505594.2023.2268496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/04/2023] [Indexed: 10/12/2023] Open
Abstract
Brucellosis is a major threat to public health and animal husbandry. Several in vivo vertebrate models, such as mice, guinea pigs, and nonhuman primates, have been used to study Brucella pathogenesis, bacteria-host interactions, and vaccine efficacy. However, these models have limitations whereas the invertebrate Galleria mellonella model is a cost-effective and ethical alternative. The aim of the present study was to examine the invertebrate G. mellonella as an in vivo infection model for Brucella. Infection assays were employed to validate the fitness of the larval model for Brucella infection and virulence evaluation. The protective efficacy of immune sera was evaluated by pre-incubated with a lethal dose of bacteria before infection. The consistency between the mouse model and the larval model was confirmed by assessing the protective efficacy of two Brucella vaccine strains. The results show that G. mellonella could be infected by Brucella strains, in a dose- and temperature-dependent way. Moreover, this larval model can effectively evaluate the virulence of Brucella strains in a manner consistent with that of mammalian infection models. Importantly, this model can assess the protective efficacy of vaccine immune sera within a day. Further investigation implied that haemolymph played a crucial role in the protective efficacy of immune sera. In conclusion, G. mellonella could serve as a quick, efficient, and reliable model for evaluating the virulence of Brucella strains and efficacy of immune sera in an ethical manner.
Collapse
Affiliation(s)
- Shuyi Wang
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Ying Yin
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Xiaodong Zai
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Yanfei Gu
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Fengyu Guo
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Fangze Shao
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Yue Zhang
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Yaohui Li
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Ruihua Li
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Jun Zhang
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Junjie Xu
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Wei Chen
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, China
| |
Collapse
|
7
|
Kaya S. Immunosuppressive effect of Plantago major on the innate immunity of Galleria mellonella. PeerJ 2023; 11:e15982. [PMID: 37753175 PMCID: PMC10519203 DOI: 10.7717/peerj.15982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 08/08/2023] [Indexed: 09/28/2023] Open
Abstract
Greater plantain (Plantago major), a medicinal plant species, is used in folk medicine for the treatment of various diseases in many countries of the world. Different studies have shown that the bioactive components contained in the plant have a dual effect. It was also reported that in vivo and in vitro studies showed different results. The aim of the study was to determine the effects of P. major extract on the hemocyte-mediated and humoral immune responses of the invertebrate model organism Galleria mellonella, which is widely used in immune studies. In the evaluation of these effects, total hemocyte count, encapsulation, melanization, phenoloxidase, superoxide dismutase, catalase, malondialdehyde and total protein parameters were evaluated. The results of the study showed that the total hemocyte count did not change, that the encapsulation responses decreased, that the melanization responses and phenoloxidase activity increased and that the superoxide dismutase activity decreased. As a result, it was determined that high doses of P. major had negative effects on cell-mediated immunity and antioxidant defence and positive effects on melanization. High doses and continuous use of P. major may have negative effects on living things.
Collapse
Affiliation(s)
- Serhat Kaya
- Department of Biology/Faculty of Science, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| |
Collapse
|
8
|
Serrano I, Verdial C, Tavares L, Oliveira M. The Virtuous Galleria mellonella Model for Scientific Experimentation. Antibiotics (Basel) 2023; 12:505. [PMID: 36978373 PMCID: PMC10044286 DOI: 10.3390/antibiotics12030505] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
The first research on the insect Galleria mellonella was published 85 years ago, and the larva is now widely used as a model to study infections caused by bacterial and fungal pathogens, for screening new antimicrobials, to study the adjacent immune response in co-infections or in host-pathogen interaction, as well as in a toxicity model. The immune system of the G. mellonella model shows remarkable similarities with mammals. Furthermore, results from G. mellonella correlate positively with mammalian models and with other invertebrate models. Unlike other invertebrate models, G. mellonella can withstand temperatures of 37 °C, and its handling and experimental procedures are simpler. Despite having some disadvantages, G. mellonella is a virtuous in vivo model to be used in preclinical studies, as an intermediate model between in vitro and mammalian in vivo studies, and is a great example on how to apply the bioethics principle of the 3Rs (Replacement, Reduction, and Refinement) in animal experimentation. This review aims to discuss the progress of the G. mellonella model, highlighting the key aspects of its use, including experimental design considerations and the necessity to standardize them. A different score in the "cocoon" category included in the G. mellonella Health Index Scoring System is also proposed.
Collapse
Affiliation(s)
- Isa Serrano
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Cláudia Verdial
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Luís Tavares
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Manuela Oliveira
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisboa, Portugal
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
Ehemann K, Mantilla MJ, Mora-Restrepo F, Rios-Navarro A, Torres M, Celis Ramírez AM. Many ways, one microorganism: Several approaches to study Malassezia in interactions with model hosts. PLoS Pathog 2022; 18:e1010784. [PMID: 36074792 PMCID: PMC9455852 DOI: 10.1371/journal.ppat.1010784] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Malassezia, a lipophilic and lipid-dependent yeast, is a microorganism of current interest to mycobiologists because of its role as a commensal or pathogen in health conditions such as dermatological diseases, fungemia, and, as discovered recently, cancer and certain neurological disorders. Various novel approaches in the study of Malassezia have led to increased knowledge of the cellular and molecular mechanisms of this yeast. However, additional efforts are needed for more comprehensive understanding of the behavior of Malassezia in interactions with the host. This article reviews advances useful in the experimental field for Malassezia.
Collapse
Affiliation(s)
- Kevin Ehemann
- Grupo de Investigación Celular y Molecular de Microorganismos Patógenos (CeMoP), Departamento de Ciencias Biológicas, Universidad de los Andes, Bogotá, Colombia
| | - María Juliana Mantilla
- Grupo de Investigación Celular y Molecular de Microorganismos Patógenos (CeMoP), Departamento de Ciencias Biológicas, Universidad de los Andes, Bogotá, Colombia
| | - Felipe Mora-Restrepo
- Grupo de Investigación Celular y Molecular de Microorganismos Patógenos (CeMoP), Departamento de Ciencias Biológicas, Universidad de los Andes, Bogotá, Colombia
| | - Andrea Rios-Navarro
- Grupo de Investigación Celular y Molecular de Microorganismos Patógenos (CeMoP), Departamento de Ciencias Biológicas, Universidad de los Andes, Bogotá, Colombia
| | - Maritza Torres
- Grupo de Investigación Celular y Molecular de Microorganismos Patógenos (CeMoP), Departamento de Ciencias Biológicas, Universidad de los Andes, Bogotá, Colombia
| | - Adriana Marcela Celis Ramírez
- Grupo de Investigación Celular y Molecular de Microorganismos Patógenos (CeMoP), Departamento de Ciencias Biológicas, Universidad de los Andes, Bogotá, Colombia
| |
Collapse
|
12
|
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: 6] [Impact Index Per Article: 2.0] [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
|
13
|
Garcia Maset R, Hapeshi A, Hall S, Dalgliesh RM, Harrison F, Perrier S. Evaluation of the Antimicrobial Activity in Host-Mimicking Media and In Vivo Toxicity of Antimicrobial Polymers as Functional Mimics of AMPs. ACS APPLIED MATERIALS & INTERFACES 2022; 14:32855-32868. [PMID: 35819416 PMCID: PMC9335526 DOI: 10.1021/acsami.2c05979] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Activity tests for synthetic antimicrobial compounds are often limited to the minimal inhibitory concentration assay using standard media and bacterial strains. In this study, a family of acrylamide copolymers that act as synthetic mimics of antimicrobial peptides were synthesized and shown to have a disruptive effect on bacterial membranes and structural integrity through microscopy techniques and membrane polarization experiments. The polymers were tested for their antimicrobial properties using media that mimic clinically relevant conditions. Additionally, their activity was compared in two different strains of the Gram-positive bacterium Staphylococcus aureus and the Gram-negative bacterium Pseudomonas aeruginosa. We showed that the medium composition can have an important influence on the polymer activity as there was a considerable reduction in minimal inhibitory concentrations against S. aureus grown in synthetic wound fluid (SWF), and against P. aeruginosa grown in synthetic cystic fibrosis sputum media (SCFM), compared to the concentrations in standard testing media. In contrast, we observed a complete loss of activity against P. aeruginosa in the serum-containing SWF. Finally, we made use of an emerging invertebrate in vivo model, using Galleria mellonella larvae, to assess toxicity of the polymeric antimicrobials, showing a good correlation with cell line toxicity measurements and demonstrating its potential in the evaluation of novel antimicrobial materials.
Collapse
Affiliation(s)
| | - Alexia Hapeshi
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
| | - Stephen Hall
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
- ISIS
Neutron and Muon Source, Rutherford Appleton
Laboratory, Didcot OX11 0DE, U.K.
| | - Robert M. Dalgliesh
- ISIS
Neutron and Muon Source, Rutherford Appleton
Laboratory, Didcot OX11 0DE, U.K.
| | - Freya Harrison
- School
of Life Sciences, University of Warwick, Coventry CV4 7AL, U.K.
| | - Sébastien Perrier
- Warwick
Medical School, University of Warwick, Coventry CV4 7AL, U.K.
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
- Faculty
of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| |
Collapse
|
14
|
O’Shaughnessy M, Piatek M, McCarron P, McCann M, Devereux M, Kavanagh K, Howe O. In Vivo Activity of Metal Complexes Containing 1,10-Phenanthroline and 3,6,9-Trioxaundecanedioate Ligands against Pseudomonas aeruginosa Infection in Galleria mellonella Larvae. Biomedicines 2022; 10:biomedicines10020222. [PMID: 35203432 PMCID: PMC8869450 DOI: 10.3390/biomedicines10020222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 11/16/2022] Open
Abstract
Drug-resistant Pseudomonas aeruginosa is rapidly developing resulting in a serious global threat. Immunocompromised patients are specifically at risk, especially those with cystic fibrosis (CF). Novel metal complexes incorporating 1,10-phenanthroline (phen) ligands have previously demonstrated antibacterial and anti-biofilm effects against resistant P. aeruginosa from CF patients in vitro. Herein, we present the in vivo efficacy of {[Cu(3,6,9-tdda)(phen)2]·3H2O·EtOH}n (Cu-tdda-phen), {[Mn(3,6,9-tdda)(phen)2]·3H2O·EtOH}n (Mn-tdda-phen) and [Ag2(3,6,9-tdda)(phen)4]·EtOH (Ag-tdda-phen) (tddaH2 = 3,6,9-trioxaundecanedioic acid). Individual treatments of these metal-tdda-phen complexes and in combination with the established antibiotic gentamicin were evaluated in vivo in larvae of Galleria mellonella infected with clinical isolates and laboratory strains of P. aeruginosa. G. mellonella were able to tolerate all test complexes up to 10 µg/larva. In addition, the immune response was affected by stimulation of immune cells (hemocytes) and genes that encode for immune-related peptides, specifically transferrin and inducible metallo-proteinase inhibitor. The amalgamation of metal-tdda-phen complexes and gentamicin further intensified this response at lower concentrations, clearing a P. aeruginosa infection that were previously resistant to gentamicin alone. Therefore this work highlights the anti-pseudomonal capabilities of metal-tdda-phen complexes alone and combined with gentamicin in an in vivo model.
Collapse
Affiliation(s)
- Megan O’Shaughnessy
- School of Biological and Health Sciences, Technological University Dublin-City Campus, D07 ADY7 Dublin, Ireland;
- Centre for Biomimetic and Therapeutic Research, FOCAS Research Institute, Technological University Dublin-City Campus, D08 CKP1 Dublin, Ireland; (P.M.); (M.D.)
| | - Magdalena Piatek
- SSPC Pharma Research Centre, Department of Biology, Maynooth University, W23 F2H6 Kildare, Ireland;
| | - Pauraic McCarron
- Centre for Biomimetic and Therapeutic Research, FOCAS Research Institute, Technological University Dublin-City Campus, D08 CKP1 Dublin, Ireland; (P.M.); (M.D.)
| | - Malachy McCann
- Chemistry Department, Maynooth University, W23 F2H6 Kildare, Ireland;
| | - Michael Devereux
- Centre for Biomimetic and Therapeutic Research, FOCAS Research Institute, Technological University Dublin-City Campus, D08 CKP1 Dublin, Ireland; (P.M.); (M.D.)
| | - Kevin Kavanagh
- SSPC Pharma Research Centre, Department of Biology, Maynooth University, W23 F2H6 Kildare, Ireland;
- Correspondence: (K.K.); (O.H.)
| | - Orla Howe
- School of Biological and Health Sciences, Technological University Dublin-City Campus, D07 ADY7 Dublin, Ireland;
- Centre for Biomimetic and Therapeutic Research, FOCAS Research Institute, Technological University Dublin-City Campus, D08 CKP1 Dublin, Ireland; (P.M.); (M.D.)
- Correspondence: (K.K.); (O.H.)
| |
Collapse
|
15
|
Xu MN, Li L, Pan W, Zheng HX, Wang ML, Peng XM, Dai SQ, Tang YM, Zeng K, Huang XW. Zinc Oxide Nanoparticles Prime a Protective Immune Response in Galleria mellonella to Defend Against Candida albicans. Front Microbiol 2021; 12:766138. [PMID: 34956129 PMCID: PMC8702860 DOI: 10.3389/fmicb.2021.766138] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 11/11/2021] [Indexed: 01/21/2023] Open
Abstract
Purpose: Zinc oxide nanoparticles (ZnO-NPs) have exerted antimicrobial properties. However, there is insufficient evaluation regarding the in vivo antifungal activity of ZnO-NPs. This study aimed to investigate the efficacy and mechanism of ZnO-NPs in controlling Candida albicans in the invertebrate Galleria mellonella. Methods: Galleria mellonella larvae were injected with different doses of ZnO-NPs to determine their in vivo toxicity. Non-toxic doses of ZnO-NPs were chosen for prophylactic injection in G. mellonella followed by C. albicans infection. Then the direct in vitro antifungal effect of ZnO-NPs against C. albicans was evaluated. In addition, the mode of action of ZnO-NPs was assessed in larvae through different assays: quantification of hemocyte density, morphology observation of hemocytes, characterization of hemocyte aggregation and phagocytosis, and measurement of hemolymph phenoloxidase (PO) activity. Results: Zinc oxide nanoparticles were non-toxic to the larvae at relatively low concentrations (≤20 mg/kg). ZnO-NP pretreatment significantly prolonged the survival of C. albicans-infected larvae and decreased the fungal dissemination and burden in the C. albicans-infected larvae. This observation was more related to the activation of host defense rather than their fungicidal capacities. Specifically, ZnO-NP treatment increased hemocyte density, promoted hemocyte aggregation, enhanced hemocyte phagocytosis, and activated PO activity in larvae. Conclusion: Prophylactic treatment with lower concentrations of ZnO-NPs protects G. mellonella from C. albicans infection. The innate immune response primed by ZnO-NPs may be part of the reason for the protective effects. This study provides new evidence of the capacity of ZnO-NPs in enhancing host immunity and predicts that ZnO-NPs will be attractive for further anti-infection applications.
Collapse
Affiliation(s)
- Mei-Nian Xu
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Li Li
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wen Pan
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Huan-Xin Zheng
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Meng-Lei Wang
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiao-Ming Peng
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Si-Qi Dai
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ying-Mei Tang
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kang Zeng
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiao-Wen Huang
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
16
|
de Andrades EO, da Costa JMAR, de Lima Neto FEM, de Araujo AR, de Oliveira Silva Ribeiro F, Vasconcelos AG, de Jesus Oliveira AC, Sobrinho JLS, de Almeida MP, Carvalho AP, Dias JN, Silva IGM, Albuquerque P, Pereira IS, do Amaral Rabello D, das Graças Nascimento Amorim A, de Souza de Almeida Leite JR, da Silva DA. Acetylated cashew gum and fucan for incorporation of lycopene rich extract from red guava (Psidium guajava L.) in nanostructured systems: Antioxidant and antitumor capacity. Int J Biol Macromol 2021; 191:1026-1037. [PMID: 34563578 DOI: 10.1016/j.ijbiomac.2021.09.116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 09/09/2021] [Accepted: 09/17/2021] [Indexed: 12/18/2022]
Abstract
Industrial application of lycopene is limited due to its chemical instability and low bioavailability. This study proposes the development of fucan-coated acetylated cashew gum nanoparticles (NFGa) and acetylated cashew gum nanoparticles (NGa) for incorporation of the lycopene-rich extract from red guava (LEG). Size, polydispersity, zeta potential, nanoparticles concentration, encapsulation efficiency, transmission electron microscopy (TEM) and atomic force microscopy (AFM) were used to characterize nanoparticles. The antioxidant activity was determinated and cell viability was evaluated in the human breast cancer cells (MCF-7) and human keratinocytes (HaCaT) by MTT assay. The toxic effect was evaluated by hemolysis test and by Galleria mellonella model. NFGa showed higher stability than NGa, having a size of 162.10 ± 3.21 nm, polydispersity of 0.348 ± 0.019, zeta potential -30.70 ± 0.53 mV, concentration of 6.4 × 109 nanoparticles/mL and 60% LEG encapsulation. Microscopic analysis revealed a spherical and smooth shape of NFGa. NFGa showed antioxidant capacity by ABTS method and ORAC assay. The NFGa presented significant cytotoxicity against MCF-7 from the lowest concentration tested (6.25-200 μg/mL) and did not affect the cell viability of the HaCaT. NFGa showed non-toxic effect in the in vitro and in vivo models. Therefore, NFGa may have a promising application in LEG stabilization for antioxidant and antitumor purposes.
Collapse
Affiliation(s)
- Eryka Oliveira de Andrades
- Programa de Pós-Graduação em Biotecnologia, RENORBIO, Brazil; Núcleo de Pesquisa em Biodiversidade e Biotecnologia, BIOTEC, Universidade Federal do Delta do Parnaíba, UFDPar, Parnaíba, PI, Brazil
| | | | | | - Alyne Rodrigues de Araujo
- Núcleo de Pesquisa em Biodiversidade e Biotecnologia, BIOTEC, Universidade Federal do Delta do Parnaíba, UFDPar, Parnaíba, PI, Brazil
| | - Fabio de Oliveira Silva Ribeiro
- Núcleo de Pesquisa em Biodiversidade e Biotecnologia, BIOTEC, Universidade Federal do Delta do Parnaíba, UFDPar, Parnaíba, PI, Brazil
| | - Andreanne Gomes Vasconcelos
- Núcleo de Pesquisa em Morfologia e Imunologia Aplicada, NuPMIA, Área de Morfologia, Faculdade de Medicina, Universidade de Brasília, UnB, Brasília, DF, Brazil
| | - Antônia Carla de Jesus Oliveira
- Núcleo de Controle de Qualidade de Medicamentos e Correlatos, NCQMC, Departamento de Ciências Farmacêuticas, Universidade Federal de Pernambuco, UFPE, Recife, PE, Brazil
| | - José Lamartine Soares Sobrinho
- Núcleo de Controle de Qualidade de Medicamentos e Correlatos, NCQMC, Departamento de Ciências Farmacêuticas, Universidade Federal de Pernambuco, UFPE, Recife, PE, Brazil
| | - Miguel Peixoto de Almeida
- LAQV/REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - Ana P Carvalho
- LAQV/REQUIMTE-GRAQ, Instituto Superior de Engenharia, Instituto Politécnico do Porto, Porto, Portugal; Centro de Biotecnologia e Química Fina, CBQF, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| | - Jhones Nascimento Dias
- Laboratório de Biologia Molecular de Fungos Patogênicos, Instituto de Biologia, Universidade de Brasília, UnB, Brasília, DF, Brazil
| | | | - Patrícia Albuquerque
- Laboratório de Biologia Molecular de Fungos Patogênicos, Instituto de Biologia, Universidade de Brasília, UnB, Brasília, DF, Brazil
| | - Ildinete Silva Pereira
- Laboratório de Biologia Molecular de Fungos Patogênicos, Instituto de Biologia, Universidade de Brasília, UnB, Brasília, DF, Brazil
| | - Doralina do Amaral Rabello
- Laboratório de Patologia Molecular do Câncer, Área de Patologia, Faculdade de Medicina, Universidade de Brasília, UnB, Brasília, DF, Brazil
| | | | - José Roberto de Souza de Almeida Leite
- Núcleo de Pesquisa em Biodiversidade e Biotecnologia, BIOTEC, Universidade Federal do Delta do Parnaíba, UFDPar, Parnaíba, PI, Brazil; Núcleo de Pesquisa em Morfologia e Imunologia Aplicada, NuPMIA, Área de Morfologia, Faculdade de Medicina, Universidade de Brasília, UnB, Brasília, DF, Brazil
| | - Durcilene Alves da Silva
- Programa de Pós-Graduação em Biotecnologia, RENORBIO, Brazil; Núcleo de Pesquisa em Biodiversidade e Biotecnologia, BIOTEC, Universidade Federal do Delta do Parnaíba, UFDPar, Parnaíba, PI, Brazil.
| |
Collapse
|
17
|
Antoine C, Laforêt F, Blasdel B, Fall A, Duprez JN, Mainil J, Delcenserie V, Thiry D. In Vitro Characterization and In Vivo Efficacy Assessment in Galleria mellonella Larvae of Newly Isolated Bacteriophages against Escherichia coli K1. Viruses 2021; 13:2005. [PMID: 34696434 PMCID: PMC8541614 DOI: 10.3390/v13102005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 01/04/2023] Open
Abstract
Extra-intestinal Escherichia coli express several virulence factors that increase their ability to colonize and survive in different localizations. The K1 capsular type is involved in several infections, including meningitis, urinary tract, and bloodstream infections. The aims of this work were to isolate, characterize, and assess the in vivo efficacy of phages targeting avian pathogenic E. coli (APEC) O18:K1, which shares many similarities with the human strains responsible for neonatal meningitis. Eleven phages were isolated against APEC O18:K1, and four of them presenting a narrow spectrum targeting E. coli K1 strains were further studied. The newly isolated phages vB_EcoS_K1-ULINTec2 were similar to the Siphoviridae family, and vB_EcoP_K1-ULINTec4, vB_EcoP_K1-ULINTec6, and vB_EcoP_K1-ULINTec7 to the Autographiviridae family. They are capsular type (K1) dependent and present several advantages characteristic of lytic phages, such as a short adsorption time and latent period. vB_EcoP_K1-ULINTec7 is able to target both K1 and K5 strains. This study shows that these phages replicate efficiently, both in vitro and in vivo in the Galleria mellonella model. Phage treatment increases the larvae survival rates, even though none of the phages were able to eliminate the bacterial load.
Collapse
Affiliation(s)
- Céline Antoine
- Bacteriology Laboratory, Department of Infectious and Parasitic Diseases, FARAH and Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium; (C.A.); (F.L.); (J.-N.D.); (J.M.)
- Food Science Department, FARAH and Faculty of Veterinary Medicine, ULiège, 4000 Liège, Belgium;
| | - Fanny Laforêt
- Bacteriology Laboratory, Department of Infectious and Parasitic Diseases, FARAH and Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium; (C.A.); (F.L.); (J.-N.D.); (J.M.)
- Food Science Department, FARAH and Faculty of Veterinary Medicine, ULiège, 4000 Liège, Belgium;
| | - Bob Blasdel
- Vésale Bioscience, Vésale Pharmaceutica, 5310 Noville-sur-Mehaigne, Belgium;
| | - Abdoulaye Fall
- Genalyse Partner SA, En Hayeneux 62, 4040 Herstal, Belgium;
| | - Jean-Noël Duprez
- Bacteriology Laboratory, Department of Infectious and Parasitic Diseases, FARAH and Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium; (C.A.); (F.L.); (J.-N.D.); (J.M.)
| | - Jacques Mainil
- Bacteriology Laboratory, Department of Infectious and Parasitic Diseases, FARAH and Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium; (C.A.); (F.L.); (J.-N.D.); (J.M.)
| | - Véronique Delcenserie
- Food Science Department, FARAH and Faculty of Veterinary Medicine, ULiège, 4000 Liège, Belgium;
| | - Damien Thiry
- Bacteriology Laboratory, Department of Infectious and Parasitic Diseases, FARAH and Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium; (C.A.); (F.L.); (J.-N.D.); (J.M.)
| |
Collapse
|
18
|
Possamai Rossatto FC, Tharmalingam N, Escobar IE, d’Azevedo PA, Zimmer KR, Mylonakis E. Antifungal Activity of the Phenolic Compounds Ellagic Acid (EA) and Caffeic Acid Phenethyl Ester (CAPE) against Drug-Resistant Candida auris. J Fungi (Basel) 2021; 7:jof7090763. [PMID: 34575801 PMCID: PMC8466507 DOI: 10.3390/jof7090763] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/06/2021] [Accepted: 09/06/2021] [Indexed: 12/17/2022] Open
Abstract
Candida auris is an emerging healthcare-associated fungal pathogen that has become a serious global health threat. Current treatment options are limited due to drug resistance. New therapeutic strategies are required to target this organism and its pathogenicity. Plant polyphenols are structurally diverse compounds that present a vast range of biological properties. In the present study, plant-derived molecules ellagic acid (EA) and caffeic acid phenethyl ester (CAPE) were investigated for their antifungal and antivirulence activities against Candida auris. We also tested against C. albicans. The minimum inhibitory concentration (MIC) for EA ranged from 0.125 to 0.25 µg/mL and for CAPE ranged from 1 to 64 µg/mL against drug-resistant C. auris strains. Killing kinetics determined that after 4 h treatment with CAPE, there was a complete reduction of viable C. auris cells compared to fluconazole. Both compounds might act by modifying the fungal cell wall. CAPE significantly reduced the biomass and the metabolic activity of C. auris biofilm and impaired C. auris adhesion to cultured human epithelial cells. Furthermore, both compounds prolonged the survival rate of Galleria mellonella infected by C. auris (p = 0.0088 for EA at 32 mg/kg and p = 0.0028 for CAPE at 4 mg/kg). In addition, EA at 4 μg/mL prolonged the survival of C. albicans-infected Caenorhabditis elegans (p < 0.0001). CAPE was not able to prolong the survival of C. albicans-infected C. elegans. These findings highlight the antifungal and antivirulence effects of EA and CAPE against C. auris, and warrant further investigation as novel antifungal agents against drug-resistant infections.
Collapse
Affiliation(s)
- Fernanda Cristina Possamai Rossatto
- Laboratory of Biofilms and Alternative Models, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, RS, Brazil; (F.C.P.R.); (P.A.d.); (K.R.Z.)
| | - Nagendran Tharmalingam
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School at Brown University, 593 Eddy Street, P.O. Box 328/330, Providence, RI 02903, USA; (N.T.); (I.E.E.)
| | - Iliana E. Escobar
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School at Brown University, 593 Eddy Street, P.O. Box 328/330, Providence, RI 02903, USA; (N.T.); (I.E.E.)
| | - Pedro Alves d’Azevedo
- Laboratory of Biofilms and Alternative Models, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, RS, Brazil; (F.C.P.R.); (P.A.d.); (K.R.Z.)
| | - Karine Rigon Zimmer
- Laboratory of Biofilms and Alternative Models, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, RS, Brazil; (F.C.P.R.); (P.A.d.); (K.R.Z.)
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School at Brown University, 593 Eddy Street, P.O. Box 328/330, Providence, RI 02903, USA; (N.T.); (I.E.E.)
- Correspondence: ; Tel.: +1-401-444-7845; Fax: +1-401-444-8179
| |
Collapse
|
19
|
Stączek S, Zdybicka-Barabas A, Wiater A, Pleszczyńska M, Cytryńska M. Activation of cellular immune response in insect model host Galleria mellonella by fungal α-1,3-glucan. Pathog Dis 2021; 78:6000214. [PMID: 33232457 PMCID: PMC7726367 DOI: 10.1093/femspd/ftaa062] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/06/2020] [Indexed: 12/26/2022] Open
Abstract
Alpha-1,3-glucan, in addition to β-1,3-glucan, is an important polysaccharide component of fungal cell walls. It is reported for many fungal species, including human pathogenic genera: Aspergillus, Blastomyces, Coccidioides, Cryptococcus, Histoplasma and Pneumocystis, plant pathogens, e.g. Magnaporthe oryzae and entomopathogens, e.g. Metarhizium acridum. In human and plant pathogenic fungi, α-1,3-glucan is considered as a shield for the β-1,3-glucan layer preventing recognition of the pathogen by the host. However, its role in induction of immune response is not clear. In the present study, the cellular immune response of the greater wax moth Galleria mellonella to Aspergillus niger α-1,3-glucan was investigated for the first time. The changes detected in the total hemocyte count (THC) and differential hemocyte count (DHC), formation of hemocyte aggregates and changes in apolipophorin III localization indicated activation of G. mellonella cellular mechanisms in response to immunization with A. niger α-1,3-glucan. Our results, which have clearly demonstrated the response of the insect immune system to this fungal cell wall component, will help in understanding the α-1,3-glucan role in immune response against fungal pathogens not only in insects but also in mammals, including humans.
Collapse
Affiliation(s)
- Sylwia Stączek
- Maria Curie-Skłodowska University, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Department of Immunobiology, Akademicka 19 St., 20-033 Lublin, Poland
| | - Agnieszka Zdybicka-Barabas
- Maria Curie-Skłodowska University, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Department of Immunobiology, Akademicka 19 St., 20-033 Lublin, Poland
| | - Adrian Wiater
- Maria Curie-Skłodowska University, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Department of Industrial and Environmental Microbiology, Akademicka 19 St., 20-033 Lublin, Poland
| | - Małgorzata Pleszczyńska
- Maria Curie-Skłodowska University, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Department of Industrial and Environmental Microbiology, Akademicka 19 St., 20-033 Lublin, Poland
| | - Małgorzata Cytryńska
- Maria Curie-Skłodowska University, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Department of Immunobiology, Akademicka 19 St., 20-033 Lublin, Poland
| |
Collapse
|
20
|
Ismail MM, Hassan M, Moawad SS, Okba MM, Ashour RM, Fayek NM, Saber FR. Exploring the Antivirulence Activity of Pulverulentone A, a Phloroglucinol-Derivative from Callistemon citrinus Leaf Extract, against Multi-Drug Resistant Pseudomonas aeruginosa. Antibiotics (Basel) 2021; 10:antibiotics10080907. [PMID: 34438957 PMCID: PMC8388764 DOI: 10.3390/antibiotics10080907] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/18/2021] [Accepted: 07/21/2021] [Indexed: 12/17/2022] Open
Abstract
(1) Background: Bacterial resistance to antibiotics is a global life-threatening issue. Antivirulence therapy is a promising approach to combat bacterial infections as it disarms the bacteria from their virulence factors with reduced selective pressure and a lower chance of resistance. (2) Methods: Callistemon citrinus leaf extract and its major constituent, Pulverulentone A, were tested for their ability to inhibit biofilm, exopolysaccharides, pyocyanin and proteases produced by MDR P. aeruginosa. In addition, a Galleria mellonella larvae model was employed to evaluate the in vivo cytotoxicity of Pulverulentone A and its ability to combat Pseudomonas infection. Docking study was further performed to investigate Pulverulentone A druggability against main quorum sensing (QS) targets expressed by P. aeruginosa; (3) Results: Both C. citrinus extract and the isolated compound could inhibit biofilm formation, extracellular polymeric substances (EPS) and pigment production by the tested isolates. Unexpectedly, no significant inhibition was observed on proteases production. The in silico docking analysis revealed good interactions of Pulverulentone A with all QS targets examined (LasR, MyfR/PqsR, QscR). Pulverulentone A was safe up to 400 µg·mL−1 in Galleria caterpillars. Moreover, pre-treatment of P. aeruginosa with Pulverulentone A slightly enhanced the survival of the infected larvae. (4) Conclusions: The present study proves Pulverulentone A safety with significant in vitro and in silico antivirulence potential against P. aeruginosa.
Collapse
Affiliation(s)
- Maha M. Ismail
- Microbiology and Immunology Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt;
- Correspondence: (M.M.I.); (F.R.S.); Tel./Fax: +20-3628426 (ext. 00202) (M.M.I.); Tel.: +20-1004405983 (F.R.S.)
| | - Mariam Hassan
- Microbiology and Immunology Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt;
| | - Sawsan S. Moawad
- Department of Pests and Plant Protection, National Research Center (NRC), Giza 12622, Egypt;
| | - Mona M. Okba
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt; (M.M.O.); (R.M.A.); (N.M.F.)
| | - Rehab M. Ashour
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt; (M.M.O.); (R.M.A.); (N.M.F.)
| | - Nesrin M. Fayek
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt; (M.M.O.); (R.M.A.); (N.M.F.)
| | - Fatema R. Saber
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt; (M.M.O.); (R.M.A.); (N.M.F.)
- Correspondence: (M.M.I.); (F.R.S.); Tel./Fax: +20-3628426 (ext. 00202) (M.M.I.); Tel.: +20-1004405983 (F.R.S.)
| |
Collapse
|
21
|
New Quinone Antibiotics against Methicillin-Resistant S. aureus. Antibiotics (Basel) 2021; 10:antibiotics10060614. [PMID: 34063846 PMCID: PMC8224091 DOI: 10.3390/antibiotics10060614] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 05/10/2021] [Accepted: 05/18/2021] [Indexed: 01/21/2023] Open
Abstract
There is an urgent need for the development of new antibiotics. Here, we describe the inhibitory activity of new quinone compounds against methicillin-resistant Staphylococcus aureus (ATCC® 43300), methicillin-sensitive S. aureus (ATCC® 29213), and two clinical isolates from Chile (ISP-213 and ISP-214). We observed 99.9% reduction in viability within 2 h of exposure without the cultures exhibiting any post-antibiotic effect, which was twice the kinetics to that observed with vancomycin. These clinical isolates did not acquire resistance to these quinone derivatives during the course of our study. We found that these compounds protected larvae of the greater wax moth, sp. Galleria mellonella, from infection by these MRSA clinical strains as effectively as vancomycin. These quinone derivatives are potential drug candidates worth further development.
Collapse
|
22
|
Moya-Andérico L, Vukomanovic M, Cendra MDM, Segura-Feliu M, Gil V, Del Río JA, Torrents E. Utility of Galleria mellonella larvae for evaluating nanoparticle toxicology. CHEMOSPHERE 2021; 266:129235. [PMID: 33316472 DOI: 10.1016/j.chemosphere.2020.129235] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 05/27/2023]
Abstract
The use of nanoparticles in consumer products is currently on the rise, so it is important to have reliable methods to predict any associated toxicity effects. Traditional in vitro assays fail to mimic true physiological responses of living organisms against nanoparticles whereas murine in vivo models are costly and ethically controversial. For these reasons, this study aimed to evaluate the efficacy of Galleria mellonella as an alternative, non-rodent in vivo model for examining nanoparticle toxicity. Silver, selenium, and functionalized gold nanoparticles were synthesized, and their toxicity was assessed in G. mellonella larvae. The degree of acute toxicity effects caused by each type of NP was efficiently detected by an array of indicators within the larvae: LD50 calculation, hemocyte proliferation, NP distribution, behavioral changes, and histological alterations. G. mellonella larvae are proposed as a nanotoxicological model that can be used as a bridge between in vitro and in vivo murine assays in order to obtain better predictions of NP toxicity.
Collapse
Affiliation(s)
- Laura Moya-Andérico
- Bacterial Infections: Antimicrobial Therapies group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Marija Vukomanovic
- Advanced Materials Department, Jozef Stefan Institute, Ljubljana, Slovenia
| | - Maria Del Mar Cendra
- Bacterial Infections: Antimicrobial Therapies group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Miriam Segura-Feliu
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain; Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - Vanessa Gil
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain; Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - José A Del Río
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain; Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
| | - Eduard Torrents
- Bacterial Infections: Antimicrobial Therapies group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; Microbiology Section, Department of Genetics, Microbiology, and Statistics, Biology Faculty, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
23
|
Zhao X, Chlebowicz-Flissikowska MA, Wang M, Vera Murguia E, de Jong A, Becher D, Maaß S, Buist G, van Dijl JM. Exoproteomic profiling uncovers critical determinants for virulence of livestock-associated and human-originated Staphylococcus aureus ST398 strains. Virulence 2020; 11:947-963. [PMID: 32726182 PMCID: PMC7550020 DOI: 10.1080/21505594.2020.1793525] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 06/20/2020] [Accepted: 06/30/2020] [Indexed: 12/24/2022] Open
Abstract
Staphylococcus aureus: with the sequence type (ST) 398 was previously associated with livestock carriage. However, in recent years livestock-independent S. aureus ST398 has emerged, representing a potential health risk for humans especially in nosocomial settings. Judged by whole-genome sequencing analyses, the livestock- and human originated strains belong to two different S. aureus ST398 clades but, to date, it was not known to what extent these clades differ in terms of actual virulence. Therefore, the objective of this study was to profile the exoproteomes of 30 representative S. aureus ST398 strains by mass spectrometry, to assess clade-specific differences in virulence factor secretion, and to correlate the identified proteins and their relative abundance to the strains' actual virulence. Although the human-originated strains are more heterogeneous at the genome level, our observations show that they are more homogeneous in terms of virulence factor production than the livestock-associated strains. To assess differences in virulence, infection models based on larvae of the wax moth Galleria mellonella and the human HeLa cell line were applied. Correlation of the exoproteome data to larval killing and toxicity toward HeLa cells uncovered critical roles of the staphylococcal Sbi, SpA, SCIN and CHIPS proteins in virulence. These findings were validated by showing that sbi or spa mutant bacteria are attenuated in G. mellonella and that the purified SCIN and CHIPS proteins are toxic for HeLa cells. Altogether, we show that exoproteome profiling allows the identification of critical determinants for virulence of livestock-associated and human-originated S. aureus ST398 strains.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Min Wang
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Elias Vera Murguia
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anne de Jong
- Department of Molecular Genetics, University of Groningen, Groningen Biomolecular Sciences and Biotechnology Institute, Groningen, The Netherlands
| | - Dörte Becher
- Institute of Microbiology, University of Greifswald, Greifswald, Germany
| | - Sandra Maaß
- Institute of Microbiology, University of Greifswald, Greifswald, Germany
| | - Girbe Buist
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan Maarten van Dijl
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
24
|
Monitoring Gene Expression during a Galleria mellonella Bacterial Infection. Microorganisms 2020; 8:microorganisms8111798. [PMID: 33207842 PMCID: PMC7697238 DOI: 10.3390/microorganisms8111798] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 12/05/2022] Open
Abstract
Galleria mellonella larvae are an alternative in vivo model that has been extensively used to study the virulence and pathogenicity of different bacteria due to its practicality and lack of ethical constraints. However, the larvae possess intrinsic autofluorescence that obstructs the use of fluorescent proteins to study bacterial infections, hence better methodologies are needed. Here, we report the construction of a promoter probe vector with bioluminescence expression as well as the optimization of a total bacterial RNA extraction protocol to enhance the monitoring of in vivo infections. By employing the vector to construct different gene promoter fusions, variable gene expression levels were efficiently measured in G. mellonella larvae at various time points during the course of infection and without much manipulation of the larvae. Additionally, our optimized RNA extraction protocol facilitates the study of transcriptional gene levels during an in vivo infection. The proposed methodologies will greatly benefit bacterial infection studies as they can contribute to a better understanding of the in vivo infection processes and pathogen–mammalian host interactions.
Collapse
|
25
|
Thomaz L, Gustavo de Almeida L, Silva FRO, Cortez M, Taborda CP, Spira B. In vivo Activity of Silver Nanoparticles Against Pseudomonas aeruginosa Infection in Galleria mellonella. Front Microbiol 2020; 11:582107. [PMID: 33240236 PMCID: PMC7680755 DOI: 10.3389/fmicb.2020.582107] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/16/2020] [Indexed: 01/30/2023] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen associated with life-threatening nosocomial and community-acquired infections. Antibiotic resistance is an immediate threat to public health and demands an urgent action to discovering new antimicrobial agents. One of the best alternatives for pre-clinical tests with animal models is the greater wax moth Galleria mellonella. Here, we evaluated the antipseudomonal activity of silver nanoparticles (AgNPs) against P. aeruginosa strain UCBPP-PA14 using G. mellonella larvae. The AgNPs were synthesized through a non-toxic biogenic process involving microorganism fermentation. The effect of AgNPs was assessed through characterization and quantification of the hemocytic response, nodulation and phenoloxidase cascade. On average, 80% of the larvae infected with P. aeruginosa and prophylactically treated with nanoparticles survived. Both the specific and total larvae hemocyte counts were restored in the treated group. In addition, the nodulation process and the phenoloxidase cascade were less exacerbated when the larvae were exposed to the silver nanoparticles. AgNPs protect the larvae from P. aeruginosa infection by directly killing the bacteria and indirectly by preventing an exacerbated immunological response against the pathogen. Our results suggest that the prophylactic use of AgNPs has a strong protective activity against P. aeruginosa infection.
Collapse
Affiliation(s)
- Luciana Thomaz
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Luiz Gustavo de Almeida
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | | | - Mauro Cortez
- Department of Parasitology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Carlos P. Taborda
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
- Laboratory of Medical Mycology/LIM53, Faculty of Medicine, Institute of Tropical Medicine of São Paulo, University of São Paulo, São Paulo, Brazil
| | - Beny Spira
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
26
|
Strong Environment-Genotype Interactions Determine the Fitness Costs of Antibiotic Resistance In Vitro and in an Insect Model of Infection. Antimicrob Agents Chemother 2020; 64:AAC.01033-20. [PMID: 32661001 DOI: 10.1128/aac.01033-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/08/2020] [Indexed: 11/20/2022] Open
Abstract
The acquisition of antibiotic resistance commonly imposes fitness costs, a reduction in the fitness of bacteria in the absence of drugs. These costs have been quantified primarily using in vitro experiments and a small number of in vivo studies in mice, and it is commonly assumed that these diverse methods are consistent. Here, we used an insect model of infection to compare the fitness costs of antibiotic resistance in vivo to those in vitro Experiments explored diverse mechanisms of resistance in a Gram-positive pathogen, Bacillus thuringiensis, and a Gram-negative intestinal symbiont, Enterobacter cloacae Rifampin resistance in B. thuringiensis showed fitness costs that were typically elevated in vivo, although these were modulated by genotype-environment interactions. In contrast, resistance to cefotaxime via derepression of AmpC β-lactamase in E. cloacae resulted in no detectable costs in vivo or in vitro, while spontaneous resistance to nalidixic acid, and carriage of the IncP plasmid RP4, imposed costs that increased in vivo Overall, fitness costs in vitro were a poor predictor of fitness costs in vivo because of strong genotype-environment interactions throughout this study. Insect infections provide a cheap and accessible means of assessing the fitness consequences of resistance mutations, data that are important for understanding the evolution and spread of resistance. This study emphasizes that the fitness costs imposed by particular mutations or different modes of resistance are extremely variable and that only a subset of these mutations is likely to be prevalent outside the laboratory.
Collapse
|
27
|
Skalska J, Andrade VM, Cena GL, Harvey PJ, Gaspar D, Mello ÉO, Henriques ST, Valle J, Gomes VM, Conceição K, Castanho MARB, Andreu D. Synthesis, Structure, and Activity of the Antifungal Plant Defensin PvD 1. J Med Chem 2020; 63:9391-9402. [PMID: 32787086 DOI: 10.1021/acs.jmedchem.0c00543] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Available treatments for invasive fungal infections have limitations, including toxicity and the emergence of resistant strains. Therefore, there is an urgent need for alternative solutions. Because of their unique mode of action and high selectivity, plant defensins (PDs) are worthy therapeutic candidates. Chemical synthesis remains a preferred method for the production of many peptide-based therapeutics. Given the relatively long sequence of PDs, as well as their complicated posttranslational modifications, the synthetic route can be considered challenging. Here, we describe a total synthesis of PvD1, the defensin from the common bean Phaseolus vulgaris. Analytical, structural, and functional characterization revealed that both natural and synthetic peptides fold into a canonical CSαβ motif stabilized by conserved disulfide bonds. Moreover, synthetic PvD1 retained the biological activity against four different Candida species and showed no toxicity in vivo. Adding the high resistance of synthetic PvD1 to proteolytic degradation, we claim that conditions are now met to consider PDs druggable biologicals.
Collapse
Affiliation(s)
- Julia Skalska
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Vitor M Andrade
- Laboratório de Bioquı́mica de Peptı́deos, Universidade Federal de São Paulo, 12231-280 São José dos Campos, Brazil
| | - Gabrielle L Cena
- Laboratório de Bioquı́mica de Peptı́deos, Universidade Federal de São Paulo, 12231-280 São José dos Campos, Brazil
| | - Peta J Harvey
- Institute for Molecular Bioscience, The University of Queensland, 4072 Brisbane, Queensland, Australia
| | - Diana Gaspar
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Érica O Mello
- Laboratório de Fisiologia e Bioquı́mica de Microrganismos, Centro de Biociências e Biotecnologı́a, Universidade Estadual do Norte Fluminense Darcy Ribeiro, 28013-602 Rio de Janeiro, Brazil
| | - Sónia T Henriques
- Institute for Molecular Bioscience, The University of Queensland, 4072 Brisbane, Queensland, Australia.,School of Biomedical Sciences, Institute of Health & Biomedical Innovation and Translational Research Institute, Queensland University of Technology, Brisbane, Queensland 4102, Australia
| | - Javier Valle
- Department of Experimental and Health Sciences, Barcelona Biomedical Research Park, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Valdirene M Gomes
- Laboratório de Fisiologia e Bioquı́mica de Microrganismos, Centro de Biociências e Biotecnologı́a, Universidade Estadual do Norte Fluminense Darcy Ribeiro, 28013-602 Rio de Janeiro, Brazil
| | - Katia Conceição
- Laboratório de Bioquı́mica de Peptı́deos, Universidade Federal de São Paulo, 12231-280 São José dos Campos, Brazil
| | - Miguel A R B Castanho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - David Andreu
- Department of Experimental and Health Sciences, Barcelona Biomedical Research Park, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| |
Collapse
|
28
|
Firacative C, Khan A, Duan S, Ferreira-Paim K, Leemon D, Meyer W. Rearing and Maintenance of Galleria mellonella and Its Application to Study Fungal Virulence. J Fungi (Basel) 2020; 6:jof6030130. [PMID: 32784766 PMCID: PMC7558789 DOI: 10.3390/jof6030130] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/31/2020] [Accepted: 08/05/2020] [Indexed: 12/18/2022] Open
Abstract
Galleria mellonella larvae have been widely used as alternative non-mammalian models for the study of fungal virulence and pathogenesis. The larvae can be acquired in small volumes from worm farms, pet stores, or other independent suppliers commonly found in the United States and parts of Europe. However, in countries with no or limited commercial availability, the process of shipping these larvae can cause them stress, resulting in decreased or altered immunity. Furthermore, the conditions used to rear these larvae including diet, humidity, temperature, and maintenance procedures vary among the suppliers. Variation in these factors can affect the response of G. mellonella larvae to infection, thereby decreasing the reproducibility of fungal virulence experiments. There is a critical need for standardized procedures and incubation conditions for rearing G. mellonella to produce quality, unstressed larvae with the least genetic variability. In order to standardize these procedures, cost-effective protocols for the propagation and maintenance of G. mellonella larvae using an artificial diet, which has been successfully used in our own laboratory, requiring minimal equipment and expertise, are herein described. Examples for the application of this model in fungal pathogenicity and gene knockout studies as feasible alternatives for traditionally used animal models are also provided.
Collapse
Affiliation(s)
- Carolina Firacative
- Molecular Mycology Research Laboratory, Centre for Infectious Diseases and Microbiology, Faculty of Medicine and Health, Sydney Medical School, Westmead Clinical School, Marie Bashir Institute for Infectious Diseases and Biosecurity, The University of Sydney, Westmead Hospital (Research and Education Network), Westmead Institute for Medical Research, Westmead 2145, NSW, Australia; (C.F.); (A.K.); (S.D.); (K.F.-P.)
- Studies in Translational Microbiology and Emerging Diseases Research Group (MICROS), School of Medicine and Health Sciences, Universidad del Rosario, Bogota 111221, Colombia
| | - Aziza Khan
- Molecular Mycology Research Laboratory, Centre for Infectious Diseases and Microbiology, Faculty of Medicine and Health, Sydney Medical School, Westmead Clinical School, Marie Bashir Institute for Infectious Diseases and Biosecurity, The University of Sydney, Westmead Hospital (Research and Education Network), Westmead Institute for Medical Research, Westmead 2145, NSW, Australia; (C.F.); (A.K.); (S.D.); (K.F.-P.)
| | - Shuyao Duan
- Molecular Mycology Research Laboratory, Centre for Infectious Diseases and Microbiology, Faculty of Medicine and Health, Sydney Medical School, Westmead Clinical School, Marie Bashir Institute for Infectious Diseases and Biosecurity, The University of Sydney, Westmead Hospital (Research and Education Network), Westmead Institute for Medical Research, Westmead 2145, NSW, Australia; (C.F.); (A.K.); (S.D.); (K.F.-P.)
| | - Kennio Ferreira-Paim
- Molecular Mycology Research Laboratory, Centre for Infectious Diseases and Microbiology, Faculty of Medicine and Health, Sydney Medical School, Westmead Clinical School, Marie Bashir Institute for Infectious Diseases and Biosecurity, The University of Sydney, Westmead Hospital (Research and Education Network), Westmead Institute for Medical Research, Westmead 2145, NSW, Australia; (C.F.); (A.K.); (S.D.); (K.F.-P.)
- Infectious Disease Department, Triangulo Mineiro Federal University, Uberaba 38025-440, Brazil
| | - Diana Leemon
- Agri Science Queensland, Department of Agriculture and Fisheries and Forestry, Brisbane 4102, QLD, Australia;
| | - Wieland Meyer
- Molecular Mycology Research Laboratory, Centre for Infectious Diseases and Microbiology, Faculty of Medicine and Health, Sydney Medical School, Westmead Clinical School, Marie Bashir Institute for Infectious Diseases and Biosecurity, The University of Sydney, Westmead Hospital (Research and Education Network), Westmead Institute for Medical Research, Westmead 2145, NSW, Australia; (C.F.); (A.K.); (S.D.); (K.F.-P.)
- Correspondence: ; Tel.: +61-2-86273430
| |
Collapse
|
29
|
Hoyle A, Cairns D, Paterson I, McMillan S, Ochoa G, Desbois AP. Optimising efficacy of antibiotics against systemic infection by varying dosage quantities and times. PLoS Comput Biol 2020; 16:e1008037. [PMID: 32745111 PMCID: PMC7467302 DOI: 10.1371/journal.pcbi.1008037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 09/02/2020] [Accepted: 06/09/2020] [Indexed: 01/02/2023] Open
Abstract
Mass production and use of antibiotics has led to the rise of resistant bacteria, a problem possibly exacerbated by inappropriate and non-optimal application. Antibiotic treatment often follows fixed-dose regimens, with a standard dose of antibiotic administered equally spaced in time. But are such fixed-dose regimens optimal or can alternative regimens be designed to increase efficacy? Yet, few mathematical models have aimed to identify optimal treatments based on biological data of infections inside a living host. In addition, assumptions to make the mathematical models analytically tractable limit the search space of possible treatment regimens (e.g. to fixed-dose treatments). Here, we aimed to address these limitations by using experiments in a Galleria mellonella (insect) model of bacterial infection to create a fully parametrised mathematical model of a systemic Vibrio infection. We successfully validated this model with biological experiments, including treatments unseen by the mathematical model. Then, by applying artificial intelligence, this model was used to determine optimal antibiotic dosage regimens to treat the host to maximise survival while minimising total antibiotic used. As expected, host survival increased as total quantity of antibiotic applied during the course of treatment increased. However, many of the optimal regimens tended to follow a large initial ‘loading’ dose followed by doses of incremental reductions in antibiotic quantity (dose ‘tapering’). Moreover, application of the entire antibiotic in a single dose at the start of treatment was never optimal, except when the total quantity of antibiotic was very low. Importantly, the range of optimal regimens identified was broad enough to allow the antibiotic prescriber to choose a regimen based on additional criteria or preferences. Our findings demonstrate the utility of an insect host to model antibiotic therapies in vivo and the approach lays a foundation for future regimen optimisation for patient and societal benefits. Research into optimal antibiotic use to improve efficacy is far behind that of cancer care, where personalised treatment is common. The integration of mathematical models with biological observations offers hope to optimise antibiotic use, and in this present study an in vivo insect model of systemic Vibrio infection was used for the first time to determine critical model parameters for optimal antibiotic treatment. By this approach, the optimal regimens tended to result from a large initial ‘loading’ dose followed by subsequent doses of incremental reductions in antibiotic quantity (dose ‘tapering’). The approach and findings of this study opens a new avenue towards optimal application of our precious antibiotic arsenal and may lead to more effective treatment regimens for patients, thus reducing the health and economic burdens associated with bacterial infections. Importantly, it can be argued that until we understand how to use a single antibiotic optimally, it is unlikely we will identify optimal ways to use multiple antibiotics simultaneously.
Collapse
Affiliation(s)
- Andy Hoyle
- Computing Science and Mathematics, University of Stirling, Stirling, United Kingdom
- * E-mail:
| | - David Cairns
- Computing Science and Mathematics, University of Stirling, Stirling, United Kingdom
| | - Iona Paterson
- Computing Science and Mathematics, University of Stirling, Stirling, United Kingdom
| | - Stuart McMillan
- Institute of Aquaculture, University of Stirling, Stirling, United Kingdom
| | - Gabriela Ochoa
- Computing Science and Mathematics, University of Stirling, Stirling, United Kingdom
| | - Andrew P. Desbois
- Institute of Aquaculture, University of Stirling, Stirling, United Kingdom
| |
Collapse
|
30
|
Siriyong T, Ontong JC, Leejae S, Suwalak S, Coote PJ, Voravuthikunchai SP. In vivo safety assessment of rhodomyrtone, a potent compound, from Rhodomyrtus tomentosa leaf extract. Toxicol Rep 2020; 7:919-924. [PMID: 32793420 PMCID: PMC7406972 DOI: 10.1016/j.toxrep.2020.07.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 07/18/2020] [Accepted: 07/21/2020] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Rhodomyrtus tomentosa (Aiton) Hassk. has been traditionally used to relieve various diseases. Rhodomyrtone, a bioactive acylphloroglucinol compound isolated from the leaves of Rhodomyrtus tomentosa, has been scientifically evidenced as a potential antibacterial agent. This study aimed to assess safety of rhodomyrtone in both invertebrate and vertebrate models. MATERIAL AND METHODS Safety of rhodomyrtone was determined in an invertebrate model, Galleria mellonella as well as vertebrate models including zebrafish (Danio rerio) and murine. In addition, toxicity to human erythrocytes was also measured. RESULTS Treatment of Galleria mellonella with rhodomyrtone at 100 mg/kg body weight up to four days showed no visible toxic effects (100 % survival). In zebrafish embryo model, at least 80 % survival of embryos was demonstrated when treated with rhodomyrtone at 0.5 μg/mL for three days. Prior to clinical trial, it is a prerequisite that rhodomyrtone has to be evaluated for its biocompatibility with human blood components. The results displayed that rhodomyrtone at 256 μg/mL did not cause any observable human erythrocyte haemolysis. Furthermore, preclinical assessment of rhodomyrtone formulation justified potential applications of rhodomyrtone in humans. Oral toxicity testing in a mouse model indicated the absence of systemic toxicity when the animals received up to 5000 mg/kg body weight of rhodomyrtone formulation for a period of fourteen days. CONCLUSIONS As the minimal inhibitory concentration of rhodomyrtone against most Gram-positive pathogens is 0.5-1 μg/mL, the results suggest that it should produce no toxic effects at concentrations used in human, thus support further development in pharmaceutical industries and public health applications.
Collapse
Affiliation(s)
- Thanyaluck Siriyong
- Faculty of Traditional Thai Medicine, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
- Natural Product Research Center of Excellence, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | - Julalak Chorachoo Ontong
- Natural Product Research Center of Excellence, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
- Cosmetic Technology and Dietary Supplement Products Program, Faculty of Agro and Bio Industry, Thaksin University, Ban Pa Phayom, Phatthalung, 93210, Thailand
| | - Sukanlaya Leejae
- Excellence Research Laboratory on Natural Products, Department of Microbiology, Faculty of Science, and Natural Product Research Center of Excellence, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | - Sakol Suwalak
- Electron Microscopy Unit, Department of Pathology, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | - Peter John Coote
- Biomedical Sciences Research Complex, School of Biology, University of St Andrews, The North Haugh, St Andrews, Fife, United Kingdom
| | - Supayang Piyawan Voravuthikunchai
- Excellence Research Laboratory on Natural Products, Department of Microbiology, Faculty of Science, and Natural Product Research Center of Excellence, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| |
Collapse
|
31
|
de Sousa AK, Ribeiro FO, de Oliveira TM, de Araújo AR, Dias JDN, Albuquerque P, Silva-Pereira I, de Jesus Oliveira AC, Quelemes PV, Leite JR, da Silva DA. Quaternization of angico gum and evaluation of anti-staphylococcal effect and toxicity of their derivatives. Int J Biol Macromol 2020; 150:1175-1183. [DOI: 10.1016/j.ijbiomac.2019.10.126] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/08/2019] [Accepted: 10/14/2019] [Indexed: 11/26/2022]
|
32
|
Gholizadeh P, Köse Ş, Dao S, Ganbarov K, Tanomand A, Dal T, Aghazadeh M, Ghotaslou R, Ahangarzadeh Rezaee M, Yousefi B, Samadi Kafil H. How CRISPR-Cas System Could Be Used to Combat Antimicrobial Resistance. Infect Drug Resist 2020; 13:1111-1121. [PMID: 32368102 PMCID: PMC7182461 DOI: 10.2147/idr.s247271] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 03/31/2020] [Indexed: 12/26/2022] Open
Abstract
Rapid emergence of antibiotic-resistant bacteria has made it harder for us to combat infectious diseases and to develop new antibiotics. The clustered regularly interspaced short palindromic repeats - CRISPR-associated (CRISPR-Cas) system, as a bacterial adaptive immune system, is recognized as one of the new strategies for controlling antibiotic-resistant strains. The programmable Cas nuclease of this system used against bacterial genomic sequences could be lethal or could help reduce resistance of bacteria to antibiotics. Therefore, this study aims to review using the CRISPR-Cas system to promote sensitizing bacteria to antibiotics. We envision that CRISPR-Cas approaches may open novel ways for the development of smart antibiotics, which could eliminate multidrug-resistant (MDR) pathogens and differentiate between beneficial and pathogenic microorganisms. These systems can be exploited to quantitatively and selectively eliminate individual bacterial strains based on a sequence-specific manner, creating opportunities in the treatment of MDR infections, the study of microbial consortia, and the control of industrial fermentation.
Collapse
Affiliation(s)
- Pourya Gholizadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Şükran Köse
- Department of Infectious Diseases and Clinical Microbiology, University of Health Sciences, Tepecik Training and Research Hospital, İzmir, Turkey
| | - Sounkalo Dao
- Faculté de Médecine, de Pharmacie et d’Odonto-Stomatologie (FMPOS), University of Bamako, Bamako, Mali
| | - Khudaverdi Ganbarov
- Department of Microbiology, Baku State University, Baku, Republic of Azerbaijan
| | - Asghar Tanomand
- Department of Basic Sciences, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Tuba Dal
- Department of Clinical Microbiology, Faculty of Medicine, Ankara Yildirim Beyazit University, Ankara, Turkey
| | - Mohammad Aghazadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Ghotaslou
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Bahman Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Samadi Kafil
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
33
|
Gandra RM, McCarron P, Viganor L, Fernandes MF, Kavanagh K, McCann M, Branquinha MH, Santos ALS, Howe O, Devereux M. In vivo Activity of Copper(II), Manganese(II), and Silver(I) 1,10-Phenanthroline Chelates Against Candida haemulonii Using the Galleria mellonella Model. Front Microbiol 2020; 11:470. [PMID: 32265890 PMCID: PMC7105610 DOI: 10.3389/fmicb.2020.00470] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/04/2020] [Indexed: 12/19/2022] Open
Abstract
Candida haemulonii is an emerging opportunistic pathogen resistant to most antifungal drugs currently used in clinical arena. Metal complexes containing 1,10-phenanthroline (phen) chelating ligands have well-established anti-Candida activity against different medically relevant species. This study utilized larvae of Galleria mellonella, a widely used model of in vivo infection, to examine C. haemulonii infection characteristics in response to different copper(II), manganese(II), and silver(I) chelates containing phen, which had demonstrated potent anti-C. haemulonii activity in a previous study. The results showed that C. haemulonii virulence was influenced by inoculum size and incubation temperature, and the host G. mellonella immune response was triggered in an inoculum-dependent manner reflected by the number of circulating immune cells (hemocytes) and observance of larval melanization process. All test chelates were non-toxic to the host in concentrations up to 10 μg/larva. The complexes also affected the G. mellonella immune system, affecting the hemocyte number and the expression of genes encoding antifungal and immune-related peptides (e.g., inducible metalloproteinase inhibitor protein, transferrin, galiomycin, and gallerimycin). Except for [Ag2(3,6,9-tdda)(phen)4].EtOH (3,6,9-tddaH2 = 3,6,9-trioxoundecanedioic acid), all chelates were capable of affecting the fungal burden of infected larvae and the virulence of C. haemulonii in a dose-dependent manner. This work shows that copper(II), manganese(II), and silver(I) chelates containing phen with anti-C. haemulonii activity are capable of (i) inhibiting fungal proliferation during in vivo infection, (ii) priming an immune response in the G. mellonella host and (iii) affecting C. haemulonii virulence.
Collapse
Affiliation(s)
- Rafael M Gandra
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto de Química, Programa de Pós-Graduação em Bioquímica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Centre for Biomimetic and Therapeutic Research, Focas Research Institute, Technological University Dublin, Dublin, Ireland
| | - Pauraic McCarron
- Centre for Biomimetic and Therapeutic Research, Focas Research Institute, Technological University Dublin, Dublin, Ireland
| | - Livia Viganor
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Centre for Biomimetic and Therapeutic Research, Focas Research Institute, Technological University Dublin, Dublin, Ireland
| | - Mariana Farias Fernandes
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kevin Kavanagh
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Ireland
| | - Malachy McCann
- Department of Chemistry, Maynooth University, National University of Ireland, Maynooth, Ireland
| | - Marta H Branquinha
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - André L S Santos
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes, Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto de Química, Programa de Pós-Graduação em Bioquímica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Orla Howe
- Centre for Biomimetic and Therapeutic Research, Focas Research Institute, Technological University Dublin, Dublin, Ireland.,School of Biological & Health Sciences, Technological University Dublin, Dublin, Ireland
| | - Michael Devereux
- Centre for Biomimetic and Therapeutic Research, Focas Research Institute, Technological University Dublin, Dublin, Ireland
| |
Collapse
|
34
|
Six A, Krajangwong S, Crumlish M, Zadoks RN, Walker D. Galleria mellonella as an infection model for the multi-host pathogen Streptococcus agalactiae reflects hypervirulence of strains associated with human invasive disease. Virulence 2020; 10:600-609. [PMID: 31230520 PMCID: PMC6592362 DOI: 10.1080/21505594.2019.1631660] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Streptococcus agalactiae, or group B Streptococcus (GBS), infects diverse hosts including humans and economically important species such as cattle and fishes. In the context of human health, GBS is a major cause of neonatal infections and an emerging cause of invasive disease in adults and of foodborne disease in Southeast Asia. Here we show that GBS is able to establish a systemic infection in Galleria mellonella larvae that is associated with extensive bacterial replication and dose-dependent larval survival. This infection model is suitable for use with GBS isolates from both homeothermic and poikilothermic hosts. Hypervirulent sequence types (ST) associated with invasive human disease in neonates (ST17) or adults (ST283) show increased virulence in this model, indicating it may be useful in studying GBS virulence determinants, albeit with limitations for some host-specific virulence factors. In addition, we demonstrate that larval survival can be afforded by antibiotic treatment and so the model may also be useful in the development of novel anti-GBS strategies. The use of G. mellonella in GBS research has the potential to provide a low-cost infection model that could reduce the number of vertebrates used in the study of GBS infection.
Collapse
Affiliation(s)
- Anne Six
- a Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences , University of Glasgow , Glasgow , UK
| | - Sakranmanee Krajangwong
- a Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences , University of Glasgow , Glasgow , UK
| | | | - Ruth N Zadoks
- c Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences , University of Glasgow , Glasgow , UK
| | - Daniel Walker
- a Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences , University of Glasgow , Glasgow , UK
| |
Collapse
|
35
|
Raaymakers C, Stijlemans B, Martin C, Zaman S, Ballet S, Martel A, Pasmans F, Roelants K. A New Family of Diverse Skin Peptides from the Microhylid Frog Genus Phrynomantis. Molecules 2020; 25:E912. [PMID: 32085597 PMCID: PMC7070584 DOI: 10.3390/molecules25040912] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/12/2020] [Accepted: 02/18/2020] [Indexed: 01/10/2023] Open
Abstract
A wide range of frogs produce skin poisons composed of bioactive peptides for defence against pathogens, parasites and predators. While several frog families have been thoroughly screened for skin-secreted peptides, others, like the Microhylidae, have remained mostly unexplored. Previous studies of microhylids found no evidence of peptide secretion, suggesting that this defence adaptation was evolutionarily lost. We conducted transcriptome analyses of the skins of Phrynomantis bifasciatus and Phrynomantis microps, two African microhylid species long suspected to be poisonous. Our analyses reveal 17 evolutionary related transcripts that diversified from to those of cytolytic peptides found in other frog families. The 19 peptides predicted to be processed from these transcripts, named phrynomantins, show a striking structural diversity that is distinct from any previously identified frog skin peptide. Functional analyses of five phrynomantins confirm the loss of a cytolytic function and the absence of insecticidal or proinflammatory activity, suggesting that they represent an evolutionary transition to a new, yet unknown function. Our study shows that peptides have been retained in the defence poison of at least one microhylid lineage and encourages research on similarly understudied taxa to further elucidate the diversity and evolution of skin defence molecules.
Collapse
Affiliation(s)
- Constantijn Raaymakers
- Amphibian Evolution Lab, Biology Department, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Elsene, Belgium; (C.R.); (S.Z.)
- Wildlife Health Ghent, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium; (A.M.); (F.P.)
| | - Benoit Stijlemans
- Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Elsene, Belgium;
- Myeloid Cell Immunology Lab, VIB Centre for Inflammation Research, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Elsene, Belgium
| | - Charlotte Martin
- Research Group of Organic Chemistry, Department of Chemistry and Department of Bio-engineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Elsene, Belgium; (C.M.); (S.B.)
| | - Shabnam Zaman
- Amphibian Evolution Lab, Biology Department, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Elsene, Belgium; (C.R.); (S.Z.)
| | - Steven Ballet
- Research Group of Organic Chemistry, Department of Chemistry and Department of Bio-engineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Elsene, Belgium; (C.M.); (S.B.)
| | - An Martel
- Wildlife Health Ghent, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium; (A.M.); (F.P.)
| | - Frank Pasmans
- Wildlife Health Ghent, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium; (A.M.); (F.P.)
| | - Kim Roelants
- Amphibian Evolution Lab, Biology Department, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Elsene, Belgium; (C.R.); (S.Z.)
| |
Collapse
|
36
|
Vellanki S, Billmyre RB, Lorenzen A, Campbell M, Turner B, Huh EY, Heitman J, Lee SC. A Novel Resistance Pathway for Calcineurin Inhibitors in the Human-Pathogenic Mucorales Mucor circinelloides. mBio 2020; 11:e02949-19. [PMID: 31992620 PMCID: PMC6989107 DOI: 10.1128/mbio.02949-19] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 12/16/2019] [Indexed: 12/24/2022] Open
Abstract
Mucormycosis is an emerging lethal fungal infection in immunocompromised patients. Mucor circinelloides is a causal agent of mucormycosis and serves as a model system to understand genetics in Mucorales. Calcineurin is a conserved virulence factor in many pathogenic fungi, and calcineurin inhibition or deletion of the calcineurin regulatory subunit (CnbR) in Mucor results in a shift from hyphal to yeast growth. We analyzed 36 calcineurin inhibitor-resistant or bypass mutants that exhibited hyphal growth in the presence of calcineurin inhibitors or in the yeast-locked cnbRΔ mutant background without carrying any mutations in known calcineurin components. We found that a majority of the mutants had altered sequence in a gene, named here bycA (bypass of calcineurin). bycA encodes an amino acid permease. We verified that both the bycAΔ single mutant and the bycAΔ cnbRΔ double mutant are resistant to calcineurin inhibitor FK506, thereby demonstrating a novel mechanism of resistance against calcineurin inhibitors. We also found that the level of expression of bycA was significantly higher in the wild-type strain treated with FK506 and in the cnbRΔ mutants but was significantly lower in the wild-type strain without FK506 treatment. These findings suggest that bycA is a negative regulator of hyphal growth and/or a positive regulator of yeast growth in Mucor and that calcineurin suppresses expression of the bycA gene at the mRNA level to promote hyphal growth. BycA is involved in the Mucor hypha-yeast transition as our data demonstrate positive correlations among bycA expression, protein kinase A activity, and Mucor yeast growth. Also, calcineurin, independently of its role in morphogenesis, contributes to virulence traits, including phagosome maturation blockade, host cell damages, and proangiogenic growth factor induction during interactions with hosts.IMPORTANCEMucor is intrinsically resistant to most known antifungals, which makes mucormycosis treatment challenging. Calcineurin is a serine/threonine phosphatase that is widely conserved across eukaryotes. When calcineurin function is inhibited in Mucor, growth shifts to a less virulent yeast growth form, which makes calcineurin an attractive target for development of new antifungal drugs. Previously, we identified two distinct mechanisms through which Mucor can become resistant to calcineurin inhibitors involving Mendelian mutations in the gene for FKBP12, including mechanisms corresponding to calcineurin A or B subunits and epimutations silencing the FKBP12 gene. Here, we identified a third novel mechanism where loss-of-function mutations in the amino acid permease corresponding to the bycA gene contribute to resistance against calcineurin inhibitors. When calcineurin activity is absent, BycA can activate protein kinase A (PKA) to promote yeast growth via a cAMP-independent pathway. Our data also show that calcineurin activity contributes to host-pathogen interactions primarily in the pathogenesis of Mucor.
Collapse
Affiliation(s)
- Sandeep Vellanki
- South Texas Center for Emerging Infectious Diseases (STCEID), Department of Biology, The University of Texas at San Antonio, San Antonio, Texas, USA
| | - R Blake Billmyre
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Alejandra Lorenzen
- South Texas Center for Emerging Infectious Diseases (STCEID), Department of Biology, The University of Texas at San Antonio, San Antonio, Texas, USA
| | - Micaela Campbell
- South Texas Center for Emerging Infectious Diseases (STCEID), Department of Biology, The University of Texas at San Antonio, San Antonio, Texas, USA
| | - Broderick Turner
- South Texas Center for Emerging Infectious Diseases (STCEID), Department of Biology, The University of Texas at San Antonio, San Antonio, Texas, USA
| | - Eun Young Huh
- South Texas Center for Emerging Infectious Diseases (STCEID), Department of Biology, The University of Texas at San Antonio, San Antonio, Texas, USA
| | - Joseph Heitman
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Soo Chan Lee
- South Texas Center for Emerging Infectious Diseases (STCEID), Department of Biology, The University of Texas at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
37
|
Larva of greater wax moth Galleria mellonella is a suitable alternative host for the fish pathogen Francisella noatunensis subsp. orientalis. BMC Microbiol 2020; 20:8. [PMID: 31918661 PMCID: PMC6953311 DOI: 10.1186/s12866-020-1695-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 12/31/2019] [Indexed: 12/26/2022] Open
Abstract
Background Francisella noatunensis subsp. orientalis (Fno) is the etiological agent of francisellosis in cultured warm water fish, such as tilapia. Antibiotics are administered to treat the disease but a better understanding of Fno infection biology will inform improved treatment and prevention measures. However, studies with native hosts are costly and considerable benefits would derive from access to a practical alternative host. Here, larvae of Galleria mellonella were assessed for suitability to study Fno virulence. Results Larvae were killed by Fno in a dose-dependent manner but the insects could be rescued from lethal doses of bacteria by antibiotic therapy. Infection progression was assessed by histopathology (haematoxylin and eosin staining, Gram Twort and immunohistochemistry) and enumeration of bacteria recovered from the larval haemolymph on selective agar. Fno was phagocytosed and could survive intracellularly, which is consistent with observations in fish. Virulence of five Fno isolates showed strong agreement between G. mellonella and red Nile tilapia hosts. Conclusions This study shows that an alternative host, G. mellonella, can be applied to understand Fno infections, which will assist efforts to identify solutions to piscine francisellosis thus securing the livelihoods of tilapia farmers worldwide and ensuring the production of this important food source.
Collapse
|
38
|
Rodrigues de Araújo A, Iles B, de Melo Nogueira K, Dias JDN, Plácido A, Rodrigues A, Albuquerque P, Silva-Pereira I, Socodatto R, Portugal CC, Relvas JB, Costa Véras LM, Dalmatti Alves Lima FC, Batagin-Neto A, Rolim Medeiros JV, Moreira Nunes PH, Eaton P, de Souza de Almeida Leite JR. Antifungal and anti-inflammatory potential of eschweilenol C-rich fraction derived from Terminalia fagifolia Mart. JOURNAL OF ETHNOPHARMACOLOGY 2019; 240:111941. [PMID: 31100435 DOI: 10.1016/j.jep.2019.111941] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/05/2019] [Accepted: 05/06/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Folk knowledge transmitted between generations allows traditional populations to maintain the use of medicinal plants for the treatment of several diseases. In this context, the species Terminalia fagifolia Mart., native to Brazil, is used for the treatment of chronic and infectious diseases. Plants rich in secondary metabolites, such as this species and their derivatives, may represent therapeutic alternatives for the treatment of diseases that reduce the quality of life of people. AIM OF THE STUDY The aim of this study was to evaluate the antifungal and anti-inflammatory potential of aqueous fraction from ethanolic extract of T. fagifolia, with in silico study of the major compound of the fraction. MATERIAL AND METHODS The phytochemical study of the aqueous fraction was performed by HPLC, LC/MS and NMR. The antifungal activity was evaluated against yeasts, by determination of the minimum inhibitory concentration and minimum fungicidal concentration. The effect on Candida albicans was analyzed by AFM. The antibiofilm potential against biofilms of C. albicans was also tested. The anti-inflammatory potential of the aqueous fraction was evaluated in vivo by the carrageenan-induced paw edema and peritonitis. A microglial model of LPS-induced neuroinflammation was also studied. Further insights on the activation mechanism were studied using quantum chemistry computer simulations. Toxicity was evaluated in the Galleria mellonella and human erythrocytes models. RESULTS Eschweilenol C was identified as the major constituent of the aqueous fraction of the ethanolic extract of T. fagifolia. The aqueous fraction was active against all Candida strains used (sensitive and resistant to Fluconazole) with MICs ranging from 1000 to 0.4 μg/mL. By AFM it was possible to observe morphological alterations in treated Candida cells. The fraction significantly (p < 0.05) inhibited paw edema and decreased levels of malondialdehyde induced by carrageenan. In a microglial cell model, aqueous fraction demonstrated the ability to inhibit NF-κB after induction with lipopolysaccharide. The theoretical studies showed structural similarity between eschweilenol C and indomethacin and an excellent antioxidant potential. The aqueous fraction did not present toxicity in the studied models. CONCLUSION The results indicate that the aqueous fraction of T. fagifolia has potential for biomedical applications with low toxicity. This finding can be attributed to the predominance of eschweilenol C in the aqueous fraction.
Collapse
Affiliation(s)
| | - Bruno Iles
- Biotechnology and Biodiversity Center Research, Biotec, Federal University of Piaui, Parnaíba, Piaui, Brazil
| | - Kerolayne de Melo Nogueira
- Biotechnology and Biodiversity Center Research, Biotec, Federal University of Piaui, Parnaíba, Piaui, Brazil
| | - Jhones do Nascimento Dias
- Laboratory of Molecular Biology of Dimorphic and Pathogenic Fungi, Institute of Biological Sciences, University of Brasilia, Brasilia, Brazil
| | - Alexandra Plácido
- Glial Cell Biology Laboratory, Institute for Research and Innovation in Health, i3S, University of Porto, Porto, Portugal; Bioprospectum, Lda, UPTEC, Porto, Portugal
| | - Artur Rodrigues
- Glial Cell Biology Laboratory, Institute for Research and Innovation in Health, i3S, University of Porto, Porto, Portugal
| | - Patrícia Albuquerque
- Laboratory of Molecular Biology of Dimorphic and Pathogenic Fungi, Institute of Biological Sciences, University of Brasilia, Brasilia, Brazil
| | - Ildinete Silva-Pereira
- Laboratory of Molecular Biology of Dimorphic and Pathogenic Fungi, Institute of Biological Sciences, University of Brasilia, Brasilia, Brazil
| | - Renato Socodatto
- Glial Cell Biology Laboratory, Institute for Research and Innovation in Health, i3S, University of Porto, Porto, Portugal
| | - Camila C Portugal
- Glial Cell Biology Laboratory, Institute for Research and Innovation in Health, i3S, University of Porto, Porto, Portugal
| | - João B Relvas
- Glial Cell Biology Laboratory, Institute for Research and Innovation in Health, i3S, University of Porto, Porto, Portugal
| | - Leiz Maria Costa Véras
- Biotechnology and Biodiversity Center Research, Biotec, Federal University of Piaui, Parnaíba, Piaui, Brazil
| | | | | | | | | | - Peter Eaton
- LAQV/REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences of the University of Porto, Porto, Portugal
| | - José Roberto de Souza de Almeida Leite
- The Northeast Biotechnology Network, RENORBIO, Federal University of Piaui, Teresina, PI, Brazil; Center for Research in Applied Morphology and Immunology, NuPMIA, University of Brasilia, Brasilia, Brazil.
| |
Collapse
|
39
|
Efficacy of oritavancin alone and in combination against vancomycin-susceptible and -resistant enterococci in an in-vivo Galleria mellonella survival model. Int J Antimicrob Agents 2019; 54:197-201. [DOI: 10.1016/j.ijantimicag.2019.04.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/03/2019] [Accepted: 04/17/2019] [Indexed: 11/19/2022]
|
40
|
Tharmalingam N, Khader R, Fuchs BB, Mylonakis E. The Anti-virulence Efficacy of 4-(1,3-Dimethyl-2,3-Dihydro-1H-Benzimidazol-2-yl)Phenol Against Methicillin-Resistant Staphylococcus aureus. Front Microbiol 2019; 10:1557. [PMID: 31379761 PMCID: PMC6653400 DOI: 10.3389/fmicb.2019.01557] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 06/21/2019] [Indexed: 12/24/2022] Open
Abstract
Antimicrobial drug discovery against drug-resistant bacteria is an urgent need. Beyond agents with direct antibacterial activity, anti-virulent molecules may also be viable compounds to defend against bacterial pathogenesis. Using a high throughput screen (HTS) that utilized Caenorhabditis elegans infected with methicillin-resistant Staphylococcus aureus (MRSA) strain of MW2, we identified 4-(1,3-dimethyl-2,3-dihydro-1H-benzimidazol-2-yl)phenol (BIP). Interestingly, BIP had no in vitro inhibition activity against MW2, at least up to 64 μg/ml. The lack of direct antimicrobial activity suggests that BIP could inhibit bacterial virulence factors. To explore the possible anti-virulence effect of the identified molecule, we first performed real-time PCR to examine changes in virulence expression. BIP was highly active against MRSA virulence factors at sub-lethal concentrations and down-regulated virulence regulator genes, such as agrA and codY. However, the benzimidazole derivatives omeprazole and pantoprazole did not down-regulate virulence genes significantly, compared to BIP. Moreover, the BIP-pretreated MW2 cells were more vulnerable to macrophage-mediated killing, as confirmed by intracellular killing and live/dead staining assays, and less efficient in establishing a lethal infection in the invertebrate host Galleria mellonella (p = 0.0131). We tested the cytotoxicity of BIP against human red blood cells (RBCs), and it did not cause hemolysis at the highest concentration tested (64 μg/ml). Taken together, our findings outline the potential anti-virulence activity of BIP that was identified through a C. elegans-based, whole animal based, screen.
Collapse
Affiliation(s)
| | | | | | - Eleftherios Mylonakis
- Department of Medicine, Division of Infectious Diseases, Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| |
Collapse
|
41
|
Zhao X, Palma Medina LM, Stobernack T, Glasner C, de Jong A, Utari P, Setroikromo R, Quax WJ, Otto A, Becher D, Buist G, van Dijl JM. Exoproteome Heterogeneity among Closely Related Staphylococcus aureus t437 Isolates and Possible Implications for Virulence. J Proteome Res 2019; 18:2859-2874. [PMID: 31119940 PMCID: PMC6617432 DOI: 10.1021/acs.jproteome.9b00179] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Staphylococcus aureus with spa-type t437 has been identified as a predominant community-associated methicillin-resistant S. aureus clone from Asia, which is also encountered in Europe. Molecular typing has previously shown that t437 isolates are highly similar regardless of geographical regions or host environments. The present study was aimed at assessing to what extent this high similarity is actually reflected in the production of secreted virulence factors. We therefore profiled the extracellular proteome, representing the main reservoir of virulence factors, of 20 representative clinical isolates by mass spectrometry. The results show that these isolates can be divided into three groups and nine subgroups based on exoproteome abundance signatures. This implies that S. aureus t437 isolates show substantial exoproteome heterogeneity. Nonetheless, 30 highly conserved extracellular proteins, of which about 50% have a predicted role in pathogenesis, were dominantly identified. To approximate the virulence of the 20 investigated isolates, we employed infection models based on Galleria mellonella and HeLa cells. The results show that the grouping of clinical isolates based on their exoproteome profile can be related to virulence. We consider this outcome important as our approach provides a tool to pinpoint differences in virulence among seemingly highly similar clinical isolates of S. aureus.
Collapse
Affiliation(s)
- Xin Zhao
- University of Groningen , University Medical Center Groningen, Department of Medical Microbiology , Hanzeplein 1 , P.O. Box 30001, 9700 RB Groningen , The Netherlands
| | - Laura M Palma Medina
- University of Groningen , University Medical Center Groningen, Department of Medical Microbiology , Hanzeplein 1 , P.O. Box 30001, 9700 RB Groningen , The Netherlands
| | - Tim Stobernack
- University of Groningen , University Medical Center Groningen, Department of Medical Microbiology , Hanzeplein 1 , P.O. Box 30001, 9700 RB Groningen , The Netherlands
| | - Corinna Glasner
- University of Groningen , University Medical Center Groningen, Department of Medical Microbiology , Hanzeplein 1 , P.O. Box 30001, 9700 RB Groningen , The Netherlands
| | - Anne de Jong
- University of Groningen , Groningen Biomolecular Sciences and Biotechnology Institute, Department of Molecular Genetics , 9747 AG Groningen , The Netherlands
| | - Putri Utari
- University of Groningen , Groningen Research Institute of Pharmacy, Department of Chemical and Pharmaceutical Biology , A. Deusinglaan 1 , 9713 AV Groningen , The Netherlands
| | - Rita Setroikromo
- University of Groningen , Groningen Research Institute of Pharmacy, Department of Chemical and Pharmaceutical Biology , A. Deusinglaan 1 , 9713 AV Groningen , The Netherlands
| | - Wim J Quax
- University of Groningen , Groningen Research Institute of Pharmacy, Department of Chemical and Pharmaceutical Biology , A. Deusinglaan 1 , 9713 AV Groningen , The Netherlands
| | - Andreas Otto
- Institut für Mikrobiologie , University of Greifswald , Felix-Hausdorff-Str. 8 , 17475 Greifswald , Germany
| | - Dörte Becher
- Institut für Mikrobiologie , University of Greifswald , Felix-Hausdorff-Str. 8 , 17475 Greifswald , Germany
| | - Girbe Buist
- University of Groningen , University Medical Center Groningen, Department of Medical Microbiology , Hanzeplein 1 , P.O. Box 30001, 9700 RB Groningen , The Netherlands
| | - Jan Maarten van Dijl
- University of Groningen , University Medical Center Groningen, Department of Medical Microbiology , Hanzeplein 1 , P.O. Box 30001, 9700 RB Groningen , The Netherlands
| |
Collapse
|
42
|
Safety assessment of Gram-negative bacteria associated with traditional French cheeses. Food Microbiol 2019; 79:1-10. [DOI: 10.1016/j.fm.2018.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 09/25/2018] [Accepted: 11/03/2018] [Indexed: 12/21/2022]
|
43
|
Gorr SU, Flory CM, Schumacher RJ. In vivo activity and low toxicity of the second-generation antimicrobial peptide DGL13K. PLoS One 2019; 14:e0216669. [PMID: 31071184 PMCID: PMC6508730 DOI: 10.1371/journal.pone.0216669] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 04/26/2019] [Indexed: 11/18/2022] Open
Abstract
Antimicrobial peptides have been evaluated as possible alternatives to traditional antibiotics. The translational potential of the antimicrobial peptide DGL13K was tested with focus on peptide toxicity and in vivo activity in two animal models. DGL13K was effective against Pseudomonas aeruginosa, Staphylococcus aureus and methicillin-resistant S. aureus with minimal bactericidal concentrations similar to the minimal inhibitory concentration. The peptide showed low toxicity to human red blood cells and HEK cells with median lethal dose around 1 mg/ml. The median lethal dose in greater wax moth larvae (Galleria mellonella) was about 125mg/kg while the peptide caused no skin toxicity in a mouse model. A novel high-throughput luminescence assay was used to test peptide activity in infected G. mellonella, thus reducing vertebrate animal use. DGL13K killed P. aeruginosa in both the G. mellonella model and a mouse burn wound infection model, with bacterial viability 3-10-fold lower than in untreated controls. Future experiments will focus on optimizing peptide delivery, dose and frequency to further improve the antibacterial effect.
Collapse
Affiliation(s)
- Sven-Ulrik Gorr
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, Minnesota, United States of America
- * E-mail:
| | - Craig M. Flory
- Center for Translational Medicine, University of Minnesota Academic Health Center, Minneapolis, Minnesota, United States of America
| | - Robert J. Schumacher
- Center for Translational Medicine, University of Minnesota Academic Health Center, Minneapolis, Minnesota, United States of America
| |
Collapse
|
44
|
New Bacteriophages against Emerging Lineages ST23 and ST258 of Klebsiella pneumoniae and Efficacy Assessment in Galleria mellonella Larvae. Viruses 2019; 11:v11050411. [PMID: 31058805 PMCID: PMC6563190 DOI: 10.3390/v11050411] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/26/2019] [Accepted: 04/29/2019] [Indexed: 12/16/2022] Open
Abstract
Klebsiella pneumoniae is a bacterial pathogen of high public health importance. Its polysaccharide capsule is highly variable but only a few capsular types are associated with emerging pathogenic sublineages. The aim of this work is to isolate and characterize new lytic bacteriophages and assess their potential to control infections by the ST23 and ST258 K. pneumoniae sublineages using a Galleria mellonella larvae model. Three selected bacteriophages, targeting lineages ST258 (bacteriophages vB_KpnP_KL106-ULIP47 and vB_KpnP_KL106-ULIP54) and ST23 (bacteriophage vB_KpnP_K1-ULIP33), display specificity for capsular types KL106 and K1, respectively. These podoviruses belong to the Autographivirinae subfamily and their genomes are devoid of lysogeny or toxin-associated genes. In a G. mellonella larvae model, a mortality rate of 70% was observed upon infection by K. pneumoniae ST258 and ST23. This number was reduced to 20% upon treatment with bacteriophages at a multiplicity of infection of 10. This work increases the number of characterized bacteriophages infecting K. pneumoniae and provides information regarding genome sequence and efficacy during preclinical phage therapy against two prominent sublineages of this bacterial species.
Collapse
|
45
|
Frawley KL, Praekunatham H, Cronican AA, Peterson J, Pearce LL. Assessing modulators of cytochrome c oxidase activity in Galleria mellonella larvae. Comp Biochem Physiol C Toxicol Pharmacol 2019; 219:77-86. [PMID: 30802621 DOI: 10.1016/j.cbpc.2019.02.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/18/2019] [Accepted: 02/20/2019] [Indexed: 12/16/2022]
Abstract
Caterpillars of the greater wax moth, Galleria mellonella, are shown to be a useful invertebrate organism for examining mitochondrial toxicants (inhibitors of electron transport) and testing putative antidotes. Administration of sodium azide, sodium cyanide, or sodium (hydro)sulfide by intra-haemocoel injection (through a proleg) results in a dose-dependent paralysed state in the larvae lasting from <1 to ~40 min. The duration of paralysis is easily monitored, because if turned onto their backs, the larvae right themselves onto their prolegs once they are able to move again. The efficacy of putative antidotes to the three toxicants can routinely be assessed by observing shortened periods of paralysis with larvae given toxicant and antidote compared to larvae administered only the same dose of toxicant. The validity of the approach is demonstrated with agents previously shown to be antidotal towards cyanide intoxication in mice; namely, sodium nitrite and CoN4[11.3.1] (cobalt(II/III) 2,12-dimethyl-3,7,11,17-tetraazabicyclo-[11.3.1]-heptadeca-1(7)2,11,13,15-pentaenyl cation). These same compounds are shown to be antidotal towards all three toxicants in the G. mellonella caterpillars; findings that may prove important in relation to azide and sulfide poisonings, for which there are currently no effective antidotes available. The observation that sodium nitrite ameliorates cyanide toxicity in the larvae is additionally interesting because it unambiguously demonstrates that the antidotal action of nitrites does not require the involvement of methemoglobin, contributing to the resolution of an ongoing controversy.
Collapse
Affiliation(s)
- Kristin L Frawley
- Department of Environmental and Occupational Health, University of Pittsburgh Graduate School of Public Health, 130 DeSoto Street, Pittsburgh, PA 15219, USA
| | - Hirunwut Praekunatham
- Department of Environmental and Occupational Health, University of Pittsburgh Graduate School of Public Health, 130 DeSoto Street, Pittsburgh, PA 15219, USA
| | - Andrea A Cronican
- Department of Environmental and Occupational Health, University of Pittsburgh Graduate School of Public Health, 130 DeSoto Street, Pittsburgh, PA 15219, USA
| | - Jim Peterson
- Department of Environmental and Occupational Health, University of Pittsburgh Graduate School of Public Health, 130 DeSoto Street, Pittsburgh, PA 15219, USA.
| | - Linda L Pearce
- Department of Environmental and Occupational Health, University of Pittsburgh Graduate School of Public Health, 130 DeSoto Street, Pittsburgh, PA 15219, USA.
| |
Collapse
|
46
|
Brust FR, Boff L, da Silva Trentin D, Pedrotti Rozales F, Barth AL, Macedo AJ. Macrocolony of NDM-1 Producing Enterobacter hormaechei subsp. oharae Generates Subpopulations with Different Features Regarding the Response of Antimicrobial Agents and Biofilm Formation. Pathogens 2019; 8:pathogens8020049. [PMID: 31014001 PMCID: PMC6631906 DOI: 10.3390/pathogens8020049] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 11/16/2022] Open
Abstract
Enterobacter cloacae complex has been increasingly recognized as a nosocomial pathogen representing the third major Enterobacteriaceae species involved with infections. This study aims to evaluate virulence and antimicrobial susceptibility of subpopulations generated from macrocolonies of NDM-1 producing Enterobacter hormaechei clinical isolates. Biofilm was quantified using crystal violet method and fimbrial genes were investigated by PCR. Susceptibility of antimicrobials, alone and combined, was determined by minimum inhibitory concentration and checkerboard assays, respectively. Virulence and efficacy of antimicrobials were evaluated in Galleria mellonella larvae. Importantly, we verified that some subpopulations that originate from the same macrocolony present different biofilm production ability and distinct susceptibility to meropenem due to the loss of blaNDM-1 encoding plasmid. A more in-depth study was performed with the 798 macrocolony subpopulations. Type 3 fimbriae were straightly related with biofilm production; however, virulence in larvae was not statistically different among subpopulations. Triple combination with meropenem-rifampicin-polymyxin B showed in vitro synergistic effect against all subpopulations; while in vivo this treatment showed different efficacy rates for 798-1S and 798-4S subpopulations. The ability of multidrug resistant E. hormaechei isolates in generating bacterial subpopulations presenting different susceptible and virulence mechanisms are worrisome and may explain why these infections are hardly overcome.
Collapse
Affiliation(s)
- Flávia Roberta Brust
- Faculty of Pharmacy and Center of Biotechnology, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul CE 90610-000, Brazil.
| | - Luana Boff
- Faculty of Pharmacy and Center of Biotechnology, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul CE 90610-000, Brazil.
| | - Danielle da Silva Trentin
- Basic Health Sciences Department, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Rio Grande do Sul CE 90050-170, Brazil.
| | - Franciele Pedrotti Rozales
- Laboratory of Research in Bacterial Resistance, Center for Experimental Research, Clinical Hospital of Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul CE 90035-007, Brazil.
| | - Afonso Luís Barth
- Laboratory of Research in Bacterial Resistance, Center for Experimental Research, Clinical Hospital of Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul CE 90035-007, Brazil.
| | - Alexandre José Macedo
- Faculty of Pharmacy and Center of Biotechnology, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul CE 90610-000, Brazil.
| |
Collapse
|
47
|
Liu Y, Leung SSY, Guo Y, Zhao L, Jiang N, Mi L, Li P, Wang C, Qin Y, Mi Z, Bai C, Gao Z. The Capsule Depolymerase Dpo48 Rescues Galleria mellonella and Mice From Acinetobacter baumannii Systemic Infections. Front Microbiol 2019; 10:545. [PMID: 30936862 PMCID: PMC6431613 DOI: 10.3389/fmicb.2019.00545] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 03/01/2019] [Indexed: 01/12/2023] Open
Abstract
The emergence of multidrug- and extensively drug-resistant Acinetobacter baumannii has made it difficult to treat and control infections caused by this bacterium. Thus, alternatives to conventional antibiotics for management of severe A. baumannii infections is urgently needed. In our previous study, we found that a capsule depolymerase Dpo48 could strip bacterial capsules, and the non-capsuled A. baumannii were significantly decreased in the presence of serum complement in vitro. Here, we further explored its potential as a therapeutic agent for controlling systemic infections caused by extensively drug-resistant A. baumannii. Prior to mammalian studies, the anti-virulence efficacy of Dpo48 was first tested in a Galleria mellonella infection model. Survival rate of Dpo48-pretreated bacteria or Dpo48 treatment group was significantly increased compared to the infective G. mellonella without treatment. Furthermore, the safety and therapeutic efficacy of Dpo48 to mice were evaluated. The mice treated with Dpo48 displayed normal serum levels of TBIL, AST, ALT, ALP, Cr, BUN and LDH, while no significant histopathology changes were observed in tissues of liver, spleen, lung, and kidney. Treatment with Dpo48 could rescue normal and immunocompromised mice from lethal peritoneal sepsis, with the bacterial counts in blood, liver, spleen, lung, and kidney significantly reduced by 1.4-3.3 log colony-forming units at 4 h posttreatment. Besides, the hemolysis and cytotoxicity assays showed that Dpo48 was non-homolytic to human red blood cells and non-toxic to human lung, liver and kidney cell lines. Overall, the present study demonstrated the promising potential of capsule depolymerases as therapeutic agents to prevent antibiotic-resistant A. baumannii infections.
Collapse
Affiliation(s)
- Yannan Liu
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China.,Department of Respiratory and Critical Care Medicine, 307th Hospital of Chinese People's Liberation Army, Beijing, China
| | | | - Yatao Guo
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Lili Zhao
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Ning Jiang
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Liyuan Mi
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Puyuan Li
- Department of Respiratory and Critical Care Medicine, 307th Hospital of Chinese People's Liberation Army, Beijing, China
| | - Can Wang
- Department of Respiratory and Critical Care Medicine, 307th Hospital of Chinese People's Liberation Army, Beijing, China
| | - Yanhong Qin
- Department of Respiratory and Critical Care Medicine, 307th Hospital of Chinese People's Liberation Army, Beijing, China
| | - Zhiqiang Mi
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Changqing Bai
- Department of Respiratory and Critical Care Medicine, 307th Hospital of Chinese People's Liberation Army, Beijing, China
| | - Zhancheng Gao
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| |
Collapse
|
48
|
Implications of the EUCAST Trailing Phenomenon in Candida tropicalis for the In Vivo Susceptibility in Invertebrate and Murine Models. Antimicrob Agents Chemother 2018; 62:AAC.01624-18. [PMID: 30224538 DOI: 10.1128/aac.01624-18] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 09/05/2018] [Indexed: 12/31/2022] Open
Abstract
Candida tropicalis isolates often display reduced but persistent growth (trailing) over a broad fluconazole concentration range during EUCAST susceptibility testing. Whereas weak trailing (<25% of the positive growth control) is common and found not to impair fluconazole efficacy, we investigated if more pronounced trailing impacted treatment efficacy. Fluconazole efficacy against two weakly (≤25% growth), two moderately (26% to 50% growth), and one heavily (>70% growth) trailing resistant isolate and one resistant (100% growth) isolate were investigated in vitro and in vivo (in a Galleria mellonella survival model and two nonlethal murine models). CDR1 expression levels and ERG11 sequences were characterized. The survival in fluconazole-treated G. mellonella was inversely correlated with the degree of trailing (71% to 9% survival in treatment groups). In mice, resistant and heavily trailing isolates responded poorly to fluconazole treatment. CDR1 expression was significantly higher in trailing and resistant isolates than in wild-type isolates (1.4-fold to 10-fold higher). All isolates exhibited ERG11 wild-type alleles. Heavily trailing isolates were less responsive to fluconazole in all in vivo models, indicating an impact on fluconazole efficacy. CDR1 upregulation may have contributed to the observed differences. Moderately trailing isolates responded less well to fluconazole in larvae only. This confirms clinical data suggesting fluconazole is effective against infections with such isolates in less severely ill patients and supports the current 50% growth endpoint for susceptibility testing. However, it is still unclear if the gradual loss of efficacy observed for moderately trailing isolates in the larva model may be a reason for concern in selected vulnerable patient populations.
Collapse
|
49
|
Li Y, Spiropoulos J, Cooley W, Khara JS, Gladstone CA, Asai M, Bossé JT, Robertson BD, Newton SM, Langford PR. Galleria mellonella - a novel infection model for the Mycobacterium tuberculosis complex. Virulence 2018; 9:1126-1137. [PMID: 30067135 PMCID: PMC6086298 DOI: 10.1080/21505594.2018.1491255] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Animal models have long been used in tuberculosis research to understand disease pathogenesis and to evaluate novel vaccine candidates and anti-mycobacterial drugs. However, all have limitations and there is no single animal model which mimics all the aspects of mycobacterial pathogenesis seen in humans. Importantly mice, the most commonly used model, do not normally form granulomas, the hallmark of tuberculosis infection. Thus there is an urgent need for the development of new alternative in vivo models. The insect larvae, Galleria mellonella has been increasingly used as a successful, simple, widely available and cost-effective model to study microbial infections. Here we report for the first time that G. mellonella can be used as an infection model for members of the Mycobacterium tuberculosis complex. We demonstrate a dose-response for G. mellonella survival infected with different inocula of bioluminescent Mycobacterium bovis BCG lux, and demonstrate suppression of mycobacterial luminesence over 14 days. Histopathology staining and transmission electron microscopy of infected G. mellonella phagocytic haemocytes show internalization and aggregation of M. bovis BCG lux in granuloma-like structures, and increasing accumulation of lipid bodies within M. bovis BCG lux over time, characteristic of latent tuberculosis infection. Our results demonstrate that G. mellonella can act as a surrogate host to study the pathogenesis of mycobacterial infection and shed light on host-mycobacteria interactions, including latent tuberculosis infection.
Collapse
Affiliation(s)
- Yanwen Li
- a Section of Paediatric Infectious Diseases and Allergy, Department of Medicine , Imperial College London , London , UK
| | - John Spiropoulos
- b Department of Pathology , Animal and Plant Health Agency , Addlestone , UK
| | - William Cooley
- b Department of Pathology , Animal and Plant Health Agency , Addlestone , UK
| | - Jasmeet Singh Khara
- a Section of Paediatric Infectious Diseases and Allergy, Department of Medicine , Imperial College London , London , UK.,c Department of Pharmacy , National University of Singapore , Singapore
| | - Camilla A Gladstone
- a Section of Paediatric Infectious Diseases and Allergy, Department of Medicine , Imperial College London , London , UK
| | - Masanori Asai
- a Section of Paediatric Infectious Diseases and Allergy, Department of Medicine , Imperial College London , London , UK
| | - Janine T Bossé
- a Section of Paediatric Infectious Diseases and Allergy, Department of Medicine , Imperial College London , London , UK
| | - Brian D Robertson
- d MRC Centre for Molecular Bacteriology and Infection, Department of Medicine , Imperial College London , London , UK
| | - Sandra M Newton
- a Section of Paediatric Infectious Diseases and Allergy, Department of Medicine , Imperial College London , London , UK
| | - Paul R Langford
- a Section of Paediatric Infectious Diseases and Allergy, Department of Medicine , Imperial College London , London , UK
| |
Collapse
|
50
|
Kathayat D, Helmy YA, Deblais L, Rajashekara G. Novel small molecules affecting cell membrane as potential therapeutics for avian pathogenic Escherichia coli. Sci Rep 2018; 8:15329. [PMID: 30333507 PMCID: PMC6193035 DOI: 10.1038/s41598-018-33587-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 09/26/2018] [Indexed: 11/09/2022] Open
Abstract
Avian pathogenic Escherichia coli (APEC), a most common bacterial pathogen of poultry, causes multiple extra-intestinal diseases in poultry which results in significant economic losses to the poultry industry worldwide. In addition, APEC are a subgroup of extra-intestinal pathogenic E. coli (ExPEC), and APEC contaminated poultry products are a potential source of foodborne ExPEC infections to humans and transfer of antimicrobial resistant genes. The emergence of multi-drug resistant APEC strains and the limited efficacy of vaccines necessitate novel APEC control approaches. Here, we screened a small molecule (SM) library and identified 11 SMs bactericidal to APEC. The identified SMs were effective against multiple APEC serotypes, biofilm embedded APEC, antimicrobials resistant APECs, and other pathogenic E. coli strains. Microscopy revealed that these SMs affect the APEC cell membrane. Exposure of SMs to APEC revealed no resistance. Most SMs showed low toxicity towards chicken and human cells and reduced the intracellular APEC load. Treatment with most SMs extended the wax moth larval survival and reduced the intra-larval APEC load. Our studies could facilitate the development of antimicrobial therapeutics for the effective management of APEC infections in poultry as well as other E. coli related foodborne zoonosis, including APEC related ExPEC infections in humans.
Collapse
Affiliation(s)
- Dipak Kathayat
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, 44691, USA
| | - Yosra A Helmy
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, 44691, USA
| | - Loic Deblais
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, 44691, USA
| | - Gireesh Rajashekara
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH, 44691, USA.
| |
Collapse
|