51
|
Giampietro L, Marinacci B, Della Valle A, D’Agostino I, Lauro A, Mori M, Carradori S, Ammazzalorso A, De Filippis B, Maccallini C, Angeli A, Capasso C, Francati S, Mollica A, Grande R, Supuran CT. Azobenzenesulfonamide Carbonic Anhydrase Inhibitors as New Weapons to Fight Helicobacter pylori: Synthesis, Bioactivity Evaluation, In Vivo Toxicity, and Computational Studies. Pharmaceuticals (Basel) 2024; 17:1027. [PMID: 39204133 PMCID: PMC11357054 DOI: 10.3390/ph17081027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/30/2024] [Accepted: 08/03/2024] [Indexed: 09/03/2024] Open
Abstract
Research into novel anti-Helicobacter pylori agents represents an important approach for the identification of new treatments for chronic gastritis and peptic ulcers, which are associated with a high risk of developing gastric carcinoma. In this respect, two series of azobenzenesulfonamides were designed, synthesized, and tested against a large panel of human and bacterial CAs to evaluate their inhibitory activity. In addition, computational studies of the novel primary benzenesulfonamides (4a-j) were performed to predict the putative binding mode to both HpCAs. Then, the antimicrobial activity versus H. pylori of the two series was also studied. The best-in-class compounds were found to be 4c and 4e among the primary azobenzenesulfonamides and 5c and 5f belonging to the secondary azobenzenesulfonamides series, showing themselves to exert a promising anti-H. pylori activity, with MIC values of 4-8 μg/mL and MBCs between 4 and 16 μg/mL. Moreover, the evaluation of their toxicity on a G. mellonella larva in vivo model indicated a safe profile for 4c,e and 5c,f. The collected results warrant considering these azobenzenesulfonamides as an interesting starting point for the development of a new class of anti-H. pylori agents.
Collapse
Affiliation(s)
- Letizia Giampietro
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (B.M.); (A.D.V.); (S.C.); (A.A.); (B.D.F.); (C.M.); (A.M.); (R.G.)
| | - Beatrice Marinacci
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (B.M.); (A.D.V.); (S.C.); (A.A.); (B.D.F.); (C.M.); (A.M.); (R.G.)
- Department of Innovative Technologies in Medicine & Dentistry, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Alice Della Valle
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (B.M.); (A.D.V.); (S.C.); (A.A.); (B.D.F.); (C.M.); (A.M.); (R.G.)
| | - Ilaria D’Agostino
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy;
| | - Aldo Lauro
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (A.L.); (M.M.)
| | - Mattia Mori
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (A.L.); (M.M.)
| | - Simone Carradori
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (B.M.); (A.D.V.); (S.C.); (A.A.); (B.D.F.); (C.M.); (A.M.); (R.G.)
| | - Alessandra Ammazzalorso
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (B.M.); (A.D.V.); (S.C.); (A.A.); (B.D.F.); (C.M.); (A.M.); (R.G.)
| | - Barbara De Filippis
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (B.M.); (A.D.V.); (S.C.); (A.A.); (B.D.F.); (C.M.); (A.M.); (R.G.)
| | - Cristina Maccallini
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (B.M.); (A.D.V.); (S.C.); (A.A.); (B.D.F.); (C.M.); (A.M.); (R.G.)
| | - Andrea Angeli
- Neurofarba Department, University of Florence, Sesto Fiorentino, 50019 Florence, Italy; (A.A.); (C.T.S.)
| | - Clemente Capasso
- Department of Biology, Agriculture and Food Sciences, National Research Council (CNR), Institute of Biosciences and Bioresources, 80131 Naples, Italy;
| | - Santolo Francati
- Department of Agricultural and Food Sciences (DISTAL), University of Bologna, 40126 Bologna, Italy;
| | - Adriano Mollica
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (B.M.); (A.D.V.); (S.C.); (A.A.); (B.D.F.); (C.M.); (A.M.); (R.G.)
| | - Rossella Grande
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (B.M.); (A.D.V.); (S.C.); (A.A.); (B.D.F.); (C.M.); (A.M.); (R.G.)
- Center for Advanced Studies and Technology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Claudiu T. Supuran
- Neurofarba Department, University of Florence, Sesto Fiorentino, 50019 Florence, Italy; (A.A.); (C.T.S.)
| |
Collapse
|
52
|
Böhm L, Schaller ME, Balczun C, Krüger A, Schummel T, Ammon A, Klein N, Helbing DL, Eming R, Fuchs F. A Case of Pseudomonas straminea Blood Stream Infection in an Elderly Woman with Cellulitis. Infect Dis Rep 2024; 16:699-706. [PMID: 39195004 DOI: 10.3390/idr16040053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
Here, we report the simultaneous isolation of Pseudomonas straminea from blood cultures and from a skin ulcer in an elderly woman who suffered from atopic dermatitis and psoriasis and developed acute cellulitis of both arms requiring hospital treatment. To the best of our knowledge, P. straminea has not been previously reported to cause invasive infections in humans. This case highlights how chronic diseases and older age increase the susceptibility to bacterial infections with environmental bacteria of low virulence. Our study describes the microbiological identification of the blood culture isolate, including morpho-molecular characterization and virulence demonstration in a Galleria mellonella model.
Collapse
Affiliation(s)
- Leopold Böhm
- Department of Microbiology and Hospital Hygiene, Bundeswehr Central Hospital Koblenz, 56072 Koblenz, Germany
| | - Marius Eberhardt Schaller
- Department of Dermatology, Venerology and Allergology, Bundeswehr Central Hospital Koblenz, 56072 Koblenz, Germany
| | - Carsten Balczun
- Department of Microbiology and Hospital Hygiene, Bundeswehr Central Hospital Koblenz, 56072 Koblenz, Germany
| | - Andreas Krüger
- Department of Microbiology and Hospital Hygiene, Bundeswehr Central Hospital Koblenz, 56072 Koblenz, Germany
| | - Timo Schummel
- Department of Microbiology and Hospital Hygiene, Bundeswehr Central Hospital Koblenz, 56072 Koblenz, Germany
| | - Alexander Ammon
- Department of Pathology, Bundeswehr Central Hospital Koblenz, 56072 Koblenz, Germany
| | - Niklas Klein
- Department of Microbiology and Hospital Hygiene, Bundeswehr Central Hospital Koblenz, 56072 Koblenz, Germany
| | - Dario Lucas Helbing
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Friedrich Schiller University Jena, Philosophenweg 3, 07743 Jena, Germany
- Leibniz Institute on Aging, Fritz Lipmann Institute, 07745 Jena, Germany
| | - Rüdiger Eming
- Department of Dermatology, Venerology and Allergology, Bundeswehr Central Hospital Koblenz, 56072 Koblenz, Germany
| | - Frieder Fuchs
- Department of Microbiology and Hospital Hygiene, Bundeswehr Central Hospital Koblenz, 56072 Koblenz, Germany
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Medical Faculty and University Hospital of Cologne, 50931 Cologne, Germany
| |
Collapse
|
53
|
Zandarco S, Günther B, Riedel M, Breitenhuber G, Kirst M, Achterhold K, Pfeiffer F, Herzen J. Speckle tracking phase-contrast computed tomography at an inverse Compton X-ray source. OPTICS EXPRESS 2024; 32:28472-28488. [PMID: 39538663 DOI: 10.1364/oe.528701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 06/10/2024] [Indexed: 11/16/2024]
Abstract
Speckle-based X-ray imaging (SBI) is a phase-contrast method developed at and for highly coherent X-ray sources, such as synchrotrons, to increase the contrast of weakly absorbing objects. Consequently, it complements the conventional attenuation-based X-ray imaging. Meanwhile, attempts to establish SBI at less coherent laboratory sources have been performed, ranging from liquid metal-jet X-ray sources to microfocus X-ray tubes. However, their lack of coherence results in interference fringes not being resolved. Therefore, algorithms were developed which neglect the interference effects. Here, we demonstrate phase-contrast computed tomography employing SBI in a laboratory-setting with an inverse Compton X-ray source. In this context, we investigate and compare also the performance of the at synchrotron conventionally used phase-retrieval algorithms for SBI, unified modulated pattern analysis (UMPA) with a phase-retrieval method developed for low coherence systems (LCS). We successfully retrieve a full computed tomography in a phantom as well as in biological specimens, such as larvae of the greater wax moth (Galleria mellonella), a model system for studies of pathogens and infections. In this context, we additionally demonstrate quantitative phase-contrast computed tomography using SBI at a low coherent set-up.
Collapse
|
54
|
Zhao Y, Mannala GK, Youf R, Rupp M, Alt V, Riool M. Development of a Galleria mellonella Infection Model to Evaluate the Efficacy of Antibiotic-Loaded Polymethyl Methacrylate (PMMA) Bone Cement. Antibiotics (Basel) 2024; 13:692. [PMID: 39199992 PMCID: PMC11350861 DOI: 10.3390/antibiotics13080692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 09/01/2024] Open
Abstract
Prosthetic joint infections (PJIs) can have disastrous consequences for patient health, including removal of the device, and placement of cemented implants is often required during surgery to eradicate PJIs. In translational research, in vivo models are widely used to assess the biocompatibility and antimicrobial efficacy of antimicrobial coatings and compounds. Here, we aim to utilize Galleria mellonella implant infection models to assess the antimicrobial activity of antibiotic-loaded bone cement (ALBC) implants. Therefore, we used commercially available bone cement loaded with either gentamicin alone (PALACOS R+G) or with a combination of gentamicin and vancomycin (COPAL G+V), compared to bone cement without antibiotics (PALACOS R). Firstly, the in vitro antimicrobial activity of ALBC was determined against Staphylococcus aureus. Next, the efficacy of ALBC implants was analyzed in both the G. mellonella hematogenous and early-stage biofilm implant infection model, by monitoring the survival of larvae over time. After 24 h, the number of bacteria on the implant surface and in the tissue was determined. Larvae receiving dual-loaded COPAL G+V implants showed higher survival rates compared to implants loaded with only gentamicin (PALACOS R+G) and the control implants without antibiotics (PALACOS R). In conclusion, G. mellonella larvae infection models with antibiotic-loaded bone cements are an excellent option to study (novel) antimicrobial approaches.
Collapse
Affiliation(s)
| | | | | | | | | | - Martijn Riool
- Department of Trauma Surgery, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany; (Y.Z.); (G.K.M.); (R.Y.); (M.R.); (V.A.)
| |
Collapse
|
55
|
Ammanath AV, Matsuo M, Wang H, Kraus F, Bleisch A, Peslalz P, Mohammad M, Deshmukh M, Grießhammer A, Purkayastha M, Vorbach A, Macek B, Brötz-Oesterhelt H, Maier L, Kretschmer D, Peschel A, Jin T, Plietker B, Götz F. Antimicrobial Evaluation of Two Polycyclic Polyprenylated Acylphloroglucinol Compounds: PPAP23 and PPAP53. Int J Mol Sci 2024; 25:8023. [PMID: 39125595 PMCID: PMC11312133 DOI: 10.3390/ijms25158023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024] Open
Abstract
Polycyclic polyprenylated acylphloroglucinols (PPAPs) comprise a large group of compounds of mostly plant origin. The best-known compound is hyperforin from St. John's wort with its antidepressant, antitumor and antimicrobial properties. The chemical synthesis of PPAP variants allows the generation of compounds with improved activity and compatibility. Here, we studied the antimicrobial activity of two synthetic PPAP-derivatives, the water-insoluble PPAP23 and the water-soluble sodium salt PPAP53. In vitro, both compounds exhibited good activity against methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococcus faecium. Both compounds had no adverse effects on Galleria mellonella wax moth larvae. However, they were unable to protect the larvae from infection with S. aureus because components of the larval coelom neutralized the antimicrobial activity; a similar effect was also seen with serum albumin. In silico docking studies with PPAP53 revealed that it binds to the F1 pocket of human serum albumin with a binding energy of -7.5 kcal/mol. In an infection model of septic arthritis, PPAP23 decreased the formation of abscesses and S. aureus load in kidneys; in a mouse skin abscess model, topical treatment with PPAP53 reduced S. aureus counts. Both PPAPs were active against anaerobic Gram-positive gut bacteria such as neurotransmitter-producing Clostridium, Enterococcus or Ruminococcus species. Based on these results, we foresee possible applications in the decolonization of pathogens.
Collapse
Affiliation(s)
- Aparna Viswanathan Ammanath
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, 72076 Tübingen, Germany
| | - Miki Matsuo
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, 72076 Tübingen, Germany
| | - Huanhuan Wang
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, 72076 Tübingen, Germany
| | - Frank Kraus
- Organic Chemistry I, Faculty of Chemistry and Food Chemistry, Technical University Dresden, 01062 Dresden, Germany (P.P.)
| | - Anton Bleisch
- Organic Chemistry I, Faculty of Chemistry and Food Chemistry, Technical University Dresden, 01062 Dresden, Germany (P.P.)
| | - Philipp Peslalz
- Organic Chemistry I, Faculty of Chemistry and Food Chemistry, Technical University Dresden, 01062 Dresden, Germany (P.P.)
| | - Majd Mohammad
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (M.M.); (M.D.)
| | - Meghshree Deshmukh
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (M.M.); (M.D.)
| | - Anne Grießhammer
- Interfakultäres Institut für Mikrobiologie und Infektionsmedizin (IMIT), University of Tübingen, 72076 Tübingen, Germany
- Excellence Cluster 2124 ‘Controlling Microbes to Fight Infections’ (CMFI), University of Tübingen, 72076 Tübingen, Germany
| | - Moushumi Purkayastha
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, 72076 Tübingen, Germany
| | - Andreas Vorbach
- Microbial Bioactive Compounds, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, 72076 Tübingen, Germany;
| | - Boris Macek
- Excellence Cluster 2124 ‘Controlling Microbes to Fight Infections’ (CMFI), University of Tübingen, 72076 Tübingen, Germany
- Quantitative Proteomics, Proteome Center Tübingen, Interfaculty Institute for Cell Biology, University of Tübingen, 72076 Tübingen, Germany
| | - Heike Brötz-Oesterhelt
- Excellence Cluster 2124 ‘Controlling Microbes to Fight Infections’ (CMFI), University of Tübingen, 72076 Tübingen, Germany
- Microbial Bioactive Compounds, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, 72076 Tübingen, Germany;
| | - Lisa Maier
- Interfakultäres Institut für Mikrobiologie und Infektionsmedizin (IMIT), University of Tübingen, 72076 Tübingen, Germany
- Excellence Cluster 2124 ‘Controlling Microbes to Fight Infections’ (CMFI), University of Tübingen, 72076 Tübingen, Germany
| | - Dorothee Kretschmer
- Interfakultäres Institut für Mikrobiologie und Infektionsmedizin (IMIT), University of Tübingen, 72076 Tübingen, Germany
- Excellence Cluster 2124 ‘Controlling Microbes to Fight Infections’ (CMFI), University of Tübingen, 72076 Tübingen, Germany
| | - Andreas Peschel
- Interfakultäres Institut für Mikrobiologie und Infektionsmedizin (IMIT), University of Tübingen, 72076 Tübingen, Germany
- Excellence Cluster 2124 ‘Controlling Microbes to Fight Infections’ (CMFI), University of Tübingen, 72076 Tübingen, Germany
| | - Tao Jin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (M.M.); (M.D.)
| | - Bernd Plietker
- Organic Chemistry I, Faculty of Chemistry and Food Chemistry, Technical University Dresden, 01062 Dresden, Germany (P.P.)
| | - Friedrich Götz
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, 72076 Tübingen, Germany
- Excellence Cluster 2124 ‘Controlling Microbes to Fight Infections’ (CMFI), University of Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
56
|
Sun N, Zhi Z, Xiao T, Deng X, He T, Dong W, Feng S, Chen S, Wong WL, Yuan W. The study of honokiol as a natural product-based antimicrobial agent and its potential interaction with FtsZ protein. Front Microbiol 2024; 15:1361508. [PMID: 39104591 PMCID: PMC11298477 DOI: 10.3389/fmicb.2024.1361508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 07/10/2024] [Indexed: 08/07/2024] Open
Abstract
Multidrug resistant bacteria have been a global health threat currently and frontline clinical treatments for these infections are very limited. To develop potent antibacterial agents with new bactericidal mechanisms is thus needed urgently to address this critical antibiotic resistance challenge. Natural products are a treasure of small molecules with high bioactive and low toxicity. In the present study, we demonstrated that a natural compound, honokiol, showed potent antibacterial activity against a number of Gram-positive bacteria including MRSA and VRE. Moreover, honokiol in combination with clinically used β-lactam antibiotics exhibits strong synergistic antimicrobial effects against drug-resistant S. aureus strains. Biochemical studies further reveal that honokiol may disrupt the GTPase activity, FtsZ polymerization, cell division. These biological impacts induced by honokiol may ultimately cause bacterial cell death. The in vivo antibacterial activity of honokiol against S. aureus infection was also verified with a biological model of G. mellonella larvae. The in vivo results support that honokiol is low toxic against the larvae and effectively increases the survival rate of the larvae infected with S. aureus. These findings demonstrate the potential of honokiol for further structural advancement as a new class of antibacterial agents with high potency against multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Ning Sun
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, China
| | - Ziling Zhi
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China
| | - Ting Xiao
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, China
| | - Xin Deng
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, China
| | - Tenghui He
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, China
| | - Wanyang Dong
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China
| | - Shuyi Feng
- Guangzhou 11th People's Hospital, Guangzhou Cadre and Talent Health Management Center, Guangzhou, China
| | - Sisi Chen
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China
| | - Wing-Leung Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Wenchang Yuan
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
57
|
Yu H, Xu Y, Imani S, Zhao Z, Ullah S, Wang Q. Navigating ESKAPE Pathogens: Considerations and Caveats for Animal Infection Models Development. ACS Infect Dis 2024; 10:2336-2355. [PMID: 38866389 PMCID: PMC11249778 DOI: 10.1021/acsinfecdis.4c00007] [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: 01/04/2024] [Revised: 05/19/2024] [Accepted: 05/29/2024] [Indexed: 06/14/2024]
Abstract
The misuse of antibiotics has led to the global spread of drug-resistant bacteria, especially multi-drug-resistant (MDR) ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species). These opportunistic bacteria pose a significant threat, in particular within hospitals, where they cause nosocomial infections, leading to substantial morbidity and mortality. To comprehensively explore ESKAPE pathogenesis, virulence, host immune response, diagnostics, and therapeutics, researchers increasingly rely on necessitate suitable animal infection models. However, no single model can fully replicate all aspects of infectious diseases. Notably when studying opportunistic pathogens in immunocompetent hosts, rapid clearance by the host immune system can limit the expression of characteristic disease symptoms. In this study, we examine the critical role of animal infection models in understanding ESKAPE pathogens, addressing limitations and research gaps. We discuss applications and highlight key considerations for effective models. Thoughtful decisions on disease replication, parameter monitoring, and data collection are crucial for model reliability. By meticulously replicating human diseases and addressing limitations, researchers maximize the potential of animal infection models. This aids in targeted therapeutic development, bridges knowledge gaps, and helps combat MDR ESKAPE pathogens, safeguarding public health.
Collapse
Affiliation(s)
- Haojie Yu
- Key
Laboratory of Artificial Organs and Computational Medicine in Zhejiang
Province, Key Laboratory of Pollution Exposure and Health Intervention
of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang China
- Stomatology
Hospital, School of Stomatology, Zhejiang University School of Medicine,
Zhejiang Provincial Clinical Research Center for Oral Diseases, Key
Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Yongchang Xu
- Key
Laboratory of Aging and Cancer Biology of Zhejiang Province, School
of Basic Medical Sciences, Hangzhou Normal
University, Hangzhou 311121, China
| | - Saber Imani
- Shulan
International Medical College, Zhejiang
Shuren University, Hangzhou 310015, Zhejiang China
| | - Zhuo Zhao
- Department
of Computer Science and Engineering, University
of Notre Dame, Notre
Dame, Indiana 46556, United States
| | - Saif Ullah
- Department
of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, United States
| | - Qingjing Wang
- Key
Laboratory of Artificial Organs and Computational Medicine in Zhejiang
Province, Key Laboratory of Pollution Exposure and Health Intervention
of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, Zhejiang China
| |
Collapse
|
58
|
Liu J, Wang Z, Zeng Y, Wang W, Tang S, Jia A. 1H-Pyrrole-2,5-dicarboxylic acid, a quorum sensing inhibitor from one endophytic fungus in Areca catechu L., acts as antibiotic accelerant against Pseudomonas aeruginosa. Front Cell Infect Microbiol 2024; 14:1413728. [PMID: 39015339 PMCID: PMC11250523 DOI: 10.3389/fcimb.2024.1413728] [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: 04/07/2024] [Accepted: 05/27/2024] [Indexed: 07/18/2024] Open
Abstract
Pseudomonas aeruginosa has already been stipulated as a "critical" pathogen, emphasizing the urgent need for researching and developing novel antibacterial agents due to multidrug resistance. Bacterial biofilm formation facilitates cystic fibrosis development and restricts the antibacterial potential of many current antibiotics. The capacity of P. aeruginosa to form biofilms and resist antibiotics is closely correlated with quorum sensing (QS). Bacterial QS is being contemplated as a promising target for developing novel antibacterial agents. QS inhibitors are a promising strategy for treating chronic infections. This study reported that the active compound PT22 (1H-pyrrole-2,5-dicarboxylic acid) isolated from Perenniporia tephropora FF2, one endophytic fungus from Areca catechu L., presents QS inhibitory activity against P. aeruginosa. Combined with gentamycin or piperacillin, PT22 functions as a novel antibiotic accelerant against P. aeruginosa. PT22 (0.50 mg/mL, 0.75 mg/mL, and 1.00 mg/mL) reduces the production of QS-related virulence factors, such as pyocyanin and rhamnolipid, and inhibits biofilm formation of P. aeruginosa PAO1 instead of affecting its growth. The architectural disruption of the biofilms was confirmed by visualization through scanning electron microscopy (SEM) and confocal laser scanning microscopy (CLSM). Real-time quantitative PCR (RT-qPCR) indicated that PT22 significantly attenuated the expression of QS-related genes followed by docking analysis of molecules against QS activator proteins. PT22 dramatically increased the survival rate of Galleria mellonella. PT22 combined with gentamycin or piperacillin presents significant inhibition of biofilm formation and eradication of mature biofilm compared to monotherapy, which was also confirmed by visualization through SEM and CLSM. After being treated with PT22 combined with gentamycin or piperacillin, the survival rates of G. mellonella were significantly increased compared to those of monotherapy. PT22 significantly enhanced the susceptibility of gentamycin and piperacillin against P. aeruginosa PAO1. Our results suggest that PT22 from P. tephropora FF2 as a potent QS inhibitor is a candidate antibiotic accelerant to combat the antibiotic resistance of P. aeruginosa.
Collapse
Affiliation(s)
- Junsheng Liu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Zhennan Wang
- Modern Industrial College of Traditional Chinese Medicine and Health, Lishui University, Lishui, China
| | - Yuexiang Zeng
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Wei Wang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Shi Tang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Aiqun Jia
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
59
|
Wang Q, Liu H, Yao Y, Chen H, Yang Z, Xie H, Cui R, Liu H, Li C, Gong W, Yu Y, Hua X, Li S. Emergence of novel hypervirulent Acinetobacter baumannii strain and herpes simplex type 1 virus in a case of community-acquired pneumonia in China. J Infect Public Health 2024; 17:102456. [PMID: 38820896 DOI: 10.1016/j.jiph.2024.05.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 04/24/2024] [Accepted: 05/16/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND A. baumannii is an important and common clinical pathogen, especially in the intensive care unit (ICU). This study aimed to characterize one hypervirulent A. baumannii strain in a patient with community-acquired pneumonia and herpes simplex type 1 virus infection. METHODS Minimum inhibitory concentrations (MICs) were determined using the Kirby-Bauer (K-B) and broth microdilution methods. Galleria mellonella infection model experiment was conducted. Whole-genome sequencing (WGS) was performed using the Illumina and Nanopore platforms. The resistance and virulence determinants were identified using the ABRicate program with ResFinder and the VFDB database. The capsular polysaccharide locus (K locus) and lipooligosaccharide outer core locus (OC locus) were identified using Kleborate with Kaptive. Phylogenetic analyses were conducted using the BacWGSTdb server. RESULTS A. baumannii XH2146 strain belongs to ST10Pas and ST447Oxf. The strain was resistant to cefazolin, ciprofloxacin, and trimethoprim/sulfamethoxazole (TMP-SMX). Bautype and Kaptive analyses showed that XH2146 contains OCL2 and KL49. WGS analysis revealed that the strain harbored blaADC-76, blaOXA-68, ant(3'')-IIa, tet(B), and sul2. Notably, tet(B) and sul2, both were located within a 114,700-bp plasmid (designated pXH2146-1). Virulence assay revealed A. baumannii XH2146 possessed higher virulence than A. baumannii AB5075 at 12 h. Comparative genomic analysis showed that A. baumannii ST447 strains were mainly isolated from the USA and exhibited a relatively close genetic relationship. Importantly, 11 strains were observed to carry blaOXA-58; blaOXA-23 was identified in 11 isolates and three ST447 A. baumannii strains harbored blaNDM-1. CONCLUSIONS Early detection of community-acquired hypervirulent Acinetobacter baumannii strains is recommended to prevent their extensive spread in hospitals.
Collapse
Affiliation(s)
- Qiujing Wang
- Department of Infectious Diseases, Zhoushan Hospital, Wenzhou Medical University, Zhoushan, Zhejiang, China
| | - Haiyang Liu
- Centre of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, 158 Shangtang Road, Hangzhou, Zhejiang 310014, China
| | - Yue Yao
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hangfei Chen
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhejuan Yang
- Department of Infectious Diseases, Zhoushan Hospital, Wenzhou Medical University, Zhoushan, Zhejiang, China
| | - Haibo Xie
- Intensive care unit (ICU), Zhoushan Women and Children Hospital, China
| | - Rongna Cui
- Intensive care unit (ICU), Zhoushan Women and Children Hospital, China
| | - Huasheng Liu
- Department of Infectious Diseases, Zhoushan Hospital, Wenzhou Medical University, Zhoushan, Zhejiang, China
| | - Chuner Li
- Microbiological laboratory, Zhoushan Hospital, Wenzhou Medical University, China
| | - Weiping Gong
- Department of dermatology, Zhoushan Hospital, Wenzhou Medical University, Zhoushan, Zhejiang, China
| | - Yunsong Yu
- Center for General Practice Medicine, Department of Infectious Diseases, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Xiaoting Hua
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Shibo Li
- Department of Infectious Diseases, Zhoushan Hospital, Wenzhou Medical University, Zhoushan, Zhejiang, China.
| |
Collapse
|
60
|
Li W, Zhang J, Fu Y, Wang J, Liu L, Long W, Yu K, Li X, Wei C, Liang X, Wang J, Li C, Zhang X. In vitro and in vivo activity of ceftazidime/avibactam and aztreonam alone or in combination against mcr-9, serine- and metallo-β-lactamases-co-producing carbapenem-resistant Enterobacter cloacae complex. Eur J Clin Microbiol Infect Dis 2024; 43:1309-1318. [PMID: 38700663 DOI: 10.1007/s10096-024-04841-8] [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: 12/04/2023] [Accepted: 04/25/2024] [Indexed: 07/20/2024]
Abstract
PURPOSE Enterobacteriaceae carrying mcr-9, in particularly those also co-containing metallo-β-lactamase (MBL) and TEM type β-lactamase, present potential transmission risks and lack adequate clinical response methods, thereby posing a major threat to global public health. The aim of this study was to assess the antimicrobial efficacy of a combined ceftazidime/avibactam (CZA) and aztreonam (ATM) regimen against carbapenem-resistant Enterobacter cloacae complex (CRECC) co-producing mcr-9, MBL and TEM. METHODS The in vitro antibacterial activity of CZA plus ATM was evaluated using a time-kill curve assay. Furthermore, the in vivo interaction between CZA plus ATM was confirmed using a Galleria mellonella (G. mellonella) infection model. RESULTS All eight clinical strains of CRECC, co-carrying mcr-9, MBL and TEM, exhibited high resistance to CZA and ATM. In vitro time-kill curve analysis demonstrated that the combination therapy of CZA + ATM exerted significant bactericidal activity against mcr-9, MBL and TEM-co-producing Enterobacter cloacae complex (ECC) isolates with a 100% synergy rate observed in our study. Furthermore, in vivo survival assay using Galleria mellonella larvae infected with CRECC strains co-harboring mcr-9, MBL and TEM revealed that the CZA + ATM combination significantly improved the survival rate compared to the drug-treatment alone and untreated control groups. CONCLUSION To our knowledge, this study represents the first report on the in vitro and in vivo antibacterial activity of CZA plus ATM against CRECC isolates co-harboring mcr-9, MBL and TEM. Our findings suggest that the combination regimen of CZA + ATM provides a valuable reference for clinicians to address the increasingly complex antibiotic resistance situation observed in clinical microorganisms.
Collapse
Affiliation(s)
- Wengang Li
- Department of Microbiology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
- Department of Pathogenic Biology, Jiamusi University School of Basic Medicine, Jiamusi, China
| | - Jisheng Zhang
- Department of Microbiology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Yanjun Fu
- Department of Clinical Laboratory, The First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Jianmin Wang
- Department of Microbiology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Longjin Liu
- Department of Microbiology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Wenzhang Long
- Department of Microbiology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Kaixin Yu
- Department of Microbiology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
- Department of Pathogenic Biology, Jiamusi University School of Basic Medicine, Jiamusi, China
| | - Xinhui Li
- Department of Microbiology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Chunli Wei
- Department of Microbiology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Xushan Liang
- Department of Microbiology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Jin Wang
- Department of Microbiology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Chunjiang Li
- Department of Life Science and Technology, Mudanjiang Normal University, Mudanjiang, China.
| | - Xiaoli Zhang
- Department of Microbiology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
61
|
Chen PJ, Tan MC, Huang WC, Hsu SY, Chen TL, Yang CY, Kuo SC. The individual contributions of blaB, blaGOB and blaCME on MICs of β-lactams in Elizabethkingia anophelis. J Antimicrob Chemother 2024; 79:1577-1580. [PMID: 38742706 PMCID: PMC11215548 DOI: 10.1093/jac/dkae137] [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: 02/06/2024] [Accepted: 04/23/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND The blaB, blaGOB and blaCME genes are thought to confer β-lactam resistance to Elizabethkingia anophelis, based on experiments conducted primarily on Escherichia coli. OBJECTIVES To determine the individual contributions of β-lactamase genes to increased MICs in E. anophelis and to assess their impact on the in vivo efficacy of carbapenem therapy. METHODS Scarless gene deletion of one or more β-lactamase gene(s) was performed in three clinical E. anophelis isolates. MICs were determined by broth microdilution. Hydrolytic activity and expressions of β-lactamase genes were measured by an enzymatic assay and quantitative RT-PCR, respectively. In vivo efficacy was determined using Galleria mellonella and murine thigh infection models. RESULTS The presence of blaB resulted in >16-fold increases, while blaGOB caused 4-16-fold increases of carbapenem MICs. Hydrolysis of carbapenems was highest in lysates of blaB-positive strains, possibly due to the constitutionally higher expression of blaB. Imipenem was ineffective against blaB-positive isolates in vivo in terms of improvement of the survival of wax moth larvae and reduction of murine bacterial load. The deletion of blaB restored the efficacy of imipenem. The blaB gene was also responsible for a >4-fold increase of ampicillin/sulbactam and piperacillin/tazobactam MICs. The presence of blaCME, but not blaB or blaGOB, increased the MICs of ceftazidime and cefepime by 8-16- and 4-8-fold, respectively. CONCLUSIONS The constitutionally and highly expressed blaB gene in E. anophelis was responsible for increased MICs of carbapenems and led to their poor in vivo efficacy. blaCME increased the MICs of ceftazidime and cefepime.
Collapse
Affiliation(s)
- Pei-Jing Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
| | - Mei-Chen Tan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
| | - Wei-Cheng Huang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
| | - Shu-Yuan Hsu
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli County, Taiwan
| | - Te-Li Chen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chiou-Ying Yang
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
| | - Shu-Chen Kuo
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
| |
Collapse
|
62
|
Curtis A, Dobes P, Marciniak J, Hurychova J, Hyrsl P, Kavanagh K. Characterization of Aspergillus fumigatus secretome during sublethal infection of Galleria mellonella larvae. J Med Microbiol 2024; 73:001844. [PMID: 38836745 PMCID: PMC11261830 DOI: 10.1099/jmm.0.001844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/17/2024] [Indexed: 06/06/2024] Open
Abstract
Introduction. The fungal pathogen Aspergillus fumigatus can induce prolonged colonization of the lungs of susceptible patients, resulting in conditions such as allergic bronchopulmonary aspergillosis and chronic pulmonary aspergillosis.Hypothesis. Analysis of the A. fumigatus secretome released during sub-lethal infection of G. mellonella larvae may give an insight into products released during prolonged human colonisation.Methodology. Galleria mellonella larvae were infected with A. fumigatus, and the metabolism of host carbohydrate and proteins and production of fungal virulence factors were analysed. Label-free qualitative proteomic analysis was performed to identify fungal proteins in larvae at 96 hours post-infection and also to identify changes in the Galleria proteome as a result of infection.Results. Infected larvae demonstrated increasing concentrations of gliotoxin and siderophore and displayed reduced amounts of haemolymph carbohydrate and protein. Fungal proteins (399) were detected by qualitative proteomic analysis in cell-free haemolymph at 96 hours and could be categorized into seven groups, including virulence (n = 25), stress response (n = 34), DNA repair and replication (n = 39), translation (n = 22), metabolism (n = 42), released intracellular (n = 28) and cellular development and cell cycle (n = 53). Analysis of the Gallerial proteome at 96 hours post-infection revealed changes in the abundance of proteins associated with immune function, metabolism, cellular structure, insect development, transcription/translation and detoxification.Conclusion. Characterizing the impact of the fungal secretome on the host may provide an insight into how A. fumigatus damages tissue and suppresses the immune response during long-term pulmonary colonization.
Collapse
Affiliation(s)
- Aaron Curtis
- Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Pavel Dobes
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Jacek Marciniak
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Jana Hurychova
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Pavel Hyrsl
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Kevin Kavanagh
- Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland
| |
Collapse
|
63
|
Naknaen A, Samernate T, Saeju P, Nonejuie P, Chaikeeratisak V. Nucleus-forming jumbophage PhiKZ therapeutically outcompetes non-nucleus-forming jumbophage Callisto. iScience 2024; 27:109790. [PMID: 38726363 PMCID: PMC11079468 DOI: 10.1016/j.isci.2024.109790] [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: 09/21/2023] [Revised: 03/21/2024] [Accepted: 04/16/2024] [Indexed: 05/12/2024] Open
Abstract
With the recent resurgence of phage therapy in modern medicine, jumbophages are currently under the spotlight due to their numerous advantages as anti-infective agents. However, most significant discoveries to date have primarily focused on nucleus-forming jumbophages, not their non-nucleus-forming counterparts. In this study, we compare the biological characteristics exhibited by two genetically diverse jumbophages: 1) the well-studied nucleus-forming jumbophage, PhiKZ; and 2) the newly discovered non-nucleus-forming jumbophage, Callisto. Single-cell infection studies further show that Callisto possesses different replication machinery, resulting in a delay in phage maturation compared to that of PhiKZ. The therapeutic potency of both phages was examined in vitro and in vivo, demonstrating that PhiKZ holds certain superior characteristics over Callisto. This research sheds light on the importance of the subcellular infection machinery and the organized progeny maturation process, which could potentially provide valuable insight in the future development of jumbophage-based therapeutics.
Collapse
Affiliation(s)
- Ampapan Naknaen
- Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Thanadon Samernate
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Panida Saeju
- Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Poochit Nonejuie
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | | |
Collapse
|
64
|
Lin Z, Zhou Z, Shuai X, Zeng G, Bao R, Chen H. Landscape of plasmids encoding β-lactamases in disinfection residual Enterobacteriaceae from wastewater treatment plants. WATER RESEARCH 2024; 255:121549. [PMID: 38564891 DOI: 10.1016/j.watres.2024.121549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/12/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
Conventional disinfection processes, such as chlorination and UV radiation, are ineffective in controling antibiotic-resistant bacteria, especially disinfection residual Enterobacteriaceae (DRE) encoding β-lactamases, some of which have been classified as "critical priority pathogens" by the World Health Organization. However, few studies have focused on the transferability, phenotype, and genetic characteristics of DRE-derived plasmids encoding β-lactamases, especially extended-spectrum β-lactamases and carbapenemases. In this study, we isolated 10 typical DRE harboring plasmid-mediated blaNDM, blaCTX-M, or blaTEM in post-disinfection effluent from two wastewater treatment plants (WWTPs), with transfer frequency ranging from 1.69 × 10-6 to 3.02 × 10-5. According to genomic maps of plasmids, all blaNDM and blaTEM were cascaded with IS26, and blaCTX-M was adjacent to ISEcp1 or IS26, indicating the important role of these elements in the movement of β-lactamase-encoding genes. The presence of intact class 1 integrons on pWTPN-01 and pWTPC-03 suggested the ability of these DRE-derived plasmids to integrate other exogenous antibiotic resistance genes (ARGs). The coexistence of antibiotic, disinfectant, and heavy metal resistance genes on the same plasmid (e.g., pWTPT-03) implied the facilitating role of disinfectants and heavy metals in the transmission of DRE-derived ARGs. Notably, two plasmid transconjugants exhibited no discernible competitive fitness cost, suggesting a heightened environmental persistence. Furthermore, enhanced virulence induced by β-lactamase-encoding plasmids in their hosts was confirmed using Galleria mellonella infection models, which might be attributed to plasmid-mediated virulence genes. Overall, this study describes the landscape of β-lactamase-encoding plasmids in DRE, and highlights the urgent need for advanced control of DRE to keep environmental and ecological security.
Collapse
Affiliation(s)
- Zejun Lin
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhenchao Zhou
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinyi Shuai
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Guangshu Zeng
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ruiqi Bao
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hong Chen
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental Resource Sciences, Zhejiang University, Hangzhou 310058, China; International Cooperation Base of Environmental Pollution and Ecological Health, Science and Technology Agency of Zhejiang, Zhejiang University, China.
| |
Collapse
|
65
|
Garrine M, Andrade M, Neves J, Mandomando I, Couto I, Costa SS. Exploring the virulence potential of Staphylococcus aureus CC121 and CC152 lineages related to paediatric community-acquired bacteraemia in Manhiça, Mozambique. Sci Rep 2024; 14:10758. [PMID: 38730020 PMCID: PMC11087594 DOI: 10.1038/s41598-024-61345-3] [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: 12/19/2023] [Accepted: 05/05/2024] [Indexed: 05/12/2024] Open
Abstract
Staphylococcus aureus is a frequent agent of bacteraemia. This bacterium has a variety of virulence traits that allow the establishment and maintenance of infection. This study explored the virulence profile of S. aureus strains causing paediatric bacteraemia (SAB) in Manhiça district, Mozambique. We analysed 336 S. aureus strains isolated from blood cultures of children younger than 5 years admitted to the Manhiça District Hospital between 2001 and 2019, previously characterized for antibiotic susceptibility and clonality. The strains virulence potential was evaluated by PCR detection of the Panton-Valentine leucocidin (PVL) encoding genes, lukS-PV/lukF-PV, assessment of the capacity for biofilm formation and pathogenicity assays in Galleria mellonella. The overall carriage of PVL-encoding genes was over 40%, although reaching ~ 70 to 100% in the last years (2014 to 2019), potentially linked to the emergence of CC152 lineage. Strong biofilm production was a frequent trait of CC152 strains. Representative CC152 and CC121 strains showed higher virulence potential in the G. mellonella model when compared to reference strains, with variations within and between CCs. Our results highlight the importance of monitoring the emergent CC152-MSSA-PVL+ and other lineages, as they display important virulence traits that may negatively impact the management of SAB paediatric patients in Manhiça district, Mozambique.
Collapse
Affiliation(s)
- Marcelino Garrine
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisbon, Portugal
| | - Mariana Andrade
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisbon, Portugal
| | - Joana Neves
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisbon, Portugal
| | - Inácio Mandomando
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisbon, Portugal
- Instituto Nacional de Saúde (INS), Ministério da Saúde, Maputo, Mozambique
- ISGlobal-Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Isabel Couto
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisbon, Portugal
| | - Sofia Santos Costa
- Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL, Rua da Junqueira 100, 1349-008, Lisbon, Portugal.
| |
Collapse
|
66
|
Thanki AM, Osei EK, Whenham N, Salter MG, Bedford MR, Masey O’Neill HV, Clokie MRJ. Broad host range phages target global Clostridium perfringens bacterial strains and clear infection in five-strain model systems. Microbiol Spectr 2024; 12:e0378423. [PMID: 38511948 PMCID: PMC11064546 DOI: 10.1128/spectrum.03784-23] [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: 10/26/2023] [Accepted: 03/06/2024] [Indexed: 03/22/2024] Open
Abstract
Clostridium perfringens is a prevalent bacterial pathogen in poultry, and due to the spread of antimicrobial resistance, alternative treatments are needed to prevent and treat infection. Bacteriophages (phages), viruses that kill bacteria, offer a viable option and can be used therapeutically to treat C. perfringens infections. The aim of this study was to isolate phages against C. perfringens strains currently circulating on farms across the world and establish their virulence and development potential using host range screening, virulence assays, and larva infection studies. We isolated 32 phages of which 19 lysed 80%-92% of our global C. perfringens poultry strain collection (n = 97). The virulence of these individual phages and 32 different phage combinations was quantified in liquid culture at multiple doses. We then developed a multi-strain C. perfringens larva infection model, to mimic an effective poultry model used by the industry. We tested the efficacy of 16/32 phage cocktails in the larva model. From this, we identified that our phage cocktail consisting of phages CPLM2, CPLM15, and CPLS41 was the most effective at reducing C. perfringens colonization in infected larvae when administered before bacterial challenge. These data suggest that phages do have significant potential to prevent and treat C. perfringens infection in poultry. IMPORTANCE Clostridium perfringens causes foodborne illness worldwide, and 95% of human infections are linked to the consumption of contaminated meat, including chicken products. In poultry, C. perfringens infection causes necrotic enteritis, and associated mortality rates can be up to 50%. However, treating infections is difficult as the bacterium is becoming antibiotic-resistant. Furthermore, the poultry industry is striving toward reduced antibiotic usage. Bacteriophages (phages) offer a promising alternative, and to progress this approach, robust suitable phages and laboratory models that mimic C. perfringens infections in poultry are required. In our study, we isolated phages targeting C. perfringens and found that many lyse C. perfringens strains isolated from chickens worldwide. Consistent with other published studies, in the model systems we assayed here, when some phages were combined as cocktails, the infection was cleared most effectively compared to individual phage use.
Collapse
Affiliation(s)
- Anisha M. Thanki
- Department of Genetics and Genome Biology, Leicester Centre for Phage Research, University of Leicester, Leicester, United Kingdom
| | - Emmanuel K. Osei
- Department of Agriculture, Food and the Marine, Teagasc Food Research Centre, Moorepark, Ireland
- APC Microbiome Ireland, University College, Cork, Ireland
| | - Natasha Whenham
- AB Agri, Innovation Way, Peterborough Business Park, Peterborough, United Kingdom
| | - Michael G. Salter
- AB Agri, Innovation Way, Peterborough Business Park, Peterborough, United Kingdom
| | - Mike R. Bedford
- AB Vista, Woodstock Court, Marlborough Business Park, Marlborough, Wiltshire, United Kingdom
| | | | - Martha R. J. Clokie
- Department of Genetics and Genome Biology, Leicester Centre for Phage Research, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
67
|
Genç TT, Kaya S, Günay M, Çakaloğlu Ç. Humoral immune response of Galleria mellonella after mono- and co-injection with Hypericum perforatum extract and Candida albicans. APMIS 2024; 132:358-370. [PMID: 38344892 DOI: 10.1111/apm.13383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 01/24/2024] [Indexed: 04/16/2024]
Abstract
Galleria mellonella is used as a model organism to study the innate immune response of insects. In this study, the humoral immune response was assessed by examining phenoloxidase activity, fungal burden, and the expression of phenoloxidase and antimicrobial peptide genes at different time point following separate and combined injections of Hypericum perforatum extract and a nonlethal dose of Candida albicans. The administration of a plant extract at low doses increased phenoloxidase activity, while higher doses had no effect. Similarly, co-injection of a low dose of the extract with the pathogen allowed half of the yeast cells to survive after 24 h. Co-injection of plant extract with the pathogen decreased the phenoloxidase activity at the end of 4 h compared to C. albicans mono-injection. The phenoloxidase gene expressions was reduced in all experimental conditions with respect to the control. When plant extracts and the pathogen were administered together, gallerimycin and hemolin gene expressions were considerably higher compared to mono-injections of plant extracts and the pathogen. The results of this study reveal that gene activation and regulatory mechanisms may change for each immune gene, and that recognition and signaling pathways may differ depending on the involved immunoregulator.
Collapse
Affiliation(s)
- Tülay Turgut Genç
- Department of Biology, Science Faculty, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Serhat Kaya
- Department of Biology, Science Faculty, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Melih Günay
- Graduate School of Natural and Applied Sciences, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Çağla Çakaloğlu
- Graduate School of Natural and Applied Sciences, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| |
Collapse
|
68
|
Castro J, Oliveira R, Fernandes L, Carvalho I, Oliveira H, Brinks E, Cho GS, Franz C, Almeida C, Silva S, Araújo D. Molecular characterization and virulence profile of Klebsiella pneumoniae and Klebsiella oxytoca isolated from ill cats and dogs in Portugal. Vet Microbiol 2024; 292:110056. [PMID: 38537400 DOI: 10.1016/j.vetmic.2024.110056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 04/10/2024]
Abstract
Klebsiella spp. are important pathogens of humans and companion animals such as cats and dogs, capable of causing severe life-threatening diseases. The aim of this study was to characterize the molecular and phenotypic properties of Klebsiella pneumoniae and Klebsiella oxytoca isolated from ill companion animals by whole genome sequencing, followed by in vitro assessment of biofilm formation and in vivo pathogenicity using the Galleria mellonella model. Two LPS O-types were identified for all the K. pneumoniae isolates tested (O3B and O1/O2v2) and only one for K. oxytoca isolates (OL104), and the most common STs found were ST11 and ST266. Furthermore, a high diversity of K-locus types was found for K. pneumoniae (KL102; KL105; KL31, and KL13). Within K. pneumoniae, one specific O/K/ST-types combination (i.e., KL105-ST11-O1/O2v2) showed results that were of concern, as it exhibited a high inflammatory response at 12 h post-infection in G. mellonella with 80% of the larvae dead at 72 h post-infection. This virulence potential, on the other hand, did not appear to be directly related to the biofilm-forming capacity. Also, virulence and resistance scores obtained for this set of strains did surpass score 1. The present study demonstrated that Klebsiella spp. isolated from companion animals belonging to STs that can cause human infections and present virulence on an invertebrate model. Thus, this study underscores the role of dogs and cats as reservoirs of resistant Klebsiella spp. that could potentially be transmitted to humans.
Collapse
Affiliation(s)
- Joana Castro
- INIAV, IP - National Institute for Agrarian and Veterinary Research, Rua dos Lagidos, Lugar da Madalena, Vairão 4485-655, Portugal; Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal
| | - Ricardo Oliveira
- INIAV, IP - National Institute for Agrarian and Veterinary Research, Rua dos Lagidos, Lugar da Madalena, Vairão 4485-655, Portugal; LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
| | - Liliana Fernandes
- Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal; LABBELS - Associate Laboratory, Braga/Guimarães, Portugal
| | - Isabel Carvalho
- Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal; LABBELS - Associate Laboratory, Braga/Guimarães, Portugal
| | - Hugo Oliveira
- Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal; LABBELS - Associate Laboratory, Braga/Guimarães, Portugal
| | - Erik Brinks
- Department of Microbiology and Biotechnology, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Kiel, Germany
| | - Gyu-Sung Cho
- Department of Microbiology and Biotechnology, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Kiel, Germany
| | - Charles Franz
- Department of Microbiology and Biotechnology, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Kiel, Germany
| | - Carina Almeida
- INIAV, IP - National Institute for Agrarian and Veterinary Research, Rua dos Lagidos, Lugar da Madalena, Vairão 4485-655, Portugal; Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal; LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
| | - Sónia Silva
- INIAV, IP - National Institute for Agrarian and Veterinary Research, Rua dos Lagidos, Lugar da Madalena, Vairão 4485-655, Portugal; Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal; LABBELS - Associate Laboratory, Braga/Guimarães, Portugal
| | - Daniela Araújo
- INIAV, IP - National Institute for Agrarian and Veterinary Research, Rua dos Lagidos, Lugar da Madalena, Vairão 4485-655, Portugal; Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal; LABBELS - Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
69
|
Yang J, Xiong Y, Barkema HW, Tong X, Lin Y, Deng Z, Kastelic JP, Nobrega DB, Wang Y, Han B, Gao J. Comparative genomic analyses of Klebsiella pneumoniae K57 capsule serotypes isolated from bovine mastitis in China. J Dairy Sci 2024; 107:3114-3126. [PMID: 37944808 DOI: 10.3168/jds.2023-23721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/21/2023] [Indexed: 11/12/2023]
Abstract
Klebsiella pneumoniae can cause severe clinical mastitis in dairy cows, with K. pneumoniae type K57 (K57-KP) being the most common capsular serotype. To identify virulence factors and antimicrobial-resistance (AMR) genes of K57-KP with varying virulence, Galleria mellonella (greater wax moth) larvae were infected as a screening model to characterize virulence of 90 K57-KP strains, with 10 and 11 strains defined as virulent or attenuated, respectively, based on larval survival rates. Next, virulence of these 21 isolates was subsequently confirmed in adhesion and lactate dehydrogenase release assays, using bovine mammary epithelial cells cultured in vitro. Finally, genes associated with virulence and AMR were characterize with whole-genome sequencing. These 21 K57-KP strains were designated into 16 sequence types based on multi-locus sequence typing and allocated in phylogenetic analysis based on single nucleotide polymorphisms. We found great genetic diversity among isolates. In addition, adhesion-associated genes (e.g., fimA, sfaA, and focA) aminoglycoside-resistance genes (aph(6)-Id, strAB) were associated with virulence. This study provided new knowledge regarding virulence of K57-KP associated with bovine mastitis, which may inform development of novel diagnostic tools and prevention strategies for bovine mastitis.
Collapse
Affiliation(s)
- Jingyue Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yindi Xiong
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Herman W Barkema
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Xiaofang Tong
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yushan Lin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Zhaoju Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - John P Kastelic
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Diego B Nobrega
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Yue Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Bo Han
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| | - Jian Gao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
70
|
Gooch HCC, Labedan M, Hall LJ, Maxwell A. Transplanting human infant gut microbiome species into Galleria mellonella. BMC Res Notes 2024; 17:123. [PMID: 38689371 PMCID: PMC11060968 DOI: 10.1186/s13104-024-06785-w] [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: 12/20/2023] [Accepted: 04/22/2024] [Indexed: 05/02/2024] Open
Abstract
OBJECTIVE Study of the human infant gut microbiome requires the use of surrogate mammalian species such as mice. We sought to investigate the usefulness of the greater wax moth larva, Galleria mellonella, as an alternative. RESULTS We have analysed the native gut microbiome of Galleria and developed methods for clearing the native microbiome and introducing species from human infant faecal samples. We find that some species, e.g. enterococci, are more successful at recolonisation, but that others, e.g. Bifidobacterium, are less so. The work paves the way for using Galleria rather than mice in this and similar work.
Collapse
Affiliation(s)
- Harriet C C Gooch
- Department of Biochemistry and Metabolism, John Innes Centre, Norwich Research Park, Norwich, NR4 7UH, UK
| | - Marjorie Labedan
- Department of Biochemistry and Metabolism, John Innes Centre, Norwich Research Park, Norwich, NR4 7UH, UK
- Present address: Department of Ecology and Evolution, University of Lausanne, Lausanne, 1015, Switzerland
| | - Lindsay J Hall
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
- Present address: Institute of Microbiology and Infection, University of Birmingham, Birmingham, B15 2TT, UK
| | - Anthony Maxwell
- Department of Biochemistry and Metabolism, John Innes Centre, Norwich Research Park, Norwich, NR4 7UH, UK.
- Department of Molecular Microbiology, John Innes Centre, Norwich Research Park, Norwich, NR4 7UH, UK.
| |
Collapse
|
71
|
Han Y, Zhang Y, Zhang X, Huang Z, Kong J, Wang X, Chen L, Wang Y, Cao J, Zhou T, Shen M. PAM-1: an antimicrobial peptide with promise against ceftazidime-avibactam resistant Escherichia coli infection. Front Microbiol 2024; 15:1291876. [PMID: 38765679 PMCID: PMC11099939 DOI: 10.3389/fmicb.2024.1291876] [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: 09/10/2023] [Accepted: 04/10/2024] [Indexed: 05/22/2024] Open
Abstract
Introduction Antibiotic misuse and overuse have led to the emergence of carbapenem-resistant bacteria. The global spread of resistance to the novel antibiotic combination ceftazidime-avibactam (CZA) is becoming a severe problem. Antimicrobial peptide PAM-1 offers a novel approach for treating infections caused by antibiotic-resistant bacteria. This study explores its antibacterial and anti-biofilm activities and mechanisms against CZA-resistant Escherichia. Coli (E. coli), evaluating its stability and biosafety as well. Methods The broth microdilution method, growth curve analysis, crystal violet staining, scanning electron microscopy, and propidium iodide staining/N-phenyl-1-naphthylamine uptake experiments were performed to explore the antibacterial action and potential mechanism of PAM-1 against CZA-resistant E. coli. The biosafety in diverse environments of PAM-1 was evaluated by red blood cell hemolysis, and cytotoxicity tests. Its stability was further assessed under different temperatures, serum concentrations, and ionic conditions using the broth microdilution method to determine its minimum inhibitory concentration (MIC). Galleria mellonella infection model and RT-qPCR were used to investigate the in vivo antibacterial and anti-inflammatory effects. Results and discussion In vitro antibacterial experiments demonstrated that the MICs of PAM-1 ranged from 2 to 8 μg/mL, with its effectiveness sustained for a duration of 24 h. PAM-1 exhibited significant antibiofilm activities against CZA-resistant E. coli (p < 0.05). Furthermore, Membrane permeability test revealed that PAM-1 may exert its antibacterial effect by disrupting membrane integrity by forming transmembrane pores (p < 0.05). Red blood cell hemolysis and cytotoxicity tests revealed that PAM-1 exerts no adverse effects at experimental concentrations (p < 0.05). Moreover, stability tests revealed its effectiveness in serum and at room temperature. The Galleria mellonella infection model revealed that PAM-1 can significantly improve the survival rate of Galleria mellonella (>50%)for in vivo treatment. Lastly, RT-qPCR revealed that PAM-1 downregulates the expression of inflammatory cytokines (p < 0.05). Overall, our study findings highlight the potential of PAM-1 as a therapeutic agent for CZA-resistant E. coli infections, offering new avenues for research and alternative antimicrobial therapy strategies.
Collapse
Affiliation(s)
- Yijia Han
- 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, China
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Yi Zhang
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Xiaodong 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, China
| | - Zeyu Huang
- 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, China
| | - Jingchun Kong
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Xiuxiu Wang
- 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, China
| | - Lijiang 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, China
| | - Yue Wang
- 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, China
| | - Jianming Cao
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Tieli 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, China
| | - Mo Shen
- 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, China
| |
Collapse
|
72
|
Wang Y, Shi D, Zou W, Jiang Y, Wang T, Chen X, Ma C, Li W, Chen T, Burrows JF, Wang L, Zhou M. An Effective Modification Strategy to Build Multifunctional Peptides Based on a Trypsin Inhibitory Peptide of the Kunitz Family. Pharmaceutics 2024; 16:597. [PMID: 38794259 PMCID: PMC11125039 DOI: 10.3390/pharmaceutics16050597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/18/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Peptides with antimicrobial activity or protease inhibitory activity are potential candidates to supplement traditional antibiotics or cancer chemotherapies. However, the potential of many peptides are limited by drawbacks such as cytotoxicity or susceptibility to hydrolysis. Therefore, strategies to modify the structure of promising peptides may represent an effective approach for developing more promising clinical candidates. In this study, the mature peptide OSTI-1949, a Kunitz-type inhibitor from Odorrana schmackeri, and four designed analogues were successfully synthesised. In contrast to the parent peptide, the analogues showed impressive multi-functionality including antimicrobial, anticancer, and trypsin inhibitory activities. In terms of safety, there were no obvious changes observed in the haemolytic activity at the highest tested concentration, and the analogue OSTI-2461 showed an increase in activity against cancer cell lines without cytotoxicity to normal cells (HaCaT). In summary, through structural modification of a natural Kunitz-type peptide, the biological activity of analogues was improved whilst retaining low cytotoxicity. The strategy of helicity enhancement by forming an artificial α-helix and ß-sheet structure provides a promising way to develop original bioactive peptides for clinical therapeutics.
Collapse
Affiliation(s)
- Ying Wang
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (Y.W.); (W.Z.); (Y.J.); (T.W.); (C.M.); (T.C.); (J.F.B.); (L.W.); (M.Z.)
| | - Daning Shi
- Chinese Academy of Agricultural Sciences, No.12 Zhongguancun South Street, Haidian District, Beijing 100081, China;
| | - Wanchen Zou
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (Y.W.); (W.Z.); (Y.J.); (T.W.); (C.M.); (T.C.); (J.F.B.); (L.W.); (M.Z.)
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Yangyang Jiang
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (Y.W.); (W.Z.); (Y.J.); (T.W.); (C.M.); (T.C.); (J.F.B.); (L.W.); (M.Z.)
| | - Tao Wang
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (Y.W.); (W.Z.); (Y.J.); (T.W.); (C.M.); (T.C.); (J.F.B.); (L.W.); (M.Z.)
| | - Xiaoling Chen
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (Y.W.); (W.Z.); (Y.J.); (T.W.); (C.M.); (T.C.); (J.F.B.); (L.W.); (M.Z.)
| | - Chengbang Ma
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (Y.W.); (W.Z.); (Y.J.); (T.W.); (C.M.); (T.C.); (J.F.B.); (L.W.); (M.Z.)
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Tianbao Chen
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (Y.W.); (W.Z.); (Y.J.); (T.W.); (C.M.); (T.C.); (J.F.B.); (L.W.); (M.Z.)
| | - James F. Burrows
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (Y.W.); (W.Z.); (Y.J.); (T.W.); (C.M.); (T.C.); (J.F.B.); (L.W.); (M.Z.)
| | - Lei Wang
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (Y.W.); (W.Z.); (Y.J.); (T.W.); (C.M.); (T.C.); (J.F.B.); (L.W.); (M.Z.)
| | - Mei Zhou
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (Y.W.); (W.Z.); (Y.J.); (T.W.); (C.M.); (T.C.); (J.F.B.); (L.W.); (M.Z.)
| |
Collapse
|
73
|
Mediati DG, Dan W, Lalaouna D, Dinh H, Pokhrel A, Rowell KN, Michie KA, Stinear TP, Cain AK, Tree JJ. The 3' UTR of vigR is required for virulence in Staphylococcus aureus and has expanded through STAR sequence repeat insertions. Cell Rep 2024; 43:114082. [PMID: 38583155 DOI: 10.1016/j.celrep.2024.114082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 01/17/2024] [Accepted: 03/25/2024] [Indexed: 04/09/2024] Open
Abstract
Infections caused by methicillin-resistant Staphylococcus aureus (MRSA) are alarmingly common, and treatment is confined to last-line antibiotics. Vancomycin is the treatment of choice for MRSA bacteremia, and treatment failure is often associated with vancomycin-intermediate S. aureus isolates. The regulatory 3' UTR of the vigR mRNA contributes to vancomycin tolerance and upregulates the autolysin IsaA. Using MS2-affinity purification coupled with RNA sequencing, we find that the vigR 3' UTR also regulates dapE, a succinyl-diaminopimelate desuccinylase required for lysine and peptidoglycan synthesis, suggesting a broader role in controlling cell wall metabolism and vancomycin tolerance. Deletion of the 3' UTR increased virulence, while the isaA mutant is completely attenuated in a wax moth larvae model. Sequence and structural analyses of vigR indicated that the 3' UTR has expanded through the acquisition of Staphylococcus aureus repeat insertions that contribute sequence for the isaA interaction seed and may functionalize the 3' UTR.
Collapse
Affiliation(s)
- Daniel G Mediati
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia; Australian Institute for Microbiology and Infection, University of Technology Sydney, Ultimo, NSW, Australia.
| | - William Dan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - David Lalaouna
- Université de Strasbourg, CNRS, ARN UPR 9002, Strasbourg, France
| | - Hue Dinh
- School of Natural Sciences, ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW, Australia
| | - Alaska Pokhrel
- Australian Institute for Microbiology and Infection, University of Technology Sydney, Ultimo, NSW, Australia; School of Natural Sciences, ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW, Australia
| | - Keiran N Rowell
- Structural Biology Facility, University of New South Wales, Sydney, NSW, Australia
| | - Katharine A Michie
- Structural Biology Facility, University of New South Wales, Sydney, NSW, Australia
| | - Timothy P Stinear
- Department of Microbiology and Immunology, Peter Doherty Institute, University of Melbourne, Melbourne, VIC, Australia
| | - Amy K Cain
- School of Natural Sciences, ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW, Australia
| | - Jai J Tree
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
74
|
Binsuwaidan R, Almuzaini O, Mercer S, Doherty C, Mokhtar J, McBain AJ, Ledder R, Humphreys GJ. Variable effects of exposure to ionic silver in wound-associated bacterial pathogens. Lett Appl Microbiol 2024; 77:ovae030. [PMID: 38533656 DOI: 10.1093/lambio/ovae030] [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: 12/20/2023] [Revised: 02/14/2024] [Accepted: 03/25/2024] [Indexed: 03/28/2024]
Abstract
Silver compounds are used in wound dressings to reduce bioburden. Where infection is not rapidly resolved, bacteria may be exposed to sub-therapeutic concentrations of antimicrobials over prolonged periods of time. In this study, a panel of chronic wound bacteria, Pseudomonas aeruginosa (two strains), Staphylococcus aureus, and Escherichia coli, were exposed to silver nitrate on agar. Phenotypic characterization was achieved using broth microdilution sensitivity testing, a crystal violet biofilm assay, and a wax moth pathogenesis model. Repeated exposure to ionic silver did not result in planktonic phenotypic silver resistance in any of the test panels, although S. aureus demonstrated reversible increases in minimum bactericidal concentration. An ulcer-derived P. aeruginosa exhibited marked reductions in biofilm eradication concentration as well as significantly increased biofilm formation and wax moth killing when compared to the same progenitor. These changes were reversible, trending towards baseline measurements following 10 passages on silver-free media. Changes in virulence and biofilm formation in the other test bacteria were generally limited. In summary, phenotypic adaptation following exposure to ionic silver was manifested other than through changes in planktonic susceptibility. Significant changes in pseudomonas biofilm formation and sensitivity could have implications for wound care regimes and therefore warrant further investigation.
Collapse
Affiliation(s)
- Reem Binsuwaidan
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Stopford Building, M13 9PT, United Kingdom
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh 13412, Saudi Arabia
| | - Osama Almuzaini
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Stopford Building, M13 9PT, United Kingdom
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj 16278, Saudi Arabia
| | - Steven Mercer
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Stopford Building, M13 9PT, United Kingdom
| | - Christopher Doherty
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Stopford Building, M13 9PT, United Kingdom
| | - Jawahir Mokhtar
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Stopford Building, M13 9PT, United Kingdom
- Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Andrew J McBain
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Stopford Building, M13 9PT, United Kingdom
| | - Ruth Ledder
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Stopford Building, M13 9PT, United Kingdom
| | - Gavin J Humphreys
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Stopford Building, M13 9PT, United Kingdom
| |
Collapse
|
75
|
Toh YH, Lin GH. Roles of DJ41_1407 and DJ41_1408 in Acinetobacter baumannii ATCC19606 Virulence and Antibiotic Response. Int J Mol Sci 2024; 25:3862. [PMID: 38612672 PMCID: PMC11011904 DOI: 10.3390/ijms25073862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
Acinetobacter baumannii is a major cause of nosocomial infections, and its highly adaptive nature and broad range of antibiotic resistance enable it to persist in hospital environments. A. baumannii often employs two-component systems (TCSs) to regulate adaptive responses and virulence-related traits. This study describes a previously uncharacterized TCS in the A. baumannii ATCC19606 strain, consisting of a transcriptional sensor, DJ41_1407, and its regulator, DJ41_1408, located adjacent to GacA of the GacSA TCS. Markerless mutagenesis was performed to construct DJ41_1407 and DJ41_1408 single and double mutants. DJ41_1408 was found to upregulate 49 genes and downregulate 43 genes, most of which were associated with carbon metabolism and other metabolic pathways, such as benzoate degradation. MEME analysis revealed a putative binding box for DJ41_1408, 5'TGTAAATRATTAYCAWTWAT3'. Colony size, motility, biofilm-forming ability, virulence, and antibiotic resistance of DJ41_1407 and DJ41_1408 single and double mutant strains were assessed against wild type. DJ41_1407 was found to enhance motility, while DJ41_1408 was found to upregulate biofilm-forming ability, and may also modulate antibiotic response. Both DJ41_1407 and DJ41_1408 suppressed virulence, based on results from a G. mellonella infection assay. These results showcase a novel A. baumannii TCS involved in metabolism, with effects on motility, biofilm-forming ability, virulence, and antibiotic response.
Collapse
Affiliation(s)
- Yee-Huan Toh
- Master Program in Biomedical Sciences, School of Medicine, Tzu Chi University, Hualien 970374, Taiwan;
| | - Guang-Huey Lin
- Department of Microbiology and Immunology, School of Medicine, Tzu Chi University, Hualien 970374, Taiwan
- International College, Tzu Chi University, Hualien 970374, Taiwan
| |
Collapse
|
76
|
Hibbert T, Krpetic Z, Latimer J, Leighton H, McHugh R, Pottenger S, Wragg C, James CE. Antimicrobials: An update on new strategies to diversify treatment for bacterial infections. Adv Microb Physiol 2024; 84:135-241. [PMID: 38821632 DOI: 10.1016/bs.ampbs.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2024]
Abstract
Ninety-five years after Fleming's discovery of penicillin, a bounty of antibiotic compounds have been discovered, modified, or synthesised. Diversification of target sites, improved stability and altered activity spectra have enabled continued antibiotic efficacy, but overwhelming reliance and misuse has fuelled the global spread of antimicrobial resistance (AMR). An estimated 1.27 million deaths were attributable to antibiotic resistant bacteria in 2019, representing a major threat to modern medicine. Although antibiotics remain at the heart of strategies for treatment and control of bacterial diseases, the threat of AMR has reached catastrophic proportions urgently calling for fresh innovation. The last decade has been peppered with ground-breaking developments in genome sequencing, high throughput screening technologies and machine learning. These advances have opened new doors for bioprospecting for novel antimicrobials. They have also enabled more thorough exploration of complex and polymicrobial infections and interactions with the healthy microbiome. Using models of infection that more closely resemble the infection state in vivo, we are now beginning to measure the impacts of antimicrobial therapy on host/microbiota/pathogen interactions. However new approaches are needed for developing and standardising appropriate methods to measure efficacy of novel antimicrobial combinations in these contexts. A battery of promising new antimicrobials is now in various stages of development including co-administered inhibitors, phages, nanoparticles, immunotherapy, anti-biofilm and anti-virulence agents. These novel therapeutics need multidisciplinary collaboration and new ways of thinking to bring them into large scale clinical use.
Collapse
Affiliation(s)
- Tegan Hibbert
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, UK
| | - Zeljka Krpetic
- School of Science, Engineering, and Environment, University of Salford, Salford, UK
| | - Joe Latimer
- School of Science, Engineering, and Environment, University of Salford, Salford, UK
| | - Hollie Leighton
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, UK
| | - Rebecca McHugh
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Sian Pottenger
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, UK
| | - Charlotte Wragg
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, UK
| | - Chloë E James
- School of Science, Engineering, and Environment, University of Salford, Salford, UK.
| |
Collapse
|
77
|
Ramírez-Sotelo U, García-Carnero LC, Martínez-Álvarez JA, Gómez-Gaviria M, Mora-Montes HM. An ELISA-based method for Galleria mellonella apolipophorin-III quantification. PeerJ 2024; 12:e17117. [PMID: 38500532 PMCID: PMC10946395 DOI: 10.7717/peerj.17117] [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: 10/31/2023] [Accepted: 02/26/2024] [Indexed: 03/20/2024] Open
Abstract
Mammalian models, such as murine, are used widely in pathophysiological studies because they have a high degree of similarity in body temperature, metabolism, and immune response with humans. However, non-vertebrate animal models have emerged as alternative models to study the host-pathogen interaction with minimal ethical concerns. Galleria mellonella is an alternative model that has proved useful in studying the interaction of the host with either bacteria or fungi, performing drug testing, and assessing the immunological response to different microorganisms. The G. mellonella immune response includes cellular and humoral components with structural and functional similarities to the immune effectors found in higher vertebrates, such as humans. An important humoral effector stimulated during infections is apolipophorin III (apoLp-III), an opsonin characterized by its lipid and carbohydrate-binding properties that participate in lipid transport, as well as immunomodulatory activity. Despite some parameters, such as the measurement of phenoloxidase activity, melanin production, hemocytes counting, and expression of antimicrobial peptides genes are already used to assess the G. mellonella immune response to pathogens with different virulence degrees, the apoLp-III quantification remains to be a parameter to assess the immune response in this invertebrate. Here, we propose an immunological tool based on an enzyme-linked immunosorbent assay that allows apoLp-III quantification in the hemolymph of larvae challenged with pathogenic agents. We tested the system with hemolymph coming from larvae infected with Escherichia coli, Candida albicans, Sporothrix schenckii, Sporothrix globosa, and Sporothrix brasiliensis. The results revealed significantly higher concentrations of apoLp-III when each microbial species was inoculated, in comparison with untouched larvae, or inoculated with phosphate-buffered saline. We also demonstrated that the apoLp-III levels correlated with the strains' virulence, which was already reported. To our knowledge, this is one of the first attempts to quantify apoLp-III, using a quick and easy-to-use serological technique.
Collapse
|
78
|
Pereira AJ, Xing H, de Campos LJ, Seleem MA, de Oliveira KMP, Obaro SK, Conda-Sheridan M. Structure-Activity Relationship Study to Develop Peptide Amphiphiles as Species-Specific Antimicrobials. Chemistry 2024; 30:e202303986. [PMID: 38221408 PMCID: PMC10939825 DOI: 10.1002/chem.202303986] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
Antimicrobial peptide amphiphiles (PAs) are a promising class of molecules that can disrupt the bacterial membrane or act as drug nanocarriers. In this study, we prepared 33 PAs to establish supramolecular structure-activity relationships. We studied the morphology and activity of the nanostructures against different Gram-positive and Gram-negative bacterial strains (such as Staphylococcus aureus, Escherichia coli, Pseudomonas aeruginosa and Acinetobacter baumannii). Next, we used principal component analysis (PCA) to determine the key contributors to activity. We found that for S. aureus, the zeta potential was the major contributor to the activity while Gram-negative bacteria were more influenced by the partition coefficient (LogP) with the following order P. aeruginosa>E. coli>A. baumannii. We also performed a study of the mechanism of action of selected PAs on the bacterial membrane assessing the membrane permeability and depolarization, changes in zeta potential and overall integrity. We studied the toxicity of the nanostructures against mammalian cells. Finally, we performed an in vivo study using the wax moth larvae to determine the therapeutic efficacy of the active PAs. This study shows cationic PA nanostructures can be an intriguing platform for the development of nanoantibacterials.
Collapse
Affiliation(s)
- Aramis J. Pereira
- A. J. Pereira, Dr. H. Xing, L. J. de Campos, Prof. Dr. M. Conda-Sheridan, Department of Pharmaceutical Sciences, University of Nebraska Medical Center (UNMC), Omaha, NE 68198 (USA)
| | - Huihua Xing
- A. J. Pereira, Dr. H. Xing, L. J. de Campos, Prof. Dr. M. Conda-Sheridan, Department of Pharmaceutical Sciences, University of Nebraska Medical Center (UNMC), Omaha, NE 68198 (USA)
| | - Luana J. de Campos
- A. J. Pereira, Dr. H. Xing, L. J. de Campos, Prof. Dr. M. Conda-Sheridan, Department of Pharmaceutical Sciences, University of Nebraska Medical Center (UNMC), Omaha, NE 68198 (USA)
| | - Mohamed A. Seleem
- Dr. M.A. Seleem, Department of Pharmaceutical Organic Chemistry, Al-Azhar University, Cairo, 4434003 (Egypt)
| | - Kelly M. P. de Oliveira
- Prof. Dr. K. M. P. de Oliveira, Department of Biological and Environmental Science, Federal University of Grande Dourados (UFGD), Dourados, MS 79804-970 (Brazil)
| | - Stephen K. Obaro
- Prof. Dr. S. K. Obaro, Division of Pediatric Infectious Diseases, University of Alabama at Birmingham (UAB), Birmingham, AL 35233 (USA), International Foundation against Infectious Diseases in Nigeria (IFAIN), Abuja, 900108 (Nigeria)
| | - Martin Conda-Sheridan
- A. J. Pereira, Dr. H. Xing, L. J. de Campos, Prof. Dr. M. Conda-Sheridan, Department of Pharmaceutical Sciences, University of Nebraska Medical Center (UNMC), Omaha, NE 68198 (USA)
| |
Collapse
|
79
|
Giammarino A, Bellucci N, Angiolella L. Galleria mellonella as a Model for the Study of Fungal Pathogens: Advantages and Disadvantages. Pathogens 2024; 13:233. [PMID: 38535576 PMCID: PMC10976154 DOI: 10.3390/pathogens13030233] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 02/11/2025] Open
Abstract
The study of pathogenicity and virulence of fungal strains, in vivo in the preclinical phase, is carried out through the use of animal models belonging to various classes of mammals (rodents, leproids, etc.). Although animals are functionally more similar to humans, these studies have some limitations in terms of ethics (animal suffering), user-friendliness, cost-effectiveness, timing (physiological response time) and logistics (need for adequately equipped laboratories). A good in vivo model must possess some optimal characteristics to be used, such as rapid growth, small size and short life cycle. For this reason, insects, such as Galleria mellonella (Lepidoptera), Drosophila melanogaster (Diptera) and Bombyx mori (Lepidoptera), have been widely used as alternative non-mammalian models. Due to their simplicity of use and low cost, the larvae of G. mellonella represent an optimal model above all to evaluate the virulence of fungal pathogens and the use of antifungal treatments (either single or in combination with biologically active compounds). A further advantage is also represented by their simple neuronal system limiting the suffering of the animal itself, their ability to survive at near-body ambient temperatures as well as the expression of proteins able to recognise combined pathogens following the three R principles (replacement, refinement and reduction). This review aims to assess the validity as well as the advantages and disadvantages of replacing mammalian classes with G. mellonella as an in vivo study model for preclinical experimentation.
Collapse
Affiliation(s)
| | | | - Letizia Angiolella
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro 5, 00100 Rome, Italy; (A.G.); (N.B.)
| |
Collapse
|
80
|
Hofkens N, Gestels Z, Abdellati S, Gabant P, Rodriguez-Villalobos H, Martin A, Kenyon C, Manoharan-Basil SS. Protective effect of microbisporicin (NAI-107) against vancomycin resistant Enterococcus faecium infection in a Galleria mellonella model. Sci Rep 2024; 14:4786. [PMID: 38413672 PMCID: PMC10899196 DOI: 10.1038/s41598-024-55262-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/21/2024] [Indexed: 02/29/2024] Open
Abstract
Increasing antimicrobial resistance in Enterococcus faecium necessitates the search for novel treatment agents, such as bacteriocins. In this study, we conducted an in vivo assessment of five bacteriocins, namely Lacticin Z, Lacticin Q, Garvicin KS (ABC), Aureocin A53 and Microbisporicin (NAI-107), against vanB-resistant Enterococcus faecium using a Galleria mellonella model. Our in vitro experiments demonstrated the efficacy of all five bacteriocins against vanB-resistant E. faecium with only NAI-107 demonstrating in vivo efficacy. Notably, NAI-107 exhibited efficacy across a range of tested doses, with the highest efficacy observed at a concentration of 16 µg/mL. Mortality rates in the group treated with 16 µg/mL NAI-107 were lower than those observed in the linezolid-treated group. These findings strongly suggest that NAI-107 holds promise as a potential alternative therapeutic agent for treating infections caused by resistant E. faecium and warrants further investigation.
Collapse
Affiliation(s)
- Nele Hofkens
- Department of Clinical Sciences, Institute of Tropical Medicine Antwerp, 2000, Antwerp, Belgium
| | - Zina Gestels
- Department of Clinical Sciences, Institute of Tropical Medicine Antwerp, 2000, Antwerp, Belgium
| | - Saïd Abdellati
- Clinical Reference Laboratory, Department of Clinical Sciences, Institute of Tropical Medicine, 2000, Antwerp, Belgium
| | | | | | | | - Chris Kenyon
- Department of Clinical Sciences, Institute of Tropical Medicine Antwerp, 2000, Antwerp, Belgium
- Department of Medicine, University of Cape Town, Cape Town, 7700, South Africa
| | | |
Collapse
|
81
|
Han P, Lin W, Fan H, Tong Y. Characterization of phage evolution and phage resistance in drug-resistant Stenotrophomonas maltophilia. J Virol 2024; 98:e0124923. [PMID: 38189285 PMCID: PMC10878236 DOI: 10.1128/jvi.01249-23] [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: 10/23/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024] Open
Abstract
Phage therapy has become a viable antimicrobial treatment as an alternative to antibiotic treatment, with an increase in antibiotic resistance. Phage resistance is a major limitation in the therapeutic application of phages, and the lack of understanding of the dynamic changes between bacteria and phages constrains our response strategies to phage resistance. In this study, we investigated the changing trends of mutual resistance between Stenotrophomonas maltophilia (S. maltophilia) and its lytic phage, BUCT603. Our results revealed that S. maltophilia resisted phage infection through mutations in the cell membrane proteins, while the evolved phage re-infected the resistant strain primarily through mutations in structure-related proteins. Compared with the wild-type strain (SMA118), the evolved phage-resistant strain (R118-2) showed reduced virulence, weakened biofilm formation ability, and reduced resistance to aminoglycosides. In addition, the evolved phage BUCT603B1 in combination with kanamycin could inhibit the development of phage-resistant S. maltophilia in vitro and significantly improve the survival rate of S. maltophilia-infected mice. Altogether, these results suggest that in vitro characterization of bacteria-phage co-evolutionary relationships is a useful research tool to optimize phages for the treatment of drug-resistant bacterial infections.IMPORTANCEPhage therapy is a promising approach to treat infections caused by drug-resistant Stenotrophomonas maltophilia (S. maltophilia). However, the rapid development of phage resistance has hindered the therapeutic application of phages. In vitro evolutionary studies of bacteria-phage co-cultures can elucidate the mechanism of resistance development between phage and its host. In this study, we investigated the resistance trends between S. maltophilia and its phage and found that inhibition of phage adsorption is the primary strategy by which bacteria resist phage infection in vitro, while phages can re-infect bacterial cells by identifying other adsorption receptors. Although the final bacterial mutants were no longer infected by phages, they incurred a fitness cost that resulted in a significant reduction in virulence. In addition, the combination treatment with phage and aminoglycoside antibiotics could prevent the development of phage resistance in S. maltophilia in vitro. These findings contribute to increasing the understanding of the co-evolutionary relationships between phages and S. maltophilia.
Collapse
Affiliation(s)
- Pengjun Han
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Wei Lin
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Huahao Fan
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Yigang Tong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China
| |
Collapse
|
82
|
Li P, Zhang S, Wang J, Al-Shamiri MM, Luo K, Liu S, Mi P, Wu X, Liu H, Tian H, Han B, Lei J, Han S, Han L. The role of type VI secretion system genes in antibiotic resistance and virulence in Acinetobacter baumannii clinical isolates. Front Cell Infect Microbiol 2024; 14:1297818. [PMID: 38384301 PMCID: PMC10879597 DOI: 10.3389/fcimb.2024.1297818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 01/23/2024] [Indexed: 02/23/2024] Open
Abstract
Introduction The type VI secretion system (T6SS) is a crucial virulence factor in the nosocomial pathogen Acinetobacter baumannii. However, its association with drug resistance is less well known. Notably, the roles that different T6SS components play in the process of antimicrobial resistance, as well as in virulence, have not been systematically revealed. Methods The importance of three representative T6SS core genes involved in the drug resistance and virulence of A. baumannii, namely, tssB, tssD (hcp), and tssM was elucidated. Results A higher ratio of the three core genes was detected in drug-resistant strains than in susceptible strains among our 114 A. baumannii clinical isolates. Upon deletion of tssB in AB795639, increased antimicrobial resistance to cefuroxime and ceftriaxone was observed, alongside reduced resistance to gentamicin. The ΔtssD mutant showed decreased resistance to ciprofloxacin, norfloxacin, ofloxacin, tetracycline, and doxycycline, but increased resistance to tobramycin and streptomycin. The tssM-lacking mutant showed an increased sensitivity to ofloxacin, polymyxin B, and furazolidone. In addition, a significant reduction in biofilm formation was observed only with the ΔtssM mutant. Moreover, the ΔtssM strain, followed by the ΔtssD mutant, showed decreased survival in human serum, with attenuated competition with Escherichia coli and impaired lethality in Galleria mellonella. Discussion The above results suggest that T6SS plays an important role, participating in the antibiotic resistance of A. baumannii, especially in terms of intrinsic resistance. Meanwhile, tssM and tssD contribute to bacterial virulence to a greater degree, with tssM being associated with greater importance.
Collapse
Affiliation(s)
- Pu Li
- School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Sirui Zhang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Jingdan Wang
- School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Mona Mohamed Al-Shamiri
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Kai Luo
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Shuyan Liu
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Peng Mi
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
- Department of Laboratory Medicine, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Xiaokang Wu
- Department of Laboratory Medicine, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Haiping Liu
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
- Department of Laboratory Medicine, Xi’an Daxing Hospital, Xi’an, China
| | - Huohuan Tian
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Bei Han
- School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Jin’e Lei
- Department of Laboratory Medicine, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Shaoshan Han
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Lei Han
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| |
Collapse
|
83
|
Tiwari V, Sharma A, Braga R, Garcia E, Appiah R, Fleeman R, Abuaita BH, Patrauchan M, Doerrler WT. Klebsiella pneumoniae DedA family proteins have redundant roles in divalent cation homeostasis and resistance to phagocytosis. Microbiol Spectr 2024; 12:e0380723. [PMID: 38214522 PMCID: PMC10846249 DOI: 10.1128/spectrum.03807-23] [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: 11/21/2023] [Accepted: 12/04/2023] [Indexed: 01/13/2024] Open
Abstract
The DedA superfamily is a highly conserved family of membrane proteins. Deletion of Escherichia coli yqjA and yghB, encoding related DedA family proteins, results in sensitivity to elevated temperature, antibiotics, and alkaline pH. The human pathogen Klebsiella pneumoniae possesses genes encoding DedA family proteins with >90% amino acid identity to E. coli YqjA and YghB. We hypothesized that the deletion of K. pneumoniae yqjA and yghB will impact its physiology and may reduce its virulence. The K. pneumoniae ΔyqjA ΔyghB mutant (strain VT101) displayed a growth defect at 42°C and alkaline pH sensitivity, not unlike its E. coli counterpart. However, VT101 retained mostly wild-type resistance to antibiotics. We found VT101 was sensitive to the chelating agent EDTA, the anionic detergent SDS, and agents capable of alkalizing the bacterial cytoplasm such as bicarbonate or chloroquine. We could restore growth at alkaline pH and at elevated temperature by addition of 0.5-2 mM Ca2+ or Mg2+ to the culture media. VT101 displayed a slower uptake of calcium, which was dependent upon calcium channel activity. VT201, with similar deletions as VT101 but derived from a virulent K. pneumoniae strain, was highly susceptible to phagocytosis by alveolar macrophages and displayed a defect in the production of capsule. These findings suggest divalent cation homeostasis and virulence are interlinked by common functions of the DedA family.IMPORTANCEKlebsiella pneumoniae is a dangerous human pathogen. The DedA protein family is found in all bacteria and is a membrane transporter often required for virulence and antibiotic resistance. K. pneumoniae possesses homologs of E. coli YqjA and YghB, with 60% amino acid identity and redundant functions, which we have previously shown to be required for tolerance to biocides and alkaline pH. A K. pneumoniae strain lacking yqjA and yghB was found to be sensitive to alkaline pH, elevated temperature, and EDTA/SDS and displayed a defect in calcium uptake. Sensitivity to these conditions was reversed by addition of calcium or magnesium to the growth medium. Introduction of ΔyqjA and ΔyghB mutations into virulent K. pneumoniae resulted in the loss of capsule, increased phagocytosis by macrophages, and a partial loss of virulence. These results show that targeting the Klebsiella DedA family results in impaired divalent cation transport and, in turn, loss of virulence.
Collapse
Affiliation(s)
- Vijay Tiwari
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Amit Sharma
- Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Reygan Braga
- Department of Microbiology and Molecular Genetics, College of Arts and Science, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Emily Garcia
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Ridhwana Appiah
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Renee Fleeman
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Basel H. Abuaita
- Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Marianna Patrauchan
- Department of Microbiology and Molecular Genetics, College of Arts and Science, Oklahoma State University, Stillwater, Oklahoma, USA
| | - William T. Doerrler
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| |
Collapse
|
84
|
Scaccaglia M, Pinelli S, Manini L, Ghezzi B, Nicastro M, Heinrich J, Kulak N, Mozzoni P, Pelosi G, Bisceglie F. Gold(III) complexes with thiosemicarbazone ligands: insights into their cytotoxic effects on lung cancer cells. J Inorg Biochem 2024; 251:112438. [PMID: 38029536 DOI: 10.1016/j.jinorgbio.2023.112438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 12/01/2023]
Abstract
Cancer continues to pose a global threat, underscoring the urgent need for more effective and safer treatment options. Gold-based compounds have recently emerged as promising candidates due to their diverse range of biological activities. In this study, three gold(III) complexes derived from thiosemicarbazone ligands have been synthesized, fully characterized, including their X-ray crystal structures. We conducted initial mode-of-action studies on DNA and BSA, followed by a comprehensive investigation into the cytotoxic effects of these novel gold(III) complexes on lung cancer cells (A549, H2052, and H28). The results demonstrated a concentration-dependent cytotoxic response, with H28 cells exhibiting the highest sensitivity to the treatment. Furthermore, the analysis of the cell cycle revealed that these compounds induce cell cycle arrest and promote apoptosis as a response to treatment. We also observed distinct morphological changes and increased oxidative stress, contributing significantly to cell death. Notably, these complexes exhibited the ability to suppress interleukin-6 production in mesothelioma cell lines, and this highlights their anti-inflammatory potential. To gain an initial understanding of cytotoxicity on healthy cells, hemolysis tests were conducted against human blood cells, with no evidence of hemolysis. Furthermore, a toxicity assessment through the in vivo Galleria mellonella model underscored the absence of detectable toxicity. These findings prove that these complexes are promising novel therapeutic agents for lung cancer.
Collapse
Affiliation(s)
- Mirco Scaccaglia
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43124 Parma, Italy.
| | - Silvana Pinelli
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Luca Manini
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43124 Parma, Italy
| | - Benedetta Ghezzi
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy; Centro Universitario di Odontoiatria, University of Parma, Via Gramsci 14, 43126 Parma, Italy; Istituto dei Materiali per l'Elettronica ed il Magnetismo, Consiglio Nazionale delle Ricerche, Parma, Italy
| | - Maria Nicastro
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Julian Heinrich
- Institute of Chemistry, Otto-von-Guericke-Universität Magdeburg, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Nora Kulak
- Institute of Chemistry, Otto-von-Guericke-Universität Magdeburg, Universitätsplatz 2, 39106 Magdeburg, Germany; Institut für Chemie, Universität Potsdam, Karl-Liebknecht-Str. 24-25, 14476 Potsdam, Golm, Germany
| | - Paola Mozzoni
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy; CERT, Centre of Excellence for Toxicological Research, University of Parma, 43124 Parma, Italy
| | - Giorgio Pelosi
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43124 Parma, Italy; CERT, Centre of Excellence for Toxicological Research, University of Parma, 43124 Parma, Italy
| | - Franco Bisceglie
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43124 Parma, Italy; CERT, Centre of Excellence for Toxicological Research, University of Parma, 43124 Parma, Italy
| |
Collapse
|
85
|
Burlacchini G, Sandri A, Papetti A, Frosi I, Boschi F, Lleo MM, Signoretto C. Evaluation of Antibacterial and Antibiofilm Activity of Rice Husk Extract against Staphylococcus aureus. Pathogens 2024; 13:80. [PMID: 38251387 PMCID: PMC10820005 DOI: 10.3390/pathogens13010080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/11/2024] [Accepted: 01/13/2024] [Indexed: 01/23/2024] Open
Abstract
Infections caused by Staphylococcus aureus are particularly difficult to treat due to the high rate of antibiotic resistance. S. aureus also forms biofilms that reduce the effects of antibiotics and disinfectants. Therefore, new therapeutic approaches are increasingly required. In this scenario, plant waste products represent a source of bioactive molecules. In this study, we evaluated the antimicrobial and antibiofilm activity of the rice husk extract (RHE) on S. aureus clinical isolates. In a biofilm inhibition assay, high concentrations of RHE counteracted the formation of biofilm by S. aureus isolates, both methicillin-resistant (MRSA) and -sensitive (MSSA). The observation of the MRSA biofilm by confocal laser scanning microscopy using live/dead cell viability staining confirmed that the bacterial viability in the RHE-treated biofilm was reduced. However, the extract showed no or little biofilm disaggregation ability. An additive effect was observed when treating S. aureus with a combination of RHE and oxacillin/cefoxitin. In Galleria mellonella larvae treated with RHE, the extract showed no toxicity even at high concentrations. Our results support that the rice husk has antimicrobial and antibiofilm properties and could potentially be used in the future in topical solutions or on medical devices to prevent biofilm formation.
Collapse
Affiliation(s)
- Gloria Burlacchini
- Diagnostic and Public Health Department, University of Verona, 37134 Verona, Italy; (G.B.); (C.S.)
| | - Angela Sandri
- Diagnostic and Public Health Department, University of Verona, 37134 Verona, Italy; (G.B.); (C.S.)
| | - Adele Papetti
- Nutraceutical and Food Chemical-Toxicological Analysis Laboratory, Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (A.P.); (I.F.)
| | - Ilaria Frosi
- Nutraceutical and Food Chemical-Toxicological Analysis Laboratory, Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (A.P.); (I.F.)
| | - Federico Boschi
- Department of Engineering for Innovation Medicine, University of Verona, 37134 Verona, Italy;
| | - Maria M. Lleo
- Diagnostic and Public Health Department, University of Verona, 37134 Verona, Italy; (G.B.); (C.S.)
| | - Caterina Signoretto
- Diagnostic and Public Health Department, University of Verona, 37134 Verona, Italy; (G.B.); (C.S.)
| |
Collapse
|
86
|
Conn BN, Lieberman JA, Chatman P, Cotton K, Essandoh MA, Ebqa’ai M, Nelson TL, Wozniak KL. Antifungal activity of eumelanin-inspired indoylenepheyleneethynylene against Cryptococcus neoformans. Front Microbiol 2024; 14:1339303. [PMID: 38293553 PMCID: PMC10826398 DOI: 10.3389/fmicb.2023.1339303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 12/19/2023] [Indexed: 02/01/2024] Open
Abstract
Cryptococcus neoformans is an opportunistic fungal pathogen that causes meningitis in >152,000 immunocompromised individuals annually, leading to 112,000 yearly deaths. The four classes of existing antifungal agents target plasma membrane sterols (ergosterol), nucleic acid synthesis, and cell wall synthesis. Existing drugs are not highly effective against Cryptococcus, and antifungal drug resistance is an increasing problem. A novel antimicrobial compound, a eumelanin-inspired indoylenepheyleneethynylene, EIPE-1, was synthesized and has antimicrobial activity against Gram-positive bacteria, including methicillin-resistant Staphylococcus aureus (MSRA), but not towards Gram-negative organisms. Based on EIPE-1's antibacterial activity, we hypothesized that EIPE-1 could have antifungal activity. For these studies, we tested EIPE-1 against C. neoformans strain H99 and 6 additional cryptococcal clinical isolates. We examined antifungal activity, cytotoxicity, effects on fungal gene expression, and mechanism of action of EIPE-1. Results showed that EIPE-1 has fungicidal effects on seven cryptococcal strains with MICs ranging from 1.56 to 3.125 μg/mL depending on the strain, and it is non-toxic to mammalian cells. We conducted scanning and transmission electron microscopy on the exposed cells to examine structural changes to the organism following EIPE-1 treatment. Cells exposed displayed structural changes to their cell wall and membranes, with internal contents leaking out of the cells. To understand the effect of EIPE-1 on fungal gene expression, RNA sequencing was conducted. Results showed that EIPE-1 affects several processes involved stress response, ergosterol biosynthesis, capsule biosynthesis, and cell wall attachment and remodeling. Therefore, our studies demonstrate that EIPE-1 has antifungal activity against C. neoformans, which affects both cellular structure and gene expression of multiple fungal pathways involved in cell membrane stability and viability.
Collapse
Affiliation(s)
- Brittney N. Conn
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, United States
| | - Jacob A. Lieberman
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, United States
| | - Priscilla Chatman
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, United States
| | - Kaitlyn Cotton
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, United States
| | - Martha A. Essandoh
- Department of Chemistry, Oklahoma State University, Stillwater, OK, United States
| | - Mohammad Ebqa’ai
- Department of Chemistry, Oklahoma State University, Stillwater, OK, United States
| | - Toby L. Nelson
- Department of Chemistry, Oklahoma State University, Stillwater, OK, United States
| | - Karen L. Wozniak
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, United States
| |
Collapse
|
87
|
Gallorini M, Marinacci B, Pellegrini B, Cataldi A, Dindo ML, Carradori S, Grande R. Immunophenotyping of hemocytes from infected Galleria mellonella larvae as an innovative tool for immune profiling, infection studies and drug screening. Sci Rep 2024; 14:759. [PMID: 38191588 PMCID: PMC10774281 DOI: 10.1038/s41598-024-51316-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/03/2024] [Indexed: 01/10/2024] Open
Abstract
In recent years, there has been a considerable increasing interest in the use of the greater wax moth Galleria mellonella as an animal model. In vivo pharmacological tests, concerning the efficacy and the toxicity of novel compounds are typically performed in mammalian models. However, the use of the latter is costly, laborious and requires ethical approval. In this context, G. mellonella larvae can be considered a valid option due to their greater ease of use and the absence of ethical rules. Furthermore, it has been demonstrated that the immune system of these invertebrates has similarity with the one of mammals, thus guaranteeing the reliability of this in vivo model, mainly in the microbiological field. To better develop the full potential of this model, we present a novel approach to characterize the hemocyte population from G. mellonella larvae and to highlight the immuno modulation upon infection and treatments. Our approach is based on the detection in isolated hemocytes from G. mellonella hemolymph of cell membrane markers typically expressed by human immune cells upon inflammation and infection, for instance CD14, CD44, CD80, CD163 and CD200. This method highlights the analogies between G. mellonella larvae and humans. Furthermore, we provide an innovative tool to perform pre-clinical evaluations of the efficacy of antimicrobial compounds in vivo to further proceed with clinical trials and support drug discovery campaigns.
Collapse
Affiliation(s)
- Marialucia Gallorini
- Department of Pharmacy, "G. d' Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy.
| | - Beatrice Marinacci
- Department of Pharmacy, "G. d' Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- Department of Innovative Technologies in Medicine & Dentistry, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Benedetta Pellegrini
- Department of Pharmacy, "G. d' Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Amelia Cataldi
- Department of Pharmacy, "G. d' Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- UdA TechLab, "G. d' Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Maria Luisa Dindo
- Department of Agricultural and Food Sciences, University of Bologna, 40127, Bologna, Italy
| | - Simone Carradori
- Department of Pharmacy, "G. d' Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Rossella Grande
- Department of Pharmacy, "G. d' Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy.
| |
Collapse
|
88
|
Blasco L, Tomás M. Use of Galleria mellonella as an Animal Model for Studying the Antimicrobial Activity of Bacteriophages with Potential Use in Phage Therapy. Methods Mol Biol 2024; 2734:171-180. [PMID: 38066369 DOI: 10.1007/978-1-0716-3523-0_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Interest in phage therapy has increased in the last decade, and animal models have become essential in this field. The larval stage of the wax moth, Galleria mellonella, represents an easy-to-handle model. The larvae have an innate immune response and survive at 37 °C, which is ideal for infection and antimicrobial studies with bacteriophages. In this chapter, we describe the procedures used to study the antimicrobial activity of bacteriophages in a G. mellonella infection model.
Collapse
Affiliation(s)
- Lucía Blasco
- Traslational and Multidisciplinary Microbiology (MicroTM), Biomedical Research Institute A Coruña (INIBIC), Microbiology Department, Hospital A Coruña (CHUAC), University of A Coruña (UDC), A Coruña, Spain
- Study Group on Mechanisms of Action and Resistance to Antimicrobials (GEMARA), Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC), Madrid, Spain
| | - María Tomás
- Traslational and Multidisciplinary Microbiology (MicroTM), Biomedical Research Institute A Coruña (INIBIC), Microbiology Department, Hospital A Coruña (CHUAC), University of A Coruña (UDC), A Coruña, Spain.
- Study Group on Mechanisms of Action and Resistance to Antimicrobials (GEMARA), Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC), Madrid, Spain.
- MePRAM, Proyecto de Medicina de Precisión contra las resistencias Antimicrobianas, CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
89
|
Kim SJ, Shin JH, Kim H, Ko KS. Roles of crrAB two-component regulatory system in Klebsiella pneumoniae: growth yield, survival in initial colistin treatment stage, and virulence. Int J Antimicrob Agents 2024; 63:107011. [PMID: 37863340 DOI: 10.1016/j.ijantimicag.2023.107011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 10/09/2023] [Accepted: 10/14/2023] [Indexed: 10/22/2023]
Abstract
OBJECTIVES Alternation of the colistin resistance-regulating two-component regulatory system (crrAB) is a colistin-resistance mechanism in Klebsiella pneumoniae (K. pneumoniae), but its role in bacteria is not fully understood. METHODS Twelve colistin-susceptible K. pneumoniae clinical isolates were included in this study: six crrAB-positive and six crrAB-negative. We deleted the crrAB genes from two crrAB-positive isolates and complemented them. We measured the growth yields by determining growth curves in lysogeny broth and minimal media with or without Fe2+. In vitro selection rates for colistin resistance were determined by exposure to colistin, and survival rates against high concentrations of colistin (20 mg/L) at the early stage of growth (20 min) were investigated. Virulence was determined using a serum bactericidal assay and Galleria mellonella larval infection. RESULTS The presence of crrAB was not associated with colistin resistance and did not increase the in vitro selection rate of colistin resistance after exposure. The growth yield of crrAB-positive isolates was higher in lysogeny broth media and increased when Fe2+ was added to minimal media. The crrAB-positive isolates showed higher survival rates in the early stages of exposure to high colistin concentrations. Decreased serum resistance was identified in the crrAB-deleted mutants. More G. mellonella larvae survived when infected by crrAB-deleted mutants, and higher survival rates of bacteria were identified within the larvae infected with wild-type than crrAB-deletant isolates. CONCLUSION Through rapid response to external signals, crrAB would provide advantages for K. pneumoniae survival by increasing the final growth yield and initial survival against colistin treatment. This may partly contribute to the bacterial virulence.
Collapse
Affiliation(s)
- Sun Ju Kim
- Department of Microbiology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jong Hyun Shin
- Department of Microbiology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Hyunkeun Kim
- Department of Microbiology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Kwan Soo Ko
- Department of Microbiology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea.
| |
Collapse
|
90
|
Takahashi R, J-Khemlani AH, Loh JMS, Radcliff FJ, Proft T, Tsai CJY. Different Group A Streptococcus pili lead to varying proinflammatory cytokine responses and virulence. Immunol Cell Biol 2024; 102:21-33. [PMID: 37795567 DOI: 10.1111/imcb.12692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 10/06/2023]
Abstract
The human pathogen Streptococcus pyogenes, or Group A Streptococcus (GAS), is associated with a variety of diseases ranging from mild skin and soft tissue infections to invasive diseases and immune sequelae such as rheumatic heart disease. We have recently reported that one of the virulence factors of this pathogen, the pilus, has inflammatory properties and strongly stimulates the innate immune system. Here we used a range of nonpathogenic Lactococcus lactis gain-of-function mutants, each expressing one of the major pilus types of GAS, to compare the immune responses generated by various types of fully assembled pili. In vitro assays indicated variability in the inflammatory response induced by different pili, with the fibronectin-binding, collagen-binding, T antigen (FCT)-1-type pilus from GAS serotype M6/T6 inducing significantly stronger cytokine secretion than other pili. Furthermore, we established that the same trend of pili-mediated immune response could be modeled in Galleria mellonella larvae, which possess a similar innate immune system to vertebrates. Counterintuitively, across the panel of pili types examined in this study, we observed a negative correlation between the intensity of the immune response demonstrated in our experiments and the disease severity observed clinically in the GAS strains associated with each pilus type. This observation suggests that pili-mediated inflammation is more likely to promote bacterial clearance instead of causing disruptive damages that intensify pathogenesis. This also indicates that pili may not be the main contributor to the inflammatory symptoms seen in GAS diseases. Rather, the immune-potentiating properties of the pilus components could potentially be exploited as a vaccine adjuvant.
Collapse
Affiliation(s)
- Risa Takahashi
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - Adrina Hema J-Khemlani
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | - Jacelyn Mei San Loh
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Fiona Jane Radcliff
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Thomas Proft
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Catherine Jia-Yun Tsai
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
91
|
Konstantinović J, Kany AM, Alhayek A, Abdelsamie AS, Sikandar A, Voos K, Yao Y, Andreas A, Shafiei R, Loretz B, Schönauer E, Bals R, Brandstetter H, Hartmann RW, Ducho C, Lehr CM, Beisswenger C, Müller R, Rox K, Haupenthal J, Hirsch AK. Inhibitors of the Elastase LasB for the Treatment of Pseudomonas aeruginosa Lung Infections. ACS CENTRAL SCIENCE 2023; 9:2205-2215. [PMID: 38161367 PMCID: PMC10755728 DOI: 10.1021/acscentsci.3c01102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Indexed: 01/03/2024]
Abstract
Infections caused by the Gram-negative pathogen Pseudomonas aeruginosa are emerging worldwide as a major threat to human health. Conventional antibiotic monotherapy suffers from rapid resistance development, underlining urgent need for novel treatment concepts. Here, we report on a nontraditional approach to combat P. aeruginosa-derived infections by targeting its main virulence factor, the elastase LasB. We discovered a new chemical class of phosphonates with an outstanding in vitro ADMET and PK profile, auspicious activity both in vitro and in vivo. We established the mode of action through a cocrystal structure of our lead compound with LasB and in several in vitro and ex vivo models. The proof of concept of a combination of our pathoblocker with levofloxacin in a murine neutropenic lung infection model and the reduction of LasB protein levels in blood as a proof of target engagement demonstrate the great potential for use as an adjunctive treatment of lung infections in humans.
Collapse
Affiliation(s)
- Jelena Konstantinović
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)−Helmholtz
Centre for Infection Research (HZI), Saarbrücken 66123, Germany
| | - Andreas M. Kany
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)−Helmholtz
Centre for Infection Research (HZI), Saarbrücken 66123, Germany
| | - Alaa Alhayek
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)−Helmholtz
Centre for Infection Research (HZI), Saarbrücken 66123, Germany
| | - Ahmed S. Abdelsamie
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)−Helmholtz
Centre for Infection Research (HZI), Saarbrücken 66123, Germany
- Department
of Chemistry of Natural and Microbial Products, Institute of Pharmaceutical and Drug Industries Research, National
Research Centre, El-Buhouth Street, Dokki, Cairo 12622, Egypt
| | - Asfandyar Sikandar
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)−Helmholtz
Centre for Infection Research (HZI), Saarbrücken 66123, Germany
| | - Katrin Voos
- Department
of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University, Saarbrücken 66123, Germany
| | - Yiwen Yao
- Department
of Internal Medicine V − Pulmonology, Allergology and Critical
Care Medicine, Saarland University, Homburg 66421, Germany
| | - Anastasia Andreas
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)−Helmholtz
Centre for Infection Research (HZI), Saarbrücken 66123, Germany
| | - Roya Shafiei
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)−Helmholtz
Centre for Infection Research (HZI), Saarbrücken 66123, Germany
- Saarland
University, Department of Pharmacy, Saarbrücken 66123, Germany
| | - Brigitta Loretz
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)−Helmholtz
Centre for Infection Research (HZI), Saarbrücken 66123, Germany
| | - Esther Schönauer
- Department
of Biosciences and Medical Biology, Division of Structural Biology, University of Salzburg, Salzburg 5020, Austria
| | - Robert Bals
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)−Helmholtz
Centre for Infection Research (HZI), Saarbrücken 66123, Germany
- Department
of Internal Medicine V − Pulmonology, Allergology and Critical
Care Medicine, Saarland University, Homburg 66421, Germany
| | - Hans Brandstetter
- Department
of Biosciences and Medical Biology, Division of Structural Biology, University of Salzburg, Salzburg 5020, Austria
| | - Rolf W. Hartmann
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)−Helmholtz
Centre for Infection Research (HZI), Saarbrücken 66123, Germany
- Saarland
University, Department of Pharmacy, Saarbrücken 66123, Germany
| | - Christian Ducho
- Department
of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University, Saarbrücken 66123, Germany
| | - Claus-Michael Lehr
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)−Helmholtz
Centre for Infection Research (HZI), Saarbrücken 66123, Germany
- Saarland
University, Department of Pharmacy, Saarbrücken 66123, Germany
| | - Christoph Beisswenger
- Department
of Internal Medicine V − Pulmonology, Allergology and Critical
Care Medicine, Saarland University, Homburg 66421, Germany
| | - Rolf Müller
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)−Helmholtz
Centre for Infection Research (HZI), Saarbrücken 66123, Germany
- Saarland
University, Department of Pharmacy, Saarbrücken 66123, Germany
- Helmholtz
International Lab for Anti-infectives, Saarbrücken 66123, Germany
| | - Katharina Rox
- Department
of Chemical Biology (CBIO), Helmholtz Centre
for Infection Research (HZI), Braunschweig 38124, Germany
- Deutsches
Zentrum für Infektionsforschung (DZIF) e.V., Braunschweig 38124, Germany
| | - Jörg Haupenthal
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)−Helmholtz
Centre for Infection Research (HZI), Saarbrücken 66123, Germany
| | - Anna K.H. Hirsch
- Helmholtz
Institute for Pharmaceutical Research Saarland (HIPS)−Helmholtz
Centre for Infection Research (HZI), Saarbrücken 66123, Germany
- Saarland
University, Department of Pharmacy, Saarbrücken 66123, Germany
- Helmholtz
International Lab for Anti-infectives, Saarbrücken 66123, Germany
- Deutsches
Zentrum für Infektionsforschung (DZIF) e.V., Braunschweig 38124, Germany
| |
Collapse
|
92
|
Shi X, Qiu S, Ji L, Lu H, Wu S, Chen Q, Zou X, Hu Q, Feng T, Chen S, Cui W, Xu S, Jiang M, Cai R, Geng Y, Bai Q, Huang D, Liu P. Pathogenetic characterization of a Micrococcus luteus strain isolated from an infant. Front Pediatr 2023; 11:1303040. [PMID: 38188910 PMCID: PMC10770869 DOI: 10.3389/fped.2023.1303040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024] Open
Abstract
Purpose To explore the clinical characteristics of Micrococcus luteus bloodstream infection in an infant and characterize the phenotype and genotype of the isolated strains, as well as seek suitable infection models for assessing virulence. Methods Clinical data was collected from an infant patient diagnosed with M. luteus bloodstream infection. Metagenomic sequencing was performed on the isolated blood sample. The strain was isolated and underwent mass spectrometry, biochemical tests, antibiotic susceptibility assays, and whole-genome sequencing. The Galleria mellonella infection model was used to assess M. luteus virulence. Results Patient responded poorly to cephalosporins, but recovered after Linezolid treatment. Metagenomic sequencing identified M. luteus as the predominant species in the sample, confirming infection. They were identified as M. luteus with a high confidence level of 98.99% using mass spectrometry. The strain showed positive results for Catalase, Oxidase, and Urea tests, and negative results for Mannose, Xylose, Lactose, Mannitol, Arginine, and Galactose tests, consistent with the biochemical profile of M. luteus reference standards. M. luteus susceptibility to 15 antibiotics was demonstrated and no resistance genes were detected. Predicted virulence genes, including clpB, were associated with metabolic pathways and the type VI secretion system. The infection model demonstrated dose-dependent survival rates. Conclusion The infant likely developed a bloodstream infection with M. luteus due to compromised immunity. Although the isolated strain is sensitive to cephalosporin antibiotics and has low pathogenicity in infection models, clinical treatment with cephalosporins was ineffective. Linezolid proved to be effective, providing valuable guidance for future clinical management of such rare infections.
Collapse
Affiliation(s)
- Xiaolu Shi
- Department of Public Health Laboratory Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Shuxiang Qiu
- Department of Public Health Laboratory Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Liyin Ji
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Huiqun Lu
- Department of Public Health Laboratory Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Shuang Wu
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Qiongcheng Chen
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Xuan Zou
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Qinghua Hu
- Department of Public Health Laboratory Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- Shenzhen Research Center for Communicable Disease Control and Prevention, Chinese Academy of Medical Sciences, Shenzhen, China
| | - Tiejian Feng
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Shiting Chen
- School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Wenkai Cui
- Department of Public Health Laboratory Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Shiqin Xu
- School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Min Jiang
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Rui Cai
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Yijie Geng
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Qinqin Bai
- Department of Public Health Laboratory Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Dingjie Huang
- School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Peihui Liu
- Pediatric Department, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, China
| |
Collapse
|
93
|
Yao A, Liu T, Cai Y, Zhou S, Chen X, Zhou M, Ma C, Chen T, Shaw C, Wang L. Progressive Design of a Ranatuerin-2 Peptide from Amolops wuyiensis: Enhancement of Bioactivity and In Vivo Efficacy. Antibiotics (Basel) 2023; 13:5. [PMID: 38275314 PMCID: PMC10812557 DOI: 10.3390/antibiotics13010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
Antimicrobial peptides (AMPs) that exert multiple functions are considered promising candidates to combat the bacterial drug resistance crisis. Nowadays, targeted peptide modification has been widely recognised to improve biological activity and make up for deficiencies in clinical applications such as toxicity. In this study, a helix-loop peptide was isolated and identified from the skin secretion of the Wuyi torrent frog Amolops wuyiensis, namely, ranatuerin-2-AW (R2AW) (GFMDTAKNVAKNVAATLLDKLKCKITGGC). Target modifications were made to R2AW to study the structure-activity relationships and to optimise its bioactivities. Five analogues were progressively designed via residue substitution and truncation and the antibacterial and anticancer activities were evaluated. We found that the serine-substitution and cyclic-domain-deletion products showed similar antibacterial activity to the natural peptide R2AW, implying that the disulphide bridge and Rana box were dispensable for the antibacterial activity of ranatuerin-2 peptides. Notably, the cationicity- and hydrophobicity-enhanced variant, [Lys4,19, Leu20]R2AW(1-22)-NH2, exhibited significantly optimised antibacterial and anticancer activities. Additionally, it killed bacteria by membrane disruption at a highly efficient rate. Moreover, [Lys4,19, Leu20]R2AW(1-22)-NH2 exerted potential in vivo efficacy in a methicillin-resistant Staphylococcus aureus (MRSA)-infected waxworm model. Overall, this study demonstrated some rational design ideas for optimising the dual antibacterial and anticancer activities of ranatuerin-2 peptides and it proposes [Lys4,19, Leu20]R2AW(1-22)-NH2 as an appealing candidate for therapeutic development.
Collapse
Affiliation(s)
- Aifang Yao
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| | - Tianxing Liu
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (T.L.); (Y.C.); (S.Z.); (M.Z.); (C.M.); (T.C.); (C.S.); (L.W.)
| | - Yuhai Cai
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (T.L.); (Y.C.); (S.Z.); (M.Z.); (C.M.); (T.C.); (C.S.); (L.W.)
| | - Siqi Zhou
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (T.L.); (Y.C.); (S.Z.); (M.Z.); (C.M.); (T.C.); (C.S.); (L.W.)
| | - Xiaoling Chen
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (T.L.); (Y.C.); (S.Z.); (M.Z.); (C.M.); (T.C.); (C.S.); (L.W.)
| | - Mei Zhou
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (T.L.); (Y.C.); (S.Z.); (M.Z.); (C.M.); (T.C.); (C.S.); (L.W.)
| | - Chengbang Ma
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (T.L.); (Y.C.); (S.Z.); (M.Z.); (C.M.); (T.C.); (C.S.); (L.W.)
| | - Tianbao Chen
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (T.L.); (Y.C.); (S.Z.); (M.Z.); (C.M.); (T.C.); (C.S.); (L.W.)
| | - Chris Shaw
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (T.L.); (Y.C.); (S.Z.); (M.Z.); (C.M.); (T.C.); (C.S.); (L.W.)
| | - Lei Wang
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (T.L.); (Y.C.); (S.Z.); (M.Z.); (C.M.); (T.C.); (C.S.); (L.W.)
| |
Collapse
|
94
|
Feng Z, Wang L, Guan Q, Chu X, Luo ZQ. Acinetobacter baumannii coordinates central metabolism, plasmid dissemination, and virulence by sensing nutrient availability. mBio 2023; 14:e0227623. [PMID: 37855599 PMCID: PMC10746170 DOI: 10.1128/mbio.02276-23] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 09/01/2023] [Indexed: 10/20/2023] Open
Abstract
IMPORTANCE Plasmid conjugation is known to be an energy-expensive process, but our understanding of the molecular linkage between conjugation and metabolism is limited. Our finding reveals that Acinetobacter baumannii utilizes a two-component system to co-regulate metabolism, plasmid transfer, and virulence by sensing reaction intermediates of key metabolic pathways, which suggests that nutrient availability dictates not only bacterial proliferation but also horizontal gene transfer. The identification of Dot/Icm-like proteins as components of a conjugation system involved in the dissemination of antibiotic-resistance genes by A. baumannii has provided important targets for the development of agents capable of inhibiting virulence and the spread of anti-microbial-resistance genes in bacterial communities.
Collapse
Affiliation(s)
- Zhengshan Feng
- Department of Respiratory Medicine, Center of Infectious Diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, The First Hospital of Jilin University, Changchun, China
| | - Lidong Wang
- Department of Respiratory Medicine, Center of Infectious Diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, The First Hospital of Jilin University, Changchun, China
| | - Qingtian Guan
- Bioinformatics Laboratory, The First Hospital of Jilin University, Changchun, China
| | - Xiao Chu
- Department of Respiratory Medicine, Center of Infectious Diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, The First Hospital of Jilin University, Changchun, China
| | - Zhao-Qing Luo
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
95
|
Yi J, Liu C, Yang P, Wu ZC, Du CJ, Shen N. Exogenous glutathione reverses meropenem resistance in carbapenem-resistant Klebsiella pneumoniae. Front Pharmacol 2023; 14:1327230. [PMID: 38174220 PMCID: PMC10762803 DOI: 10.3389/fphar.2023.1327230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
Background: The rate of carbapenem-resistant Klebsiella pneumoniae (CRKP) infection has been increasing rapidly worldwide and, poses a significant risk to human health. Effective methods are urgently needed to address treatment failures related to antibiotic resistance. Recent research has reported that some drugs in combination with antibiotics have displayed synergistic killing of resistant bacteria. Here, we investigated whether glutathione (GSH) can synergize with meropenem, and enhance its effectiveness against CRKP. Methods: Synergistic activity was assessed by checkerboard and time-killing assays. The mechanism of these combinations was assessed by total ROS and membrane permeability assays. The bacterial metabolites were assessed by LC‒MS/MS. Results: The FICIs of GSH and meropenem were approximately 0.5 and the combined treatment with GSH and meropenem resulted in a more than 2log10 CFU/mL reduction in bacteria compared to the individual treatments. These findings indicated the synergistic effect of the two drugs. Moreover, the meropenem MIC of CRKP was reduced to less than 4 mg/L when combined with 6 mg/mL GSH, indicating that GSH could significantly reverse resistance to meropenem in bacteria. The production of ROS in bacteria was determined by flow cytometry. After adding GSH, the ROS in the GSH group and the combined group was significantly higher than that in the control and meropenem groups, but there was no significant difference between the combined and GSH groups. The metabolic disturbance caused by GSH alone and in combination with meropenem was significant intracellularly and extracellularly, especially in terms of glycerophospholipid metabolism, indicating that the synergistic effect of the combined use of GSH and meropenem was relevant to glycerophospholipid metabolism. In addition, we measured the cell membrane permeability. The cell membrane permeability of the combination group was significantly higher than that of the blank control or monotreatment groups. This confirmed that the GSH can serve as a meropenem enhancers by disturbing glycerophospholipid metabolism and increasing cell membrane permeability. Conclusion: GSH and meropenem display a synergistic effect, wherein GSH increases the sensitivity of CRKP to meropenem. The synergy and susceptibility effects are thought to related to the increased membrane permeability resulting from the perturbations in glycerophospholipid metabolism, presenting a novel avenue for CRKP treatment.
Collapse
Affiliation(s)
- Juan Yi
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Chao Liu
- Department of Infectious Disease, Peking University Third Hospital, Beijing, China
- Center of Infectious Disease, Peking University Third Hospital, Beijing, China
| | - Ping Yang
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Zhen-chao Wu
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
- Center of Infectious Disease, Peking University Third Hospital, Beijing, China
| | - Chun-jing Du
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
- Center of Infectious Disease, Peking University Third Hospital, Beijing, China
| | - Ning Shen
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
- Department of Infectious Disease, Peking University Third Hospital, Beijing, China
- Center of Infectious Disease, Peking University Third Hospital, Beijing, China
| |
Collapse
|
96
|
Bina XR, Weng Y, Budnick J, Van Allen ME, Bina JE. Klebsiella pneumoniae TolC contributes to antimicrobial resistance, exopolysaccharide production, and virulence. Infect Immun 2023; 91:e0030323. [PMID: 37982617 PMCID: PMC10715176 DOI: 10.1128/iai.00303-23] [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: 08/04/2023] [Accepted: 10/26/2023] [Indexed: 11/21/2023] Open
Abstract
Klebsiella pneumoniae is a Gram-negative bacterium that causes a variety of human diseases, ranging from pneumonia to urinary tract infections and invasive diseases. The emergence of K. pneumoniae strains that are resistant to multiple antibiotics has made treatment more complex and led to K. pneumoniae becoming a global health threat. Addressing this threat necessitates the development of new therapeutic strategies to combat this pathogen, including strategies to overcome antimicrobial resistance and therapeutics for novel targets such as antivirulence. Here, we investigated the function of TolC, an outer membrane protein essential for the function of tripartite transporters, in K. pneumoniae. Mutation of tolC rendered K. pneumoniae hypersensitive to multiple antibiotics. Moreover, the tolC mutation impaired capsule production and affected the expression of key capsule biosynthetic genes, indicating a regulatory role for TolC in capsule biosynthesis. Additionally, TolC was essential for growth under iron-limiting conditions, suggesting its involvement in iron acquisition. The tolC mutant exhibited increased adherence to human enterocytes and enhanced serum sensitivity. In the Galleria mellonella infection model, the tolC mutant displayed reduced virulence compared to the wild type. Our findings highlight the pleiotropic role of TolC in K. pneumoniae pathobiology, influencing antimicrobial resistance, capsule production, iron homeostasis, adherence to host cells, and virulence. Understanding the multifaceted role of TolC in K. pneumoniae may guide the development of new therapeutic strategies against this pathogen. .
Collapse
Affiliation(s)
- X. Renee Bina
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yuding Weng
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - James Budnick
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mia E. Van Allen
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - James E. Bina
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
97
|
Hofkens N, Gestels Z, Abdellati S, De Baetselier I, Gabant P, Martin A, Kenyon C, Manoharan-Basil SS. Microbisporicin (NAI-107) protects Galleria mellonella from infection with Neisseria gonorrhoeae. Microbiol Spectr 2023; 11:e0282523. [PMID: 37823634 PMCID: PMC10715042 DOI: 10.1128/spectrum.02825-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/06/2023] [Indexed: 10/13/2023] Open
Abstract
IMPORTANCE We screened 66 bacteriocins to see if they exhibited anti-gonococcal activity. We found 12 bacteriocins with anti-gonococcal effects, and 4 bacteriocins showed higher anti-gonococcal activity. Three bacteriocins, lacticin Z, lacticin Q, and Garvicin KS (ABC), showed in vitro anti-gonococcal activity but no in vivo inhibitory effects against the Neisseria gonorrhoeae (WHO-P) isolate. On the other hand, NAI-107 showed in vivo anti-gonococcal activity. The findings suggest that NAI-107 is a promising alternative to treat gonorrhea infections.
Collapse
Affiliation(s)
- Nele Hofkens
- Department of Clinical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Zina Gestels
- Department of Clinical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Said Abdellati
- Clinical Reference Laboratory, Department of Clinical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Irith De Baetselier
- Clinical Reference Laboratory, Department of Clinical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | | | | | - Christopher Kenyon
- Department of Clinical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
- Department of Medicine, University of Cape Town, Cape Town, South Africa
| | | |
Collapse
|
98
|
Simanek KA, Schumacher ML, Mallery CP, Shen S, Li L, Paczkowski JE. Quorum-sensing synthase mutations re-calibrate autoinducer concentrations in clinical isolates of Pseudomonas aeruginosa to enhance pathogenesis. Nat Commun 2023; 14:7986. [PMID: 38042853 PMCID: PMC10693556 DOI: 10.1038/s41467-023-43702-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 11/17/2023] [Indexed: 12/04/2023] Open
Abstract
Quorum sensing is a mechanism of bacterial communication that controls virulence gene expression. Pseudomonas aeruginosa regulates virulence via two synthase/transcription factor receptor pairs: LasI/R and RhlI/R. LasR is considered the master transcriptional regulator of quorum sensing, as it upregulates rhlI/R. However, clinical isolates often have inactivating mutations in lasR, while maintaining Rhl-dependent signaling. We sought to understand how quorum sensing progresses in isolates with lasR mutations, specifically via activation of RhlR. We find that clinical isolates with lasR inactivating mutations often harbor concurrent mutations in rhlI. Using ultra-high-performance liquid chromatography coupled with high-resolution mass spectrometry, we discover that strains lacking lasR overproduce the RhlI-synthesized autoinducer and that RhlI variants re-calibrate autoinducer concentrations to wild-type levels, restoring virulent phenotypes. These findings provide a mechanism for the plasticity of quorum sensing progression in an acute infection niche.
Collapse
Affiliation(s)
- Kayla A Simanek
- Department of Biomedical Sciences, University at Albany, School of Public Health, Albany, New York, 12201, USA
| | - Megan L Schumacher
- Department of Biomedical Sciences, University at Albany, School of Public Health, Albany, New York, 12201, USA
| | - Caleb P Mallery
- Department of Biomedical Sciences, University at Albany, School of Public Health, Albany, New York, 12201, USA
| | - Stella Shen
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, New York, 12208, USA
| | - Lingyun Li
- Division of Environmental Health Sciences, Wadsworth Center, New York State Department of Health, Albany, New York, 12208, USA
| | - Jon E Paczkowski
- Department of Biomedical Sciences, University at Albany, School of Public Health, Albany, New York, 12201, USA.
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, New York, 12208, USA.
| |
Collapse
|
99
|
Alves LA, Naveed H, Franco EM, Garcia MT, Freitas VA, Junqueira JC, Bastos DC, Araujo TLS, Chen T, Mattos-Graner RO. PepO and CppA modulate Streptococcus sanguinis susceptibility to complement immunity and virulence. Virulence 2023; 14:2239519. [PMID: 37563831 PMCID: PMC10424592 DOI: 10.1080/21505594.2023.2239519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/12/2023] [Accepted: 07/15/2023] [Indexed: 08/12/2023] Open
Abstract
Streptococcus sanguinis is a ubiquitous commensal species of the oral cavity commonly involved as an opportunistic pathogen in cardiovascular infections. In this study, we investigated the functions of endopeptidase O (PepO) and a C3-degrading protease (CppA) in the systemic virulence of S. sanguinis. Isogenic mutants of pepO and cppA obtained in strain SK36 showed increased susceptibility to C3b deposition and to opsonophagocytosis by human polymorphonuclear neutrophils (PMN). These mutants differ, however, in their profiles of binding to serum amyloid P component (SAP) and C1q, whereas both showed reduced interaction with C4b-binding protein (C4BP) and/or factor H (FH) regulators as compared to SK36. The two mutants showed defects in ex vivo persistence in human blood, serum-mediated invasion of HCAEC endothelial cells, and virulence in a Galleria mellonella infection model. The transcriptional activities of pepO and cppA, assessed by RT-qPCR in nine wild-type strains, further indicated strain-specific profiles of pepO/cppA expression. Moreover, non-conserved amino acid substitutions were detected among the strains, mostly in CppA. Phylogenetic comparisons with homologues of streptococcal species of the oral and oropharyngeal sites suggested that S. sanguinis PepO and CppA have independent ancestralities. Thus, this study showed that PepO and CppA are complement evasion proteins expressed by S. sanguinis in a strain-specific manner, which are required for multiple functions associated with cardiovascular virulence.
Collapse
Affiliation(s)
- Lívia A. Alves
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, SP, Brazil
| | - Hassan Naveed
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, SP, Brazil
| | - Eduardo M. Franco
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, SP, Brazil
| | - Maíra Terra Garcia
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos, SP, Brazil
| | - Victor A. Freitas
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, SP, Brazil
| | - Juliana C. Junqueira
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos, SP, Brazil
| | - Débora C. Bastos
- Department of Biosciences, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, SP, Brazil
- Department of Cell Biology, São Leopoldo Mandic Medical School, Campinas, SP, Brazil
| | - Thaís L. S. Araujo
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Tsute Chen
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, USA
| | - Renata O. Mattos-Graner
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, SP, Brazil
| |
Collapse
|
100
|
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
|