1
|
Yuan S, Liu B, Quan Y, Gao S, Zuo J, Jin W, Shen Y, Li Y, Wang Y, Wang Y. Streptococcus suis regulates central carbon fluxes in response to environment to balance drug resistance and virulence. Microbiol Res 2025; 296:128157. [PMID: 40174362 DOI: 10.1016/j.micres.2025.128157] [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: 10/01/2024] [Revised: 02/28/2025] [Accepted: 03/25/2025] [Indexed: 04/04/2025]
Abstract
Streptococcus suis, a zoonotic pathogen, must adapt to the distinct nutritional environment of the host microhabitat during infection and the establishment of invasive disease, primarily by modulating its metabolic pathways. Metabolic plasticity endows S. suis with an enhanced capacity for environmental adaptation. Multidrug-resistant S. suis is increasingly prevalent due to the extensive use of antibiotics in swine production. In this study, an environment-dependent evolutionary model demonstrated that S. suis could modulate its metabolism in response to environmental changes, thereby altering its drug resistance and virulence. The central carbon flux regulated by pyruvate dehydrogenase (PDH) was identified as a pivotal node in balancing drug resistance and virulence in S. suis. Within the in vivo host environment, increased carbon flux through PDH enhances the production of capsular polysaccharide (CPS), thereby improving immune evasion. Conversely, in the antibiotic environment, reduced carbon flux through PDH downregulates the bacterial metabolic state, which diminishes the induction of toxic metabolites by antibiotics, thereby augmenting drug resistance. This concept provides a reasonable explanation for the puzzling phenomena observed with S. suis in clinical settings. For instance, antibiotic-resistant S. suis has a survival advantage in pig farms where antibiotics are frequently used but is less frequently associated with invasive infections. Furthermore, this study demonstrates that exogenous pyruvate can enhance the bactericidal effect of gentamicin against clinically multidrug-resistant S. suis, offering new insights and potential strategies for controlling clinical multidrug-resistant S. suis infections.
Collapse
Affiliation(s)
- Shuo Yuan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China.
| | - Baobao Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China.
| | - Yingying Quan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China.
| | - Shuji Gao
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China.
| | - Jing Zuo
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China; College of Life Sciences, Sichuan University, Chengdu 610000, China.
| | - Wenjie Jin
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China.
| | - Yamin Shen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China.
| | - Yue Li
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China.
| | - Yuxin Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China.
| | - Yang Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang 471003, China.
| |
Collapse
|
2
|
Xiao J, Wang Y, Xu X, Zhou H. An auto-excision system for rapid and efficient genetic manipulation in Glaesserella parasuis. Microbiol Res 2025; 298:128235. [PMID: 40413914 DOI: 10.1016/j.micres.2025.128235] [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: 12/16/2024] [Revised: 05/20/2025] [Accepted: 05/20/2025] [Indexed: 05/27/2025]
Abstract
Site-specific recombination systems are widely used in bacterial gene editing due to their precision and efficiency. However, traditional gene editing methods often require labor-intensive plasmid construction and multiple transformation steps, which can be time-consuming and inefficient. In this study, we developed an Auto-Excision (AE) system that overcomes these limitations by optimizing the entire process-from the preparation of targeting sequences to the screening of marker-free mutants. The AE system simplifies the knockout process by eliminating the need to construct targeting plasmids for each target gene, requiring only a single transformation, and allowing for the direct selection of markerless mutants in the presence of antibiotics. We validated the AE system's ability to enable rapid and efficient gene knockout in Glaesserella parasuis (G. parasuis), demonstrating its potential as a rapid and labor-efficient gene manipulation tool. This method reduces the overall timeline to as little as one day, with a hands-on time of less than one hour, while achieving a knockout efficiency greater than 90 %. Additionally, the system successfully performed multi-gene knockouts, targeting five genes in succession. This approach offers substantial time and labor savings, with the entire process achievable within a single bacterial colony growth cycle. This positions the AE system as a rapid bacterial genetic manipulation method currently known, with broad potential applications across diverse bacterial species.
Collapse
Affiliation(s)
- Jing Xiao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Yuxin Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaojuan Xu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China.
| | - Hongbo Zhou
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, Hubei, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China.
| |
Collapse
|
3
|
Suh K, Lee Y, Baek S, Kim J, Seo J, Yang YH, Kim JS, Lee W, Seo EK, Kim W. Bakuchiol kills Staphylococcus aureus persisters and potentiates colistin activity against Acinetobacter baumannii persisters. Front Pharmacol 2025; 16:1592183. [PMID: 40444041 PMCID: PMC12119637 DOI: 10.3389/fphar.2025.1592183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Accepted: 04/28/2025] [Indexed: 06/02/2025] Open
Abstract
Infections associated with bacterial persisters are challenging to cure because they can evade antibiotics and regrow, often resulting in relapse. Current antibiotics are not optimized to target persisters, highlighting the urgent need for new therapeutics. Here, we report that bakuchiol, a plant-derived natural product, exhibits anti-persister and adjuvant properties. Bakuchiol eradicates persisters formed by the gram-positive bacterium Staphylococcus aureus at 8 μg/mL and, in combination with 1 μg/mL colistin, completely eliminates persisters formed by the gram-negative bacterium Acinetobacter baumannii. Mechanistic analyses revealed that bakuchiol selectively disrupted bacterial membrane phospholipids while sparing mammalian membranes and exhibited low cytotoxicity. In Acinetobacter baumannii persisters, bakuchiol likely damages phospholipid patches in the outer membrane, causing nominal lethality but facilitating membrane permeabilization. This activity synergizes with colistin, which targets the lipooligosaccharide layer, resulting in the mutual reinforcement of their bactericidal effects. These findings highlight the potential of dual glycolipid-phospholipid targeting as a strategy to combat gram-negative persisters and highlight natural products as valuable sources for anti-persister therapeutics with membrane selectivity.
Collapse
Affiliation(s)
- Kayeong Suh
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Yeju Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Seongeun Baek
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Jin Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Jinbeom Seo
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yung-Hun Yang
- Advanced Materials Program, Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, Republic of Korea
| | - Jae-Seok Kim
- Department of Laboratory Medicine, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Wonsik Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Eun Kyoung Seo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Wooseong Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
4
|
Patton S, Silva DP, Fuques E, Klinges G, Muller EM, Thurber RLV. Antibiotic type and dose variably affect microbiomes of a disease-resistant Acropora cervicornis genotype. ENVIRONMENTAL MICROBIOME 2025; 20:46. [PMID: 40317056 PMCID: PMC12049008 DOI: 10.1186/s40793-025-00709-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 04/16/2025] [Indexed: 05/04/2025]
Abstract
BACKGROUND As coral diseases become more prevalent and frequent, the need for new intervention strategies also increases to counteract the rapid spread of disease. Recent advances in coral disease mitigation have resulted in increased use of antibiotics on reefs, as their application may halt disease lesion progression. Although efficacious, consequences of deliberate microbiome manipulation resulting from antibiotic administration are less well-understood- especially in non-diseased corals that appear visually healthy. Therefore, to understand how apparently healthy corals are affected by antibiotics, we investigated how three individual antibiotics, and a mixture of the three, impact the microbiome structure and diversity of a disease-resistant Caribbean staghorn coral (Acropora cervicornis) genotype. Over a 96-hour, aquarium-based antibiotic exposure experiment, we collected and processed coral tissue and water samples for 16S rRNA gene analysis. RESULTS We found that antibiotic type and dose distinctively impact microbiome alpha diversity, beta diversity, and community composition. In experimental controls, microbiome composition was dominated by an unclassified bacterial taxon from the order Campylobacterales, while each antibiotic treatment significantly reduced the relative abundance of this taxon. Those taxa that persisted following antibiotic treatment largely differed by antibiotic type and dose, thereby indicating that antibiotic treatment may result in varying potential for opportunist establishment. CONCLUSION Together, these data suggest that antibiotics induce microbiome dysbiosis- hallmarked by the loss of a dominant bacterium and the increase in taxa associated with coral stress responses. Understanding the off-target consequences of antibiotic administration is critical not only for informed, long-term coral restoration practices, but also for highlighting the importance of responsible antibiotic dissemination into natural environments.
Collapse
Affiliation(s)
- Sunni Patton
- Ecology, Evolution, and Marine Biology, University of California, Santa Barbara, Santa Barbara, CA, 93106-9620, USA.
- Department of Microbiology, Oregon State University, 226 Nash Hall, Corvallis, OR, 97331, USA.
| | - Denise P Silva
- Ecology, Evolution, and Marine Biology, University of California, Santa Barbara, Santa Barbara, CA, 93106-9620, USA
- Department of Microbiology, Oregon State University, 226 Nash Hall, Corvallis, OR, 97331, USA
| | - Eddie Fuques
- Ecology, Evolution, and Marine Biology, University of California, Santa Barbara, Santa Barbara, CA, 93106-9620, USA
- Department of Microbiology, Oregon State University, 226 Nash Hall, Corvallis, OR, 97331, USA
| | - Grace Klinges
- Center for Global Discovery and Conservation Science, Arizona State University, Hilo, HI, 96720, USA
- Mote Marine Laboratory, 1600 Ken Thompson Pkwy, Sarasota, FL, 34236, USA
- Mote Marine Laboratory International Center for Coral Reef Research and Restoration, 24244 Overseas Hwy, Summerland Key, FL, 33042, USA
| | - Erinn M Muller
- Mote Marine Laboratory, 1600 Ken Thompson Pkwy, Sarasota, FL, 34236, USA
- Mote Marine Laboratory International Center for Coral Reef Research and Restoration, 24244 Overseas Hwy, Summerland Key, FL, 33042, USA
| | - Rebecca L Vega Thurber
- Ecology, Evolution, and Marine Biology, University of California, Santa Barbara, Santa Barbara, CA, 93106-9620, USA
- Department of Microbiology, Oregon State University, 226 Nash Hall, Corvallis, OR, 97331, USA
| |
Collapse
|
5
|
Kaplan E, Chaloin L, Guichou J, Berrou K, Rahimova R, Labesse G, Lionne C. APH Inhibitors that Reverse Aminoglycoside Resistance in Enterococcus casseliflavus. ChemMedChem 2025; 20:e202400842. [PMID: 39801466 PMCID: PMC12005471 DOI: 10.1002/cmdc.202400842] [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: 10/24/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/28/2025]
Abstract
Aminoglycoside-phosphotransferases (APHs) are a class of bacterial enzymes that mediate acquired resistance to aminoglycoside antibiotics. Here we report the identification of small molecules counteracting aminoglycoside resistance in Enterococcus casseliflavus. Molecular dynamics simulations were performed to identify an allosteric pocket in three APH enzymes belonging to 3' and 2'' subfamilies in which we then screened, in silico, 12,000 small molecules. From a subset of only 14 high-scored molecules tested in vitro, we identified a compound, named here EK3, able to non-competitively inhibit the APH(2'')-IVa, an enzyme mediating clinical gentamicin resistance. Structure-activity relationship (SAR) exploration of this hit compound allowed us to identify a molecule with improved enzymatic inhibition. By measuring bacterial sensitivity, we found that the three best compounds in this series restored bactericidal activity of various aminoglycosides, including gentamicin, without exhibiting toxicity to HeLa cells. This work not only provides a basis to fight aminoglycoside resistance but also highlights a proof-of-concept for the search of allosteric modulators by using in silico methods.
Collapse
Affiliation(s)
- Elise Kaplan
- Institut de Recherche en Infectiologie de Montpellier – IRIMUniversity of MontpellierCNRS UMR 90041919 route de Mende34293Montpellier cedex 5France
- Current address: University of LyonCNRS, UMR5086, Molecular Microbiology and Structural Biochemistry, IBCP7 Passage du Vercors69367LyonFrance
| | - Laurent Chaloin
- Institut de Recherche en Infectiologie de Montpellier – IRIMUniversity of MontpellierCNRS UMR 90041919 route de Mende34293Montpellier cedex 5France
| | - Jean‐François Guichou
- Centre de Biologie Structurale – CBSUniversity of MontpellierCNRS UMR 5048INSERM U 105429 rue de Navacelles34090MontpellierFrance
| | - Kévin Berrou
- Institut de Recherche en Infectiologie de Montpellier – IRIMUniversity of MontpellierCNRS UMR 90041919 route de Mende34293Montpellier cedex 5France
| | - Rahila Rahimova
- Centre de Biologie Structurale – CBSUniversity of MontpellierCNRS UMR 5048INSERM U 105429 rue de Navacelles34090MontpellierFrance
- Current address: University of Grenoble Alpes, CEA, CNRS, IBS, Metalloproteins Unit, 71 avenue des MartyrsCS 10090, 38000GrenobleFrance
| | - Gilles Labesse
- Centre de Biologie Structurale – CBSUniversity of MontpellierCNRS UMR 5048INSERM U 105429 rue de Navacelles34090MontpellierFrance
| | - Corinne Lionne
- Institut de Recherche en Infectiologie de Montpellier – IRIMUniversity of MontpellierCNRS UMR 90041919 route de Mende34293Montpellier cedex 5France
- Centre de Biologie Structurale – CBSUniversity of MontpellierCNRS UMR 5048INSERM U 105429 rue de Navacelles34090MontpellierFrance
- Current address: University of Montpellier, CNRS UMR 5048, INSERM U 1054, CBS, 29 rue de Navacelles34090MontpellierFrance
| |
Collapse
|
6
|
Li X, Zhao J, Liu X, Song Z, Xu W, Li Z. Probing the interaction mechanism of tigecycline with γ-globulin and hemoglobin in the absence and presence of amikacin. Int J Biol Macromol 2025; 291:139109. [PMID: 39722387 DOI: 10.1016/j.ijbiomac.2024.139109] [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: 10/21/2024] [Revised: 12/02/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024]
Abstract
The interaction mechanism of tigecycline with γ-globulin and hemoglobin in the absence and presence of amikacin was investigated through multipectral, molecular docking and molecular dynamics simulation. The results show that tigecycline and γ-globulin/hemoglobin forms a ground state complex without or with amikacin. The presence of amikacin slightly increases the binding constant of tigecycline to γ-globulin/hemoglobin, but all are of moderate binding affinity, at 104 L mol-1. The equilibrium fraction of unbound tigecycline fu is >90 %, but the presence of amikacin reduces the free concentration of tigecycline in γ-globulin and hemoglobin. Whether amikacin is present or not, the interaction between tigecycline and γ-globulin/hemoglobin is a synergistic interaction driven by enthalpy and entropy. Non-covalent forces are primarily hydrophobic interactions, but also include electrostatic forces and hydrogen bonds. In the presence of amikacin, the effect of tigecycline on the skeleton structure of γ-globulin/hemoglobin is more significant. The effect of tigecycline and/or amikacin on the secondary structure of γ-globulin/hemoglobin is not significant, while the secondary structure changes in different systems are not the same. Molecular docking shows that γ-globulin/hemoglobin-tigecycline (first)-amikacin ternary system is the most stable. Molecular dynamics simulation explores the stability and dynamic behavior of γ-globulin/hemoglobin-tigecycline complex without or with amikacin.
Collapse
Affiliation(s)
- Xiangrong Li
- Department of Medical Chemistry, Xinxiang Engineering Technology Research Center of Functional Medical Nanomaterials, Key Laboratory of Medical Molecular Probes, School of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan 453003, PR China.
| | - Jingjing Zhao
- Department of Medical Chemistry, Xinxiang Engineering Technology Research Center of Functional Medical Nanomaterials, Key Laboratory of Medical Molecular Probes, School of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan 453003, PR China; Clinical Pharmacy Room, Pharmacy Department, Xinmi Hospital of Traditional Chinese Medicine, Xinmi, Henan 452370, PR China
| | - Xianfei Liu
- Department of Medical Chemistry, Xinxiang Engineering Technology Research Center of Functional Medical Nanomaterials, Key Laboratory of Medical Molecular Probes, School of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan 453003, PR China; School of Life Science, Ludong University, Yantai, Shandong 264025, PR China
| | - Zhizhi Song
- Grade 2020, Clinical Medicine, School of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan 453003, PR China
| | - Wanqing Xu
- Grade 2022, Clinical Medicine, School of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan 453003, PR China
| | - Zheng Li
- Grade 2022, Clinical Medicine, School of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan 453003, PR China
| |
Collapse
|
7
|
Dubois E, Spasovski V, Plésiat P, Llanes C. Role of the two-component system AmgRS in early resistance of Pseudomonas aeruginosa to cinnamaldehyde. Microbiol Spectr 2025; 13:e0169924. [PMID: 39656006 PMCID: PMC11705830 DOI: 10.1128/spectrum.01699-24] [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: 07/24/2024] [Accepted: 11/09/2024] [Indexed: 01/11/2025] Open
Abstract
Exposure of Pseudomonas aeruginosa to cinnamaldehyde (CNA), a natural electrophilic antimicrobial often used as self-medication to treat mild infections, triggers overproduction of the MexAB-OprM efflux system, leading to multidrug resistance. In this study, we demonstrate that CNA exposure induces expression of genes regulated by the two-component system AmgRS. AmgRS activates MexAB-OprM production, independent of repressors MexR and NalD. In addition to the essential role played by the NalC-ArmR pathway in this adaptive process, AmgRS is critical for the survival of P. aeruginosa challenged with CNA. Altogether, these data suggest that efflux-dependent and -independent mechanisms are activated in the early phase of CNA exposure, allowing for progressive enzymatic reduction of the biocide to non-toxic cinnamic alcohol.IMPORTANCEExposure of Pseudomonas aeruginosa to cinnamaldehyde (CNA), an antimicrobial used in self-medication, induces overproduction of the MexAB-OprM efflux system, leading to multidrug resistance. Our study demonstrates that the AmgRS two-component system aids in the survival of P. aeruginosa strain PA14 under CNA exposure through both MexAB-OprM-dependent and -independent mechanisms until the enzymatic reduction of CNA into the less toxic cinnamic alcohol. This discovery highlights the pivotal role of AmgRS in mediating defense against aldehyde biocides, emphasizing its significance in the persistence of P. aeruginosa, a pathogen associated with hospital-acquired infections and cystic fibrosis, and underscores the potential impact on clinical treatment strategies.
Collapse
Affiliation(s)
- Eline Dubois
- UMR CNRS 6249 Chrono-Environnement, UFR Santé, Université Bourgogne Franche-Comté, Besançon, France
| | - Vladimir Spasovski
- UMR CNRS 6249 Chrono-Environnement, UFR Santé, Université Bourgogne Franche-Comté, Besançon, France
| | - Patrick Plésiat
- UMR CNRS 6249 Chrono-Environnement, UFR Santé, Université Bourgogne Franche-Comté, Besançon, France
| | - Catherine Llanes
- UMR CNRS 6249 Chrono-Environnement, UFR Santé, Université Bourgogne Franche-Comté, Besançon, France
| |
Collapse
|
8
|
Choi AJ, Bennison DJ, Kulkarni E, Azar H, Sun H, Li H, Bradshaw J, Yeap HW, Lim N, Mishra V, Crespo-Puig A, Mills EA, Davies F, Sriskandan S, Shenoy AR. Aminoglycoside heteroresistance in Enterobacter cloacae is driven by the cell envelope stress response. mBio 2024; 15:e0169924. [PMID: 39475244 PMCID: PMC11633387 DOI: 10.1128/mbio.01699-24] [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: 06/06/2024] [Accepted: 10/02/2024] [Indexed: 12/12/2024] Open
Abstract
Enterobacter cloacae is a Gram-negative nosocomial pathogen of the ESKAPE (Enterococcus, Staphylococcus, Klebsiella, Acinetobacter, Pseudomonas, and Enterobacter spp.) priority group with increasing multi-drug resistance via the acquisition of resistance plasmids. However, E. cloacae can also display forms of antibiotic refractoriness, such as heteroresistance and tolerance. Here, we report that E. cloacae displays transient heteroresistance to aminoglycosides, which is accompanied with the formation of small colony variants (SCVs) with increased minimum inhibitor concentration (MIC) of gentamicin and other aminoglycosides used in the clinic, but not other antibiotic classes. To explore the underlying mechanisms, we performed RNA sequencing of heteroresistant bacteria, which revealed global gene expression changes and a signature of the CpxRA cell envelope stress response. Deletion of the cpxRA two-component system abrogated aminoglycoside heteroresistance and SCV formation, pointing to its indispensable role in these processes. The introduction of a constitutively active allele of cpxA led to high aminoglycoside MICs, consistent with cell envelope stress response driving these behaviors in E. cloacae. Cell envelope stress can be caused by environmental cues, including heavy metals. Indeed, bacterial exposure to copper increased gentamicin MIC in the wild-type but not in the ΔcpxRA mutant. Moreover, copper exposure also elevated the gentamicin MICs of clinical isolates from bloodstream infections, suggesting that CpxRA- and copper-dependent aminoglycoside resistance is broadly conserved in E. cloacae strains. Altogether, we establish that E. cloacae relies on transcriptional reprogramming via the envelope stress response pathway for transient resistance to a major class of frontline antibiotic.IMPORTANCEEnterobacter cloacae is a bacterium that belongs to the WHO high-priority group and an increasing threat worldwide due its multi-drug resistance. E. cloacae can also display heteroresistance, which has been linked to treatment failure. We report that E. cloacae shows heteroresistance to aminoglycoside antibiotics. These are important frontline microbicidal drugs used against Gram-negative bacterial infections; therefore, understanding how resistance develops among sensitive strains is important. We show that aminoglycoside resistance is driven by the activation of the cell envelope stress response and transcriptional reprogramming via the CpxRA two-component system. Furthermore, heterologous activation of envelope stress via copper, typically a heavy metal with antimicrobial actions, also increased aminoglycoside MICs of the E. cloacae type strain and clinical strains isolated from bloodstream infections. Our study suggests aminoglycoside recalcitrance in E. cloacae could be broadly conserved and cautions against the undesirable effects of copper.
Collapse
Affiliation(s)
- Ana J. Choi
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Daniel J. Bennison
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Esha Kulkarni
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Hibah Azar
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Haoyu Sun
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Hanqi Li
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Jonathan Bradshaw
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Hui Wen Yeap
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Nicholas Lim
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Vishwas Mishra
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Anna Crespo-Puig
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Ewurabena A. Mills
- NIHR Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, Imperial College London, London, United Kingdom
| | - Frances Davies
- NIHR Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, Imperial College London, London, United Kingdom
| | - Shiranee Sriskandan
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
- NIHR Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, Imperial College London, London, United Kingdom
| | - Avinash R. Shenoy
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
9
|
Shankar G, Akhter Y. Stealing survival: Iron acquisition strategies of Mycobacteriumtuberculosis. Biochimie 2024; 227:37-60. [PMID: 38901792 DOI: 10.1016/j.biochi.2024.06.006] [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: 05/06/2024] [Revised: 06/07/2024] [Accepted: 06/18/2024] [Indexed: 06/22/2024]
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), faces iron scarcity within the host due to immune defenses. This review explores the importance of iron for Mtb and its strategies to overcome iron restriction. We discuss how the host limits iron as an innate immune response and how Mtb utilizes various iron acquisition systems, particularly the siderophore-mediated pathway. The review illustrates the structure and biosynthesis of mycobactin, a key siderophore in Mtb, and the regulation of its production. We explore the potential of targeting siderophore biosynthesis and uptake as a novel therapeutic approach for TB. Finally, we summarize current knowledge on Mtb's iron acquisition and highlight promising directions for future research to exploit this pathway for developing new TB interventions.
Collapse
Affiliation(s)
- Gauri Shankar
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh, 226 025, India
| | - Yusuf Akhter
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh, 226 025, India.
| |
Collapse
|
10
|
Chatupale V, Pohnerkar J. Genetics of conditional extended mycelial cell viability of Streptomyces minutiscleroticus in deep starvation phase implicates the involvement of (p)ppGpp, clpX, and a histidine kinase sasA. Front Microbiol 2024; 15:1495007. [PMID: 39611085 PMCID: PMC11604128 DOI: 10.3389/fmicb.2024.1495007] [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: 09/11/2024] [Accepted: 10/18/2024] [Indexed: 11/30/2024] Open
Abstract
Bacterial lifespan ranges from a few hours to geological timescales. The prolonged survival trait under extreme energy starvation is essential for the perpetuation of their existence. The theme for long-term survival [long-term stationary phase (LTSP)] in the non-growing state may be dependent on the diversity in the environmental niche and the lifestyle of the bacteria, exemplified by longevity studies, albeit few, with model organisms. In the present study, we characterized the LTSP of mycelial cells of Streptomyces Minutiscleroticus, which remain metabolically active, demonstrate ongoing protein synthesis-killed by protein synthesis inhibitors-and remarkably by the cell-wall synthesis inhibitors, vancomycin, and ampicillin, suggesting "growth." Their rapid turnover is also evident in ~10-fold loss of colony-forming unit (CFU) over a year, suggesting that for the death of one "old" cell, slightly less than one "new" cell is born. This longevity is consequent to (i) induction of the gene expression program effected by non-metabolizable, non-ionic osmolyte, sucrose, thus conditional, and (ii) possibly rendering this carbon utilizable by the production of a slow hydrolytic activity generating glucose, reinforcing the relevance of low-level energy resource for long term survival in the starvation phase. The viability parameters of LTSP cells measured through up to 90 days suggest that the stationary phase transitioning into LTSP following nutrient exhaustion is nearly quantitative. Expectedly, the viability in LTSP is (p)ppGpp/RelA dependent. Whereas mutation in chaperone clpX, negatively affects survival in stationary phase, overexpression of signal sensor-transducer histidine kinase, SasA8, enhances cell survivability. The relevance of longevity functions identified here requires further deduction of the genetic program.
Collapse
|
11
|
Wegrzynowicz AK, Heelan WJ, Demas SP, McLean MS, Peters JM, Henzler-Wildman KA. Substrate dependence of transport coupling and phenotype of a small multidrug resistance transporter in Pseudomonas aeruginosa. J Bacteriol 2024; 206:e0015124. [PMID: 39258918 PMCID: PMC11500531 DOI: 10.1128/jb.00151-24] [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: 04/11/2024] [Accepted: 08/15/2024] [Indexed: 09/12/2024] Open
Abstract
Small multidrug resistance (SMR) transporters are key players in the defense of multidrug-resistant pathogens to toxins and other homeostasis-perturbing compounds. However, recent evidence demonstrates that EmrE, an SMR from Escherichia coli and a model for understanding transport, can also induce susceptibility to some compounds by drug-gated proton leak. This runs down the ∆pH component of the proton-motive force (PMF), reducing the viability of the affected bacteria. Proton leak may provide an unexplored drug target distinct from the targets of most known antibiotics. Activating proton leak requires an SMR to be merely present, rather than be the primary resistance mechanism, and dissipates the energy source for many other efflux pumps. PAsmr, an EmrE homolog from Pseudomonas aeruginosa, transports many EmrE substrates in cells and purified systems. We hypothesized that PAsmr, like EmrE, may confer susceptibility to some compounds via drug-gated proton leak. Growth assays of E. coli expressing PAsmr displayed substrate-dependent resistance and susceptibility phenotypes, and in vitro solid-supported membrane electrophysiology experiments revealed that PAsmr performs both antiport and substrate-gated proton uniport, demonstrating the same functional promiscuity observed in EmrE. Growth assays of P. aeruginosa strain PA14 demonstrated that PAsmr contributes resistance to some antimicrobial compounds, but no growth defect is observed with susceptibility substrates, suggesting P. aeruginosa can compensate for the proton leak occurring through PAsmr. These phenotypic differences between P. aeruginosa and E. coli advance our understanding of the underlying resistance mechanisms in P. aeruginosa and prompt further investigation into the role that SMRs play in antibiotic resistance in pathogens. IMPORTANCE Small multidrug resistance (SMR) transporters are a class of efflux pumps found in many pathogens, although their contributions to antibiotic resistance are not fully understood. We hypothesize that these transporters may confer not only resistance but also susceptibility, by dissipating the proton-motive force. This means to use an SMR transporter as a target; it merely needs to be present (as opposed to being the primary resistance mechanism). Here, we test this hypothesis with an SMR transporter found in Pseudomonas aeruginosa and find that it can perform both antiport (conferring resistance) and substrate-gated proton leak. Proton leak is detrimental to growth in Escherichia coli but not P. aeruginosa, suggesting that P. aeruginosa responds differently to or can altogether prevent ∆pH dissipation.
Collapse
Affiliation(s)
- Andrea K. Wegrzynowicz
- Department of Biochemistry, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - William J. Heelan
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Sydnye P. Demas
- Department of Biochemistry, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Maxwell S. McLean
- Department of Biochemistry, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Jason M. Peters
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin—Madison, Madison, Wisconsin, USA
- DOE Great Lakes Bioenergy Research Center, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Department of Bacteriology, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Center for Genomic Science Innovation, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Katherine A. Henzler-Wildman
- Department of Biochemistry, University of Wisconsin—Madison, Madison, Wisconsin, USA
- National Magnetic Resonance Facility at Madison, Madison, Wisconsin, USA
| |
Collapse
|
12
|
Ahmed S, Hulbert AK, Xin X, Neff MM. The ability of Arabidopsis to recover from Basta and its application in isolating Cas9-free mutants. FRONTIERS IN PLANT SCIENCE 2024; 15:1408230. [PMID: 39479542 PMCID: PMC11521829 DOI: 10.3389/fpls.2024.1408230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 09/25/2024] [Indexed: 11/02/2024]
Abstract
After successfully performing Agrobacterium-mediated CRISPR-Cas9-based gene editing in plants, isolation of the Cas9 T-DNA is essential for the stable inheritance of induced mutations. Here, we report a simple technique that allows the isolation of Cas9-free mutants, eliminating the need for outcrossing or other intricate methods. This method is based on the ability of Basta-sensitive Arabidopsis thaliana seedlings, which generally perish, to recover and grow once transplanted to Basta-free growth media. By growing gene-edited heterozygous populations of single-locus insertion Basta-resistant plants on Basta selection media, plants lacking the Cas9 T-DNA can be identified. These pale-looking plants lacking Cas9 are then rescued on media lacking the Basta to recover Cas9-free plants. The ability of seedlings to recover from Basta selection was also studied in camelina, canola, and wheat. All three crops showed different recovery rates, with wheat demonstrating the highest recovery once transplanted from Basta to normal growth media. In summary, our findings demonstrate that by harnessing the recovery capability of Basta-sensitive seedlings, we can effectively identify and rescue plants lacking the Cas9 T-DNA, enabling the isolation of Cas9-free mutants in Arabidopsis and potentially extending to other crops.
Collapse
Affiliation(s)
- Shahbaz Ahmed
- The Department of Crop and Soil Sciences, Washington State University, Pullman, WA, United States
| | - Anna K. Hulbert
- The Department of Crop and Soil Sciences, Washington State University, Pullman, WA, United States
| | - Xin Xin
- Graduate Program in Molecular Plant Sciences, Washington State University, Pullman, WA, United States
| | - Michael M. Neff
- The Department of Crop and Soil Sciences, Washington State University, Pullman, WA, United States
- Graduate Program in Molecular Plant Sciences, Washington State University, Pullman, WA, United States
| |
Collapse
|
13
|
Krawczyk SJ, Leśniczak-Staszak M, Gowin E, Szaflarski W. Mechanistic Insights into Clinically Relevant Ribosome-Targeting Antibiotics. Biomolecules 2024; 14:1263. [PMID: 39456196 PMCID: PMC11505993 DOI: 10.3390/biom14101263] [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: 09/08/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/28/2024] Open
Abstract
Antibiotics targeting the bacterial ribosome are essential to combating bacterial infections. These antibiotics bind to various sites on the ribosome, inhibiting different stages of protein synthesis. This review provides a comprehensive overview of the mechanisms of action of clinically relevant antibiotics that target the bacterial ribosome, including macrolides, lincosamides, oxazolidinones, aminoglycosides, tetracyclines, and chloramphenicol. The structural and functional details of antibiotic interactions with ribosomal RNA, including specific binding sites, interactions with rRNA nucleotides, and their effects on translation processes, are discussed. Focus is placed on the diversity of these mechanisms and their clinical implications in treating bacterial infections, particularly in the context of emerging resistance. Understanding these mechanisms is crucial for developing novel therapeutic agents capable of overcoming bacterial resistance.
Collapse
Affiliation(s)
- Szymon J. Krawczyk
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznań, Poland; (S.J.K.); (M.L.-S.)
| | - Marta Leśniczak-Staszak
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznań, Poland; (S.J.K.); (M.L.-S.)
| | - Ewelina Gowin
- Department of Health Promotion, Poznan University of Medical Sciences, 60-781 Poznań, Poland;
- Department of Immunology, Poznan University of Medical Sciences, 60-806 Poznań, Poland
| | - Witold Szaflarski
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznań, Poland; (S.J.K.); (M.L.-S.)
| |
Collapse
|
14
|
Mikami M, Shimizu H, Iwama N, Yajima M, Kuwasako K, Ogura Y, Himeno H, Kurita D, Nameki N. Stalled ribosome rescue factors exert different roles depending on types of antibiotics in Escherichia coli. NPJ ANTIMICROBIALS AND RESISTANCE 2024; 2:22. [PMID: 39843510 PMCID: PMC11721466 DOI: 10.1038/s44259-024-00039-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 07/08/2024] [Indexed: 01/24/2025]
Abstract
Escherichia coli possesses three stalled-ribosome rescue factors, tmRNA·SmpB (primary factor), ArfA (alternative factor to tmRNA·SmpB), and ArfB. Here, we examined the susceptibility of rescue factor-deficient strains from E. coli SE15 to various ribosome-targeting antibiotics. Aminoglycosides specifically decreased the growth of the ΔssrA (tmRNA gene) strain, in which the levels of reactive oxygen species were elevated. The decrease in growth of ΔssrA could not be complemented by plasmid-borne expression of arfA, arfB, or ssrAAA to DD mutant gene possessing a proteolysis-resistant tag sequence. These results highlight the significance of tmRNA·SmpB-mediated proteolysis during growth under aminoglycoside stress. In contrast, tetracyclines or amphenicols decreased the growth of the ΔarfA strain despite the presence of tmRNA·SmpB. Quantitative RT-PCR revealed that tetracyclines and amphenicols, but not aminoglycosides, considerably induced mRNA expression of arfA. These findings indicate that tmRNA·SmpB, and ArfA exert differing functions during stalled-ribosome rescue depending on the type of ribosome-targeting antibiotic.
Collapse
Affiliation(s)
- Mayu Mikami
- Division of Molecular Science, Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjin-cho, Kiryu-shi, Gunma, 376-8515, Japan
| | - Hidehiko Shimizu
- Division of Molecular Science, Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjin-cho, Kiryu-shi, Gunma, 376-8515, Japan
| | - Norika Iwama
- Division of Molecular Science, Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjin-cho, Kiryu-shi, Gunma, 376-8515, Japan
| | - Mihono Yajima
- Division of Molecular Science, Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjin-cho, Kiryu-shi, Gunma, 376-8515, Japan
| | - Kanako Kuwasako
- Faculty of Pharmacy and Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo, 202-8585, Japan
| | - Yoshitoshi Ogura
- Division of Microbiology, Department of Infectious Medicine, Kurume University School of Medicine, Kurume, Fukuoka, 830-0011, Japan
| | - Hyouta Himeno
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki, Japan
| | - Daisuke Kurita
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki, Japan
| | - Nobukazu Nameki
- Division of Molecular Science, Graduate School of Science and Technology, Gunma University, 1-5-1 Tenjin-cho, Kiryu-shi, Gunma, 376-8515, Japan.
| |
Collapse
|
15
|
Gonçalves ASC, Leitão MM, Fernandes JR, Saavedra MJ, Pereira C, Simões M, Borges A. Photodynamic activation of phytochemical-antibiotic combinations for combatting Staphylococcus aureus from acute wound infections. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 258:112978. [PMID: 39002192 DOI: 10.1016/j.jphotobiol.2024.112978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/15/2024]
Abstract
Staphylococcus aureus is characterized by its high resistance to conventional antibiotics, particularly methicillin-resistant (MRSA) strains, making it a predominant pathogen in acute and chronic wound infections. The persistence of acute S. aureus wound infections poses a threat by increasing the incidence of their chronicity. This study investigated the potential of photodynamic activation using phytochemical-antibiotic combinations to eliminate S. aureus under conditions representative of acute wound infections, aiming to mitigate the risk of chronicity. The strategy applied takes advantage of the promising antibacterial and photosensitising properties of phytochemicals, and their ability to act as antibiotic adjuvants. The antibacterial activity of selected phytochemicals (berberine, curcumin, farnesol, gallic acid, and quercetin; 6.25-1000 μg/mL) and antibiotics (ciprofloxacin, tetracycline, fusidic acid, oxacillin, gentamicin, mupirocin, methicillin, and tobramycin; 0.0625-1024 μg/mL) was screened individually and in combination against two S. aureus clinical strains (methicillin-resistant and -susceptible-MRSA and MSSA). The photodynamic activity of the phytochemicals was assessed using a light-emitting diode (LED) system with blue (420 nm) or UV-A (365 nm) variants, at 30 mW/cm2 (light doses of 9, 18, 27 J/cm2) and 5.5 mW/cm2 (light doses of 1.5, 3.3 and 5.0 J/cm2), respectively. Notably, all phytochemicals restored antibiotic activity, with 9 and 13 combinations exhibiting potentiating effects on MSSA and MRSA, respectively. Photodynamic activation with blue light (420 nm) resulted in an 8- to 80-fold reduction in the bactericidal concentration of berberine against MSSA and MRSA, while curcumin caused 80-fold reduction for both strains at the light dose of 18 J/cm2. Berberine and curcumin-antibiotic combinations when subjected to photodynamic activation (420 nm light, 10 min, 18 J/cm2) reduced S. aureus culturability by ≈9 log CFU/mL. These combinations lowered the bactericidal concentration of antibiotics, achieving a 2048-fold reduction for gentamicin and 512-fold reduction for tobramycin. Overall, the dual approach involving antimicrobial photodynamic inactivation and selected phytochemical-antibiotic combinations demonstrated a synergistic effect, drastically reducing the culturability of S. aureus and restoring the activity of gentamicin and tobramycin.
Collapse
Affiliation(s)
- Ariana S C Gonçalves
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal; Environmental Health Department, Portuguese National Health Institute Doutor Ricardo Jorge, Porto, Portugal
| | - Miguel M Leitão
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal; CIQUP-IMS-Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre, 4169-007, Porto, Portugal
| | - José R Fernandes
- CQVR-Vila Real Chemistry Center, University of Trás-os-Montes e Alto Douro, Portugal; Physical Department, University of Trás-os-Montes and Alto Douro, Quinta dos Prados, 5000-801 Vila Real, Portugal
| | - Maria José Saavedra
- Antimicrobials, Biocides and Biofilms Unit (AB2Unit), Laboratory of Medical Microbiology, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; Animal and Veterinary Research Center (CECAV)-Al4AnimalS, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; Center Interdisciplinar of Marine and Environmental Research (CIIMAR), University of Porto, 4450-208 Matosinhos, Portugal; Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB)-Inov4Agro, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
| | - Cristiana Pereira
- Environmental Health Department, Portuguese National Health Institute Doutor Ricardo Jorge, Porto, Portugal; Environmental Hygiene and Human Biomonitoring Unit, Department of Health Protection, Laboratoire National de Santé, Luxembourg
| | - Manuel Simões
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal; DEQ-Department of Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
| | - Anabela Borges
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALICE-Associate Laboratory for Innovation in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal; DEQ-Department of Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal.
| |
Collapse
|
16
|
Lyu Z, Ling Y, van Hoof A, Ling J. Inactivation of the ribosome assembly factor RimP causes streptomycin resistance and impairs motility in Salmonella. Antimicrob Agents Chemother 2024; 68:e0000224. [PMID: 38629858 PMCID: PMC11620500 DOI: 10.1128/aac.00002-24] [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: 01/02/2024] [Accepted: 03/27/2024] [Indexed: 05/02/2024] Open
Abstract
The ribosome is the central hub for protein synthesis and the target of many antibiotics. Although the majority of ribosome-targeting antibiotics inhibit protein synthesis and are bacteriostatic, aminoglycosides promote protein mistranslation and are bactericidal. Understanding the resistance mechanisms of bacteria against aminoglycosides is not only vital for improving the efficacy of this critically important group of antibiotics but also crucial for studying the molecular basis of translational fidelity. In this work, we analyzed Salmonella mutants evolved in the presence of the aminoglycoside streptomycin (Str) and identified a novel gene rimP to be involved in Str resistance. RimP is a ribosome assembly factor critical for the maturation of the 30S small subunit that binds Str. Deficiency in RimP increases resistance against Str and facilitates the development of even higher resistance. Deleting rimP decreases mistranslation and cellular uptake of Str and further impairs flagellar motility. Our work thus highlights a previously unknown mechanism of aminoglycoside resistance via defective ribosome assembly.
Collapse
Affiliation(s)
- Zhihui Lyu
- Department of Cell Biology and Molecular Genetics, The University of Maryland, College Park, Maryland, USA
| | - Yunyi Ling
- Department of Cell Biology and Molecular Genetics, The University of Maryland, College Park, Maryland, USA
| | - Ambro van Hoof
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Jiqiang Ling
- Department of Cell Biology and Molecular Genetics, The University of Maryland, College Park, Maryland, USA
| |
Collapse
|
17
|
Uneme M, Ishikawa K, Furuta K, Yamashita A, Kaito C. Overexpression of the flagellar motor protein MotB sensitizes Bacillus subtilis to aminoglycosides in a motility-independent manner. PLoS One 2024; 19:e0300634. [PMID: 38669243 PMCID: PMC11051680 DOI: 10.1371/journal.pone.0300634] [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: 10/13/2023] [Accepted: 03/02/2024] [Indexed: 04/28/2024] Open
Abstract
The flagellar motor proteins, MotA and MotB, form a complex that rotates the flagella by utilizing the proton motive force (PMF) at the bacterial cell membrane. Although PMF affects the susceptibility to aminoglycosides, the effect of flagellar motor proteins on the susceptibility to aminoglycosides has not been investigated. Here, we found that MotB overexpression increased susceptibility to aminoglycosides, such as kanamycin and gentamicin, in Bacillus subtilis without affecting swimming motility. MotB overexpression did not affect susceptibility to ribosome-targeting antibiotics other than aminoglycosides, cell wall-targeting antibiotics, DNA synthesis-inhibiting antibiotics, or antibiotics inhibiting RNA synthesis. Meanwhile, MotB overexpression increased the susceptibility to aminoglycosides even in the motA-deletion mutant, which lacks swimming motility. Overexpression of the MotB mutant protein carrying an amino acid substitution at the proton-binding site (D24A) resulted in the loss of the enhanced aminoglycoside-sensitive phenotype. These results suggested that MotB overexpression sensitizes B. subtilis to aminoglycosides in a motility-independent manner. Notably, the aminoglycoside-sensitive phenotype induced by MotB requires the proton-binding site but not the MotA/MotB complex formation.
Collapse
Affiliation(s)
- Mio Uneme
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kazuya Ishikawa
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kazuyuki Furuta
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Atsuko Yamashita
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Chikara Kaito
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
18
|
Hinnu M, Putrinš M, Kogermann K, Kaldalu N, Tenson T. Fluorescent reporters give new insights into antibiotics-induced nonsense and frameshift mistranslation. Sci Rep 2024; 14:6883. [PMID: 38519558 PMCID: PMC10959953 DOI: 10.1038/s41598-024-57597-8] [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: 11/15/2023] [Accepted: 03/20/2024] [Indexed: 03/25/2024] Open
Abstract
We developed a reporter system based on simultaneous expression of two fluorescent proteins: GFP as a reporter of the capacity of protein synthesis and mutated mScarlet-I as a reporter of translational errors. Because of the unique stop codons or frameshift mutations introduced into the mScarlet-I gene, red fluorescence was produced only after a mistranslation event. These reporters allowed us to estimate mistranslation at a single cell level using either flow cytometry or fluorescence microscopy. We found that laboratory strains of Escherichia coli are more prone to mistranslation compared to the clinical isolates. As relevant for uropathogenic E. coli, growth in human urine elevated translational frameshifting compared to standard laboratory media, whereas different standard media had a small effect on translational fidelity. Antibiotic-induced mistranslation was studied by using amikacin (aminoglycoside family) and azithromycin (macrolide family). Bactericidal amikacin induced preferably stop-codon readthrough at a moderate level. Bacteriostatic azithromycin on the other hand induced both frameshifting and stop-codon readthrough at much higher level. Single cell analysis revealed that fluorescent reporter-protein signal can be lost due to leakage from a fraction of bacteria in the presence of antibiotics, demonstrating the complexity of the antimicrobial activity.
Collapse
Affiliation(s)
- Mariliis Hinnu
- Institute of Technology, University of Tartu, 50411, Tartu, Estonia.
| | - Marta Putrinš
- Institute of Technology, University of Tartu, 50411, Tartu, Estonia
- Institute of Pharmacy, University of Tartu, 50411, Tartu, Estonia
| | - Karin Kogermann
- Institute of Pharmacy, University of Tartu, 50411, Tartu, Estonia
| | - Niilo Kaldalu
- Institute of Technology, University of Tartu, 50411, Tartu, Estonia
| | - Tanel Tenson
- Institute of Technology, University of Tartu, 50411, Tartu, Estonia
| |
Collapse
|
19
|
Heo HY, Zou G, Baek S, Kim J, Mylonakis E, Ausubel FM, Gao H, Kim W. A Methylazanediyl Bisacetamide Derivative Sensitizes Staphylococcus aureus Persisters to a Combination of Gentamicin And Daptomycin. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306112. [PMID: 38126676 PMCID: PMC10916567 DOI: 10.1002/advs.202306112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 12/01/2023] [Indexed: 12/23/2023]
Abstract
Infections caused by Staphylococcus aureus, notably methicillin-resistant S. aureus (MRSA), pose treatment challenges due to its ability to tolerate antibiotics and develop antibiotic resistance. The former, a mechanism independent of genetic changes, allows bacteria to withstand antibiotics by altering metabolic processes. Here, a potent methylazanediyl bisacetamide derivative, MB6, is described, which selectively targets MRSA membranes over mammalian membranes without observable resistance development. Although MB6 is effective against growing MRSA cells, its antimicrobial activity against MRSA persisters is limited. Nevertheless, MB6 significantly potentiates the bactericidal activity of gentamicin against MRSA persisters by facilitating gentamicin uptake. In addition, MB6 in combination with daptomycin exhibits enhanced anti-persister activity through mutual reinforcement of their membrane-disrupting activities. Crucially, the "triple" combination of MB6, gentamicin, and daptomycin exhibits a marked enhancement in the killing of MRSA persisters compared to individual components or any double combinations. These findings underscore the potential of MB6 to function as a potent and selective membrane-active antimicrobial adjuvant to enhance the efficacy of existing antibiotics against persister cells. The molecular mechanisms of MB6 elucidated in this study provide valuable insights for designing anti-persister adjuvants and for developing new antimicrobial combination strategies to overcome the current limitations of antibiotic treatments.
Collapse
Affiliation(s)
- Hee Young Heo
- College of PharmacyGraduate School of Pharmaceutical SciencesEwha Womans UniversitySeoul03760Republic of Korea
| | - Guijin Zou
- Institute of High Performance Computing (IHPC)Agency for ScienceTechnology and Research (A*STAR)Singapore138632Republic of Singapore
| | - Seongeun Baek
- College of PharmacyGraduate School of Pharmaceutical SciencesEwha Womans UniversitySeoul03760Republic of Korea
| | - Jae‐Seok Kim
- Department of Laboratory MedicineKangdong Sacred Heart HospitalHallym University College of MedicineSeoul05355Republic of Korea
| | | | - Frederick M. Ausubel
- Department of Molecular BiologyMassachusetts General HospitalBostonMA02114USA
- Department of GeneticsHarvard Medical SchoolBostonMA02115USA
| | - Huajian Gao
- Institute of High Performance Computing (IHPC)Agency for ScienceTechnology and Research (A*STAR)Singapore138632Republic of Singapore
- School of Mechanical and Aerospace EngineeringCollege of EngineeringNanyang Technological UniversitySingapore639789Republic of Singapore
| | - Wooseong Kim
- College of PharmacyGraduate School of Pharmaceutical SciencesEwha Womans UniversitySeoul03760Republic of Korea
| |
Collapse
|
20
|
Kim YJ, Choi HS, Park DH. Persistence and viable but non-culturable state induced by streptomycin in Erwinia amylovora. Front Microbiol 2024; 15:1346300. [PMID: 38450169 PMCID: PMC10914980 DOI: 10.3389/fmicb.2024.1346300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/08/2024] [Indexed: 03/08/2024] Open
Abstract
Persister cell and viable but non-culturable (VBNC) state of bacteria are survival strategies against antibiotics and various environmental stresses, respectively, but they tend to be ignored in agriculture fields, even though bacteria can regain their abilities to survive and produce disease once those stresses disappear. This study was carried out to determine whether persister cell and VBNC state in Erwinia amylovora are present after exposures to streptomycin, the length of their persistence, and the steps needed to decrease the inoculum. Persister cells were observed using biphasic killed growth curve for 4-8 h when the late stationary phase cells of E. amylovora were cultured in liquid medium containing streptomycin. This state was maintained for up to 12 h based on the colony forming units (CFUs) of the colonies that grew on the mannitol glutamate yeast extract (MGY) medium after streptomycin was removed. The CFUs on the MGY medium were lower than the total count determined using the LIVE/DEAD Kit, suggesting that persister cells and VBNC state might co-exist for up to 12 h after exposure to streptomycin. However, after 12 h, E. amylovora cells did not continue to grow on the medium for 9 days, suggesting that they entered a VBNC state at that time and remained in a persistent state. In addition, based on the Redox Sensor Green staining method, the presence of both states was confirmed for up to 12 h, and only then did the VBNC state became apparent. Furthermore, persister cells were observed for up to 24 h, and damaged cells reduced when E. amylovora cells were culture in distilled water with streptomycin, indicating that the uptake of lower nutrients in E. amylovora led to prolonged persister cells and VBNC state, which are more likely to survive after streptomycin treatments. The addition of sucrose and oxytetracycline to distilled water containing streptomycin reduced persister cells than other sources did. Thus, to inhibit the spread of fire blight, management techniques must consider the hazards of using streptomycin treatments that induce dormancy, such as persister cells and VBNC state, beyond the development of resistant strain.
Collapse
Affiliation(s)
- Yeon Ju Kim
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon-si, Republic of Korea
| | - Hyun Seo Choi
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon-si, Republic of Korea
| | - Duck Hwan Park
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon-si, Republic of Korea
- Plant Medicine Program, Division of Bioresource Sciences, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon-si, Republic of Korea
| |
Collapse
|
21
|
Hershko Y, Rannon E, Adler A, Burstein D, Barkan D. WarA, a remote homolog of NpmA and KamB from Nocardia wallacei, confers broad spectrum aminoglycoside resistance in Nocardia and Mycobacteria. Int J Antimicrob Agents 2024; 63:107089. [PMID: 38218322 DOI: 10.1016/j.ijantimicag.2024.107089] [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: 08/09/2023] [Revised: 12/24/2023] [Accepted: 01/07/2024] [Indexed: 01/15/2024]
Abstract
OBJECTIVES Aminoglycoside resistance in bacteria is typically conferred by specific drug-modifying enzymes. Infrequently, such resistance is achieved through 16S ribosomal RNA methyltransferases, such as NpmA and KamB encoded by Escherichia coli and Streptoalloteichus tenebrarius, respectively. These enzymes are not widespread and have not been described in Nocardia species to date. METHODS We report the genomic mining of 18 Nocardia wallacei isolates that were found to be specifically and substantially resistant to amikacin. RESULTS We identified a gene coding for a protein with very distant homology to NpmA and KamB. However, 3-D modeling revealed that the tertiary structure of these three proteins was highly similar. Cloning and expressing this gene in two susceptible bacteria Nocardia asteroides, and Mycobacterium smegmatis (another Actinobacterium) led to high-level, pan-aminoglycoside resistance in both cases. We named this gene warA (Wallacei Amikacin Resistance A). CONCLUSIONS This is the first description and experimental characterization of a gene of this family in Nocardia, and the first demonstration that such activity could lead to pan-aminoglycoside resistance in Mycobacteria as well. The discovery of this novel gene has important biotechnology and clinical implications.
Collapse
Affiliation(s)
- Yizhak Hershko
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel; Clinical Microbiology Laboratory, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Ella Rannon
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel
| | - Amos Adler
- Clinical Microbiology Laboratory, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Department of Epidemiology and Preventive Medicine, School of Public Health, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - David Burstein
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel
| | - Daniel Barkan
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel.
| |
Collapse
|
22
|
Lee EB, Lee K. A Pharmacodynamic Study of Aminoglycosides against Pathogenic E. coli through Monte Carlo Simulation. Pharmaceuticals (Basel) 2023; 17:27. [PMID: 38256861 PMCID: PMC10819079 DOI: 10.3390/ph17010027] [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: 11/24/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
This research focuses on combating the increasing problem of antimicrobial resistance, especially in Escherichia coli (E. coli), by assessing the efficacy of aminoglycosides. The study specifically addresses the challenge of developing new therapeutic approaches by integrating experimental data with mathematical modeling to better understand the action of aminoglycosides. It involves testing various antibiotics like streptomycin (SMN), kanamycin (KMN), gentamicin (GMN), tobramycin (TMN), and amikacin (AKN) against the O157:H7 strain of E. coli. The study employs a pharmacodynamics (PD) model to analyze how different antibiotic concentrations affect bacterial growth, utilizing minimum inhibitory concentration (MIC) to gauge the effective bactericidal levels of the antibiotics. The study's approach involved transforming bacterial growth rates, as obtained from time-kill curve data, into logarithmic values. A model was then developed to correlate these log-transformed values with their respective responses. To generate additional data points, each value was systematically increased by an increment of 0.1. To simulate real-world variability and randomness in the data, a Gaussian scatter model, characterized by parameters like κ and EC50, was employed. The mathematical modeling was pivotal in uncovering the bactericidal properties of these antibiotics, indicating different PD MIC (zMIC) values for each (SMN: 1.22; KMN: 0.89; GMN: 0.21; TMN: 0.32; AKN: 0.13), which aligned with MIC values obtained through microdilution methods. This innovative blend of experimental and mathematical approaches in the study marks a significant advancement in formulating strategies to combat the growing threat of antimicrobial-resistant E. coli, offering a novel pathway to understand and tackle antimicrobial resistance more effectively.
Collapse
Affiliation(s)
- Eon-Bee Lee
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea;
| | - Kyubae Lee
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seoul 03722, Republic of Korea
| |
Collapse
|
23
|
Lang M, Carvalho A, Baharoglu Z, Mazel D. Aminoglycoside uptake, stress, and potentiation in Gram-negative bacteria: new therapies with old molecules. Microbiol Mol Biol Rev 2023; 87:e0003622. [PMID: 38047635 PMCID: PMC10732077 DOI: 10.1128/mmbr.00036-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023] Open
Abstract
SUMMARYAminoglycosides (AGs) are long-known molecules successfully used against Gram-negative pathogens. While their use declined with the discovery of new antibiotics, they are now classified as critically important molecules because of their effectiveness against multidrug-resistant bacteria. While they can efficiently cross the Gram-negative envelope, the mechanism of AG entry is still incompletely understood, although this comprehension is essential for the development of new therapies in the face of the alarming increase in antibiotic resistance. Increasing antibiotic uptake in bacteria is one strategy to enhance effective treatments. This review aims, first, to consolidate old and recent knowledge about AG uptake; second, to explore the connection between AG-dependent bacterial stress and drug uptake; and finally, to present new strategies of potentiation of AG uptake for more efficient antibiotic therapies. In particular, we emphasize on the connection between sugar transport and AG potentiation.
Collapse
Affiliation(s)
- Manon Lang
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| | - André Carvalho
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| | - Zeynep Baharoglu
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| | - Didier Mazel
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| |
Collapse
|
24
|
Bren A, Glass DS, Kohanim YK, Mayo A, Alon U. Tradeoffs in bacterial physiology determine the efficiency of antibiotic killing. Proc Natl Acad Sci U S A 2023; 120:e2312651120. [PMID: 38096408 PMCID: PMC10742385 DOI: 10.1073/pnas.2312651120] [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: 07/26/2023] [Accepted: 11/02/2023] [Indexed: 12/18/2023] Open
Abstract
Antibiotic effectiveness depends on a variety of factors. While many mechanistic details of antibiotic action are known, the connection between death rate and bacterial physiology is poorly understood. A common observation is that death rate in antibiotics rises linearly with growth rate; however, it remains unclear how other factors, such as environmental conditions and whole-cell physiological properties, affect bactericidal activity. To address this, we developed a high-throughput assay to precisely measure antibiotic-mediated death. We found that death rate is linear in growth rate, but the slope depends on environmental conditions. Growth under stress lowers death rate compared to nonstressed environments with similar growth rate. To understand stress's role, we developed a mathematical model of bacterial death based on resource allocation that includes a stress-response sector; we identify this sector using RNA-seq. Our model accurately predicts the minimal inhibitory concentration (MIC) with zero free parameters across a wide range of growth conditions. The model also quantitatively predicts death and MIC when sectors are experimentally modulated using cyclic adenosine monophosphate (cAMP), including protection from death at very low cAMP levels. The present study shows that different conditions with equal growth rate can have different death rates and establishes a quantitative relation between growth, death, and MIC that suggests approaches to improve antibiotic efficacy.
Collapse
Affiliation(s)
- Anat Bren
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - David S. Glass
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Yael Korem Kohanim
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT06520
| | - Avi Mayo
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Uri Alon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| |
Collapse
|
25
|
Krin E, Carvalho A, Lang M, Babosan A, Mazel D, Baharoglu Z. RavA-ViaA antibiotic response is linked to Cpx and Zra2 envelope stress systems in Vibrio cholerae. Microbiol Spectr 2023; 11:e0173023. [PMID: 37861314 PMCID: PMC10848872 DOI: 10.1128/spectrum.01730-23] [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: 04/25/2023] [Accepted: 09/08/2023] [Indexed: 10/21/2023] Open
Abstract
IMPORTANCE The RavA-ViaA complex was previously found to sensitize Escherichia coli to aminoglycosides (AGs) in anaerobic conditions, but the mechanism is unknown. AGs are antibiotics known for their high efficiency against Gram-negative bacteria. In order to elucidate how the expression of the ravA-viaA genes increases bacterial susceptibility to aminoglycosides, we aimed at identifying partner functions necessary for increased tolerance in the absence of RavA-ViaA, in Vibrio cholerae. We show that membrane stress response systems Cpx and Zra2 are required in the absence of RavA-ViaA, for the tolerance to AGs and for outer membrane integrity. In the absence of these systems, the ∆ravvia strain's membrane becomes permeable to external agents such as the antibiotic vancomycin.
Collapse
Affiliation(s)
- Evelyne Krin
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| | - André Carvalho
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
- Sorbonne Université, Collège doctoral, Paris, France
| | - Manon Lang
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
- Sorbonne Université, Collège doctoral, Paris, France
| | - Anamaria Babosan
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| | - Didier Mazel
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| | - Zeynep Baharoglu
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| |
Collapse
|
26
|
Rivetti S, Romano A, Mastrangelo S, Attinà G, Maurizi P, Ruggiero A. Aminoglycosides-Related Ototoxicity: Mechanisms, Risk Factors, and Prevention in Pediatric Patients. Pharmaceuticals (Basel) 2023; 16:1353. [PMID: 37895824 PMCID: PMC10610175 DOI: 10.3390/ph16101353] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/17/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
Aminoglycosides are broad-spectrum antibiotics largely used in children, but they have potential toxic side effects, including ototoxicity. Ototoxicity from aminoglycosides is permanent and is a consequence of its action on the inner ear cells via multiple mechanisms. Both uncontrollable risk factors and controllable risk factors are involved in the pathogenesis of aminoglycoside-related ototoxicity and, because of the irreversibility of ototoxicity, an important undertaking for preventing ototoxicity includes antibiotic stewardship to limit the use of aminoglycosides. Aminoglycosides are fundamental in the treatment of numerous infectious conditions at neonatal and pediatric age. In childhood, normal auditory function ensures adequate neurocognitive and social development. Hearing damage from aminoglycosides can therefore strongly affect the normal growth of the child. This review describes the molecular mechanisms of aminoglycoside-related ototoxicity and analyzes the risk factors and the potential otoprotective strategies in pediatric patients.
Collapse
Affiliation(s)
- Serena Rivetti
- Pediatric Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.R.); (A.R.); (S.M.); (G.A.); (P.M.)
| | - Alberto Romano
- Pediatric Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.R.); (A.R.); (S.M.); (G.A.); (P.M.)
| | - Stefano Mastrangelo
- Pediatric Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.R.); (A.R.); (S.M.); (G.A.); (P.M.)
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Giorgio Attinà
- Pediatric Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.R.); (A.R.); (S.M.); (G.A.); (P.M.)
| | - Palma Maurizi
- Pediatric Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.R.); (A.R.); (S.M.); (G.A.); (P.M.)
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonio Ruggiero
- Pediatric Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (S.R.); (A.R.); (S.M.); (G.A.); (P.M.)
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
27
|
Magaña AJ, Sklenicka J, Pinilla C, Giulianotti M, Chapagain P, Santos R, Ramirez MS, Tolmasky ME. Restoring susceptibility to aminoglycosides: identifying small molecule inhibitors of enzymatic inactivation. RSC Med Chem 2023; 14:1591-1602. [PMID: 37731693 PMCID: PMC10507813 DOI: 10.1039/d3md00226h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/21/2023] [Indexed: 09/22/2023] Open
Abstract
Growing resistance to antimicrobial medicines is a critical health problem that must be urgently addressed. Adding to the increasing number of patients that succumb to infections, there are other consequences to the rise in resistance like the compromise of several medical procedures and dental work that are heavily dependent on infection prevention. Since their introduction in the clinics, aminoglycoside antibiotics have been a critical component of the armamentarium to treat infections. Still, the increase in resistance and their side effects led to a decline in their utilization. However, numerous current factors, like the urgent need for antimicrobials and their favorable properties, led to renewed interest in these drugs. While efforts to design new classes of aminoglycosides refractory to resistance mechanisms and with fewer toxic effects are starting to yield new promising molecules, extending the useful life of those already in use is essential. For this, numerous research projects are underway to counter resistance from different angles, like inhibition of expression or activity of resistance components. This review focuses on selected examples of one aspect of this quest, the design or identification of small molecule inhibitors of resistance caused by enzymatic modification of the aminoglycoside. These compounds could be developed as aminoglycoside adjuvants to overcome resistant infections.
Collapse
Affiliation(s)
- Angel J Magaña
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton Fullerton CA 92831 USA
| | - Jan Sklenicka
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton Fullerton CA 92831 USA
| | - Clemencia Pinilla
- Center for Translational Science, Florida International University Port St. Lucie FL 34987 USA
| | - Marc Giulianotti
- Center for Translational Science, Florida International University Port St. Lucie FL 34987 USA
| | - Prem Chapagain
- Department of Physics, Florida International University Miami FL 33199 USA
- Biomolecular Sciences Institute, Florida International University Miami FL 33199 USA
| | - Radleigh Santos
- Department of Mathematics, Nova Southeastern University Fort Lauderdale FL 33314 USA
| | - Maria Soledad Ramirez
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton Fullerton CA 92831 USA
| | - Marcelo E Tolmasky
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton Fullerton CA 92831 USA
| |
Collapse
|
28
|
Maziero M, Lane D, Polard P, Bergé M. Fever-like temperature bursts promote competence development via an HtrA-dependent pathway in Streptococcus pneumoniae. PLoS Genet 2023; 19:e1010946. [PMID: 37699047 PMCID: PMC10516426 DOI: 10.1371/journal.pgen.1010946] [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: 04/17/2023] [Revised: 09/22/2023] [Accepted: 08/30/2023] [Indexed: 09/14/2023] Open
Abstract
Streptococcus pneumoniae (the pneumococcus) is well known for its ability to develop competence for natural DNA transformation. Competence development is regulated by an autocatalytic loop driven by variations in the basal level of transcription of the comCDE and comAB operons. These genes are part of the early gene regulon that controls expression of the late competence genes known to encode the apparatus of transformation. Several stressful conditions are known to promote competence development, although the induction pathways are remain poorly understood. Here we demonstrate that transient temperature elevation induces an immediate increase in the basal expression level of the comCDE operon and early genes that, in turn, stimulates its full induction, including that of the late competence regulon. This thermal regulation depends on the HtrA chaperone/protease and its proteolytic activity. We find that other competence induction stimulus, like norfloxacin, is not conveyed by the HtrA-dependent pathway. This finding strongly suggests that competence can be induced by at least two independent pathways and thus reinforces the view that competence is a general stress response system in the pneumococcus.
Collapse
Affiliation(s)
- Mickaël Maziero
- Laboratoire de Microbiologie et Génétique Moléculaires (LMGM), UMR5100, Centre de Biologie Intégrative (CBI), Centre Nationale de la Recherche Scientifique (CNRS), Toulouse, France
- Université Paul Sabatier (Toulouse III), Toulouse, France
| | - David Lane
- Laboratoire de Microbiologie et Génétique Moléculaires (LMGM), UMR5100, Centre de Biologie Intégrative (CBI), Centre Nationale de la Recherche Scientifique (CNRS), Toulouse, France
- Université Paul Sabatier (Toulouse III), Toulouse, France
| | - Patrice Polard
- Laboratoire de Microbiologie et Génétique Moléculaires (LMGM), UMR5100, Centre de Biologie Intégrative (CBI), Centre Nationale de la Recherche Scientifique (CNRS), Toulouse, France
- Université Paul Sabatier (Toulouse III), Toulouse, France
| | - Mathieu Bergé
- Laboratoire de Microbiologie et Génétique Moléculaires (LMGM), UMR5100, Centre de Biologie Intégrative (CBI), Centre Nationale de la Recherche Scientifique (CNRS), Toulouse, France
- Université Paul Sabatier (Toulouse III), Toulouse, France
| |
Collapse
|
29
|
Marasini S, Craig JP, Dean SJ, Leanse LG. Managing Corneal Infections: Out with the old, in with the new? Antibiotics (Basel) 2023; 12:1334. [PMID: 37627753 PMCID: PMC10451842 DOI: 10.3390/antibiotics12081334] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/24/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
There have been multiple reports of eye infections caused by antibiotic-resistant bacteria, with increasing evidence of ineffective treatment outcomes from existing therapies. With respect to corneal infections, the most commonly used antibiotics (fluoroquinolones, aminoglycosides, and cephalosporines) are demonstrating reduced efficacy against bacterial keratitis isolates. While traditional methods are losing efficacy, several novel technologies are under investigation, including light-based anti-infective technology with or without chemical substrates, phage therapy, and probiotics. Many of these methods show non-selective antimicrobial activity with potential development as broad-spectrum antimicrobial agents. Multiple preclinical studies and a limited number of clinical case studies have confirmed the efficacy of some of these novel methods. However, given the rapid evolution of corneal infections, their treatment requires rapid institution to limit the impact on vision and prevent complications such as scarring and corneal perforation. Given their rapid effects on microbial viability, light-based technologies seem particularly promising in this regard.
Collapse
Affiliation(s)
- Sanjay Marasini
- Department of Ophthalmology, New Zealand National Eye Centre, The University of Auckland, Auckland 1142, New Zealand; (S.M.); (J.P.C.); (S.J.D.)
| | - Jennifer P. Craig
- Department of Ophthalmology, New Zealand National Eye Centre, The University of Auckland, Auckland 1142, New Zealand; (S.M.); (J.P.C.); (S.J.D.)
| | - Simon J. Dean
- Department of Ophthalmology, New Zealand National Eye Centre, The University of Auckland, Auckland 1142, New Zealand; (S.M.); (J.P.C.); (S.J.D.)
| | - Leon G. Leanse
- Health and Sports Sciences Hub, Europa Point Campus, University of Gibraltar, Gibraltar GX11 1AA, Gibraltar
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
30
|
Wang J, Sijs B, Bakker W, de Haan L, Bouwmeester H. Ribotoxin deoxynivalenol induces taurocholic acid malabsorption in an in vitro human intestinal model. Toxicol Lett 2023; 383:S0378-4274(23)00201-1. [PMID: 37315771 DOI: 10.1016/j.toxlet.2023.06.001] [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: 03/11/2023] [Revised: 05/23/2023] [Accepted: 06/10/2023] [Indexed: 06/16/2023]
Abstract
The trichothecene toxin deoxynivalenol (DON) is a ribotoxic mycotoxin that contaminates cereal-based food. DON binds to ribosomes, thereby inhibiting protein translation and activating stress mitogen-activated protein kinases (MAPK). The activation of MAPK induces pro-inflammatory cytokine production. Emerging evidence showed that DON decreased bile acid reabsorption and apical sodium-dependent bile acid transporter (ASBT) expression in Caco-2 cell layers. We hypothesized that the effect of DON on decreased ASBT mRNA expression is regulated via pro-inflammatory cytokines. We observed that MAPK inhibitors prevented DON to induce IL-8 secretion and prevented the DON-induced downregulation of ASBT mRNA expression. However, DON-induced taurocholic acid (TCA) transport reduction was not prevent by the MAPK inhibitors. We next observed a similarity between the activity of the non-inflammatory ribotoxin cycloheximide and DON to decrease TCA transport, which is consistent with their common ability to inhibit protein synthesis. Together, our results suggest that DON-induced TCA malabsorption is regulated by MAPK activation-induced pro-inflammatory cytokine production and protein synthesis inhibition, both of which are initiated by DON binding to the ribosomes which therefore is the molecular initiating event for the adverse outcome of bile acid malabsorption. This study provides insights into the mechanism of ribotoxins-induced bile acid malabsorption in human intestine.
Collapse
Affiliation(s)
- Jingxuan Wang
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE Wageningen, the Netherlands.
| | - Bas Sijs
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - Wouter Bakker
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - Laura de Haan
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - Hans Bouwmeester
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| |
Collapse
|
31
|
Papadopoulou V, Sidders AE, Lu KY, Velez AZ, Durham PG, Bui DT, Angeles-Solano M, Dayton PA, Rowe SE. Overcoming biological barriers to improve treatment of a Staphylococcus aureus wound infection. Cell Chem Biol 2023; 30:513-526.e5. [PMID: 37148883 PMCID: PMC10198964 DOI: 10.1016/j.chembiol.2023.04.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/19/2023] [Accepted: 04/17/2023] [Indexed: 05/08/2023]
Abstract
Chronic wounds frequently become infected with bacterial biofilms which respond poorly to antibiotic therapy. Aminoglycoside antibiotics are ineffective at treating deep-seated wound infections due to poor drug penetration, poor drug uptake into persister cells, and widespread antibiotic resistance. In this study, we combat the two major barriers to successful aminoglycoside treatment against a biofilm-infected wound: limited antibiotic uptake and limited biofilm penetration. To combat the limited antibiotic uptake, we employ palmitoleic acid, a host-produced monounsaturated fatty acid that perturbs the membrane of gram-positive pathogens and induces gentamicin uptake. This novel drug combination overcomes gentamicin tolerance and resistance in multiple gram-positive wound pathogens. To combat biofilm penetration, we examined the ability of sonobactericide, a non-invasive ultrasound-mediated-drug delivery technology to improve antibiotic efficacy using an in vivo biofilm model. This dual approach dramatically improved antibiotic efficacy against a methicillin-resistant Staphylococcus aureus (MRSA) wound infection in diabetic mice.
Collapse
Affiliation(s)
- Virginie Papadopoulou
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA.
| | - Ashelyn E Sidders
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kuan-Yi Lu
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Amanda Z Velez
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Phillip G Durham
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA; Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Duyen T Bui
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Michelle Angeles-Solano
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Paul A Dayton
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA; Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Sarah E Rowe
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
32
|
Zhang N, Zheng W, Bakker W, van Ravenzwaay B, Rietjens IMCM. In vitro models to measure effects on intestinal deconjugation and transport of mixtures of bile acids. Chem Biol Interact 2023; 375:110445. [PMID: 36889625 DOI: 10.1016/j.cbi.2023.110445] [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: 01/10/2023] [Revised: 03/01/2023] [Accepted: 03/05/2023] [Indexed: 03/08/2023]
Abstract
Bile acid metabolism and transport are critical to maintain bile acid homeostasis and host health. In this study, it was investigated if effects on intestinal bile acid deconjugation and transport can be quantified in vitro model systems using mixtures of bile acids instead of studying individual bile acids. To this end deconjugation of mixtures of selected bile acids in anaerobic rat or human fecal incubations and the effect of the antibiotic tobramycin on these reactions was studied. In addition, the effect of tobramycin on the transport of the bile acids in isolation or in a mixture across Caco-2 cell layers was characterized. The results demonstrate that both the reduction of bile acid deconjugation and transport by tobramycin can be adequately detected in vitro systems using a mixture of bile acids, thus eliminating the need to characterize the effects for each bile acid in separate experiments. Subtle differences between the experiments with single or combined bile acids point at mutual competitive interactions and indicate that the use of bile acid mixtures is preferred over use of single bile acid given that also in vivo bile acids occurs in mixtures.
Collapse
Affiliation(s)
- Nina Zhang
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708, WE Wageningen, the Netherlands.
| | - Weijia Zheng
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708, WE Wageningen, the Netherlands
| | - Wouter Bakker
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708, WE Wageningen, the Netherlands
| | - Bennard van Ravenzwaay
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708, WE Wageningen, the Netherlands
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708, WE Wageningen, the Netherlands
| |
Collapse
|
33
|
Usui M, Yoshii Y, Thiriet-Rupert S, Ghigo JM, Beloin C. Intermittent antibiotic treatment of bacterial biofilms favors the rapid evolution of resistance. Commun Biol 2023; 6:275. [PMID: 36928386 PMCID: PMC10020551 DOI: 10.1038/s42003-023-04601-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 02/16/2023] [Indexed: 03/18/2023] Open
Abstract
Bacterial antibiotic resistance is a global health concern of increasing importance and intensive study. Although biofilms are a common source of infections in clinical settings, little is known about the development of antibiotic resistance within biofilms. Here, we use experimental evolution to compare selection of resistance mutations in planktonic and biofilm Escherichia coli populations exposed to clinically relevant cycles of lethal treatment with the aminoglycoside amikacin. Consistently, mutations in sbmA, encoding an inner membrane peptide transporter, and fusA, encoding the essential elongation factor G, are rapidly selected in biofilms, but not in planktonic cells. This is due to a combination of enhanced mutation rate, increased adhesion capacity and protective biofilm-associated tolerance. These results show that the biofilm environment favors rapid evolution of resistance and provide new insights into the dynamic evolution of antibiotic resistance in biofilms.
Collapse
Affiliation(s)
- Masaru Usui
- Laboratory of Food Microbiology and Food Safety, Department of Health and Environmental Sciences, School of Veterinary Medicine, Rakuno Gakuen University, Hokkaido, Japan.
- Institut Pasteur, Université de Paris Cité, UMR CNRS 6047, Genetics of Biofilms Laboratory, 75015, Paris, France.
| | - Yutaka Yoshii
- Institut Pasteur, Université de Paris Cité, UMR CNRS 6047, Genetics of Biofilms Laboratory, 75015, Paris, France
| | - Stanislas Thiriet-Rupert
- Institut Pasteur, Université de Paris Cité, UMR CNRS 6047, Genetics of Biofilms Laboratory, 75015, Paris, France
| | - Jean-Marc Ghigo
- Institut Pasteur, Université de Paris Cité, UMR CNRS 6047, Genetics of Biofilms Laboratory, 75015, Paris, France
| | - Christophe Beloin
- Institut Pasteur, Université de Paris Cité, UMR CNRS 6047, Genetics of Biofilms Laboratory, 75015, Paris, France.
| |
Collapse
|
34
|
Duran S, Anwar J, Moin ST. Interaction of gentamicin and gentamicin-AOT with poly-(lactide-co-glycolate) in a drug delivery system - density functional theory calculations and molecular dynamics simulation. Biophys Chem 2023; 294:106958. [PMID: 36682087 DOI: 10.1016/j.bpc.2023.106958] [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: 10/29/2022] [Revised: 11/23/2022] [Accepted: 01/12/2023] [Indexed: 01/15/2023]
Abstract
Gentamicin is used to treat brucellosis, an infectious disease caused by the Brucella species but the drug faces several issues such as low efficacy, instability, low solubility, and toxicity. It also has a very short half-life, therefore, requiring frequent dosing. Consequently, several other antibiotics are also being used for the treatment of brucellosis as a single dose as well as in combination with other antibiotics but none of these therapies are satisfactory. Nanoparticles in particular polymer-based ones utilizing polymers that are biodegradable and biocompatible for instance PLGA are a method of choice to overcome such drug delivery issues and enable potential targeted delivery. The current study focuses on the evaluation of the structural and dynamical properties of a drug-polymer system consisting of gentamicin drug and PLGA polymer nanoparticles in the water representing a targeted drug delivery system for the treatment of brucellosis. For this purpose, all-atom molecular dynamics simulations were carried out on the drug-polymer systems in the absence and presence of the surfactant bis(2-Ethylhexyl) sulfosuccinate (AOT) to determine the structural and dynamical properties as well as the effect of the surfactant on these properties. We also investigated systems in which the polymer constituents were in the form of monomeric units toward decoupling the primary interactions of the monomer units and polymer effects. The simulation results explain the nature of the interactions between the drug and the polymer as well as transport properties in terms of drug diffusion coefficients, which characterize the molecular behavior of gentamicin-polymer nanoparticles for use in brucellosis.
Collapse
Affiliation(s)
- Shahid Duran
- Third World Center for Science and Technology, H.E.J. Research, Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Jamshed Anwar
- Department of Chemistry, Lancaster University, Lancaster LA1 4YW, United Kingdom.
| | - Syed Tarique Moin
- Third World Center for Science and Technology, H.E.J. Research, Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| |
Collapse
|
35
|
Bie L, Zhang M, Wang J, Fang M, Li L, Xu H, Wang M. Comparative Analysis of Transcriptomic Response of Escherichia coli K-12 MG1655 to Nine Representative Classes of Antibiotics. Microbiol Spectr 2023; 11:e0031723. [PMID: 36853057 PMCID: PMC10100721 DOI: 10.1128/spectrum.00317-23] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 02/05/2023] [Indexed: 03/01/2023] Open
Abstract
The use of antibiotics leads to strong stresses to bacteria, leading to profound impact on cellular physiology. Elucidating how bacteria respond to antibiotic stresses not only helps us to decipher bacteria's strategies to resistant antibiotics but also assists in proposing targets for antibiotic development. In this work, a comprehensive comparative transcriptomic analysis on how Escherichia coli responds to nine representative classes of antibiotics (tetracycline, mitomycin C, imipenem, ceftazidime, kanamycin, ciprofloxacin, polymyxin E, erythromycin, and chloramphenicol) was performed, aimed at determining and comparing the responses of this model organism to antibiotics at the transcriptional level. On average, 39.71% of genes were differentially regulated by antibiotics at concentrations that inhibit 50% growth. Kanamycin leads to the strongest transcriptomic response (76.4% of genes regulated), whereas polymyxin E led to minimal transcriptomic response (4.7% of genes regulated). Further GO, KEGG, and EcoCyc enrichment analysis found significant transcriptomic changes in carbon metabolism, amino acid metabolism, nutrient assimilation, transport, stress response, nucleotide metabolism, protein biosynthesis, cell wall biosynthesis, energy conservation, mobility, and cell-environmental communications. Analysis of coregulated genes led to the finding of significant reduction of sulfur metabolism by all antibiotics, and analysis of transcription factor-coding genes suggested clustered regulatory patterns implying coregulation. In-depth analysis of regulated pathways revealed shared and unique strategies of E. coli resisting antibiotics, leading to the proposal of four different strategies (the pessimistic, the ignorant, the defensive, and the invasive). In conclusion, this work provides a comprehensive analysis of E. coli's transcriptomic response to antibiotics, which paves the road for further physiological investigation. IMPORTANCE Antibiotics are among the most important inventions in the history of humankind. They are the ultimate reason why bacterial infections are no longer the number one threat to people's lives. However, the wide application of antibiotics in the last half a century has led to aggravating antibiotic resistance, weakening the efficacy of antibiotics. To better comprehend the ways bacteria deal with antibiotics that may eventually turn into resistance mechanisms, and to identify good targets for potential antibiotics, knowledge on how bacteria regulate their physiology in response to different classes of antibiotics is needed. This work aimed to fill this knowledge gap by identifying changes of bacterial functions at the transcription level and suggesting strategies of bacteria to resist antibiotics.
Collapse
Affiliation(s)
- Luyao Bie
- State Key Laboratory of Microbial Technology, Microbial Technology Research Institute, Shandong University, Qingdao, China
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Mengge Zhang
- State Key Laboratory of Microbial Technology, Microbial Technology Research Institute, Shandong University, Qingdao, China
| | - Juan Wang
- State Key Laboratory of Microbial Technology, Microbial Technology Research Institute, Shandong University, Qingdao, China
- No.3 Middle School of Huimin, Binzhou, China
| | - Meng Fang
- State Key Laboratory of Microbial Technology, Microbial Technology Research Institute, Shandong University, Qingdao, China
| | - Ling Li
- State Key Laboratory of Microbial Technology, Microbial Technology Research Institute, Shandong University, Qingdao, China
| | - Hai Xu
- State Key Laboratory of Microbial Technology, Microbial Technology Research Institute, Shandong University, Qingdao, China
| | - Mingyu Wang
- State Key Laboratory of Microbial Technology, Microbial Technology Research Institute, Shandong University, Qingdao, China
| |
Collapse
|
36
|
Risk Factors of Clonally Related, Multi, and Extensively Drug-Resistant Acinetobacter baumannii in Severely Ill COVID-19 Patients. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2023; 2023:3139270. [PMID: 36814503 PMCID: PMC9940951 DOI: 10.1155/2023/3139270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/30/2022] [Accepted: 02/06/2023] [Indexed: 02/15/2023]
Abstract
Background The secondary infection of multi and extensively drug-resistant "Acinetobacter baumannii" in severely ill COVID-19 individuals is usually associated with extended hospitalisation and a high mortality rate. The current study aimed to assess the exact incidence rate of A. baumannii coinfection in severely ill COVID-19 patients admitted to intensive care unit (ICUs), to identify the possible mechanism of A. baumannii transfer to COVID-19 patients and to find out their resistance rate against different antibiotics. Methods Fifty severely ill "COVID-19" individuals on respiratory support were selected with samples being collected from the pharynx. In addition, another 60 samples were collected from the surrounding environment. Bacterial isolates were diagnosed by microbiological cultures and confirmed by "Vitek 2 system" and real-time PCR. The "Vitek 2 Compact system" was used to evaluate these isolates for antimicrobial susceptibility. The recovered isolates' DNA fingerprints and genetic similarities were performed using ERIC-PCR. Results Twenty-six samples were tested positive for A. baumannii (20 out of 50 samples taken from patients, 40%; 6 out of 60 swabs from a nosocomial setting, 10%). All A. baumannii strains isolated from the nosocomial sites were clonally related (have the same genetic lineage) to some strains isolated from patients. However, the majority of the patients' strains were categorised as belonging to the same genetic lineage. Furthermore, "the multi and extensively drug" resistance patterns were seen in all isolates. In addition, total isolates showed resistance to the most commonly tested antibiotics, while none of them was found to be resistant to tigecycline. Conclusion Secondary "A. baumannii" infection in severely ill "COVID-19" patients is a serious matter, especially when it has one spot of transmission in the ICU as well as when it is extensively drug-resistant, necessitating an immediate and tactical response to secure the issue.
Collapse
|
37
|
Dove AS, Dzurny DI, Dees WR, Qin N, Nunez Rodriguez CC, Alt LA, Ellward GL, Best JA, Rudawski NG, Fujii K, Czyż DM. Silver nanoparticles enhance the efficacy of aminoglycosides against antibiotic-resistant bacteria. Front Microbiol 2023; 13:1064095. [PMID: 36798870 PMCID: PMC9927651 DOI: 10.3389/fmicb.2022.1064095] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/30/2022] [Indexed: 02/04/2023] Open
Abstract
As the threat of antimicrobial-resistant bacteria compromises the safety and efficacy of modern healthcare practices, the search for effective treatments is more urgent than ever. For centuries, silver (Ag) has been known to have antibacterial properties and, over the past two decades, Ag-based nanoparticles have gained traction as potential antimicrobials. The antibacterial efficacy of Ag varies with structure, size, and concentration. In the present study, we examined Ag nanoparticles (AgNPs) for their antimicrobial activity and safety. We compared different commercially-available AgNPs against gram-negative Escherichia coli, Pseudomonas aeruginosa, Acinetobacter baumannii, and gram-positive Staphylococcus aureus methicillin-resistant and susceptible strains. The most effective formula of AgNPs tested had single-digit (μg/mL) minimum inhibitory concentrations against gram-negative multidrug-resistant clinical bacterial isolates with novel and emerging mechanisms of resistance. The mode of killing was assessed in E. coli and was found to be bactericidal, which is consistent with previous studies using other AgNP formulations. We evaluated cytotoxicity by measuring physiological readouts using the Caenorhabditis elegans model and found that motility was affected, but not the lifespan. Furthermore, we found that at their antibacterial concentrations, AgNPs were non-cytotoxic to any of the mammalian cell lines tested, including macrophages, stem cells, and epithelial cells. More interestingly, our experiments revealed synergy with clinically relevant antibiotics. We found that a non-toxic and non-effective concentration of AgNPs reduced the minimum inhibitory concentrations of aminoglycoside by approximately 22-fold. Because both aminoglycosides and Ag are known to target the bacterial ribosome, we tested whether Ag could also target eukaryotic ribosomes. We measured the rate of mistranslation at bactericidal concentration and found no effect, indicating that AgNPs are not proteotoxic to the host at the tested concentrations. Collectively, our results suggest that AgNPs could have a promising clinical application as a potential stand-alone therapy or antibiotic adjuvants.
Collapse
Affiliation(s)
- Autumn S. Dove
- Department of Microbiology and Cell Science, College of Agricultural and Life Sciences, University of Florida, Gainesville, FL, United States
| | - Dominika I. Dzurny
- Department of Microbiology and Cell Science, College of Agricultural and Life Sciences, University of Florida, Gainesville, FL, United States
| | - Wren R. Dees
- Department of Microbiology and Cell Science, College of Agricultural and Life Sciences, University of Florida, Gainesville, FL, United States
| | - Nan Qin
- Natural Immunogenics Corporation, Sarasota, FL, United States
| | | | - Lauren A. Alt
- Natural Immunogenics Corporation, Sarasota, FL, United States
| | - Garrett L. Ellward
- Department of Microbiology and Cell Science, College of Agricultural and Life Sciences, University of Florida, Gainesville, FL, United States
| | - Jacob A. Best
- Department of Microbiology and Cell Science, College of Agricultural and Life Sciences, University of Florida, Gainesville, FL, United States
| | - Nicholas G. Rudawski
- Research Service Centers, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, United States
| | - Kotaro Fujii
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, United States
- Center for NeuroGenetics, University of Florida, Gainesville, FL, United States
| | - Daniel M. Czyż
- Department of Microbiology and Cell Science, College of Agricultural and Life Sciences, University of Florida, Gainesville, FL, United States
| |
Collapse
|
38
|
Revitt‐Mills SA, Wright EK, Vereker M, O'Flaherty C, McPherson F, Dawson C, van Oijen AM, Robinson A. Defects in DNA double-strand break repair resensitize antibiotic-resistant Escherichia coli to multiple bactericidal antibiotics. Microbiologyopen 2022; 11:e1316. [PMID: 36314749 PMCID: PMC9500592 DOI: 10.1002/mbo3.1316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 09/03/2022] [Accepted: 09/03/2022] [Indexed: 11/11/2022] Open
Abstract
Antibiotic resistance is becoming increasingly prevalent amongst bacterial pathogens and there is an urgent need to develop new types of antibiotics with novel modes of action. One promising strategy is to develop resistance-breaker compounds, which inhibit resistance mechanisms and thus resensitize bacteria to existing antibiotics. In the current study, we identify bacterial DNA double-strand break repair as a promising target for the development of resistance-breaking co-therapies. We examined genetic variants of Escherichia coli that combined antibiotic-resistance determinants with DNA repair defects. We observed that defects in the double-strand break repair pathway led to significant resensitization toward five bactericidal antibiotics representing different functional classes. Effects ranged from partial to full resensitization. For ciprofloxacin and nitrofurantoin, sensitization manifested as a reduction in the minimum inhibitory concentration. For kanamycin and trimethoprim, sensitivity manifested through increased rates of killing at high antibiotic concentrations. For ampicillin, repair defects dramatically reduced antibiotic tolerance. Ciprofloxacin, nitrofurantoin, and trimethoprim induce the promutagenic SOS response. Disruption of double-strand break repair strongly dampened the induction of SOS by these antibiotics. Our findings suggest that if break-repair inhibitors can be developed they could resensitize antibiotic-resistant bacteria to multiple classes of existing antibiotics and may suppress the development of de novo antibiotic-resistance mutations.
Collapse
Affiliation(s)
- Sarah A. Revitt‐Mills
- School of Chemistry and Molecular Bioscience, Molecular Horizons InstituteUniversity of WollongongWollongongNew South WalesAustralia
- Illawarra Health and Medical Research InstituteWollongongNew South WalesAustralia
| | - Elizabeth K. Wright
- School of Chemistry and Molecular Bioscience, Molecular Horizons InstituteUniversity of WollongongWollongongNew South WalesAustralia
- Illawarra Health and Medical Research InstituteWollongongNew South WalesAustralia
| | - Madaline Vereker
- School of Chemistry and Molecular Bioscience, Molecular Horizons InstituteUniversity of WollongongWollongongNew South WalesAustralia
- Illawarra Health and Medical Research InstituteWollongongNew South WalesAustralia
| | - Callum O'Flaherty
- School of Chemistry and Molecular Bioscience, Molecular Horizons InstituteUniversity of WollongongWollongongNew South WalesAustralia
- Illawarra Health and Medical Research InstituteWollongongNew South WalesAustralia
| | - Fairley McPherson
- School of Chemistry and Molecular Bioscience, Molecular Horizons InstituteUniversity of WollongongWollongongNew South WalesAustralia
- Illawarra Health and Medical Research InstituteWollongongNew South WalesAustralia
| | - Catherine Dawson
- School of Chemistry and Molecular Bioscience, Molecular Horizons InstituteUniversity of WollongongWollongongNew South WalesAustralia
- Illawarra Health and Medical Research InstituteWollongongNew South WalesAustralia
| | - Antoine M. van Oijen
- School of Chemistry and Molecular Bioscience, Molecular Horizons InstituteUniversity of WollongongWollongongNew South WalesAustralia
- Illawarra Health and Medical Research InstituteWollongongNew South WalesAustralia
| | - Andrew Robinson
- School of Chemistry and Molecular Bioscience, Molecular Horizons InstituteUniversity of WollongongWollongongNew South WalesAustralia
- Illawarra Health and Medical Research InstituteWollongongNew South WalesAustralia
| |
Collapse
|
39
|
Zhou J, Cai Y, Liu Y, An H, Deng K, Ashraf MA, Zou L, Wang J. Breaking down the cell wall: Still an attractive antibacterial strategy. Front Microbiol 2022; 13:952633. [PMID: 36212892 PMCID: PMC9544107 DOI: 10.3389/fmicb.2022.952633] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/18/2022] [Indexed: 11/17/2022] Open
Abstract
Since the advent of penicillin, humans have known about and explored the phenomenon of bacterial inhibition via antibiotics. However, with changes in the global environment and the abuse of antibiotics, resistance mechanisms have been selected in bacteria, presenting huge threats and challenges to the global medical and health system. Thus, the study and development of new antimicrobials is of unprecedented urgency and difficulty. Bacteria surround themselves with a cell wall to maintain cell rigidity and protect against environmental insults. Humans have taken advantage of antibiotics to target the bacterial cell wall, yielding some of the most widely used antibiotics to date. The cell wall is essential for bacterial growth and virulence but is absent from humans, remaining a high-priority target for antibiotic screening throughout the antibiotic era. Here, we review the extensively studied targets, i.e., MurA, MurB, MurC, MurD, MurE, MurF, Alr, Ddl, MurI, MurG, lipid A, and BamA in the cell wall, starting from the very beginning to the latest developments to elucidate antimicrobial screening. Furthermore, recent advances, including MraY and MsbA in peptidoglycan and lipopolysaccharide, and tagO, LtaS, LspA, Lgt, Lnt, Tol-Pal, MntC, and OspA in teichoic acid and lipoprotein, have also been profoundly discussed. The review further highlights that the application of new methods such as macromolecular labeling, compound libraries construction, and structure-based drug design will inspire researchers to screen ideal antibiotics.
Collapse
Affiliation(s)
- Jingxuan Zhou
- The People’s Hospital of China Three Gorges University, Yichang, Hubei, China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China
- The Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China
| | - Yi Cai
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China
- The Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China
| | - Ying Liu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China
- The Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China
| | - Haoyue An
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China
- The Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China
| | - Kaihong Deng
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China
- The Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China
| | - Muhammad Awais Ashraf
- Department of Microbiology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Lili Zou
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China
- The Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China
| | - Jun Wang
- The People’s Hospital of China Three Gorges University, Yichang, Hubei, China
- *Correspondence: Jun Wang,
| |
Collapse
|
40
|
Activation of the SigE-SigB signaling pathway by inhibition of the respiratory electron transport chain and its effect on rifampicin resistance in Mycobacterium smegmatis. J Microbiol 2022; 60:935-947. [DOI: 10.1007/s12275-022-2202-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 10/16/2022]
|
41
|
Núñez-Batalla F, Jáudenes-Casaubón C, Sequí-Canet JM, Vivanco-Allende A, Zubicaray-Ugarteche J. Ototoxicity in childhood: Recommendations of the CODEPEH (Commission for the Early Detection of Childhood Hearing Loss) for prevention and early diagnosis. ACTA OTORRINOLARINGOLOGICA ESPANOLA 2022; 73:255-265. [PMID: 35872300 DOI: 10.1016/j.otoeng.2022.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/18/2021] [Indexed: 11/22/2022]
Abstract
Ototoxicity is defined as the damage, reversible or irreversible, produced in the inner ear by various substances that are called ototoxic and that can cause hearing loss and/or an alteration of the vestibular system. Permanent hearing loss significantly affects quality of life and is especially important in children. The lack or delay in its detection is frequent, since it often progresses in an inconspicuous manner until it affects communication and overall development. This impact can be minimized by following a strategy of audiological monitoring of ototoxicity, which allows for its early detection and treatment. This document recommends that children who are going to be treated with cisplatin or aminoglycosides be monitored. This CODEPEH review and recommendation document focuses on the early detection, prophylaxis, otoprotection, monitoring and treatment of ototoxicity caused by aminoglycosides and platinum-based antineoplastics in the paediatric population.
Collapse
|
42
|
Ototoxicidad en la edad pediátrica: recomendaciones de la CODEPEH (Comisión para la Detección Precoz de la Hipoacusia infantil) para su prevención y diagnóstico precoz. ACTA OTORRINOLARINGOLOGICA ESPANOLA 2022. [DOI: 10.1016/j.otorri.2021.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
43
|
Abreu RBV, Gomes TT, Nepomuceno TC, Li X, Fuchshuber-Moraes M, De Gregoriis G, Suarez-Kurtz G, Monteiro ANA, Carvalho MA. Functional Restoration of BRCA1 Nonsense Mutations by Aminoglycoside-Induced Readthrough. Front Pharmacol 2022; 13:935995. [PMID: 35837282 PMCID: PMC9273842 DOI: 10.3389/fphar.2022.935995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
BRCA1 is a major tumor suppressor that functions in the accurate repair of DNA double-strand breaks via homologous recombination (HR). Nonsense mutations in BRCA1 lead to inactive truncated protein products and are associated with high risk of breast and ovarian cancer. These mutations generate premature termination codons (PTCs). Different studies have shown that aminoglycosides can induce PTC suppression by promoting stop codon readthrough and restoring full-length (FL) protein expression. The use of these compounds has been studied in clinical trials for genetic diseases such as cystic fibrosis and Duchenne muscular dystrophy, with encouraging results. Here we show proof-of-concept data demonstrating that the aminoglycoside G418 can induce BRCA1 PTC readthrough and restore FL protein synthesis and function. We first demonstrate that G418 treatment restores BRCA1 FL protein synthesis in HCC1395, a human breast tumor cell line carrying the R1751X mutation. HCC1395 cells treated with G418 also recover HR DNA repair and restore cell cycle checkpoint activation. A set of naturally occurring BRCA1 nonsense variants encoding different PTCs was evaluated in a GFP C-terminal BRCA1 construct model and BRCA1 PTC readthrough levels vary depending on the stop codon context. Because PTC readthrough could generate FL protein carrying pathogenic missense mutations, variants representing the most probable acquired amino acid substitutions in consequence of readthrough were functionally assessed by a validated transcription activation assay. Overall, this is the first study that evaluates the readthrough of PTC variants with clinical relevance in the breast and ovarian cancer-predisposing gene BRCA1.
Collapse
Affiliation(s)
- Renata B. V. Abreu
- Divisão de Pesquisa Clínica, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Thiago T. Gomes
- Divisão de Pesquisa Clínica, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Thales C. Nepomuceno
- Divisão de Pesquisa Clínica, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
- Cancer Epidemiology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Xueli Li
- Cancer Epidemiology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | | | | | | | - Alvaro N. A. Monteiro
- Cancer Epidemiology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Marcelo A. Carvalho
- Divisão de Pesquisa Clínica, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
- Instituto Federal do Rio de Janeiro—IFRJ, Rio de Janeiro, Brazil
- *Correspondence: Marcelo A. Carvalho,
| |
Collapse
|
44
|
Wijers CDM, Pham L, Douglass MV, Skaar EP, Palmer LD, Noto MJ. Gram-negative bacteria act as a reservoir for aminoglycoside antibiotics that interact with host factors to enhance bacterial killing in a mouse model of pneumonia. FEMS MICROBES 2022; 3:xtac016. [PMID: 35909464 PMCID: PMC9326624 DOI: 10.1093/femsmc/xtac016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/04/2022] [Accepted: 05/11/2022] [Indexed: 01/28/2023] Open
Abstract
In vitro exposure of multiple Gram-negative bacteria to an aminoglycoside (AG) antibiotic has previously been demonstrated to result in bacterial alterations that interact with host factors to suppress Gram-negative pneumonia. However, the mechanisms resulting in suppression are not known. Here, the hypothesis that Gram-negative bacteria bind and retain AGs, which are introduced into the lung and interact with host defenses to affect bacterial killing, was tested. Following in vitro exposure of one of several, pathogenic Gram-negative bacteria to the AG antibiotics kanamycin or gentamicin, AGs were detected in bacterial cell pellets (up to 208 μg/mL). Using inhibitors of AG binding and internalization, the bacterial outer membrane was implicated as the predominant kanamycin and gentamicin reservoir. Following intranasal administration of gentamicin-bound bacteria or gentamicin solution at the time of infection with live, AG-naïve bacteria, gentamicin was detected in the lungs of infected mice (up to 8 μg/g). Co-inoculation with gentamicin-bound bacteria resulted in killing of AG-naïve bacteria by up to 3-log10, mirroring the effects of intranasal gentamicin treatment. In vitro killing of AG-naïve bacteria mediated by kanamycin-bound bacteria required the presence of detergents or pulmonary surfactant, suggesting that increased bacterial killing inside the murine lung is facilitated by the detergent component of pulmonary surfactant. These findings demonstrate that Gram-negative bacteria bind and retain AGs that can interact with host-derived pulmonary surfactant to enhance bacterial killing in the lung. This may help explain why AGs appear to have unique efficacy in the lung and might expand their clinical utility.
Collapse
Affiliation(s)
- Christiaan D M Wijers
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, United States
| | - Ly Pham
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, United States
| | - Martin V Douglass
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, United States
| | - Eric P Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, United States
| | - Lauren D Palmer
- Department of Microbiology and Immunology, University of Illinois Chicago, 835 South Wolcott Avenue, Chicago, IL 60612, United States
| | - Michael J Noto
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, United States
- Department of Medicine, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, United States
| |
Collapse
|
45
|
Rodriguez GM, Sharma N, Biswas A, Sharma N. The Iron Response of Mycobacterium tuberculosis and Its Implications for Tuberculosis Pathogenesis and Novel Therapeutics. Front Cell Infect Microbiol 2022; 12:876667. [PMID: 35646739 PMCID: PMC9132128 DOI: 10.3389/fcimb.2022.876667] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/25/2022] [Indexed: 12/16/2022] Open
Abstract
Most pathogenic bacteria require iron for growth. However, this metal is not freely available in the mammalian host. Due to its poor solubility and propensity to catalyze the generation of reactive oxygen species, host iron is kept in solution bound to specialized iron binding proteins. Access to iron is an important factor in the outcome of bacterial infections; iron limitation frequently induces virulence and drives pathogenic interactions with host cells. Here, we review the response of Mycobacterium tuberculosis to changes in iron availability, the relevance of this response to TB pathogenesis, and its potential for the design of new therapeutic interventions.
Collapse
|
46
|
Milicaj J, Hassan BA, Cote JM, Ramirez-Mondragon CA, Jaunbocus N, Rafalowski A, Patel KR, Castro CD, Muthyala R, Sham YY, Taylor EA. Discovery of first-in-class nanomolar inhibitors of heptosyltransferase I reveals a new aminoglycoside target and potential alternative mechanism of action. Sci Rep 2022; 12:7302. [PMID: 35508636 PMCID: PMC9068772 DOI: 10.1038/s41598-022-10776-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 04/04/2022] [Indexed: 11/08/2022] Open
Abstract
A clinically relevant inhibitor for Heptosyltransferase I (HepI) has been sought after for many years because of its critical role in the biosynthesis of lipopolysaccharides on bacterial cell surfaces. While many labs have discovered or designed novel small molecule inhibitors, these compounds lacked the bioavailability and potency necessary for therapeutic use. Extensive characterization of the HepI protein has provided valuable insight into the dynamic motions necessary for catalysis that could be targeted for inhibition. Structural inspection of Kdo2-lipid A suggested aminoglycoside antibiotics as potential inhibitors for HepI. Multiple aminoglycosides have been experimentally validated to be first-in-class nanomolar inhibitors of HepI, with the best inhibitor demonstrating a Ki of 600 ± 90 nM. Detailed kinetic analyses were performed to determine the mechanism of inhibition while circular dichroism spectroscopy, intrinsic tryptophan fluorescence, docking, and molecular dynamics simulations were used to corroborate kinetic experimental findings. While aminoglycosides have long been described as potent antibiotics targeting bacterial ribosomes' protein synthesis leading to disruption of the stability of bacterial cell membranes, more recently researchers have shown that they only modestly impact protein production. Our research suggests an alternative and novel mechanism of action of aminoglycosides in the inhibition of HepI, which directly leads to modification of LPS production in vivo. This finding could change our understanding of how aminoglycoside antibiotics function, with interruption of LPS biosynthesis being an additional and important mechanism of aminoglycoside action. Further research to discern the microbiological impact of aminoglycosides on cells is warranted, as inhibition of the ribosome may not be the sole and primary mechanism of action. The inhibition of HepI by aminoglycosides may dramatically alter strategies to modify the structure of aminoglycosides to improve the efficacy in fighting bacterial infections.
Collapse
Affiliation(s)
- Jozafina Milicaj
- Department of Chemistry, Wesleyan University, Middletown, CT, 06459, USA
| | - Bakar A Hassan
- Department of Chemistry, Wesleyan University, Middletown, CT, 06459, USA
| | - Joy M Cote
- Department of Chemistry, Wesleyan University, Middletown, CT, 06459, USA
| | | | - Nadiya Jaunbocus
- Department of Chemistry, Wesleyan University, Middletown, CT, 06459, USA
| | | | - Kaelan R Patel
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Colleen D Castro
- Department of Chemistry, Wesleyan University, Middletown, CT, 06459, USA
| | - Ramaiah Muthyala
- Department of Experimental and Clinical Pharmacology, College Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Yuk Y Sham
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, MN, 55455, USA.
- Bioinformatics and Computational Biology Program, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - Erika A Taylor
- Department of Chemistry, Wesleyan University, Middletown, CT, 06459, USA.
| |
Collapse
|
47
|
Obszynski J, Loidon H, Blanc A, Weibel JM, Pale P. Targeted modifications of neomycin and paromomycin: Towards resistance-free antibiotics? Bioorg Chem 2022; 126:105824. [DOI: 10.1016/j.bioorg.2022.105824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 04/10/2022] [Accepted: 04/19/2022] [Indexed: 12/01/2022]
|
48
|
Kim J, Hemachandran S, Cheng AG, Ricci AJ. Identifying targets to prevent aminoglycoside ototoxicity. Mol Cell Neurosci 2022; 120:103722. [PMID: 35341941 PMCID: PMC9177639 DOI: 10.1016/j.mcn.2022.103722] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/14/2022] [Accepted: 03/19/2022] [Indexed: 12/21/2022] Open
Abstract
Aminoglycosides are potent antibiotics that are commonly prescribed worldwide. Their use carries significant risks of ototoxicity by directly causing inner ear hair cell degeneration. Despite their ototoxic side effects, there are currently no approved antidotes. Here we review recent advances in our understanding of aminoglycoside ototoxicity, mechanisms of drug transport, and promising sites for intervention to prevent ototoxicity.
Collapse
Affiliation(s)
- Jinkyung Kim
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sriram Hemachandran
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alan G Cheng
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Anthony J Ricci
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
49
|
Tawde M, Bior A, Feiss M, Teng F, Freimuth P. A polypeptide model for toxic aberrant proteins induced by aminoglycoside antibiotics. PLoS One 2022; 17:e0258794. [PMID: 35486612 PMCID: PMC9053816 DOI: 10.1371/journal.pone.0258794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 03/01/2022] [Indexed: 11/24/2022] Open
Abstract
Aminoglycoside antibiotics interfere with the selection of cognate tRNAs during translation, resulting in the synthesis of aberrant proteins that are the ultimate cause of cell death. However, the toxic potential of aberrant proteins and how they avoid degradation by the cell’s protein quality control (QC) machinery are not understood. Here we report that levels of the heat shock (HS) transcription factor σ32 increased sharply following exposure of Escherichia coli to the aminoglycoside kanamycin (Kan), suggesting that at least some of the aberrant proteins synthesized in these cells were recognized as substrates by DnaK, a molecular chaperone that regulates the HS response, the major protein QC pathway in bacteria. To further investigate aberrant protein toxic potential and interaction with cell QC factors, we studied an acutely toxic 48-residue polypeptide (ARF48) that is encoded by an alternate reading frame in a plant cDNA. As occurred in cells exposed to Kan, σ32 levels were strongly elevated following ARF48 expression, suggesting that ARF48 was recognized as a substrate by DnaK. Paradoxically, an internal 10-residue region that was tightly bound by DnaK in vitro also was required for the ARF48 toxic effect. Despite the increased levels of σ32, levels of several HS proteins were unchanged following ARF48 expression, suggesting that the HS response had been aborted. Nucleoids were condensed and cell permeability increased rapidly following ARF48 expression, together suggesting that ARF48 disrupts DNA-membrane interactions that could be required for efficient gene expression. Our results are consistent with earlier studies showing that aberrant proteins induced by aminoglycoside antibiotics disrupt cell membrane integrity. Insights into the mechanism for this effect could be gained by further study of the ARF48 model system.
Collapse
Affiliation(s)
- Mangala Tawde
- Department of Biological Sciences and Geology, Queensborough Community College, City University of New York, Bayside, New York, United States of America
| | - Abdelaziz Bior
- Department of Natural and Applied Sciences, Cheyney University of Pennsylvania, Cheyney, Pennsylvania, United States of America
| | - Michael Feiss
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Feiyue Teng
- Center for Functional Nanomaterials, Brookhaven National Laboratory, Upton, New York, United States of America
| | - Paul Freimuth
- Biology Department, Brookhaven National Laboratory, Upton, New York, United States of America
- * E-mail:
| |
Collapse
|
50
|
Lu C, Zhang N, Kou S, Gao L, Peng B, Dai Y, Zheng J. Sanguinarine synergistically potentiates aminoglycoside-mediated bacterial killing. Microb Biotechnol 2022; 15:2055-2070. [PMID: 35318794 PMCID: PMC9249330 DOI: 10.1111/1751-7915.14017] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 02/01/2022] [Indexed: 11/30/2022] Open
Abstract
Aminoglycosides are one of the oldest classes of antimicrobials that are being used in current clinical practice, especially on multi‐drug resistant Gram‐negative pathogenic bacteria. However, the serious side effects at high dosage such as ototoxicity, neuropathy and nephrotoxicity limit their applications in clinical practice. Approaches that potentiate aminoglycoside killing could lower down their effective concentrations to a non‐toxic dosage for clinical treatment. In this research, we screened a compound library and identified sanguinarine that acts synergistically with various aminoglycosides. By checkerboard and dynamical killing assay, we found that sanguinarine effectively potentiated aminoglycoside killing on diverse bacterial pathogens, including Escherichia coli, Acinetobacter baumannii, Klebsiella pneumonia and Pseudomonas aeruginosa. The mechanistic studies showed an elevated intracellular ROS and DNA oxidative level in the bacterial cells treated by a combination of sanguinarine with aminoglycosides. Furthermore, an enhanced level of sanguinarine was observed in bacteria in the presence of aminoglycosides, suggesting that aminoglycosides promote the uptake of sanguinarine. Importantly, sanguinarine was shown to promote the elimination of persister cells and established biofilm cells both in vivo and in vitro. Our study provides a novel insight for approaches to lower down the clinical dosages of aminoglycosides.
Collapse
Affiliation(s)
- Chang Lu
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Nian Zhang
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Sihoi Kou
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Liangliang Gao
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Bo Peng
- School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, China
| | - Yunlu Dai
- Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translational Medicine, University of Macau, Macau, China
| | - Jun Zheng
- Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translational Medicine, University of Macau, Macau, China
| |
Collapse
|