1
|
Farazandehnia N, Sotoudegan F, Sepahy AA, Fazeli MR. Antibacterial and antioxidant properties of sumac extract on methicillin-resistant Staphylococcus aureus. AMB Express 2024; 14:111. [PMID: 39361209 PMCID: PMC11450108 DOI: 10.1186/s13568-024-01759-4] [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: 02/23/2024] [Accepted: 08/27/2024] [Indexed: 10/06/2024] Open
Abstract
The research aimed to evaluate the antioxidative and antibacterial characteristics of aqueous sumac extract on methicillin-resistant Staphylococcus aureus through in-vitro and in-vivo study. Sumac extract has been obtained through the soaking method, and its antioxidant properties were gauged using the DPPH free radical scavenging method. The minimum inhibitory concentration (MIC) of sumac extract was determined on S. aureus obtained from hospitalized patients, as well as an assessment of biofilm-formation and the release of bacterial intracellular compounds. in vivo experimentation involved injecting bacteria (108 cfu/ml) into mice, which subsequently manifested indicators of symptoms of infection, and the number of bacteria within their bloodstream was quantified. The Sumac extract demonstrated strong antioxidant properties at concentrations of 1000 mg/ml. Furthermore, the agar tests for the gram staining, mannitol, coagulase, and DNase revealed that 190 cultured bacteria samples were identified as Staphylococcus aureus. These bacteria were resistant to clindamycin, ciprofloxacin, and methicillin antibiotics, but sensitive to erythromycin and penicillin antibiotics. Additionally, the bacteria displayed significant methicillin resistance and formed a strong biofilm (65.78%). The sumac extract showed a MIC range of 125-1000 µg/ml against Staphylococcus aureus. Treatment with concentrations above the MIC was found to prevent the formation of biofilm and increase the release of bacterial intracellular compounds. Sumac extract led to a decrease in bacterial count in the blood of mice and reduced signs of infection. Sumac extract demonstrated powerful antioxidant and antibacterial effects against resistant microorganisms, suggesting its potential as a promising compound for the treatment of resistant infections in future research.
Collapse
Affiliation(s)
- Nafiseh Farazandehnia
- Department of Microbiology, Faculty of Biological Science, North of Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Farzaneh Sotoudegan
- Quality Control of Medicines and Supplements Group, Pharmaceutical Quality Assurance Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Akhavan Sepahy
- Department of Microbiology, Faculty of Biological Science, North of Tehran Branch, Islamic Azad University, Tehran, Iran.
| | - Mohamad Reza Fazeli
- Quality Control of Medicines and Supplements Group, Pharmaceutical Quality Assurance Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.
- Department of Drug and Food Control, Pharmaceutical Quality Assurance Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Abbas Akhavan Sepahy, Iran.
| |
Collapse
|
2
|
Jandl B, Dighe S, Gasche C, Makristathis A, Muttenthaler M. Intestinal biofilms: pathophysiological relevance, host defense, and therapeutic opportunities. Clin Microbiol Rev 2024; 37:e0013323. [PMID: 38995034 PMCID: PMC11391705 DOI: 10.1128/cmr.00133-23] [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] [Indexed: 07/13/2024] Open
Abstract
SUMMARYThe human intestinal tract harbors a profound variety of microorganisms that live in symbiosis with the host and each other. It is a complex and highly dynamic environment whose homeostasis directly relates to human health. Dysbiosis of the gut microbiota and polymicrobial biofilms have been associated with gastrointestinal diseases, including irritable bowel syndrome, inflammatory bowel diseases, and colorectal cancers. This review covers the molecular composition and organization of intestinal biofilms, mechanistic aspects of biofilm signaling networks for bacterial communication and behavior, and synergistic effects in polymicrobial biofilms. It further describes the clinical relevance and diseases associated with gut biofilms, the role of biofilms in antimicrobial resistance, and the intestinal host defense system and therapeutic strategies counteracting biofilms. Taken together, this review summarizes the latest knowledge and research on intestinal biofilms and their role in gut disorders and provides directions toward the development of biofilm-specific treatments.
Collapse
Affiliation(s)
- Bernhard Jandl
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Vienna, Austria
- Vienna Doctoral School in Chemistry (DoSChem), University of Vienna, Vienna, Austria
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Satish Dighe
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Christoph Gasche
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
- Loha for Life, Center for Gastroenterology and Iron Deficiency, Vienna, Austria
| | - Athanasios Makristathis
- Department of Laboratory Medicine, Division of Clinical Microbiology, Medical University of Vienna, Vienna, Austria
| | - Markus Muttenthaler
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Vienna, Austria
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
3
|
Alsamhary K. Evaluating the Expression of Efflux Pumps in Pseudomonas aeruginosa in Exposure to Sodium Dodecyl Sulfate, Didecyldimethylammonium Chloride, and Octenidine Dihydrochloride. Microb Drug Resist 2024; 30:385-390. [PMID: 39082183 DOI: 10.1089/mdr.2024.0070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024] Open
Abstract
Emerging resistance of Gram-negative bacteria, including Pseudomonas aeruginosa, to commonly used detergents and disinfectant is encountering us with hazard. Inappropriate use of disinfectants has forced bacteria to gain resistance. The ability of bacteria to extrude substrates from the cellular interior to the external environment has enabled them to persist in exposure to toxic compounds, which is due to existence of transport proteins. Efflux pumps, in Gram-negative bacteria, are proteins responsible for exporting molecules outside of the cell, by crossing the two membranes. In this study, 40 P. aeruginosa strains from hospitals, clinics, and burn center laundries and 40 P. aeruginosa strains from urban laundries were collected. This study evaluated the minimum inhibitory concentration (MIC) level of sodium dodecyl sulfate (SDS), didecyldimethylammonium chloride (DDAC), and octenidine dihydrochloride (Od) in P. aeruginosa strains. The real-time PCR was carried out to evaluate the expression of MexAB-OprM, MexCD-OprJ, and MexXY-OprM efflux system. The obtained results indicated a higher MIC level for SDS, DDAC, and Od in medical laundries. The sub-MIC level of DDAC and Od increased the expression level of MexAB-OprM, MexCD-OprJ, and MexXY-OprM in P. aeruginosa strains, suggesting that efflux pumps contribute to disinfectant resistance in P. aeruginosa.
Collapse
Affiliation(s)
- Khawla Alsamhary
- Department of Biology, College of Science and Humanities in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| |
Collapse
|
4
|
Belay WY, Getachew M, Tegegne BA, Teffera ZH, Dagne A, Zeleke TK, Abebe RB, Gedif AA, Fenta A, Yirdaw G, Tilahun A, Aschale Y. Mechanism of antibacterial resistance, strategies and next-generation antimicrobials to contain antimicrobial resistance: a review. Front Pharmacol 2024; 15:1444781. [PMID: 39221153 PMCID: PMC11362070 DOI: 10.3389/fphar.2024.1444781] [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: 06/06/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Antibacterial drug resistance poses a significant challenge to modern healthcare systems, threatening our ability to effectively treat bacterial infections. This review aims to provide a comprehensive overview of the types and mechanisms of antibacterial drug resistance. To achieve this aim, a thorough literature search was conducted to identify key studies and reviews on antibacterial resistance mechanisms, strategies and next-generation antimicrobials to contain antimicrobial resistance. In this review, types of resistance and major mechanisms of antibacterial resistance with examples including target site modifications, decreased influx, increased efflux pumps, and enzymatic inactivation of antibacterials has been discussed. Moreover, biofilm formation, and horizontal gene transfer methods has also been included. Furthermore, measures (interventions) taken to control antimicrobial resistance and next-generation antimicrobials have been discussed in detail. Overall, this review provides valuable insights into the diverse mechanisms employed by bacteria to resist the effects of antibacterial drugs, with the aim of informing future research and guiding antimicrobial stewardship efforts.
Collapse
Affiliation(s)
- Wubetu Yihunie Belay
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Melese Getachew
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Bantayehu Addis Tegegne
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Zigale Hibstu Teffera
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Abebe Dagne
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Tirsit Ketsela Zeleke
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Rahel Belete Abebe
- Department of clinical pharmacy, College of medicine and health sciences, University of Gondar, Gondar, Ethiopia
| | - Abebaw Abie Gedif
- Department of Pharmacy, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Abebe Fenta
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Getasew Yirdaw
- Department of environmental health science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Adane Tilahun
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Yibeltal Aschale
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
5
|
Zuberi A, Ahmad N, Ahmad H, Saeed M, Ahmad I. Beyond antibiotics: CRISPR/Cas9 triumph over biofilm-associated antibiotic resistance infections. Front Cell Infect Microbiol 2024; 14:1408569. [PMID: 39035353 PMCID: PMC11257871 DOI: 10.3389/fcimb.2024.1408569] [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: 03/28/2024] [Accepted: 05/27/2024] [Indexed: 07/23/2024] Open
Abstract
A complex structure known as a biofilm is formed when a variety of bacterial colonies or a single type of cell in a group sticks to a surface. The extracellular polymeric compounds that encase these cells, often consisting of proteins, eDNA, and polysaccharides, exhibit strong antibiotic resistance. Concerns about biofilm in the pharmaceutical industry, public health, and medical fields have sparked a lot of interest, as antibiotic resistance is a unique capacity exhibited by these biofilm-producing bacteria, which increases morbidity and death. Biofilm formation is a complicated process that is controlled by several variables. Insights into the processes to target for the therapy have been gained from multiple attempts to dissect the biofilm formation process. Targeting pathogens within a biofilm is profitable because the bacterial pathogens become considerably more resistant to drugs in the biofilm state. Although biofilm-mediated infections can be lessened using the currently available medications, there has been a lot of focus on the development of new approaches, such as bioinformatics tools, for both treating and preventing the production of biofilms. Technologies such as transcriptomics, metabolomics, nanotherapeutics and proteomics are also used to develop novel anti-biofilm agents. These techniques help to identify small compounds that can be used to inhibit important biofilm regulators. The field of appropriate control strategies to avoid biofilm formation is expanding quickly because of this spurred study. As a result, the current article addresses our current knowledge of how biofilms form, the mechanisms by which bacteria in biofilms resist antibiotics, and cutting-edge treatment approaches for infections caused by biofilms. Furthermore, we have showcased current ongoing research utilizing the CRISPR/Cas9 gene editing system to combat bacterial biofilm infections, particularly those brought on by lethal drug-resistant pathogens, concluded the article with a novel hypothesis and aspirations, and acknowledged certain limitations.
Collapse
Affiliation(s)
- Azna Zuberi
- Department of Molecular, Cellular & Developmental Biology, University of Colorado Boulder, Boulder, CO, United States
- Department of Obs & Gynae, Northwestern University, Chicago, IL, United States
| | - Nayeem Ahmad
- Department of Biophysics, All India Institute of Medical Science, New Delhi, India
- Department of Microbiology, Immunology, and Infectious Diseases, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Hafiz Ahmad
- Department of Medical Microbiology & Immunology, Ras Al Khaimah (RAK) College of Medical Sciences, Ras Al Khaimah (RAK) Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Mohd Saeed
- Department of Biology, College of Science University of Hail, Hail, Saudi Arabia
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
6
|
Banerjee A, Singh P, Sheikh PA, Kumar A, Koul V, Bhattacharyya J. A multifunctional silk-hyaluronic acid self-healing hydrogel laden with alternatively activated macrophage-derived exosomes reshape microenvironment of diabetic wound and accelerate healing. Int J Biol Macromol 2024; 270:132384. [PMID: 38754682 DOI: 10.1016/j.ijbiomac.2024.132384] [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: 01/25/2024] [Revised: 04/14/2024] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
The impairment of phenotype switching of pro-inflammatory M1 to pro-healing M2 macrophage induced by hyperglycemic microenvironment often elevates oxidative stress, impairs angiogenesis, and leads to chronic non-healing wounds in diabetic patients. Administration of M2 macrophage-derived exosomes (M2Exo) at wound site is known to polarize M1 to M2 macrophage and can accelerate wound healing by enhancing collagen deposition, angiogenesis, and re-epithelialization. In the present study, M2Exo were conjugated with oxidized hyaluronic acid and mixed with PEGylated silk fibroin to develop self-healing Exo-gel to achieve an efficient therapy for diabetic wounds. Exo-gel depicted porous networked morphology with self-healing and excellent water retention behaviour. Fibroblast cells treated with Exo-gel showed significant uptake of M2Exo that increased their proliferation and migration in vitro. Interestingly, in a diabetic wound model of wistar rats, Exo-gel treatment induced 75 % wound closure within 7 days with complete epithelial layer regeneration by modulating cytokine levels, stimulating fibroblast-keratinocyte interaction and migration, angiogenesis, and organized collagen deposition. Taken together, this study suggests that Exo-gel depict properties of an excellent wound healing matrix and can be used as a therapeutic alternative to treat chronic non-healing diabetic wounds.
Collapse
Affiliation(s)
- Ahana Banerjee
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi-110016, India; Department of Biomedical Engineering, All India Institute of Medical Science, Delhi, New Delhi-110029, India
| | - Prerna Singh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kalyanpur, Kanpur, Uttar Pradesh-208016, India; Centre for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kalyanpur, Kanpur, Uttar Pradesh-208016, India
| | - Parvaiz A Sheikh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi-110016, India
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kalyanpur, Kanpur, Uttar Pradesh-208016, India; Centre for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kalyanpur, Kanpur, Uttar Pradesh-208016, India; The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kalyanpur, Kanpur, Uttar Pradesh-208016, India; Centre of Excellence for Orthopedics and Prosthetics, Indian Institute of Technology Kanpur, Kalyanpur, Kanpur, Uttar Pradesh-208016, India; Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kalyanpur, Kanpur, Uttar Pradesh-208016, India
| | - Veena Koul
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi-110016, India; Department of Biomedical Engineering, All India Institute of Medical Science, Delhi, New Delhi-110029, India
| | - Jayanta Bhattacharyya
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi-110016, India; Department of Biomedical Engineering, All India Institute of Medical Science, Delhi, New Delhi-110029, India.
| |
Collapse
|
7
|
Otite SV, Lag-Brotons AJ, Ezemonye LI, Martin AD, Pickup RW, Semple KT. Volatile Fatty Acids Effective as Antibacterial Agents against Three Enteric Bacteria during Mesophilic Anaerobic Incubation. Molecules 2024; 29:1908. [PMID: 38731399 PMCID: PMC11085169 DOI: 10.3390/molecules29091908] [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: 03/20/2024] [Revised: 04/19/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024] Open
Abstract
The antibacterial effects of a selection of volatile fatty acids (acetic, propionic, butyric, valeric, and caproic acids) relevant to anaerobic digestion were investigated at 1, 2 and 4 g/L. The antibacterial effects were characterised by the dynamics of Enterococcus faecalis NCTC 00775, Escherichia coli JCM 1649 and Klebsiella pneumoniae A17. Mesophilic anaerobic incubation to determine the minimum bactericidal concentration (MBC) and median lethal concentration of the VFAs was carried out in Luria Bertani broth at 37 °C for 48 h. Samples collected at times 0, 3, 6, 24 and 48 h were used to monitor bacterial kinetics and pH. VFAs at 4 g/L demonstrated the highest bactericidal effect (p < 0.05), while 1 g/L supported bacterial growth. The VFA cocktail was the most effective, while propionic acid was the least effective. Enterococcus faecalis NCTC 00775 was the most resistant strain with the VFAs MBC of 4 g/L, while Klebsiella pneumoniae A17 was the least resistant with the VFAs MBC of 2 g/L. Allowing a 48 h incubation period led to more log decline in the bacterial numbers compared to earlier times. The VFA cocktail, valeric, and caproic acids at 4 g/L achieved elimination of the three bacteria strains, with over 7 log10 decrease within 48 h.
Collapse
Affiliation(s)
- Saanu Victoria Otite
- Lancaster Environment Centre, Library Avenue, Lancaster University, Lancaster LA1 4YQ, UK;
| | | | - Lawrence I. Ezemonye
- Centre for Global Eco-Innovation Nigeria, University of Benin, Benin City PMB 300313, Nigeria
- Vice Chancellor’s Office, Igbinedion University Okada, Benin City PMB 0006, Nigeria
| | - Alastair D. Martin
- Engineering Department, Gillow Avenue, Lancaster University, Lancaster LA1 4YW, UK
| | - Roger W. Pickup
- Division of Biomedical and Life Sciences, Furness Building, Lancaster University, Lancaster LA1 4YG, UK
| | - Kirk T. Semple
- Lancaster Environment Centre, Library Avenue, Lancaster University, Lancaster LA1 4YQ, UK;
| |
Collapse
|
8
|
Chatterjee D, Panda AP, Daya Manasi AR, Ghosh AS. P-type ATPase zinc transporter Rv3270 of Mycobacterium tuberculosis enhances multi-drug efflux activity. MICROBIOLOGY (READING, ENGLAND) 2024; 170:001441. [PMID: 38373028 PMCID: PMC10924464 DOI: 10.1099/mic.0.001441] [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: 08/14/2023] [Accepted: 02/08/2024] [Indexed: 02/20/2024]
Abstract
Metal homeostasis is maintained by the uptake, storage and efflux of metal ions that are necessary for the survival of the bacterium. Homeostasis is mostly regulated by a group of transporters categorized as ABC transporters and P-type ATPases. On the other hand, efflux pumps often play a role in drug-metal cross-resistance. Here, with the help of antibiotic sensitivity, antibiotic/dye accumulation and semi-quantitative biofilm formation assessments we report the ability of Rv3270, a P-type ATPase known for its role in combating Mn2+ and Zn2+ metal ion toxicity in Mycobacterium tuberculosis, in influencing the extrusion of multiple structurally unrelated drugs and enhancing the biofilm formation of Escherichia coli and Mycobacterium smegmatis. Overexpression of Rv3270 increased the tolerance of host cells to norfloxacin, ofloxacin, sparfloxacin, ampicillin, oxacillin, amikacin and isoniazid. A significantly lower accumulation of norfloxacin, ethidium bromide, bocillin FL and levofloxacin in cells harbouring Rv3270 as compared to host cells indicated its role in enhancing efflux activity. Although over-expression of Rv3270 did not alter the susceptibility levels of levofloxacin, rifampicin and apramycin, the presence of a sub-inhibitory concentration of Zn2+ resulted in low-level tolerance towards these drugs. Of note, the expression of Rv3270 enhanced the biofilm-forming ability of the host cells strengthening its role in antimicrobial resistance. Therefore, the study indicated that the over-expression of Rv3270 enhances the drug efflux activity of the micro-organism where zinc might facilitate drug-metal cross-resistance for some antibiotics.
Collapse
Affiliation(s)
- Debasmita Chatterjee
- Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| | - Aditya Prasad Panda
- Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| | - A. R. Daya Manasi
- Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| | - Anindya S. Ghosh
- Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| |
Collapse
|
9
|
Lazarus E, Meyer AS, Ikuma K, Rivero IV. Three dimensional printed biofilms: Fabrication, design and future biomedical and environmental applications. Microb Biotechnol 2024; 17:e14360. [PMID: 38041693 PMCID: PMC10832517 DOI: 10.1111/1751-7915.14360] [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: 07/01/2023] [Revised: 10/02/2023] [Accepted: 10/11/2023] [Indexed: 12/03/2023] Open
Abstract
Three dimensional printing has emerged as a widely acceptable strategy for the fabrication of mammalian cell laden constructs with complex microenvironments for tissue engineering and regenerative medicine. More recently 3D printed living materials containing microorganisms have been developed and matured into living biofilms. The potential for engineered 3D biofilms as in vitro models for biomedical applications, such as antimicrobial susceptibility testing, and environmental applications, such as bioleaching, bioremediation, and wastewater purification, is extensive but the need for an in-depth understanding of the structure-function relationship between the complex construct and the microorganism response still exists. This review discusses 3D printing fabrication methods for engineered biofilms with specific structural features. Next, it highlights the importance of bioink compositions and 3D bioarchitecture design. Finally, a brief overview of current and potential applications of 3D printed biofilms in environmental and biomedical fields is discussed.
Collapse
Affiliation(s)
- Emily Lazarus
- Department Industrial and Systems EngineeringRochester Institute of TechnologyRochesterNew YorkUSA
| | - Anne S. Meyer
- Department of BiologyUniversity of RochesterRochesterNew YorkUSA
| | - Kaoru Ikuma
- Department of Civil, Construction, and Environmental EngineeringIowa State UniversityAmesIowaUSA
| | - Iris V. Rivero
- Department Industrial and Systems EngineeringRochester Institute of TechnologyRochesterNew YorkUSA
- Department of Biomedical EngineeringRochester Institute of TechnologyRochesterNew YorkUSA
- Department of Industrial and Systems EngineeringUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
10
|
Karimzadeh Barenji E, Beglari S, Tahghighi A, Azerang P, Rohani M. Evaluation of Anti-Bacterial and Anti-Biofilm Activity of Native Probiotic Strains of Lactobacillus Extracts. IRANIAN BIOMEDICAL JOURNAL 2023; 28:102-12. [PMID: 38850020 PMCID: PMC11186614 DOI: 10.61186/ibj.4043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 12/12/2023] [Indexed: 06/09/2024]
Abstract
Background Lactic acid bacteria produce various beneficial metabolites, including antimicrobial agents. Owing to the fast-rising antibiotic resistance among pathogenic microbes, scientists are exploring antimicrobials beyond antibiotics. In this study, we examined four Lactobacillus strains, namely L. plantarum 42, L. brevis 205, L. rhamnosus 239, and L. delbrueckii 263, isolated from healthy human microbiota, to evaluate their antibacterial and antifungal activity. Methods Lactobacillus strains were cultivated, and the conditioned media were obtained. The supernatant was then used to treat pathogenic bacteria and applied to the growth media containing fungal and bacterial strains. Additionally, the supernatant was separated to achieve the organic and aqueous phases. The two phases were then examined in terms of bacterial and fungal growth rates. Disk diffusion and MIC tests were conducted to determine strains with the most growth inhibition potential. Finally, the potent strains identified through the MIC test were tested on the pathogenic microorganisms to assess their effects on the formation of pathogenic biofilms. Results The organic phase of L. rhamnosus 239 extracts exhibited the highest antibacterial and antibiofilm effects, while that of L. brevis 205 demonstrated the most effective antifungal impact, with a MIC of 125 µg/mL against Saccharomyces cerevisiae. Conclusion This study confirms the significant antimicrobial impacts of the lactic acid bacteria strains on pathogenic bacteria and fungi; hence, they could serve as a reliable alternative to antibiotics for a safe and natural protection against pathogenic microorganisms.
Collapse
Affiliation(s)
- Elmira Karimzadeh Barenji
- Department of Biology, Science and Research branch, Islamic Azad University, Tehran, Iran
- Medicinal Chemistry Laboratory, Department of Clinical Research, Pasteur Institute of Iran, Tehran, Iran
| | - Shokufeh Beglari
- Department of Biology, Science and Research branch, Islamic Azad University, Tehran, Iran
| | - Azar Tahghighi
- Medicinal Chemistry Laboratory, Department of Clinical Research, Pasteur Institute of Iran, Tehran, Iran
| | - Parisa Azerang
- Medicinal Chemistry Laboratory, Department of Clinical Research, Pasteur Institute of Iran, Tehran, Iran
| | - Mahdi Rohani
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
- Research Center for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
11
|
Stevenson P, Marguet M, Regulski M. Biofilm and Hospital-Acquired Infections in Older Adults. Crit Care Nurs Clin North Am 2023; 35:375-391. [PMID: 37838413 DOI: 10.1016/j.cnc.2023.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2023]
Abstract
Biofilm infections are a serious threat to public health, resistant to traditional treatments and host immune defenses. Biofilm infections are often polymicrobial, related to chronic wounds, medical devices (eg, knee replacements, catheters, tubes, contact lenses, or prosthetic valves) and chronic recurring diseases. Biofilms are more complex than nonadhered planktonic bacteria and produce a structure that prevents damage to the bacteria within the biofilm structure. The structure provides a hidden route to feed and nurture the bacteria allowing for ongoing spread of the bacteria.
Collapse
Affiliation(s)
- Patricia Stevenson
- Next Science™ LLC, 10550 Deerwood Park Boulevard, Suite 300, Jacksonville, FL 32256, USA.
| | - Melissa Marguet
- Next Science™ LLC, 10550 Deerwood Park Boulevard, Suite 300, Jacksonville, FL 32256, USA
| | - Matthew Regulski
- Next Science™ LLC, 10550 Deerwood Park Boulevard, Suite 300, Jacksonville, FL 32256, USA; The Wound Institute of Ocean County, 54 Bey Lea Road Tom's River, NJ 08759, USA
| |
Collapse
|
12
|
Yaeger LN, French S, Brown ED, Côté JP, Burrows LL. Central metabolism is a key player in E. coli biofilm stimulation by sub-MIC antibiotics. PLoS Genet 2023; 19:e1011013. [PMID: 37917668 PMCID: PMC10645362 DOI: 10.1371/journal.pgen.1011013] [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: 07/22/2023] [Revised: 11/14/2023] [Accepted: 10/10/2023] [Indexed: 11/04/2023] Open
Abstract
Exposure of Escherichia coli to sub-inhibitory antibiotics stimulates biofilm formation through poorly characterized mechanisms. Using a high-throughput Congo Red binding assay to report on biofilm matrix production, we screened ~4000 E. coli K12 deletion mutants for deficiencies in this biofilm stimulation response. We screened using three different antibiotics to identify core components of the biofilm stimulation response. Mutants lacking acnA, nuoE, or lpdA failed to respond to sub-MIC cefixime and novobiocin, implicating central metabolism and aerobic respiration in biofilm stimulation. These genes are members of the ArcA/B regulon-controlled by a respiration-sensitive two-component system. Mutants of arcA and arcB had a 'pre-activated' phenotype, where biofilm formation was already high relative to wild type in vehicle control conditions, and failed to increase further with the addition of sub-MIC cefixime. Using a tetrazolium dye and an in vivo NADH sensor, we showed spatial co-localization of increased metabolic activity with sub-lethal concentrations of the bactericidal antibiotics cefixime and novobiocin. Supporting a role for respiratory stress, the biofilm stimulation response to cefixime and novobiocin was inhibited when nitrate was provided as an alternative electron acceptor. Deletion of a gene encoding part of the machinery for respiring nitrate abolished its ameliorating effects, and nitrate respiration increased during growth with sub-MIC cefixime. Finally, in probing the generalizability of biofilm stimulation, we found that the stimulation response to translation inhibitors, unlike other antibiotic classes, was minimally affected by nitrate supplementation, suggesting that targeting the ribosome stimulates biofilm formation in distinct ways. By characterizing the biofilm stimulation response to sub-MIC antibiotics at a systems level, we identified multiple avenues for design of therapeutics that impair bacterial stress management.
Collapse
Affiliation(s)
- Luke N. Yaeger
- Department of Biochemistry and Biomedical Sciences, and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Shawn French
- Department of Biochemistry and Biomedical Sciences, and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Eric D. Brown
- Department of Biochemistry and Biomedical Sciences, and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Jean Philippe Côté
- Department of Biochemistry and Biomedical Sciences, and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Département de Biologie, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Lori L. Burrows
- Department of Biochemistry and Biomedical Sciences, and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
13
|
Coluzzi C, Guillemet M, Mazzamurro F, Touchon M, Godfroid M, Achaz G, Glaser P, Rocha EPC. Chance Favors the Prepared Genomes: Horizontal Transfer Shapes the Emergence of Antibiotic Resistance Mutations in Core Genes. Mol Biol Evol 2023; 40:msad217. [PMID: 37788575 PMCID: PMC10575684 DOI: 10.1093/molbev/msad217] [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: 07/04/2023] [Revised: 09/08/2023] [Accepted: 09/19/2023] [Indexed: 10/05/2023] Open
Abstract
Bacterial lineages acquire novel traits at diverse rates in part because the genetic background impacts the successful acquisition of novel genes by horizontal transfer. Yet, how horizontal transfer affects the subsequent evolution of core genes remains poorly understood. Here, we studied the evolution of resistance to quinolones in Escherichia coli accounting for population structure. We found 60 groups of genes whose gain or loss induced an increase in the probability of subsequently becoming resistant to quinolones by point mutations in the gyrase and topoisomerase genes. These groups include functions known to be associated with direct mitigation of the effect of quinolones, with metal uptake, cell growth inhibition, biofilm formation, and sugar metabolism. Many of them are encoded in phages or plasmids. Although some of the chronologies may reflect epidemiological trends, many of these groups encoded functions providing latent phenotypes of antibiotic low-level resistance, tolerance, or persistence under quinolone treatment. The mutations providing resistance were frequent and accumulated very quickly. Their emergence was found to increase the rate of acquisition of other antibiotic resistances setting the path for multidrug resistance. Hence, our findings show that horizontal gene transfer shapes the subsequent emergence of adaptive mutations in core genes. In turn, these mutations further affect the subsequent evolution of resistance by horizontal gene transfer. Given the substantial gene flow within bacterial genomes, interactions between horizontal transfer and point mutations in core genes may be a key to the success of adaptation processes.
Collapse
Affiliation(s)
- Charles Coluzzi
- Institut Pasteur, Université Paris Cité, CNRS, UMR3525, Microbial Evolutionary Genomics, Paris, France
| | - Martin Guillemet
- Institut Pasteur, Université Paris Cité, CNRS, UMR3525, Microbial Evolutionary Genomics, Paris, France
| | - Fanny Mazzamurro
- Institut Pasteur, Université Paris Cité, CNRS, UMR3525, Microbial Evolutionary Genomics, Paris, France
- Collège Doctoral, Sorbonne Université, Paris, France
| | - Marie Touchon
- Institut Pasteur, Université Paris Cité, CNRS, UMR3525, Microbial Evolutionary Genomics, Paris, France
| | - Maxime Godfroid
- SMILE Group, Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | - Guillaume Achaz
- SMILE Group, Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | - Philippe Glaser
- Institut Pasteur, Université de Paris Cité, CNRS, UMR6047, Unité EERA, Paris, France
| | - Eduardo P C Rocha
- Institut Pasteur, Université Paris Cité, CNRS, UMR3525, Microbial Evolutionary Genomics, Paris, France
| |
Collapse
|
14
|
Kauser A, Parisini E, Suarato G, Castagna R. Light-Based Anti-Biofilm and Antibacterial Strategies. Pharmaceutics 2023; 15:2106. [PMID: 37631320 PMCID: PMC10457815 DOI: 10.3390/pharmaceutics15082106] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/29/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Biofilm formation and antimicrobial resistance pose significant challenges not only in clinical settings (i.e., implant-associated infections, endocarditis, and urinary tract infections) but also in industrial settings and in the environment, where the spreading of antibiotic-resistant bacteria is on the rise. Indeed, developing effective strategies to prevent biofilm formation and treat infections will be one of the major global challenges in the next few years. As traditional pharmacological treatments are becoming inadequate to curb this problem, a constant commitment to the exploration of novel therapeutic strategies is necessary. Light-triggered therapies have emerged as promising alternatives to traditional approaches due to their non-invasive nature, precise spatial and temporal control, and potential multifunctional properties. Here, we provide a comprehensive overview of the different biofilm formation stages and the molecular mechanism of biofilm disruption, with a major focus on the quorum sensing machinery. Moreover, we highlight the principal guidelines for the development of light-responsive materials and photosensitive compounds. The synergistic effects of combining light-triggered therapies with conventional treatments are also discussed. Through elegant molecular and material design solutions, remarkable results have been achieved in the fight against biofilm formation and antibacterial resistance. However, further research and development in this field are essential to optimize therapeutic strategies and translate them into clinical and industrial applications, ultimately addressing the global challenges posed by biofilm and antimicrobial resistance.
Collapse
Affiliation(s)
- Ambreen Kauser
- Department of Biotechnology, Latvian Institute of Organic Synthesis, Aizkraukles 21, LV-1006 Riga, Latvia; (A.K.); (E.P.)
- Faculty of Materials Science and Applied Chemistry, Riga Technical University, Paula Valdena 3, LV-1048 Riga, Latvia
| | - Emilio Parisini
- Department of Biotechnology, Latvian Institute of Organic Synthesis, Aizkraukles 21, LV-1006 Riga, Latvia; (A.K.); (E.P.)
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Giulia Suarato
- Istituto di Elettronica e di Ingegneria dell’Informazione e delle Telecomunicazioni, Consiglio Nazionale delle Ricerche, CNR-IEIIT, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
| | - Rossella Castagna
- Department of Biotechnology, Latvian Institute of Organic Synthesis, Aizkraukles 21, LV-1006 Riga, Latvia; (A.K.); (E.P.)
- Dipartimento di Chimica, Materiali e Ingegneria Chimica “G. Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
| |
Collapse
|
15
|
Sharma S, Mohler J, Mahajan SD, Schwartz SA, Bruggemann L, Aalinkeel R. Microbial Biofilm: A Review on Formation, Infection, Antibiotic Resistance, Control Measures, and Innovative Treatment. Microorganisms 2023; 11:1614. [PMID: 37375116 PMCID: PMC10305407 DOI: 10.3390/microorganisms11061614] [Citation(s) in RCA: 83] [Impact Index Per Article: 83.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/15/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Biofilm is complex and consists of bacterial colonies that reside in an exopolysaccharide matrix that attaches to foreign surfaces in a living organism. Biofilm frequently leads to nosocomial, chronic infections in clinical settings. Since the bacteria in the biofilm have developed antibiotic resistance, using antibiotics alone to treat infections brought on by biofilm is ineffective. This review provides a succinct summary of the theories behind the composition of, formation of, and drug-resistant infections attributed to biofilm and cutting-edge curative approaches to counteract and treat biofilm. The high frequency of medical device-induced infections due to biofilm warrants the application of innovative technologies to manage the complexities presented by biofilm.
Collapse
Affiliation(s)
- Satish Sharma
- Department of Urology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14260, USA; (S.S.); (S.A.S.)
| | - James Mohler
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA;
| | - Supriya D. Mahajan
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA;
| | - Stanley A. Schwartz
- Department of Urology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14260, USA; (S.S.); (S.A.S.)
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA;
- Department of Medicine, VA Western New York Healthcare System, Buffalo, NY 14215, USA
| | - Liana Bruggemann
- Department of Biomedical Informatics, University at Buffalo, Buffalo, NY 14260, USA;
| | - Ravikumar Aalinkeel
- Department of Urology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14260, USA; (S.S.); (S.A.S.)
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA;
- Department of Medicine, VA Western New York Healthcare System, Buffalo, NY 14215, USA
| |
Collapse
|
16
|
Goswami AG, Basu S, Banerjee T, Shukla VK. Biofilm and wound healing: from bench to bedside. Eur J Med Res 2023; 28:157. [PMID: 37098583 PMCID: PMC10127443 DOI: 10.1186/s40001-023-01121-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 04/14/2023] [Indexed: 04/27/2023] Open
Abstract
The bubbling community of microorganisms, consisting of diverse colonies encased in a self-produced protective matrix and playing an essential role in the persistence of infection and antimicrobial resistance, is often referred to as a biofilm. Although apparently indolent, the biofilm involves not only inanimate surfaces but also living tissue, making it truly ubiquitous. The mechanism of biofilm formation, its growth, and the development of resistance are ever-intriguing subjects and are yet to be completely deciphered. Although an abundance of studies in recent years has focused on the various ways to create potential anti-biofilm and antimicrobial therapeutics, a dearth of a clear standard of clinical practice remains, and therefore, there is essentially a need for translating laboratory research to novel bedside anti-biofilm strategies that can provide a better clinical outcome. Of significance, biofilm is responsible for faulty wound healing and wound chronicity. The experimental studies report the prevalence of biofilm in chronic wounds anywhere between 20 and 100%, which makes it a topic of significant concern in wound healing. The ongoing scientific endeavor to comprehensively understand the mechanism of biofilm interaction with wounds and generate standardized anti-biofilm measures which are reproducible in the clinical setting is the challenge of the hour. In this context of "more needs to be done", we aim to explore various effective and clinically meaningful methods currently available for biofilm management and how these tools can be translated into safe clinical practice.
Collapse
Affiliation(s)
| | - Somprakas Basu
- All India Institute of Medical Sciences, Rishikesh, 249203, India.
| | | | | |
Collapse
|
17
|
Lamba A, Kopel J, Westenberg D, Kapila S. Fatty acids, esters, and biogenic oil disinfectants: novel agents against bacteria. Proc AMIA Symp 2023; 36:375-379. [PMID: 37091766 PMCID: PMC10120445 DOI: 10.1080/08998280.2023.2167191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
For hundreds of years, disinfectants have comprised a variety of active chemical agents that destroy microorganisms through a wide spectrum of mechanisms. In recent years, there has been growing interest in novel disinfectants. One novel method for disinfectant is aerosols. Since the beginning of the 20th century, aerosols produced by the volatilization and subsequent recondensation of oil vapors have been utilized as obscurants (smoke) screens during military operations. Specifically, a petroleum middle distillate, known as the FOG oil, has been used in the US military battlefield to create obscurant smoke screens. Biogenic oils are non-petroleum-based oils that resemble FOG oil in terms of their physical characteristics. Furthermore, FOG and biogenic oils have characteristics that make them preferable to other disinfectants that are frequently employed. In this review, we examine the antimicrobial activities of mineral oils and biogenic oil esters aerosols/vapors as novel disinfectants against bacteria and other microorganisms.
Collapse
Affiliation(s)
- Aruna Lamba
- Department of Microbiology, Missouri University of Science and Technology, Rolla, Missouri
| | - Jonathan Kopel
- School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - David Westenberg
- Department of Microbiology, Missouri University of Science and Technology, Rolla, Missouri
| | - Shubhender Kapila
- Department of Chemistry, Missouri University of Science and Technology, Rolla, Missouri
| |
Collapse
|
18
|
Selvarajan R, Obize C, Sibanda T, Abia ALK, Long H. Evolution and Emergence of Antibiotic Resistance in Given Ecosystems: Possible Strategies for Addressing the Challenge of Antibiotic Resistance. Antibiotics (Basel) 2022; 12:28. [PMID: 36671228 PMCID: PMC9855083 DOI: 10.3390/antibiotics12010028] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022] Open
Abstract
Antibiotics were once considered the magic bullet for all human infections. However, their success was short-lived, and today, microorganisms have become resistant to almost all known antimicrobials. The most recent decade of the 20th and the beginning of the 21st century have witnessed the emergence and spread of antibiotic resistance (ABR) in different pathogenic microorganisms worldwide. Therefore, this narrative review examined the history of antibiotics and the ecological roles of antibiotics, and their resistance. The evolution of bacterial antibiotic resistance in different environments, including aquatic and terrestrial ecosystems, and modern tools used for the identification were addressed. Finally, the review addressed the ecotoxicological impact of antibiotic-resistant bacteria and public health concerns and concluded with possible strategies for addressing the ABR challenge. The information provided in this review will enhance our understanding of ABR and its implications for human, animal, and environmental health. Understanding the environmental dimension will also strengthen the need to prevent pollution as the factors influencing ABR in this setting are more than just antibiotics but involve others like heavy metals and biocides, usually not considered when studying ABR.
Collapse
Affiliation(s)
- Ramganesh Selvarajan
- Laboratory of Extraterrestrial Ocean Systems (LEOS), Institute of Deep-Sea Science and Engineering, Chinese Academy of Sciences, Sanya 572000, China
| | - Chinedu Obize
- Centre d’étude de la Forêt, Institut de Biologie Intégrative et des Systèmes, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Timothy Sibanda
- School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, Johannesburg 2050, South Africa
| | - Akebe Luther King Abia
- Department of Microbiology, Venda University, Thohoyando 1950, South Africa
- Environmental Research Foundation, Westville 3630, South Africa
| | - Haijun Long
- Laboratory of Extraterrestrial Ocean Systems (LEOS), Institute of Deep-Sea Science and Engineering, Chinese Academy of Sciences, Sanya 572000, China
| |
Collapse
|
19
|
An Initial Report of the Antimicrobial Activities of Volatiles Produced during Rapid Volatilization of Oils. Antibiotics (Basel) 2022; 11:antibiotics11121742. [PMID: 36551399 PMCID: PMC9774546 DOI: 10.3390/antibiotics11121742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 10/29/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Aerosols generated through volatilization and subsequent recondensation of oil vapors have been used as obscurant (smoke) screens during military operations since the early twentieth century. Specifically, a petroleum middle distillate known as the fog oil (FO) has been used in US military battlefields to create obscurant smoke screens. During a study on the feasibility of replacing petroleum-derived FO with vegetable oil-derived esters such as methyl soyate (MS), it was observed that that FO and MS aerosols and vapors did not exhibit detectable mutagenic activity but were lethal to Ames strains bacteria even after very short exposure periods. These results opened the potential use of oil-derived vapors as antimicrobial agents. Subsequent studies showed that optimal aerosol/vapor production conditions could further enhance disinfectant efficiency. For this purpose, we examined the antimicrobial activities of mineral oils and biogenic oil ester aerosols/vapors against a wide range of Gram-positive and Gram-negative bacteria. The results of the study showed that the aerosols/vapors obtained from mineral oil or vegetable oil ester under proper conditions can serve as an excellent antibacterial disinfectant.
Collapse
|
20
|
Patel H, Buchad H, Gajjar D. Pseudomonas aeruginosa persister cell formation upon antibiotic exposure in planktonic and biofilm state. Sci Rep 2022; 12:16151. [PMID: 36168027 PMCID: PMC9515113 DOI: 10.1038/s41598-022-20323-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/09/2022] [Indexed: 11/09/2022] Open
Abstract
Persister cell (PC) is dormant, tolerant to antibiotics, and a transient reversible phenotype. These phenotypes are observed in P. aeruginosa and cause bacterial chronic infection as well as recurrence of biofilm-mediated infection. PC formation requires stringent response and toxin-antitoxin (TA) modules. This study shows the P. aeruginosa PC formation in planktonic and biofilm stages on ceftazidime, gentamicin, and ciprofloxacin treatments. The PC formation was studied using persister assay, flow cytometry using Redox Sensor Green, fluorescence as well as Confocal Laser Scanning Microscopy, and gene expression of stringent response and TA genes. In the planktonic stage, ceftazidime showed a high survival fraction, high redox activity, and elongation of cells was observed followed by ciprofloxacin and gentamicin treatment having redox activity and rod-shaped cells. The gene expression of stringent response and TA genes were upregulated on gentamicin followed by ceftazidime treatment and varied among the isolates. In the biofilm stage, gentamicin and ciprofloxacin showed the biphasic killing pattern, redox activity, gene expression level of stringent response and TA varied across the isolates. Ceftazidime treatment showed higher persister cells in planktonic growth while all three antibiotics were able to induce persister cell formation in the biofilm stage.
Collapse
Affiliation(s)
- Hiral Patel
- Department of Microbiology and Biotechnology Centre, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390002, India
| | - Hasmatbanu Buchad
- Department of Microbiology and Biotechnology Centre, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390002, India
| | - Devarshi Gajjar
- Department of Microbiology and Biotechnology Centre, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390002, India.
| |
Collapse
|
21
|
Oluwole OM. BIOFILM: FORMATION AND NATURAL PRODUCTS' APPROACH TO CONTROL - A REVIEW. Afr J Infect Dis 2022; 16:59-71. [PMID: 36124328 PMCID: PMC9480886 DOI: 10.21010/ajid.v16i2s.7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 11/09/2022] Open
Abstract
Biofilm formation, especially on indwelling medical devices such as catheters, can result in infections and substantially affect patients' quality of life. Biofilm-associated infections have led to increased morbidity and mortality, increased cost of treatment, and length of hospital stay. However, all of the identified consequences of the biofilm-associated infections had been attributed to the reduced susceptibility of biofilm to conventional antimicrobial agents which has necessitated the development of a new strategy for biofilm infections control, thereby making a search for more effective antimicrobial agents from plant source inevitable. So far, some antimicrobial agents (crude or isolated compounds) from plant sources affect a specific stage of biofilm development while a few of them have been developed into a suitable dosage form for biofilm control. In this review, an attempt is made to look into some definitions of biofilm by "biofilmologists", stages in biofilm formation, mechanisms of resistance in biofilm, biofilm control strategies, the use of some natural products in biofilm control and concepts of probiotics as agents of biofilm control.
Collapse
Affiliation(s)
- Osungunna Michael Oluwole
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria,Corresponding Author’s E-mail:
| |
Collapse
|
22
|
Chislett M, Yu Z, Donose BC, Guo J, Yuan Z. Understanding the Effect of Free Nitrous Acid on Biofilms. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:11625-11634. [PMID: 35913828 DOI: 10.1021/acs.est.2c01156] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Free nitrous acid (FNA, i.e., HNO2) has been recently applied to biofilm control in wastewater management. The mechanism triggering biofilm detachment upon exposure to FNA still remains largely unknown. In this work, we aim to prove that FNA induces biofilm dispersal via extracellular polymeric matrix breakdown and cell lysis. Biofilms formed by a model organism, Pseudomonas aeruginosa PAO1, were treated with FNA at concentrations ranging from 0.2 to 15 mg N/L for 24 h (conditions typically used in applications). The biofilms and suspended biomass were monitored both before and after FNA treatment using a range of methods including optical density measurements, viability assays, confocal laser scanning microscopy, and atomic force microscopy. It was revealed that FNA treatment caused substantial and concentration-dependent biofilm detachment. The addition of a reactive nitrogen species (RNS) scavenger, that is, 2-4-carboxyphenyl-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide, substantially reduced biofilm dispersal, suggesting that the nitrosative decomposition species of HNO2 (i.e., RNS, e.g., •NO + •NO2) were mainly responsible for the effects. The study provides insight into and support for the use of FNA for biofilm control in wastewater treatment.
Collapse
Affiliation(s)
- Mariella Chislett
- Australian Centre for Water and Environmental Biotechnology (ACWEB, formerly AWMC), The University of Queensland, St. Lucia, Brisbane, Queensland 4072, Australia
| | - Zhigang Yu
- Australian Centre for Water and Environmental Biotechnology (ACWEB, formerly AWMC), The University of Queensland, St. Lucia, Brisbane, Queensland 4072, Australia
| | - Bogdan C Donose
- School of Information Technology and Electrical Engineering, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Jianhua Guo
- Australian Centre for Water and Environmental Biotechnology (ACWEB, formerly AWMC), The University of Queensland, St. Lucia, Brisbane, Queensland 4072, Australia
| | - Zhiguo Yuan
- Australian Centre for Water and Environmental Biotechnology (ACWEB, formerly AWMC), The University of Queensland, St. Lucia, Brisbane, Queensland 4072, Australia
| |
Collapse
|
23
|
Malyshev D, Robinson NF, Öberg R, Dahlberg T, Andersson M. Reactive oxygen species generated by infrared laser light in optical tweezers inhibits the germination of bacterial spores. JOURNAL OF BIOPHOTONICS 2022; 15:e202200081. [PMID: 35538633 DOI: 10.1002/jbio.202200081] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/28/2022] [Accepted: 05/08/2022] [Indexed: 06/14/2023]
Abstract
Bacterial spores are highly resistant to heat, radiation and various disinfection chemicals. The impact of these on the biophysical and physicochemical properties of spores can be studied on the single-cell level using optical tweezers. However, the effect of the trapping laser on spores' germination rate is not fully understood. In this work, we assess the impact of 1064 nm laser light on the germination of Bacillus thuringiensis spores. The results show that the germination rate of spores after laser exposure follows a sigmoid dose-response relationship, with only 15% of spores germinating after 20 J of laser light. Under anaerobic growth conditions, the percentage of germinating spores at 20 J increased to 65%. The results thereby indicate that molecular oxygen is a major contributor to the germination-inhibiting effect observed. Thus, our study highlights the risk for optical trapping of spores and ways to mitigate it.
Collapse
Affiliation(s)
| | | | - Rasmus Öberg
- Department of Physics, Umeå University, Umeå, Sweden
| | | | - Magnus Andersson
- Department of Physics, Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| |
Collapse
|
24
|
Biofilm- i: A Platform for Predicting Biofilm Inhibitors Using Quantitative Structure-Relationship (QSAR) Based Regression Models to Curb Antibiotic Resistance. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27154861. [PMID: 35956807 PMCID: PMC9369795 DOI: 10.3390/molecules27154861] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/16/2022] [Accepted: 07/17/2022] [Indexed: 11/19/2022]
Abstract
Antibiotic drug resistance has emerged as a major public health threat globally. One of the leading causes of drug resistance is the colonization of microorganisms in biofilm mode. Hence, there is an urgent need to design novel and highly effective biofilm inhibitors that can work either synergistically with antibiotics or individually. Therefore, we have developed a recursive regression-based platform “Biofilm-i” employing a quantitative structure–activity relationship approach for making generalized predictions, along with group and species-specific predictions of biofilm inhibition efficiency of chemical(s). The platform encompasses eight predictors, three analysis tools, and data visualization modules. The experimentally validated biofilm inhibitors for model development were retrieved from the “aBiofilm” resource and processed using a 10-fold cross-validation approach using the support vector machine and andom forest machine learning techniques. The data was further sub-divided into training/testing and independent validation sets. From training/testing data sets the Pearson’s correlation coefficient of overall chemicals, Gram-positive bacteria, Gram-negative bacteria, fungus, Pseudomonas aeruginosa, Staphylococcus aureus, Candida albicans, and Escherichia coli was 0.60, 0.77, 0.62, 0.77, 0.73, 0.83, 0.70, and 0.71 respectively via Support Vector Machine. Further, all the QSAR models performed equally well on independent validation data sets. Additionally, we also checked the performance of the random forest machine learning technique for the above datasets. The integrated analysis tools can convert the chemical structure into different formats, search for a similar chemical in the aBiofilm database and design the analogs. Moreover, the data visualization modules check the distribution of experimentally validated biofilm inhibitors according to their common scaffolds. The Biofilm-i platform would be of immense help to researchers engaged in designing highly efficacious biofilm inhibitors for tackling the menace of antibiotic drug resistance.
Collapse
|
25
|
The Association between Biofilm Formation and Antimicrobial Resistance with Possible Ingenious Bio-Remedial Approaches. Antibiotics (Basel) 2022; 11:antibiotics11070930. [PMID: 35884186 PMCID: PMC9312340 DOI: 10.3390/antibiotics11070930] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/03/2022] [Accepted: 07/06/2022] [Indexed: 02/01/2023] Open
Abstract
Biofilm has garnered a lot of interest due to concerns in various sectors such as public health, medicine, and the pharmaceutical industry. Biofilm-producing bacteria show a remarkable drug resistance capability, leading to an increase in morbidity and mortality. This results in enormous economic pressure on the healthcare sector. The development of biofilms is a complex phenomenon governed by multiple factors. Several attempts have been made to unravel the events of biofilm formation; and, such efforts have provided insights into the mechanisms to target for the therapy. Owing to the fact that the biofilm-state makes the bacterial pathogens significantly resistant to antibiotics, targeting pathogens within biofilm is indeed a lucrative prospect. The available drugs can be repurposed to eradicate the pathogen, and as a result, ease the antimicrobial treatment burden. Biofilm formers and their infections have also been found in plants, livestock, and humans. The advent of novel strategies such as bioinformatics tools in treating, as well as preventing, biofilm formation has gained a great deal of attention. Development of newfangled anti-biofilm agents, such as silver nanoparticles, may be accomplished through omics approaches such as transcriptomics, metabolomics, and proteomics. Nanoparticles’ anti-biofilm properties could help to reduce antimicrobial resistance (AMR). This approach may also be integrated for a better understanding of biofilm biology, guided by mechanistic understanding, virtual screening, and machine learning in silico techniques for discovering small molecules in order to inhibit key biofilm regulators. This stimulated research is a rapidly growing field for applicable control measures to prevent biofilm formation. Therefore, the current article discusses the current understanding of biofilm formation, antibiotic resistance mechanisms in bacterial biofilm, and the novel therapeutic strategies to combat biofilm-mediated infections.
Collapse
|
26
|
Adhikari BR, Dummer J, Gordon KC, Das SC. An expert opinion on respiratory delivery of high dose powders for lung infections. Expert Opin Drug Deliv 2022; 19:795-813. [PMID: 35695722 DOI: 10.1080/17425247.2022.2089111] [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] [Indexed: 12/30/2022]
Abstract
INTRODUCTION High dose powder inhalation is evolving as an important approach to to treat lung infections. It is important to its identify applications, consider the factors affecting high dose powder delivery, and assess the effect of high dose drugs in patients. AREA COVERED Both current and pipeline high dose inhalers and their applications have been summarized. Challenges and opportunities to high dose delivery have been highlighted after reviewing formulation techniques in the context of factors affecting aerosolization, devices, and patient factors. EXPERT OPINION High dose inhaled delivery of antimicrobials is an innovative way to increase treatment efficacy of respiratory infections, tackle drug resistance, and the scarcity of new antimicrobials. The high dose inhaled technology also has potential for systemic action; however, innovations in formulation strategies and devices are required to realize its full potential. Advances in formulation strategies include the use of excipients or the engineering of particles to decrease the cohesive property of microparticles and their packing density. Similarly, selection of a synergistic drug instead of an excipient can be considered to increase aerosolization and stability. Device development focused on improving dispersion and loading capacity is also important, and modification of existing devices for high dose delivery can also be considered.
Collapse
Affiliation(s)
| | - Jack Dummer
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Keith C Gordon
- The Dodd-Walls Centre for Photonic and Quantum Technologies, Department of Chemistry, University of Otago, Dunedin, New Zealand
| | - Shyamal C Das
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
27
|
Laborda P, Hernando-Amado S, Martínez JL, Sanz-García F. Antibiotic Resistance in Pseudomonas. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1386:117-143. [DOI: 10.1007/978-3-031-08491-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
28
|
Flores-Vargas G, Bergsveinson J, Lawrence JR, Korber DR. Environmental Biofilms as Reservoirs for Antimicrobial Resistance. Front Microbiol 2022; 12:766242. [PMID: 34970233 PMCID: PMC8713029 DOI: 10.3389/fmicb.2021.766242] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 11/22/2021] [Indexed: 12/17/2022] Open
Abstract
Characterizing the response of microbial communities to a range of antibiotic concentrations is one of the strategies used to understand the impact of antibiotic resistance. Many studies have described the occurrence and prevalence of antibiotic resistance in microbial communities from reservoirs such as hospitals, sewage, and farm feedlots, where bacteria are often exposed to high and/or constant concentrations of antibiotics. Outside of these sources, antibiotics generally occur at lower, sub-minimum inhibitory concentrations (sub-MICs). The constant exposure to low concentrations of antibiotics may serve as a chemical "cue" that drives development of antibiotic resistance. Low concentrations of antibiotics have not yet been broadly described in reservoirs outside of the aforementioned environments, nor is the transfer and dissemination of antibiotic resistant bacteria and genes within natural microbial communities fully understood. This review will thus focus on low antibiotic-concentration environmental reservoirs and mechanisms that are important in the dissemination of antibiotic resistance to help identify key knowledge gaps concerning the environmental resistome.
Collapse
Affiliation(s)
| | | | - John R Lawrence
- Environment and Climate Change Canada, Saskatoon, SK, Canada
| | - Darren R Korber
- Food and Bioproduct Sciences, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
29
|
Xu Q, Hu X, Wang Y. Alternatives to Conventional Antibiotic Therapy: Potential Therapeutic Strategies of Combating Antimicrobial-Resistance and Biofilm-Related Infections. Mol Biotechnol 2021; 63:1103-1124. [PMID: 34309796 DOI: 10.1007/s12033-021-00371-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/08/2021] [Indexed: 12/14/2022]
Abstract
Antibiotics have been denoted as the orthodox therapeutic agents for fighting bacteria-related infections in clinical practices for decades. Nevertheless, overuse of antibiotics has led to the upsurge of species with antimicrobial resistance (AMR) or multi-drug resistance. Bacteria can also grow into the biofilm, which accounts for at least two-thirds of infections. Distinct gene expression and self-produced heterogeneous hydrated extracellular polymeric substance matrix architecture of biofilm contribute to their tolerance and externally manifest as antibiotic resistance. In this review, the difficulties in combating biofilm formation and AMR are introduced, and novel alternatives to antibiotics such as metal nanoparticles and quaternary ammonium compounds, chitosan and its derivatives, antimicrobial peptides, stimuli-responsive materials, phage therapy and other therapeutic strategies, from compounds to hydrogel, from inorganic to biological, are discussed. We expect to provide useful information for the readers who are seeking for solutions to the problem of AMR and biofilm-related infections.
Collapse
Affiliation(s)
- Qian Xu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, Sichuan, People's Republic of China
| | - Xuefeng Hu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, Sichuan, People's Republic of China.
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, Sichuan, People's Republic of China.
| |
Collapse
|
30
|
Duan X, Pan Y, Cai Z, Liu Y, Zhang Y, Liu M, Liu Y, Wang K, Zhang L, Yang L. rpoS-mutation variants are selected in Pseudomonas aeruginosa biofilms under imipenem pressure. Cell Biosci 2021; 11:138. [PMID: 34289907 PMCID: PMC8293535 DOI: 10.1186/s13578-021-00655-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/11/2021] [Indexed: 12/14/2022] Open
Abstract
Background Pseudomonas aeruginosa is a notorious opportunistic pathogen causing various types of biofilm-related infections. Biofilm formation is a unique microbial strategy that allows P. aeruginosa to survive adverse conditions such as antibiotic treatment and human immune clearance. Results In this study, we experimentally evolved P. aeruginosa PAO1 biofilms for cyclic treatment in the presence of high dose of imipenem, and enriched hyperbiofilm mutants within six cycles in two independent lineages. The competition assay showed that the evolved hyperbiofilm mutants can outcompete the ancestral strain within biofilms but not in planktonic cultures. Whole-genome sequencing analysis revealed the hyperbiofilm phenotype is caused by point mutations in rpoS gene in all independently evolved mutants and the same mutation was found in P. aeruginosa clinical isolates. We further showed that mutation in rpoS gene increased the intracellular c-di-GMP level by turning on the expression of the diguanylate cyclases. Mutation in rpoS increased pyocyanin production and virulence in hyperbiofilm variants. Conclusion Here, our study revealed that antibiotic treatment of biofilm-related P. aeruginosa infections might induce a hyperbiofilm phenotype via rpoS mutation, which might partially explain antimicrobial treatment failure of many P. aeruginosa biofilm-related infections. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00655-9.
Collapse
Affiliation(s)
- Xiangke Duan
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research Center, South China Agricultural University, Guangzhou, 510642, Guangdong, People's Republic of China.,School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Yanrong Pan
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Zhao Cai
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Yumei Liu
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Yingdan Zhang
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Moxiao Liu
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Yang Liu
- Southern University of Science and Technology Hospital, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Ke Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, People's Republic of China
| | - Lianhui Zhang
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control, Integrative Microbiology Research Center, South China Agricultural University, Guangzhou, 510642, Guangdong, People's Republic of China.
| | - Liang Yang
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, People's Republic of China.,Shenzhen Key Laboratory for Gene Regulation and Systems Biology, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, People's Republic of China
| |
Collapse
|
31
|
Persister Escherichia coli Cells Have a Lower Intracellular pH than Susceptible Cells but Maintain Their pH in Response to Antibiotic Treatment. mBio 2021; 12:e0090921. [PMID: 34281389 PMCID: PMC8406257 DOI: 10.1128/mbio.00909-21] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Persister and viable but non-culturable (VBNC) cells are two clonal subpopulations that can survive multidrug exposure via a plethora of putative molecular mechanisms. Here, we combine microfluidics, time-lapse microscopy, and a plasmid-encoded fluorescent pH reporter to measure the dynamics of the intracellular pH of individual persister, VBNC, and susceptible Escherichia coli cells in response to ampicillin treatment. We found that even before antibiotic exposure, persisters have a lower intracellular pH than those of VBNC and susceptible cells. We then investigated the molecular mechanisms underlying the observed differential pH regulation in persister E. coli cells and found that this is linked to the activity of the enzyme tryptophanase, which is encoded by tnaA. In fact, in a ΔtnaA strain, we found no difference in intracellular pH between persister, VBNC, and susceptible E. coli cells. Whole-genome transcriptomic analysis revealed that, besides downregulating tryptophan metabolism, the ΔtnaA strain downregulated key pH homeostasis pathways, including the response to pH, oxidation reduction, and several carboxylic acid catabolism processes, compared to levels of expression in the parental strain. Our study sheds light on pH homeostasis, proving that the regulation of intracellular pH is not homogeneous within a clonal population, with a subset of cells displaying a differential pH regulation to perform dedicated functions, including survival after antibiotic treatment. IMPORTANCE Persister and VBNC cells can phenotypically survive environmental stressors, such as antibiotic treatment, limitation of nutrients, and acid stress, and have been linked to chronic infections and antimicrobial resistance. It has recently been suggested that pH regulation might play a role in an organism's phenotypic survival to antibiotics; however, this hypothesis remains to be tested. Here, we demonstrate that even before antibiotic treatment, cells that will become persisters have a more acidic intracellular pH than clonal cells that will be either susceptible or VBNC upon antibiotic treatment. Moreover, after antibiotic treatment, persisters become more alkaline than VBNC and susceptible E. coli cells. This newly found phenotypic feature is remarkable because it distinguishes persister and VBNC cells that have often been thought to display the same dormant phenotype. We then show that this differential pH regulation is abolished in the absence of the enzyme tryptophanase via a major remodeling of bacterial metabolism and pH homeostasis. These new whole-genome transcriptome data should be taken into account when modeling bacterial metabolism at the crucial transition from exponential to stationary phase. Overall, our findings indicate that the manipulation of the intracellular pH represents a bacterial strategy for surviving antibiotic treatment. In turn, this suggests a strategy for developing persister-targeting antibiotics by interfering with cellular components, such as tryptophanase, that play a major role in pH homeostasis.
Collapse
|
32
|
Gonzaga ZJC, Merakou C, DiGiandomenico A, Priebe GP, Rehm BHA. A Pseudomonas aeruginosa-Derived Particulate Vaccine Protects against P. aeruginosa Infection. Vaccines (Basel) 2021; 9:803. [PMID: 34358220 PMCID: PMC8309987 DOI: 10.3390/vaccines9070803] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/12/2021] [Accepted: 07/16/2021] [Indexed: 01/05/2023] Open
Abstract
Despite numerous efforts to develop an effective vaccine against Pseudomonas aeruginosa, no vaccine has yet been approved for human use. This study investigates the utility of the P. aeruginosa inherently produced polyhydroxyalkanaote (PHA) inclusions and associated host-cell proteins (HCP) as a particulate vaccine platform. We further engineered PHA inclusions to display epitopes derived from the outer membrane proteins OprF/OprI/AlgE (Ag) or the type III secretion system translocator PopB. PHA and engineered PHA beads induced antigen-specific humoral, cell-mediated immune responses, anti-HCP and anti-polysaccharide Psl responses in mice. Antibodies mediated opsonophagocytic killing and serotype-independent protective immunity as shown by 100% survival upon challenge with P. aeruginosa in an acute pneumonia murine model. Vaccines were stable at 4 °C for at least one year. Overall, our data suggest that vaccination with subcellular empty PHA beads was sufficient to elicit multiple immune effectors that can prevent P. aeruginosa infection.
Collapse
Affiliation(s)
- Zennia Jean C. Gonzaga
- Centre for Cell Factories and Biopolymers (CCFB), Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, QLD 4111, Australia;
| | - Christina Merakou
- Division of Critical Care Medicine, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA 02115, USA; (C.M.); (G.P.P.)
- Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA
| | - Antonio DiGiandomenico
- Discovery Microbiome, Microbial Sciences, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 34321, USA;
| | - Gregory P. Priebe
- Division of Critical Care Medicine, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA 02115, USA; (C.M.); (G.P.P.)
- Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA
- Division of Infectious Diseases, Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Bernd H. A. Rehm
- Centre for Cell Factories and Biopolymers (CCFB), Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, QLD 4111, Australia;
- Menzies Health Institute Queensland (MHIQ), Griffith University, Gold Coast, QLD 4222, Australia
| |
Collapse
|
33
|
Gan BH, Gaynord J, Rowe SM, Deingruber T, Spring DR. The multifaceted nature of antimicrobial peptides: current synthetic chemistry approaches and future directions. Chem Soc Rev 2021; 50:7820-7880. [PMID: 34042120 PMCID: PMC8689412 DOI: 10.1039/d0cs00729c] [Citation(s) in RCA: 184] [Impact Index Per Article: 61.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Indexed: 12/13/2022]
Abstract
Bacterial infections caused by 'superbugs' are increasing globally, and conventional antibiotics are becoming less effective against these bacteria, such that we risk entering a post-antibiotic era. In recent years, antimicrobial peptides (AMPs) have gained significant attention for their clinical potential as a new class of antibiotics to combat antimicrobial resistance. In this review, we discuss several facets of AMPs including their diversity, physicochemical properties, mechanisms of action, and effects of environmental factors on these features. This review outlines various chemical synthetic strategies that have been applied to develop novel AMPs, including chemical modifications of existing peptides, semi-synthesis, and computer-aided design. We will also highlight novel AMP structures, including hybrids, antimicrobial dendrimers and polypeptides, peptidomimetics, and AMP-drug conjugates and consider recent developments in their chemical synthesis.
Collapse
Affiliation(s)
- Bee Ha Gan
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| | - Josephine Gaynord
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| | - Sam M Rowe
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| | - Tomas Deingruber
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| | - David R Spring
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| |
Collapse
|
34
|
Lewis K. At the Crossroads of Bioenergetics and Antibiotic Discovery. BIOCHEMISTRY (MOSCOW) 2021; 85:1469-1483. [PMID: 33705287 DOI: 10.1134/s0006297920120019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Dr. Vladimir Skulachev was my mentor, and his pioneering work in the field of bioenergetics inspired the discoveries described in this review, written in the form of a personal account of events. Examining basic mechanisms of chemiosmotic coupling unexpectedly led us to transenvelope multidrug resistance pumps (MDR pumps) that severely limit development of novel antibiotics. One of the major advances of Skulachev and his group was the discovery of the mitochondrial membrane potential with the use of permeant cations such as TPP+, which served as electric probes. We describe our finding of their natural counterparts in plants, where they act as antimicrobials. The most challenging problems in antimicrobial drug discovery are antibiotic tolerance of chronic infections caused by dormant persister cells; antibiotic resistance, responsible for the current antimicrobial resistance crisis (AMR); and finding novel compounds acting against Gram-negative bacteria, protected by their powerful multidrug resistance pumps. Our study of persisters shows that these are rare cells formed by stochastic fluctuation in expression of Krebs cycle enzymes, leading to a drop in ATP, target shutdown, and antibiotic tolerance. Searching for compounds that can corrupt targets in the absence of ATP, we identified acyldepsipeptide (ADEP) that activates the ClpP protease, forcing cells to self-digest. Growing previously uncultured bacteria led us to teixobactin, a novel cell wall acting antibiotic. Teixobactin avoids efflux by targeting lipid II and lipid III, precursors of peptidoglycan and wall teichoic acid, located on the surface. The targets are immutable, and teixobactin is the first antibiotic with no detectable resistance. Our search for compounds acting against Gram-negative bacteria led to the discovery of darobactins, which also hit a surface target, the essential chaperone BamA.
Collapse
Affiliation(s)
- K Lewis
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
35
|
Guzmán-Soto I, McTiernan C, Gonzalez-Gomez M, Ross A, Gupta K, Suuronen EJ, Mah TF, Griffith M, Alarcon EI. Mimicking biofilm formation and development: Recent progress in in vitro and in vivo biofilm models. iScience 2021; 24:102443. [PMID: 34013169 PMCID: PMC8113887 DOI: 10.1016/j.isci.2021.102443] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Biofilm formation in living organisms is associated to tissue and implant infections, and it has also been linked to the contribution of antibiotic resistance. Thus, understanding biofilm development and being able to mimic such processes is vital for the successful development of antibiofilm treatments and therapies. Several decades of research have contributed to building the foundation for developing in vitro and in vivo biofilm models. However, no such thing as an "all fit" in vitro or in vivo biofilm models is currently available. In this review, in addition to presenting an updated overview of biofilm formation, we critically revise recent approaches for the improvement of in vitro and in vivo biofilm models.
Collapse
Affiliation(s)
- Irene Guzmán-Soto
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
| | - Christopher McTiernan
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
| | - Mayte Gonzalez-Gomez
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
| | - Alex Ross
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, K1H8M5, Canada
| | - Keshav Gupta
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
| | - Erik J. Suuronen
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
| | - Thien-Fah Mah
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, K1H8M5, Canada
| | - May Griffith
- Centre de Recherche Hôpital Maisonneuve-Rosemont, Montréal, QC, H1T 2M4, Canada
- Département d'ophtalmologie, Université de Montréal, Montréal, QC, H3T1J4, Canada
| | - Emilio I. Alarcon
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, K1H8M5, Canada
| |
Collapse
|
36
|
Kim J, Ren D, Gilbert JL. Cytotoxic effect of galvanically coupled magnesium-titanium particles on Escherichia coli. J Biomed Mater Res B Appl Biomater 2021; 109:2162-2173. [PMID: 33979012 DOI: 10.1002/jbm.b.34864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 02/04/2021] [Accepted: 04/28/2021] [Indexed: 11/06/2022]
Abstract
Orthopedic device-related infections (ODRIs) are difficult to control due to microbial biofilm formation and associated with high-level resistance to conventional antibiotics. In many cases, the only treatment option for ODRI is explantation. Previous studies have shown that application of cathodic potentials at the metal surface can eradicate biofilms, and Mg and Mg-Ti particles have the same effect as cathodic potentials. This study investigated the effects of Mg and Mg-Ti particles on established biofilms and planktonic cells E. coli. Bacterial cultures with developed biofilms or planktonic cells were treated with Mg or Mg-Ti particles, and the viability were assessed using flow cytometry or visual assessment methods (i.e., observation from SEM images and opacity of the solution). It was found that viability of biofilms treated with 16.67 mg/ml of Mg was 2.8 ± 0.96% at the end of 6-hr killing compared to untreated controls. This extent of killing was more significant compared to 24-hr grown biofilms treated with ofloxacin, an antibiotic known to be effective against these bacteria. Biofilms treated with 50 and 100 μg/ml of ofloxacin had 62 ± 4.6% and 52 ± 19.3% survival, respectively, where ofloxacin at these concentrations is known to kill planktonic counterparts very effectively. Inhibition zone tests revealed that biofilms within 2 mm of Mg or Mg-Ti particle clusters were effectively killed. These results demonstrated the potential of Mg or Mg-Ti particles in killing microbial biofilms and potential for controlling ODRI.
Collapse
Affiliation(s)
- Jua Kim
- Department of Biomedical and Chemical Engineering, College of Engineering and Computer Science, Syracuse University, Syracuse, New York, USA.,Syracuse Biomaterials Institute, College of Engineering and Computer Science, Syracuse University, Syracuse, New York, USA.,Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Dacheng Ren
- Department of Biomedical and Chemical Engineering, College of Engineering and Computer Science, Syracuse University, Syracuse, New York, USA.,Syracuse Biomaterials Institute, College of Engineering and Computer Science, Syracuse University, Syracuse, New York, USA
| | - Jeremy L Gilbert
- Department of Biomedical and Chemical Engineering, College of Engineering and Computer Science, Syracuse University, Syracuse, New York, USA.,Syracuse Biomaterials Institute, College of Engineering and Computer Science, Syracuse University, Syracuse, New York, USA.,Department of Bioengineering, Clemson University, Clemson, South Carolina, USA.,Clemson-Medical University of South Carolina Bioengineering Program, Charleston, South Carolina, USA
| |
Collapse
|
37
|
Lopes SP, Jorge P, Sousa AM, Pereira MO. Discerning the role of polymicrobial biofilms in the ascent, prevalence, and extent of heteroresistance in clinical practice. Crit Rev Microbiol 2021; 47:162-191. [PMID: 33527850 DOI: 10.1080/1040841x.2020.1863329] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Antimicrobial therapy is facing a worrisome and underappreciated challenge, the phenomenon of heteroresistance (HR). HR has been gradually documented in clinically relevant pathogens (e.g. Pseudomonas aeruginosa, Staphylococcus aureus, Burkholderia spp., Acinetobacter baumannii, Klebsiella pneumoniae, Candida spp.) towards several drugs and is believed to complicate the clinical picture of chronic infections. This type of infections are typically mediated by polymicrobial biofilms, wherein microorganisms inherently display a wide range of physiological states, distinct metabolic pathways, diverging refractory levels of stress responses, and a complex network of chemical signals exchange. This review aims to provide an overview on the relevance, prevalence, and implications of HR in clinical settings. Firstly, related terminologies (e.g. resistance, tolerance, persistence), sometimes misunderstood and overlapped, were clarified. Factors generating misleading HR definitions were also uncovered. Secondly, the recent HR incidences reported in clinically relevant pathogens towards different antimicrobials were annotated. The potential mechanisms underlying such occurrences were further elucidated. Finally, the link between HR and biofilms was discussed. The focus was to recognize the presence of heterogeneous levels of resistance within most biofilms, as well as the relevance of polymicrobial biofilms in chronic infectious diseases and their role in resistance spreading. These topics were subject of a critical appraisal, gaining insights into the ascending clinical implications of HR in antimicrobial resistance spreading, which could ultimately help designing effective therapeutic options.
Collapse
Affiliation(s)
- Susana Patrícia Lopes
- CEB - Centre of Biological Engineering, LIBRO - Laboratory of Research in Biofilms Rosário Oliveira, University of Minho, Braga, Portugal
| | - Paula Jorge
- CEB - Centre of Biological Engineering, LIBRO - Laboratory of Research in Biofilms Rosário Oliveira, University of Minho, Braga, Portugal
| | - Ana Margarida Sousa
- CEB - Centre of Biological Engineering, LIBRO - Laboratory of Research in Biofilms Rosário Oliveira, University of Minho, Braga, Portugal
| | - Maria Olívia Pereira
- CEB - Centre of Biological Engineering, LIBRO - Laboratory of Research in Biofilms Rosário Oliveira, University of Minho, Braga, Portugal
| |
Collapse
|
38
|
Jara J, Alarcón F, Monnappa AK, Santos JI, Bianco V, Nie P, Ciamarra MP, Canales Á, Dinis L, López-Montero I, Valeriani C, Orgaz B. Self-Adaptation of Pseudomonas fluorescens Biofilms to Hydrodynamic Stress. Front Microbiol 2021; 11:588884. [PMID: 33510716 PMCID: PMC7835673 DOI: 10.3389/fmicb.2020.588884] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/14/2020] [Indexed: 11/26/2022] Open
Abstract
In some conditions, bacteria self-organize into biofilms, supracellular structures made of a self-produced embedding matrix, mainly composed of polysaccharides, DNA, proteins, and lipids. It is known that bacteria change their colony/matrix ratio in the presence of external stimuli such as hydrodynamic stress. However, little is still known about the molecular mechanisms driving this self-adaptation. In this work, we monitor structural features of Pseudomonas fluorescens biofilms grown with and without hydrodynamic stress. Our measurements show that the hydrodynamic stress concomitantly increases the cell density population and the matrix production. At short growth timescales, the matrix mediates a weak cell-cell attractive interaction due to the depletion forces originated by the polymer constituents. Using a population dynamics model, we conclude that hydrodynamic stress causes a faster diffusion of nutrients and a higher incorporation of planktonic bacteria to the already formed microcolonies. This results in the formation of more mechanically stable biofilms due to an increase of the number of crosslinks, as shown by computer simulations. The mechanical stability also relies on a change in the chemical compositions of the matrix, which becomes enriched in carbohydrates, known to display adhering properties. Overall, we demonstrate that bacteria are capable of self-adapting to hostile hydrodynamic stress by tailoring the biofilm chemical composition, thus affecting both the mesoscale structure of the matrix and its viscoelastic properties that ultimately regulate the bacteria-polymer interactions.
Collapse
Affiliation(s)
- Josué Jara
- Departamento de Farmacia Galénica y Tecnología Alimentaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Francisco Alarcón
- Departamento de Estructura de la Materia, Física Térmica y Electrónica, Universidad Complutense de Madrid, Madrid, Spain.,Departamento de Ingeniería Física, Universidad de Guanajuato, León, Mexico
| | - Ajay K Monnappa
- Instituto de Investigación Biomédica Hospital 12 de Octubre (imas12), Madrid, Spain
| | | | - Valentino Bianco
- Departamento de Química Física, Universidad Complutense de Madrid, Madrid, Spain
| | - Pin Nie
- Nanyang Technological University, Singapore, Singapore
| | | | - Ángeles Canales
- Departamento de Química Orgánica, Universidad Complutense de Madrid, Madrid, Spain
| | - Luis Dinis
- Departamento de Estructura de la Materia, Física Térmica y Electrónica, Universidad Complutense de Madrid, Madrid, Spain
| | - Iván López-Montero
- Instituto de Investigación Biomédica Hospital 12 de Octubre (imas12), Madrid, Spain.,Departamento de Química Física, Universidad Complutense de Madrid, Madrid, Spain
| | - Chantal Valeriani
- Departamento de Estructura de la Materia, Física Térmica y Electrónica, Universidad Complutense de Madrid, Madrid, Spain
| | - Belén Orgaz
- Departamento de Farmacia Galénica y Tecnología Alimentaria, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
39
|
Balakrishnan J, Ganapathi P, Kannan S, Marudhamuthu M, Shanmugam K. Anti-listerial activity of microalgal fatty acid methyl esters and their possible applications as chicken marinade. Int J Food Microbiol 2020; 339:109027. [PMID: 33412385 DOI: 10.1016/j.ijfoodmicro.2020.109027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/02/2020] [Accepted: 12/08/2020] [Indexed: 11/30/2022]
Abstract
Fatty acid methyl esters (FAMEs) from marine microalgae have been reported to possess antimicrobial activities against several Gram positive and Gram negative bacteria, but a majority of them needs to be explored. The objective of this study was to investigate the antibacterial activity, mechanism of FAMEs from selected marine microalgae against Listeria monocytogenes, and to elucidate its efficacy in food model. The minimum inhibitory concentration of FAMEs was calculated to be 155 μg/mL for Chromulina sp. and 162 μg/mL for Nannochloropsis sp. against L. monocytogenes. Time-killing kinetics showed that FAMEs efficiently inhibited the growth of L. monocytogenes in a time and concentration dependent manner. The mechanism of action of FAMEs was studied by analysing its effects at a MIC on the cellular metabolism, membrane permeability, and membrane integrity of L. monocytogenes. Transmission Electron Microscopy (TEM) results showed that cells exposed to FAMEs showed damaged cell membrane structure with leakage of the internal contents in the cells of L. monocytogenes. Fluorescence microscopy images showed that L. monocytogenes cells treated with FAMEs showed high dead cell population corresponding with propidium iodide positive cells. Furthermore, FAMEs significantly down regulated quorum sensing and biofilm related genes (DegU, FlaE, and FlaD). In vivo therapeutic potential of FAMEs revealed improved Caenorhabditis elegans survival and reduced intestinal colonization during L. monocytogenes infection. Growth of listeria was abolished in chicken meat during the cold storage of 9 days when the samples were pre-treated with FAMEs. These results suggest anti-L. monocytogenes activity of FAMEs and elucidated its use in food control of chicken meat at refrigerated conditions.
Collapse
Affiliation(s)
- Jeyakumar Balakrishnan
- Department of Molecular Biology, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, Tamil Nadu, India
| | - Prakash Ganapathi
- Department of Molecular Biology, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, Tamil Nadu, India
| | - Suganya Kannan
- Department of Microbial Technology, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, Tamil Nadu, India
| | - Murugan Marudhamuthu
- Department of Microbial Technology, School of Biological Sciences, Madurai Kamaraj University, Madurai 625021, Tamil Nadu, India
| | - Kathiresan Shanmugam
- Department of Life Sciences, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur 610005, Tamil Nadu, India.
| |
Collapse
|
40
|
Huemer M, Mairpady Shambat S, Brugger SD, Zinkernagel AS. Antibiotic resistance and persistence-Implications for human health and treatment perspectives. EMBO Rep 2020; 21:e51034. [PMID: 33400359 PMCID: PMC7726816 DOI: 10.15252/embr.202051034] [Citation(s) in RCA: 255] [Impact Index Per Article: 63.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/13/2020] [Accepted: 11/02/2020] [Indexed: 12/24/2022] Open
Abstract
Antimicrobial resistance (AMR) and persistence are associated with an elevated risk of treatment failure and relapsing infections. They are thus important drivers of increased morbidity and mortality rates resulting in growing healthcare costs. Antibiotic resistance is readily identifiable with standard microbiological assays, and the threat imposed by antibiotic resistance has been well recognized. Measures aiming to reduce resistance development and spreading of resistant bacteria are being enforced. However, the phenomenon of bacteria surviving antibiotic exposure despite being fully susceptible, so-called antibiotic persistence, is still largely underestimated. In contrast to antibiotic resistance, antibiotic persistence is difficult to measure and therefore often missed, potentially leading to treatment failures. In this review, we focus on bacterial mechanisms allowing evasion of antibiotic killing and discuss their implications on human health. We describe the relationship between antibiotic persistence and bacterial heterogeneity and discuss recent studies that link bacterial persistence and tolerance with the evolution of antibiotic resistance. Finally, we review persister detection methods, novel strategies aiming at eradicating bacterial persisters and the latest advances in the development of new antibiotics.
Collapse
Affiliation(s)
- Markus Huemer
- Department of Infectious Diseases and Hospital EpidemiologyUniversity Hospital ZurichUniversity of ZurichZurichSwitzerland
| | - Srikanth Mairpady Shambat
- Department of Infectious Diseases and Hospital EpidemiologyUniversity Hospital ZurichUniversity of ZurichZurichSwitzerland
| | - Silvio D Brugger
- Department of Infectious Diseases and Hospital EpidemiologyUniversity Hospital ZurichUniversity of ZurichZurichSwitzerland
| | - Annelies S Zinkernagel
- Department of Infectious Diseases and Hospital EpidemiologyUniversity Hospital ZurichUniversity of ZurichZurichSwitzerland
| |
Collapse
|
41
|
Abstract
The formation of microbial biofilms enables single planktonic cells to assume a multicellular mode of growth. During dispersion, the final step of the biofilm life cycle, single cells egress from the biofilm to resume a planktonic lifestyle. As the planktonic state is considered to be more vulnerable to antimicrobial agents and immune responses, dispersion is being considered a promising avenue for biofilm control. In this Review, we discuss conditions that lead to dispersion and the mechanisms by which native and environmental cues contribute to dispersion. We also explore recent findings on the role of matrix degradation in the dispersion process, and the distinct phenotype of dispersed cells. Last, we discuss the translational and therapeutic potential of dispersing bacteria during infection.
Collapse
Affiliation(s)
- Kendra P Rumbaugh
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of the TTUHSC Surgery Burn Center of Research Excellence, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Karin Sauer
- Department of Biological Sciences, Binghamton University, Binghamton, NY, USA.
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, NY, USA.
| |
Collapse
|
42
|
Gonzalez A, Miñán AG, Grillo CA, Prieto ED, Schilardi PL, Fernández Lorenzo de Mele MA. Characterization and antimicrobial effect of a bioinspired thymol coating formed on titanium surface by one-step immersion treatment. Dent Mater 2020; 36:1495-1507. [PMID: 32988646 DOI: 10.1016/j.dental.2020.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/03/2020] [Accepted: 09/04/2020] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To develop an antimicrobial and anti-adherent thymol (TOH)-containing coating on titanium (Ti) by a bioinspired one-step biocompatible method. METHODS A nanolayer of adsorbed TOH (TOH-NL-Ti) was formed by an easy deep coating method on Ti surface. The treatment consists in a simple one-step immersion process in a TOH-containing solution. Attenuated Total Reflection Fourier Transform Infrared Spectroscopy (ATR-FTIR), potentiodynamic electrochemical technique, open circuit potential records, Atomic Force Microscopy (AFM) and measurements of TOH release were used to characterize TOH-NL-Ti. Live/Dead staining and plate counting were employed to quantify attached and living adhered bacteria, respectively. Biocompatibility and cytotoxicity in fibroblastic and pre-osteoblastic cell lines were evaluated by acridine orange staining and MTT assay, respectively. RESULTS TOH adsorbed on TOH-NL-Ti was detected by ATR-FTIR and electrochemical techniques. ATR-FTIR results showed that TOH nanofilms development involves spontaneous production of ketonic structures on Ti surface. AFM analysis revealed that the thickness of the TOH-NL was below 80 nm. Finally, microbiological assays confirmed that TOH-NL-Ti can inhibit the adhesion and kill attached bacteria leading to the eradication of leaving cells on its surface. After 24 h of biocidal release, the antimicrobial effect is also significant (a decrease of 3 orders in the number of attached bacteria). SIGNIFICANCE The formation of TOH-NL-Ti nanolayer is a simple strategy able to be applied by not specially trained personnel, to reduce implant infection risks, ensure highly effective antimicrobial action and inhibition of bacterial adhesion on Ti surfaces without showing toxic effects for pre-osteoblastic and fibroblastic cells.
Collapse
Affiliation(s)
- Ariel Gonzalez
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA) Facultad de Ciencias Exactas, Universidad Nacional de La Plata- CONICET, Casilla de Correo 16, Sucursal 4, 1900 La Plata, Argentina.
| | - Alejandro Guillermo Miñán
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA) Facultad de Ciencias Exactas, Universidad Nacional de La Plata- CONICET, Casilla de Correo 16, Sucursal 4, 1900 La Plata, Argentina.
| | - Claudia Alejandra Grillo
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA) Facultad de Ciencias Exactas, Universidad Nacional de La Plata- CONICET, Casilla de Correo 16, Sucursal 4, 1900 La Plata, Argentina.
| | - Eduardo Daniel Prieto
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA) Facultad de Ciencias Exactas, Universidad Nacional de La Plata- CONICET, Casilla de Correo 16, Sucursal 4, 1900 La Plata, Argentina.
| | - Patricia Laura Schilardi
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA) Facultad de Ciencias Exactas, Universidad Nacional de La Plata- CONICET, Casilla de Correo 16, Sucursal 4, 1900 La Plata, Argentina.
| | - Mónica Alicia Fernández Lorenzo de Mele
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA) Facultad de Ciencias Exactas, Universidad Nacional de La Plata- CONICET, Casilla de Correo 16, Sucursal 4, 1900 La Plata, Argentina; Facultad de Ingeniería, Universidad Nacional de La Plata, Calle 47 y 1, 1900 La Plata, Argentina.
| |
Collapse
|
43
|
Mediati DG, Wu S, Wu W, Tree JJ. Networks of Resistance: Small RNA Control of Antibiotic Resistance. Trends Genet 2020; 37:35-45. [PMID: 32951948 DOI: 10.1016/j.tig.2020.08.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/13/2020] [Accepted: 08/20/2020] [Indexed: 12/20/2022]
Abstract
The golden age of antibiotics has passed, and the threat of untreatable antimicrobial resistant infections is now a reality for many individuals. Understanding how bacteria resist antimicrobial treatment and regulate gene expression in response to antibiotics is an important step towards combating resistance. In this review we focus on a ubiquitous class of bacterial gene regulators termed regulatory small RNAs (sRNAs) and how they contribute to antimicrobial resistance and tolerance. Small RNAs have notable roles in modulating the composition of the bacterial envelope, and through these functions control intrinsic antimicrobial resistance in many human pathogens. Recent technical advances that allow profiling of the 'sRNA interactome' have revealed a complex post-transcriptional network of sRNA interactions that can be used to identify network hubs and regulatory bottlenecks. Sequence-specific inhibition of these sRNAs with programmable RNA-targeting therapeutics may present avenues for treating antimicrobial resistant pathogens or resensitizing to our current antibiotics.
Collapse
Affiliation(s)
- Daniel G Mediati
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Sylvania Wu
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Winton Wu
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Jai J Tree
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
44
|
Baral B, Mozafari MR. Strategic Moves of "Superbugs" Against Available Chemical Scaffolds: Signaling, Regulation, and Challenges. ACS Pharmacol Transl Sci 2020; 3:373-400. [PMID: 32566906 PMCID: PMC7296549 DOI: 10.1021/acsptsci.0c00005] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Indexed: 12/12/2022]
Abstract
Superbugs' resistivity against available natural products has become an alarming global threat, causing a rapid deterioration in public health and claiming tens of thousands of lives yearly. Although the rapid discovery of small molecules from plant and microbial origin with enhanced bioactivity has provided us with some hope, a rapid hike in the resistivity of superbugs has proven to be the biggest therapeutic hurdle of all times. Moreover, several distinct mechanisms endowed by these notorious superbugs make them immune to these antibiotics subsequently causing our antibiotic wardrobe to be obsolete. In this unfortunate situation, though the time frame for discovering novel "hit molecules" down the line remains largely unknown, our small hope and untiring efforts injected in hunting novel chemical scaffolds with unique molecular targets using high-throughput technologies may safeguard us against these life-threatening challenges to some extent. Amid this crisis, the current comprehensive review highlights the present status of knowledge, our search for bacteria Achilles' heel, distinct molecular signaling that an opportunistic pathogen bestows to trespass the toxicity of antibiotics, and facile strategies and appealing therapeutic targets of novel drugs. Herein, we also discuss multidimensional strategies to combat antimicrobial resistance.
Collapse
Affiliation(s)
- Bikash Baral
- Department
of Biochemistry, University of Turku, Tykistökatu 6, Turku, Finland
| | - M. R. Mozafari
- Australasian
Nanoscience and Nanotechnology Initiative, 8054 Monash University LPO, Clayton, Victoria 3168, Australia
| |
Collapse
|
45
|
Daboor SM, Rohde JR, Cheng Z. Disruption of the extracellular polymeric network of Pseudomonas aeruginosa biofilms by alginate lyase enhances pathogen eradication by antibiotics. J Cyst Fibros 2020; 20:264-270. [PMID: 32482592 DOI: 10.1016/j.jcf.2020.04.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/15/2020] [Accepted: 04/18/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Pseudomonas aeruginosa forms antibiotic-resistant biofilms that are responsible for the treatment failure or relapses of the bacterial infections in the lungs of patients with cystic fibrosis (CF). The alginate lyases that target extracellular polysaccharide alginate of P. aeruginosa biofilms are promising therapeutic candidates for treatment of P. aeruginosa biofilm infections. METHODS Immunofluorescent staining and thin layer chromatography were used to demonstrate the alginolytic activity of the alginate lyase enzyme (AlyP1400) purified from a marine Pseudoalteromonas bacterium. Anti-biofilm activities of AlyP1400 were tested alone or in combination with antibiotics on the biofilms of a mucoid Pseudomonas aeruginosa clinical isolate CF27 that were cultivated in 96-well plates and a flow cell. RESULTS We showed that AlyP1400 facilitated antibiotic activities to eliminate CF27 biofilms. The combination of AlyP1400 with antibiotics reduced the biofilm biomass and boosted bactericidal activity of antibiotics. Importantly, we demonstrated that the enzymatic activity of AlyP1400 was required for its biofilm disruption activity and its synergy with antibiotics to eradicate biofilm cells. CONCLUSION This work shed new light on the potential mechanisms of the therapeutic activity for the combinational use of alginate lyase and antibiotics to treat P. aeruginosa infections in CF lungs or other P. aeruginosa biofilm-related infections.
Collapse
Affiliation(s)
- Said M Daboor
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, B3H 4R2, Canada; National Institute of Oceanography and Fisheries, Cairo, Egypt
| | - John R Rohde
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, B3H 4R2, Canada
| | - Zhenyu Cheng
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, B3H 4R2, Canada.
| |
Collapse
|
46
|
Investigating the Effects of Osmolytes and Environmental pH on Bacterial Persisters. Antimicrob Agents Chemother 2020; 64:AAC.02393-19. [PMID: 32094133 DOI: 10.1128/aac.02393-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/14/2020] [Indexed: 01/21/2023] Open
Abstract
Bacterial persisters are phenotypic variants that temporarily demonstrate an extraordinary tolerance toward antibiotics. Persisters have been linked to the recalcitrance of biofilm-related infections; hence, a complete understanding of their physiology can lead to improvement of therapeutic strategies for such infections. Mechanisms pertaining to persister formation are thought to be associated with stress response pathways triggered by intra- or extracellular stress factors. Unfortunately, studies demonstrating the effects of osmolyte- and/or pH-induced stresses on bacterial persistence are largely missing. To fill this knowledge gap within the field, we studied the effects of various osmolytes and pH conditions on Escherichia coli persistence with the use of phenotype microarrays and antibiotic tolerance assays. Although we found that a number of chemicals and pH environments, including urea, sodium nitrite, and acidic pH, significantly reduced persister formation in E. coli compared to no-osmolyte/no-buffer controls, this reduction in persister levels was less pronounced in late-stationary-phase cultures. Our results further demonstrated a positive correlation between cell growth and persister formation, which challenges the general notion in the field that slow-growing cultures have more persister cells than fast-growing cultures.
Collapse
|
47
|
Readel E, Portillo A, Talebi M, Armstrong DW. Enantiomeric separation of quorum sensing autoinducer homoserine lactones using GC-MS and LC-MS. Anal Bioanal Chem 2020; 412:2927-2937. [DOI: 10.1007/s00216-020-02534-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/12/2020] [Accepted: 02/18/2020] [Indexed: 10/24/2022]
|
48
|
Peppoloni S, Pericolini E, Colombari B, Pinetti D, Cermelli C, Fini F, Prati F, Caselli E, Blasi E. The β-Lactamase Inhibitor Boronic Acid Derivative SM23 as a New Anti- Pseudomonas aeruginosa Biofilm. Front Microbiol 2020; 11:35. [PMID: 32117094 PMCID: PMC7018986 DOI: 10.3389/fmicb.2020.00035] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 01/09/2020] [Indexed: 12/15/2022] Open
Abstract
Pseudomonas aeruginosa is a Gram-negative nosocomial pathogen, often causative agent of severe device-related infections, given its great capacity to form biofilm. P. aeruginosa finely regulates the expression of numerous virulence factors, including biofilm production, by Quorum Sensing (QS), a cell-to-cell communication mechanism used by many bacteria. Selective inhibition of QS-controlled pathogenicity without affecting bacterial growth may represent a novel promising strategy to overcome the well-known and widespread drug resistance of P. aeruginosa. In this study, we investigated the effects of SM23, a boronic acid derivate specifically designed as β-lactamase inhibitor, on biofilm formation and virulence factors production by P. aeruginosa. Our results indicated that SM23: (1) inhibited biofilm development and production of several virulence factors, such as pyoverdine, elastase, and pyocyanin, without affecting bacterial growth; (2) decreased the levels of 3-oxo-C12-HSL and C4-HSL, two QS-related autoinducer molecules, in line with a dampened lasR/lasI system; (3) failed to bind to bacterial cells that had been preincubated with P. aeruginosa-conditioned medium; and (4) reduced both biofilm formation and pyoverdine production by P. aeruginosa onto endotracheal tubes, as assessed by a new in vitro model closely mimicking clinical settings. Taken together, our results indicate that, besides inhibiting β-lactamase, SM23 can also act as powerful inhibitor of P. aeruginosa biofilm, suggesting that it may have a potential application in the prevention and treatment of biofilm-associated P. aeruginosa infections.
Collapse
Affiliation(s)
- Samuele Peppoloni
- Department of Surgical, Medical, Dental and Morphological Sciences With Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Eva Pericolini
- Department of Surgical, Medical, Dental and Morphological Sciences With Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Bruna Colombari
- Department of Surgical, Medical, Dental and Morphological Sciences With Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Diego Pinetti
- Centro Interdipartimentale "Grandi Strumenti" (CIGS), University of Modena and Reggio Emilia, Modena, Italy
| | - Claudio Cermelli
- Department of Surgical, Medical, Dental and Morphological Sciences With Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesco Fini
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Fabio Prati
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Emilia Caselli
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisabetta Blasi
- Department of Surgical, Medical, Dental and Morphological Sciences With Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
49
|
Machineni L. Effects of biotic and abiotic factors on biofilm growth dynamics and their heterogeneous response to antibiotic challenge. J Biosci 2020. [DOI: 10.1007/s12038-020-9990-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
50
|
Vadlamani RA, Dhanabal A, Detwiler DA, Pal R, McCarthy J, Seleem MN, Garner AL. Nanosecond electric pulses rapidly enhance the inactivation of Gram-negative bacteria using Gram-positive antibiotics. Appl Microbiol Biotechnol 2020; 104:2217-2227. [PMID: 31965221 DOI: 10.1007/s00253-020-10365-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 01/01/2020] [Accepted: 01/09/2020] [Indexed: 12/16/2022]
Abstract
Physically disrupting microorganism membranes to enable antibiotics to overcome resistance mechanisms that inhibit or excrete antibiotics has great potential for reducing antibiotic doses and rendering resistance mechanisms inert. We demonstrate the synergistic inactivation of a Gram-positive (Staphylococcus aureus) and two Gram-negative (Escherichia coli and Pseudomonas aeruginosa) bacteria by combining 222 30 kV/cm electric pulses (EPs) or 500 20 kV/cm EPs with 300-ns EP duration with various antibiotics with different mechanisms of action is demonstrated. Doses of antibiotics that produced no inactivation in 10 min of exposure in solution with bacteria induced several log reductions under the influence of nanosecond EPs. Combining 2 μg/L or 20 μg/mL of rifampicin with the 30 kV/cm EPs enhanced Staphylococcus aureus inactivation compared with EPs alone, while only a few of the other combinations demonstrated improvement. Combining 2 μg/L or 20 μg/mL of mupirocin or rifampicin with either EP train enhanced E. coli inactivation compared with EPs alone. Combining 2 μg/L or 20 μg/mL of erythromycin or vancomycin with the 30 kV/cm EPs enhanced E. coli inactivation compared with EPs alone. These results indicate that EPs can make Gram-positive antibiotics efficient for inactivating Gram-negative bacteria with future studies required to optimize EP parameters for other antibiotics and Gram-negative bacteria.
Collapse
Affiliation(s)
| | - Agni Dhanabal
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, USA
| | | | - Rusha Pal
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | | | - Mohamed N Seleem
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Allen L Garner
- School of Nuclear Engineering, Purdue University, West Lafayette, IN, USA. .,Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, USA. .,School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|