201
|
Lagha R, Abdallah FB, Mezni A, Alzahrani OM. Effect of Plasmonic Gold Nanoprisms on Biofilm Formation and Heat Shock Proteins Expression in Human Pathogenic Bacteria. Pharmaceuticals (Basel) 2021; 14:1335. [PMID: 34959736 PMCID: PMC8703320 DOI: 10.3390/ph14121335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/13/2021] [Accepted: 12/17/2021] [Indexed: 11/17/2022] Open
Abstract
Gold nanoparticles have gained interest in biomedical sciences in the areas of nano-diagnostics, bio-labeling, drug delivery, and bacterial infection. In this study, we examined, for the first time, the antibacterial and antibiofilm properties of plasmonic gold nanoprisms against human pathogenic bacteria using MIC and crystal violet. In addition, the expression level of GroEL/GroES heat shock proteins was also investigated by western blot. Gold nanoparticles were characterized by TEM and EDX, which showed equilateral triangular prisms with an average edge length of 150 nm. Antibacterial activity testing showed a great effect of AuNPs against pathogenic bacteria with MICs values ranging from 50 μg/mL to 100 μg/mL. Nanoparticles demonstrated strong biofilm inhibition action with a percentage of inhibition ranging from 40.44 to 82.43%. Western blot analysis revealed that GroEL was an AuNPs-inducible protein with an increase of up to 66.04%, but GroES was down-regulated with a reduction of up to 46.81%. Accordingly, plasmonic gold nanoprisms, could be a good candidate for antibiotics substitution in order to treat bacterial infections.
Collapse
Affiliation(s)
- Rihab Lagha
- Department of Biology, College of Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (R.L.); (O.M.A.)
- Research Unit UR17ES30: Virology and Antiviral Strategies, Higher Institute of Biotechnology, University of Monastir, Monastir 5000, Tunisia
| | - Fethi Ben Abdallah
- Department of Biology, College of Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (R.L.); (O.M.A.)
- Research Unit UR17ES30: Virology and Antiviral Strategies, Higher Institute of Biotechnology, University of Monastir, Monastir 5000, Tunisia
| | - Amine Mezni
- Department of Chemistry, College of Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Othman M. Alzahrani
- Department of Biology, College of Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (R.L.); (O.M.A.)
| |
Collapse
|
202
|
Stenger KS, Wikmark OG, Bezuidenhout CC, Molale-Tom LG. Microplastics pollution in the ocean: Potential carrier of resistant bacteria and resistance genes. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 291:118130. [PMID: 34562691 DOI: 10.1016/j.envpol.2021.118130] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 08/24/2021] [Accepted: 09/06/2021] [Indexed: 06/13/2023]
Abstract
Microplastics pollution in marine environments is concerning. Microplastics persist and accumulate in various sections of the ocean where they present opportunity for micropollutant accumulation and microbial colonisation. Even though biofilm formation on plastics was first reported in the 1970's, it is only in recent years were plastic associated biofilms have gained research attention. Plastic surfaces pose a problem as they are a niche ready for colonisation by diverse biofilm assemblages, composed of specific bacterial communities and putative pathogens prone to acquiring ARGs and resistance in the biofilm. However, the nature of antibiotic resistance on aquatic plastic debris is not yet fully understood and remains a concern. Given the inevitable increase of plastic production and waste generation, microplastics released into the environment may prove to be problematic. This review explores microplastic waste in the ocean and possible concerns that may arise from the presence of microplastics in conjunction with favourable conditions for the development and dispersal of antibiotic resistance in the ocean and food web.
Collapse
Affiliation(s)
- K S Stenger
- Unit for Environmental Sciences and Management - Microbiology, North-West University, Potchefstroom, South Africa.
| | - O G Wikmark
- Unit for Environmental Sciences and Management - Microbiology, North-West University, Potchefstroom, South Africa; GenØk - Centre for Biosafety, Tromsø, Norway.
| | - C C Bezuidenhout
- Unit for Environmental Sciences and Management - Microbiology, North-West University, Potchefstroom, South Africa.
| | - L G Molale-Tom
- Unit for Environmental Sciences and Management - Microbiology, North-West University, Potchefstroom, South Africa.
| |
Collapse
|
203
|
Maan H, Povolotsky TL, Porat Z, Itkin M, Malitsky S, Kolodkin-Gal I. Imaging flow cytometry reveals a dual role for exopolysaccharides in biofilms: To promote self-adhesion while repelling non-self-community members. Comput Struct Biotechnol J 2021; 20:15-25. [PMID: 34976308 PMCID: PMC8666610 DOI: 10.1016/j.csbj.2021.11.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022] Open
Abstract
In nature, bacteria frequently reside in differentiated communities or biofilms. These multicellular communities are held together by self-produced polymers that allow the community members to adhere to the surface as well as to neighbor bacteria. Here, we report that exopolysaccharides prevent Bacillus subtilis from co-aggregating with a distantly related bacterium Bacillus mycoides, while maintaining their role in promoting self-adhesion and co-adhesion with phylogenetically related bacterium, Bacillus atrophaeus. The defensive role of the exopolysaccharides is due to the specific regulation of bacillaene. Single cell analysis of biofilm and free-living bacterial cells using imaging flow cytometry confirmed a specific role for the exopolysaccharides in microbial competition repelling B. mycoides. Unlike exopolysaccharides, the matrix protein TasA induced bacillaene but inhibited the expression of the biosynthetic clusters for surfactin, and therefore its overall effect on microbial competition during floating biofilm formation was neutral. Thus, the exopolysaccharides provide a dual fitness advantage for biofilm-forming cells, as it acts to promote co-aggregation of related species, as well as, a secreted cue for chemical interference with non-compatible partners. These results experimentally demonstrate a general assembly principle of complex communities and provides an appealing explanation for how closely related species are favored during community assembly. Furthermore, the differential regulation of surfactin and bacillaene by the extracellular matrix may explain the spatio-temporal gradients of antibiotic production within biofilms.
Collapse
Affiliation(s)
- Harsh Maan
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | | | - Ziv Porat
- Flow Cytometry Unit, Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Maxim Itkin
- Life Science Core Facilities Weizmann Institute of Science, 234 Herzl Street, Rehovot, Israel
| | - Sergey Malitsky
- Life Science Core Facilities Weizmann Institute of Science, 234 Herzl Street, Rehovot, Israel
| | - Ilana Kolodkin-Gal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
204
|
Zarif ME, Yehia SA, Biță B, Sătulu V, Vizireanu S, Dinescu G, Holban AM, Marinescu F, Andronescu E, Grumezescu AM, Bîrcă AC, Farcașiu AT. Atmospheric Pressure Plasma Activation of Hydroxyapatite to Improve Fluoride Incorporation and Modulate Bacterial Biofilm. Int J Mol Sci 2021; 22:13103. [PMID: 34884908 PMCID: PMC8658314 DOI: 10.3390/ijms222313103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/28/2021] [Accepted: 11/30/2021] [Indexed: 11/27/2022] Open
Abstract
Despite the technological progress of the last decade, dental caries is still the most frequent oral health threat in children and adults alike. Such a condition has multiple triggers and is caused mainly by enamel degradation under the acidic attack of microbial cells, which compose the biofilm of the dental plaque. The biofilm of the dental plaque is a multispecific microbial consortium that periodically develops on mammalian teeth. It can be partially removed through mechanical forces by individual brushing or in specialized oral care facilities. Inhibition of microbial attachment and biofilm formation, as well as methods to strengthen dental enamel to microbial attack, represent the key factors in caries prevention. The purpose of this study was to elaborate a cold plasma-based method in order to modulate microbial attachment and biofilm formation and to improve the retention of fluoride (F-) in an enamel-like hydroxyapatite (HAP) model sample. Our results showed improved F retention in the HAP model, which correlated with an increased antimicrobial and antibiofilm effect. The obtained cold plasma with a dual effect exhibited through biofilm modulation and enamel strengthening through fluoridation is intended for dental application, such as preventing and treating dental caries and enamel deterioration.
Collapse
Affiliation(s)
- Maria Elena Zarif
- Low Temperature Plasma Laboratory, National Institute for Laser, Plasma and Radiation Physics, 077125 Măgurele, Romania
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 011061 Bucharest, Romania
| | - Sașa Alexandra Yehia
- Low Temperature Plasma Laboratory, National Institute for Laser, Plasma and Radiation Physics, 077125 Măgurele, Romania
- Faculty of Physics, University of Bucharest, 077125 Măgurele, Romania
| | - Bogdan Biță
- Low Temperature Plasma Laboratory, National Institute for Laser, Plasma and Radiation Physics, 077125 Măgurele, Romania
- Faculty of Physics, University of Bucharest, 077125 Măgurele, Romania
| | - Veronica Sătulu
- Low Temperature Plasma Laboratory, National Institute for Laser, Plasma and Radiation Physics, 077125 Măgurele, Romania
| | - Sorin Vizireanu
- Low Temperature Plasma Laboratory, National Institute for Laser, Plasma and Radiation Physics, 077125 Măgurele, Romania
| | - Gheorghe Dinescu
- Low Temperature Plasma Laboratory, National Institute for Laser, Plasma and Radiation Physics, 077125 Măgurele, Romania
- Faculty of Physics, University of Bucharest, 077125 Măgurele, Romania
| | - Alina Maria Holban
- Microbiology & Immunology Department, Faculty of Biology, University of Bucharest, 77206 Bucharest, Romania
- Research Institute of the University of Bucharest-ICUB, University of Bucharest, 050657 Bucharest, Romania
| | - Florica Marinescu
- Microbiology & Immunology Department, Faculty of Biology, University of Bucharest, 77206 Bucharest, Romania
- Research Institute of the University of Bucharest-ICUB, University of Bucharest, 050657 Bucharest, Romania
| | - Ecaterina Andronescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 011061 Bucharest, Romania
- Academy of Romanian Scientists, Ilfov no. 3, 050045 Bucharest, Romania
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 011061 Bucharest, Romania
- Research Institute of the University of Bucharest-ICUB, University of Bucharest, 050657 Bucharest, Romania
- Academy of Romanian Scientists, Ilfov no. 3, 050045 Bucharest, Romania
| | - Alexandra Cătălina Bîrcă
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 011061 Bucharest, Romania
| | - Alexandru Titus Farcașiu
- Department of Removable Prosthodontics, Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, 032799 Bucharest, Romania
| |
Collapse
|
205
|
Rather MA, Gupta K, Mandal M. Microbial biofilm: formation, architecture, antibiotic resistance, and control strategies. Braz J Microbiol 2021; 52:1701-1718. [PMID: 34558029 PMCID: PMC8578483 DOI: 10.1007/s42770-021-00624-x] [Citation(s) in RCA: 225] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 09/19/2021] [Indexed: 01/08/2023] Open
Abstract
The assembly of microorganisms over a surface and their ability to develop resistance against available antibiotics are major concerns of interest. To survive against harsh environmental conditions including known antibiotics, the microorganisms form a unique structure, referred to as biofilm. The mechanism of biofilm formation is triggered and regulated by quorum sensing, hostile environmental conditions, nutrient availability, hydrodynamic conditions, cell-to-cell communication, signaling cascades, and secondary messengers. Antibiotic resistance, escape of microbes from the body's immune system, recalcitrant infections, biofilm-associated deaths, and food spoilage are some of the problems associated with microbial biofilms which pose a threat to humans, veterinary, and food processing sectors. In this review, we focus in detail on biofilm formation, its architecture, composition, genes and signaling cascades involved, and multifold antibiotic resistance exhibited by microorganisms dwelling within biofilms. We also highlight different physical, chemical, and biological biofilm control strategies including those based on plant products. So, this review aims at providing researchers the knowledge regarding recent advances on the mechanisms involved in biofilm formation at the molecular level as well as the emergent method used to get rid of antibiotic-resistant and life-threatening biofilms.
Collapse
Affiliation(s)
- Muzamil Ahmad Rather
- Department of Molecular Biology and Biotechnology, Tezpur University (A Central University), Napaam, Tezpur, 784028, Assam, India
| | - Kuldeep Gupta
- Department of Molecular Biology and Biotechnology, Tezpur University (A Central University), Napaam, Tezpur, 784028, Assam, India
| | - Manabendra Mandal
- Department of Molecular Biology and Biotechnology, Tezpur University (A Central University), Napaam, Tezpur, 784028, Assam, India.
| |
Collapse
|
206
|
Carter CJ, Pillai K, Badar S, Mekkawy AH, Akhter J, Jefferies T, Valle SJ, Morris DL. Dissolution of Biofilm Secreted by Three Different Strains of Pseudomonas aeruginosa with Bromelain, N-Acetylcysteine, and Their Combinations. APPLIED SCIENCES 2021; 11:11388. [DOI: 10.3390/app112311388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
Bacterial infection of hernia mesh with the formation of biofilms presents a barrier to antibiotic treatment with subsequent surgical intervention and hospitalization. Hence, in the current study, we examined the effect of BromAc, a mucolytic agent, on the dissolution of biofilm formed by three different strains of Pseudomonas aeruginosa. Pseudomonas aeruginosa was carefully grown on hernia mesh and treated with various concentrations of bromelain, NAC, and their combinations at 37 °C over 4 h in vitro. Then, the biofilm dissolution activities of the agents were evaluated. Moreover, the combination index (CI) was analyzed to determine the synergy of the bromelain and NAC combination. The results indicated that biofilms were more susceptible to degradation by bromelain, whilst NAC showed growth enhancement in two of the strains. However, in combination (BromAc), the three strains were dramatically affected by the agents, with more than 80% debridement fir a suitable combination of bromelain and NAC that was also strain-specific. Hence, the current study shows that the biofilms formed by these three strains of Pseudomonas aeruginosa were adversely affected by a single treatment of BromAc, with more than 80% debridement, indicating that subsequent treatment may abolish the biofilm completely.
Collapse
Affiliation(s)
- Carly J. Carter
- Department of Surgery, St George Hospital, Sydney, NSW 2217, Australia
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia
| | - Krishna Pillai
- Department of Surgery, St George Hospital, Sydney, NSW 2217, Australia
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia
| | - Samina Badar
- Department of Surgery, St George Hospital, Sydney, NSW 2217, Australia
- St George & Sutherland Clinical School, University of New South Wales, Sydney, NSW 2217, Australia
| | - Ahmed H. Mekkawy
- Department of Surgery, St George Hospital, Sydney, NSW 2217, Australia
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia
| | - Javed Akhter
- Department of Surgery, St George Hospital, Sydney, NSW 2217, Australia
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia
| | - Thomas Jefferies
- School of Science, Western Sydney University, Richmond, NSW 2753, Australia
| | - Sarah J. Valle
- Department of Surgery, St George Hospital, Sydney, NSW 2217, Australia
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia
| | - David L. Morris
- Department of Surgery, St George Hospital, Sydney, NSW 2217, Australia
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia
- St George & Sutherland Clinical School, University of New South Wales, Sydney, NSW 2217, Australia
| |
Collapse
|
207
|
Sindeldecker D, Stoodley P. The many antibiotic resistance and tolerance strategies of Pseudomonas aeruginosa. Biofilm 2021; 3:100056. [PMID: 34471871 PMCID: PMC8387898 DOI: 10.1016/j.bioflm.2021.100056] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 12/15/2022] Open
Abstract
Pseudomonas aeruginosa is a bacterial pathogen associated with a wide range of infections and utilizes several strategies to establish and maintain infection including biofilm production, multidrug resistance, and antibiotic tolerance. Multidrug resistance in P. aeruginosa, as well as in all other bacterial pathogens, is a growing concern. Aminoglycoside resistance, in particular, is a major concern in P. aeruginosa infections and must be better understood in order to maintain effective clinical treatment. In this review, the various antibiotic resistance and tolerance mechanisms of Pseudomonas are explored including: classic mutation driven resistance, adaptive resistance, persister cells, small colony variants, phoenix colonies, and biofilms. It is important to further characterize each of these phenotypes and continue to evaluate antibiotic surviving isolates for novel driving mechanisms, so that we are better prepared to combat the rising number of recurrent and recalcitrant infections.
Collapse
Affiliation(s)
- Devin Sindeldecker
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Paul Stoodley
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
- Department of Orthopaedics, The Ohio State University, Columbus, OH, USA
- National Center for Advanced Tribology at Southampton (nCATS), Mechanical Engineering, University of Southampton, Southampton, UK
| |
Collapse
|
208
|
Jayasena Kaluarachchi TD, Campbell PM, Wickremasinghe R, Ranasinghe S, Wickremasinghe R, Yasawardene S, De Silva H, Menike C, Jayarathne MCK, Jayathilake S, Dilhari A, McBain AJ, Weerasekera MM. Distinct microbiome profiles and biofilms in Leishmania donovani-driven cutaneous leishmaniasis wounds. Sci Rep 2021; 11:23181. [PMID: 34848752 PMCID: PMC8633208 DOI: 10.1038/s41598-021-02388-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 11/15/2021] [Indexed: 11/09/2022] Open
Abstract
The endemic strain of Leishmania donovani in Sri Lanka causes cutaneous leishmaniasis (CL) rather than more common visceral form. We have visualized biofilms and profiled the microbiome of lesions and unaffected skin in thirty-nine CL patients. Twenty-four lesions (61.5%) were biofilm-positive according to fluorescence in situ hybridization. Biopsies of biofilm-positive lesions were dominated by Pseudomonas, class Bacilli and Enterobacteriaceae and distinguished by significantly lower community evenness. Higher relative abundance of a class Bacilli OTU was detected in wound swabs versus contralateral skin. Wound swabs and biopsies had significantly distinct microbiome profiles and lower diversity compared to unaffected skin. Greater abundances of potentially pathogenic organisms were observed in wet ulcers, lesions with high parasite loads and large wounds. In summary, more than half of L. donovani associated CL wounds harboured biofilms and the wounds exhibited a distinct, less diverse, microbiome than unaffected skin.
Collapse
Affiliation(s)
- T D Jayasena Kaluarachchi
- Department of Parasitology, Faculty of Medical Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka.
| | - Paul M Campbell
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Rajitha Wickremasinghe
- Department of Public Health, Faculty of Medicine, University of Kelaniya, Kelaniya, Sri Lanka
| | - Shalindra Ranasinghe
- Department of Parasitology, Faculty of Medical Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka
| | - Renu Wickremasinghe
- Department of Parasitology, Faculty of Medical Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka
| | - Surangi Yasawardene
- Department of Anatomy, Faculty of Medical Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka
| | | | - Chandrani Menike
- Department of Parasitology, Faculty of Medical Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka
| | - M C K Jayarathne
- Department of Family Medicine, Faculty of Medical Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka
| | - Subodha Jayathilake
- Department of Pathology, Faculty of Medical Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka
| | - Ayomi Dilhari
- Department of Basic Sciences, Faculty of Allied Health Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka
| | - Andrew J McBain
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Manjula M Weerasekera
- Department of Microbiology, Faculty of Medical Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka
| |
Collapse
|
209
|
Genomic and Metabolic Characteristics of the Pathogenicity in Pseudomonas aeruginosa. Int J Mol Sci 2021; 22:ijms222312892. [PMID: 34884697 PMCID: PMC8657582 DOI: 10.3390/ijms222312892] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/26/2021] [Accepted: 11/27/2021] [Indexed: 01/22/2023] Open
Abstract
In recent years, the effectiveness of antimicrobials in the treatment of Pseudomonas aeruginosa infections has gradually decreased. This pathogen can be observed in several clinical cases, such as pneumonia, urinary tract infections, sepsis, in immunocompromised hosts, such as neutropenic cancer, burns, and AIDS patients. Furthermore, Pseudomonas aeruginosa causes diseases in both livestock and pets. The highly flexible and versatile genome of P. aeruginosa allows it to have a high rate of pathogenicity. The numerous secreted virulence factors, resulting from its numerous secretion systems, the multi-resistance to different classes of antibiotics, and the ability to produce biofilms are pathogenicity factors that cause numerous problems in the fight against P. aeruginosa infections and that must be better understood for an effective treatment. Infections by P. aeruginosa represent, therefore, a major health problem and, as resistance genes can be disseminated between the microbiotas associated with humans, animals, and the environment, this issue needs be addressed on the basis of an One Health approach. This review intends to bring together and describe in detail the molecular and metabolic pathways in P. aeruginosa's pathogenesis, to contribute for the development of a more targeted therapy against this pathogen.
Collapse
|
210
|
Junghans S, Rojas SV, Skusa R, Püschel A, Grambow E, Kohlen J, Warnke P, Gummert J, Gross J. Bacteriophages for the Treatment of Graft Infections in Cardiovascular Medicine. Antibiotics (Basel) 2021; 10:antibiotics10121446. [PMID: 34943658 PMCID: PMC8698116 DOI: 10.3390/antibiotics10121446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 11/22/2022] Open
Abstract
Bacterial infections of vascular grafts represent a major burden in cardiovascular medicine, which is related to an increase in morbidity and mortality. Different factors that are associated with this medical field such as patient frailty, biofilm formation, or immunosuppression negatively influence antibiotic treatment, inhibiting therapy success. Thus, further treatment strategies are required. Bacteriophage antibacterial properties were discovered 100 years ago, but the focus on antibiotics in Western medicine since the mid-20th century slowed the further development of bacteriophage therapy. Therefore, the experience and knowledge gained until then in bacteriophage mechanisms of action, handling, clinical uses, and limitations were largely lost. However, the parallel emergence of antimicrobial resistance and individualized medicine has provoked a radical reassessment of this approach and cardiovascular surgery is one area in which phages may play an important role to cope with this new scenario. In this context, bacteriophages might be applicable for both prophylactic and therapeutic use, serving as a stand-alone therapy or in combination with antibiotics. From another perspective, standardization of phage application is also required. The ideal surgical bacteriophage application method should be less invasive, enabling highly localized concentrations, and limiting bacteriophage distribution to the infection site during a prolonged time lapse. This review describes the latest reports of phage therapy in cardiovascular surgery and discusses options for their use in implant and vascular graft infections.
Collapse
Affiliation(s)
- Simon Junghans
- G. Pohl-Boskamp GmbH & Co. KG, 25551 Hohenlockstedt, Germany;
| | - Sebastian V. Rojas
- Department of Cardio-Thoracic Surgery, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany; (S.V.R.); (J.G.)
| | - Romy Skusa
- Department for General, Visceral, Thoracic, Vascular and Transplantation Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (R.S.); (A.P.); (E.G.); (J.K.)
- Institute for Medical Microbiology, Virology and Hygiene, University Medicine Rostock, 18057 Rostock, Germany;
| | - Anja Püschel
- Department for General, Visceral, Thoracic, Vascular and Transplantation Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (R.S.); (A.P.); (E.G.); (J.K.)
| | - Eberhard Grambow
- Department for General, Visceral, Thoracic, Vascular and Transplantation Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (R.S.); (A.P.); (E.G.); (J.K.)
| | - Juliane Kohlen
- Department for General, Visceral, Thoracic, Vascular and Transplantation Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (R.S.); (A.P.); (E.G.); (J.K.)
| | - Philipp Warnke
- Institute for Medical Microbiology, Virology and Hygiene, University Medicine Rostock, 18057 Rostock, Germany;
| | - Jan Gummert
- Department of Cardio-Thoracic Surgery, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany; (S.V.R.); (J.G.)
| | - Justus Gross
- Department for General, Visceral, Thoracic, Vascular and Transplantation Surgery, Rostock University Medical Center, 18057 Rostock, Germany; (R.S.); (A.P.); (E.G.); (J.K.)
- Correspondence: ; Tel.:+49-381-494-146007
| |
Collapse
|
211
|
Park NH, Lee SJ, Lee EB, Birhanu BT, Park SC. Colistin Induces Resistance through Biofilm Formation, via Increased phoQ Expression, in Avian Pathogenic Escherichia coli. Pathogens 2021; 10:pathogens10111525. [PMID: 34832681 PMCID: PMC8620993 DOI: 10.3390/pathogens10111525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 11/16/2022] Open
Abstract
This study aimed to optimize the colistin-based antibacterial therapy to prevent antimicrobial resistance related to biofilm formation in avian pathogenic Escherichia coli (APEC) in chicken. Of all the bacterial isolates (n = 136), 69 were identified as APEC by polymerase chain reaction (PCR). Through a series of antibiotic susceptibility tests, susceptibility to colistin (<2 μg/mL) was confirmed in all isolates. Hence, a mutant selection window (MSW) was determined to obtain colistin-induced resistant bacteria. The minimum inhibitory concentration (MIC) of colistin against the colistin-induced resistant APEC strains ranged from 8 to 16 μg/mL. To identify the inhibitory activity of colistin against the resistant strains, the mutant prevention concentration (MPC) was investigated for 72 h, and the single and multi-dose colistin activities were determined through the time-kill curve against APEC strains. Bacterial regrowth occurred after 12 h at a double MIC50 concentration (1.00 μg/mL), and regrowth was not inhibited even during multiple exposures. However, upon exposure to 8 μg/mL—a concentration that was close to the MPC—the growth of APEC was inhibited, including in the resistant strains. Additionally, colistin-induced resistant strains showed a slower growth compared with the susceptible ones. Colistin-induced resistant APEC strains did not show colistin resistance gene (mcr-1). However, the expression of higher mgrB and phoQ levels was observed in the resistant strains. Furthermore, these strains showed increased formation of biofilm. Hence, the present study indicated that colistin could induce resistance through the increased formation of biofilm in APEC strains by enhancing the expression of phoQ.
Collapse
Affiliation(s)
- Na-Hye Park
- Laboratory Animal Center, Daegu Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea;
| | - Seung-Jin Lee
- Reproductive and Developmental Toxicology Research Group, Korea Institute of Toxicology, Daejeon 34114, Korea;
| | - Eon-Bee Lee
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea;
| | - Biruk Tesfaye Birhanu
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea;
- Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Korea
- Correspondence: (B.T.B.); (S.-C.P.); Tel.: +82-53-950-5964 (B.T.B. & S.-C.P.)
| | - Seung-Chun Park
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea;
- Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Korea
- Correspondence: (B.T.B.); (S.-C.P.); Tel.: +82-53-950-5964 (B.T.B. & S.-C.P.)
| |
Collapse
|
212
|
Patel N, Curtis JC, Plotkin BJ. Insulin Regulation of Escherichia coli Abiotic Biofilm Formation: Effect of Nutrients and Growth Conditions. Antibiotics (Basel) 2021; 10:antibiotics10111349. [PMID: 34827287 PMCID: PMC8615133 DOI: 10.3390/antibiotics10111349] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/21/2021] [Accepted: 11/02/2021] [Indexed: 11/23/2022] Open
Abstract
Escherichia coli plays an important role in biofilm formation across a wide array of disease and ecological settings. Insulin can function as an adjuvant in the regulation of biofilm levels. The modulation of insulin-regulated biofilm formation by environmental conditions has not been previously described. In the present study, the effects that various environmental growth conditions and nutrients have on insulin-modulated levels of biofilm production were measured. Micropipette tips were incubated with E. coli ATCC® 25922™ in a Mueller Hinton broth (MH), or a yeast nitrogen base with 1% peptone (YNBP), which was supplemented with glucose, lactose, galactose and/or insulin (Humulin®-R). The incubation conditions included a shaking or static culture, at 23 °C or 37 °C. After incubation, the biofilm production was calculated per CFU. At 23 °C, the presence of insulin increased biofilm formation. The amount of biofilm formation was highest in glucose > galactose >> lactose, while the biofilm levels decreased in shaking cultures, except for galactose (3-fold increase; 0.1% galactose and 20 μU insulin). At 37 °C, regardless of condition, there was more biofilm formation/CFU under static conditions in YNBP than in MH, except for the MH containing galactose. E. coli biofilm formation is influenced by aeration, temperature, and insulin concentration in combination with the available sugars.
Collapse
Affiliation(s)
- Nina Patel
- Department of Microbiology and Immunology, College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA; (N.P.); (J.C.C.)
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60625, USA
| | - Jeremy C. Curtis
- Department of Microbiology and Immunology, College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA; (N.P.); (J.C.C.)
| | - Balbina J. Plotkin
- Department of Microbiology and Immunology, College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA; (N.P.); (J.C.C.)
- Correspondence:
| |
Collapse
|
213
|
Roy J, Pandey V, Gupta I, Shekhar H. Antibacterial Sonodynamic Therapy: Current Status and Future Perspectives. ACS Biomater Sci Eng 2021; 7:5326-5338. [PMID: 34714638 DOI: 10.1021/acsbiomaterials.1c00587] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Multidrug-resistant bacteria have emerged in both community and hospital settings, partly due to the misuse of antibiotics. The inventory of viable antibiotics is rapidly declining, and efforts toward discovering newer antibiotics are not yielding the desired outcomes. Therefore, alternate antibacterial therapies based on physical mechanisms such as light and ultrasound are being explored. Sonodynamic therapy (SDT) is an emerging therapeutic approach that involves exposing target tissues to a nontoxic sensitizing chemical and low-intensity ultrasound. SDT can enable site-specific cytotoxicity by producing reactive oxygen species (ROS) in response to ultrasound, which can be harnessed for treating bacterial infections. This approach can potentially be used for both superficial and deep-seated microbial infections. The majority of the sonosensitizers reported are nonpolar, exhibiting limited bioavailability and a high clearance rate in the body. Therefore, targeted delivery agents such as nanoparticle composites, liposomes, and microbubbles are being investigated. This article reviews recent developments in antibacterial sonodynamic therapy, emphasizing biophysical and chemical mechanisms, novel delivery agents, ultrasound exposure and image guidance strategies, and the challenges in the pathway to clinical translation.
Collapse
Affiliation(s)
- Jayishnu Roy
- Discipline of Biological Engineering, Indian Institute of Technology (IIT) Gandhinagar, Gandhinagar, Gujarat 382355, India
| | - Vijayalakshmi Pandey
- Discipline of Chemistry, Indian Institute of Technology (IIT) Gandhinagar, Gandhinagar, Gujarat 382355, India
| | - Iti Gupta
- Discipline of Chemistry, Indian Institute of Technology (IIT) Gandhinagar, Gandhinagar, Gujarat 382355, India
| | - Himanshu Shekhar
- Discipline of Electrical Engineering, Indian Institute of Technology (IIT) Gandhinagar, Gandhinagar, Gujarat 382355, India
| |
Collapse
|
214
|
Wu Y, Yang G, van der Mei HC, Shi L, Busscher HJ, Ren Y. Synergy between "Probiotic" Carbon Quantum Dots and Ciprofloxacin in Eradicating Infectious Biofilms and Their Biosafety in Mice. Pharmaceutics 2021; 13:1809. [PMID: 34834224 PMCID: PMC8620463 DOI: 10.3390/pharmaceutics13111809] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/23/2021] [Accepted: 10/25/2021] [Indexed: 11/17/2022] Open
Abstract
Orally administrated probiotic bacteria can aid antibiotic treatment of intestinal infections, but their arrival at their intestinal target site is hampered by killing in the gastrointestinal tract and by antibiotics solely intended for pathogen killing. Carbon-quantum-dots are extremely small nanoparticles and can be derived from different sources, including bacteria. Here, we hypothesize that carbon-quantum-dots inherit antibacterial activity from probiotic source bacteria to fulfill a similar role as live probiotics in intestinal infection therapy. Physico-chemical analyses indicated that carbon-quantum-dots, hydrothermally derived from Bifidobacterium breve (B-C-dots), inherited proteins and polysaccharides from their source-bacteria. B-C-dots disrupted biofilm matrices of Escherichia coli and Salmonella typhimurium biofilms through extensive reactive-oxygen-species (ROS)-generation, causing a decrease in volumetric bacterial-density in biofilms. Decreased bacterial densities leave more open space in biofilms and have enhanced ciprofloxacin penetration and killing potential in an E. coli biofilm pre-exposed to probiotic B-C-dots. Pathogenic carbon-quantum-dots hydrothermally derived from E. coli (E-C-dots) did not disrupt pathogenic biofilms nor enhance E. coli killing potential by ciprofloxacin. B-C-dots were biosafe in mice upon daily administration, while E-C-dots demonstrated a decrease in white blood cell and platelet counts and an increase in C-reactive protein levels. Therefore, the way is paved for employing probiotic carbon-quantum-dots instead of viable, probiotic bacteria for synergistic use with existing antibiotics in treating intestinal infections.
Collapse
Affiliation(s)
- Yanyan Wu
- University of Groningen and University Medical Center of Groningen, Department of Orthodontics, Hanzeplein 1, 9700 RB Groningen, The Netherlands; (Y.W.); (Y.R.)
| | - Guang Yang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China; (G.Y.); (L.S.)
- University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands;
| | - Henny C. van der Mei
- University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands;
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China; (G.Y.); (L.S.)
| | - Henk J. Busscher
- University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands;
| | - Yijin Ren
- University of Groningen and University Medical Center of Groningen, Department of Orthodontics, Hanzeplein 1, 9700 RB Groningen, The Netherlands; (Y.W.); (Y.R.)
| |
Collapse
|
215
|
Exploration of the Pharmacodynamics for Pseudomonas aeruginosa Biofilm Eradication by Tobramycin. Antimicrob Agents Chemother 2021; 66:e0137121. [PMID: 34694882 DOI: 10.1128/aac.01371-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pseudomonas aeruginosa is a Gram-negative, opportunistic pathogen which is involved in numerous infections. It is of growing concern within the field of antibiotic resistant and tolerance and often exhibits multi-drug resistance. Previous studies have shown the emergence of antibiotic resistant and tolerant variants within the zone of clearance of a biofilm lawn after exposure to aminoglycosides. As concerning as the tolerant variant emergence is, there was also a zone of killing (ZOK) immediately surrounding the antibiotic source from which no detectable bacteria emerged or were cultured. In this study, the ZOK was analyzed using both in vitro and in silico methods to determine if there was a consistent antibiotic concentration versus time constraint (area under the curve, (AUC)) which is able to completely kill all bacteria in the lawn biofilms in our in vitro model. Our studies revealed that by achieving an average AUC of 4,372.5 μg*hr/mL, complete eradication of biofilms grown on both agar and hydroxyapatite was possible. These findings show that appropriate antibiotic concentrations and treatment duration may be able to treat antibiotic resistant and tolerant biofilm infections.
Collapse
|
216
|
Koide T, Tamura M. Effect of diglyceryl dicaprylate on Candida albicans growth and pathogenicity. Biosci Biotechnol Biochem 2021; 85:2334-2342. [PMID: 34508624 DOI: 10.1093/bbb/zbab159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/07/2021] [Indexed: 01/06/2023]
Abstract
The antifungal effect of diglyceryl dicaprylate, one of the emulsifiers used as a food additive, on Candida albicans that is a pathogenic fungus that is predominant in the oral cavity was investigated. This component did not affect C. albicans growth; however, it suppressed some virulence factors in a concentration-dependent manner. Furthermore, the suppression of pathogenic factors, such as biofilm formation, adhesion, highly pathogenic dimorphism, and ability to produce proteolytic enzymes, was due to reduction in mRNA expression levels of genes involved in fungal pathogenicities. From these results, this emulsifier could potentially prevent the development of intraoral and extraoral diseases involving C. albicans and could potentially use in oral care and improvement of quality of life.
Collapse
Affiliation(s)
- Tomojiro Koide
- Department of Food Ingredients Development, Riken Vitamin Co., Ltd., Mihama-ku Chiba-City, Chiba, Japan
| | - Muneaki Tamura
- Department of Microbiology, Nihon University School of Dentistry, Kanda-surugadai Chiyoda-ku, Tokyo, Japan
| |
Collapse
|
217
|
Niegowska M, Sanseverino I, Navarro A, Lettieri T. Knowledge gaps in the assessment of antimicrobial resistance in surface waters. FEMS Microbiol Ecol 2021; 97:fiab140. [PMID: 34625810 PMCID: PMC8528692 DOI: 10.1093/femsec/fiab140] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 10/06/2021] [Indexed: 11/26/2022] Open
Abstract
The spread of antibiotic resistance in the water environment has been widely described. However, still many knowledge gaps exist regarding the selection pressure from antibiotics, heavy metals and other substances present in surface waters as a result of anthropogenic activities, as well as the extent and impact of this phenomenon on aquatic organisms and humans. In particular, the relationship between environmental concentrations of antibiotics and the acquisition of ARGs by antibiotic-sensitive bacteria as well as the impact of heavy metals and other selective agents on antimicrobial resistance (AMR) need to be defined. Currently, established safety values are based on the effects of antibiotic toxicity neglecting the question of AMR spread. In turn, risk assessment of antibiotics in waterbodies remains a complex question implicating multiple variables and unknowns reinforced by the lack of harmonized protocols and official guidelines. In the present review, we discussed current state-of-the-art and the knowledge gaps related to pressure exerted by antibiotics and heavy metals on aquatic environments and their relationship to the spread of AMR. Along with this latter, we reflected on (i) the risk assessment in surface waters, (ii) selective pressures contributing to its transfer and propagation and (iii) the advantages of metagenomics in investigating AMR. Furthermore, the role of microplastics in co-selection for metal and antibiotic resistance, together with the need for more studies in freshwater are highlighted.
Collapse
Affiliation(s)
- Magdalena Niegowska
- European Commission, Joint Research Centre (JRC), Via Enrico Fermi 2749, 21027 Ispra, Italy
| | - Isabella Sanseverino
- European Commission, Joint Research Centre (JRC), Via Enrico Fermi 2749, 21027 Ispra, Italy
| | - Anna Navarro
- European Commission, Joint Research Centre (JRC), Via Enrico Fermi 2749, 21027 Ispra, Italy
| | - Teresa Lettieri
- European Commission, Joint Research Centre (JRC), Via Enrico Fermi 2749, 21027 Ispra, Italy
| |
Collapse
|
218
|
Silva C, Requicha JF, Martins JJ, Duarte A, Dias IR, Viegas CA, Saavedra MJ. Black-and-White Ruffed Lemur ( Varecia variegata) in Captivity: Analysis of the Oral Microbiota in a One Health Perspective. Animals (Basel) 2021; 11:2905. [PMID: 34679926 PMCID: PMC8533010 DOI: 10.3390/ani11102905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/24/2021] [Accepted: 09/29/2021] [Indexed: 11/16/2022] Open
Abstract
This study aimed to characterize the susceptibility profile to antibiotics and biofilm formation of Gram-negative bacterial isolates obtained from the oral cavity of the black-and-white ruffed lemur (Varecia variegata). From eight individuals from a zoo located in Portugal, samples of the oral microbiota were collected with sterile swabs and then placed in closed tubes with a transport medium. Culture was carried out for media of Gram-negative bacteria. Twenty-two isolates were obtained and subjected to susceptibility tests to twenty-five antimicrobial agents belonging to seven different classes. All tested isolates demonstrated resistance to, at least, one antibiotic, and it was possible to observe multidrug resistance in 11 of the 22 isolates (50%). It should be noted that an isolate showed phenotypic resistance to imipenem, an antibiotic for exclusive use in a hospital environment. All the isolates showed an increasing ability of biofilm formation over time. The obtained results show that wild mammals in captivity could be reservoirs and potential sources of multi-resistant pathogens. In view of this fact and considering the One Health concept, it will be advisable to establish local monitoring programs worldwide that benefit and protect human, animal and environmental health.
Collapse
Affiliation(s)
- Carolina Silva
- Department of Veterinary Sciences, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; (C.S.); (J.F.R.); (I.R.D.); (C.A.V.)
| | - João F. Requicha
- Department of Veterinary Sciences, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; (C.S.); (J.F.R.); (I.R.D.); (C.A.V.)
- Animal and Veterinary Research Center (CECAV) and AL4AnimalS, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal;
| | - José J. Martins
- Animal and Veterinary Research Center (CECAV) and AL4AnimalS, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal;
- Department of Animal Science, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal
| | - Aida Duarte
- Department of Microbiology and Immunology, Faculty of Pharmacy and Research, University of Lisbon, 1640-042 Lisbon, Portugal;
- Center of Egas Moniz (CiiEM), 2825-001 Monte da Caparica, Portugal
| | - Isabel R. Dias
- Department of Veterinary Sciences, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; (C.S.); (J.F.R.); (I.R.D.); (C.A.V.)
- Animal and Veterinary Research Center (CECAV) and AL4AnimalS, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal;
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB) and Inov4Agro, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
| | - Carlos A. Viegas
- Department of Veterinary Sciences, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; (C.S.); (J.F.R.); (I.R.D.); (C.A.V.)
- Animal and Veterinary Research Center (CECAV) and AL4AnimalS, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal;
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB) and Inov4Agro, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
| | - Maria J. Saavedra
- Department of Veterinary Sciences, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; (C.S.); (J.F.R.); (I.R.D.); (C.A.V.)
- Animal and Veterinary Research Center (CECAV) and AL4AnimalS, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal;
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB) and Inov4Agro, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
| |
Collapse
|
219
|
Patenall BL, Hathaway HJ, Laabei M, Young AE, Thet NT, Jenkins ATA, Short RD, Allinson SL. Assessment of mutations induced by cold atmospheric plasma jet treatment relative to known mutagens in Escherichia coli. Mutagenesis 2021; 36:380-387. [PMID: 34459491 DOI: 10.1093/mutage/geab030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 08/29/2021] [Indexed: 11/12/2022] Open
Abstract
The main bactericidal components of cold atmospheric plasma (CAP) are thought to be reactive oxygen and nitrogen species (RONS) and UV-radiation, both of which have the capacity to cause DNA damage and mutations. Here, the mutagenic effects of CAP on Escherichia coli were assessed in comparison to X- and UV-irradiation. DNA damage and mutagenesis were screened for using a diffusion-based DNA fragmentation assay and modified Ames test, respectively. Mutant colonies obtained from the latter were quantitated and sequenced. CAP was found to elicit a similar mutation spectrum to X-irradiation, which did not resemble that for UV implying that CAP-produced RONS are more likely the mutagenic component of CAP. CAP treatment was also shown to promote resistance to the antibiotic ciprofloxacin. Our data suggest that CAP treatment has mutagenic effects that may have important phenotypic consequences.
Collapse
Affiliation(s)
| | - Hollie J Hathaway
- Department of Chemistry, Faculty of Science and Technology, Lancaster University, Lancaster, LA1 4YB, UK
| | - Maisem Laabei
- Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AX, UK
| | - Amber E Young
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 1UD, UK
| | - Naing T Thet
- Department of Chemistry, University of Bath, Bath, BA2 7AY, UK
| | | | - Robert D Short
- Department of Chemistry, Faculty of Science and Technology, Lancaster University, Lancaster, LA1 4YB, UK
| | - Sarah L Allinson
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, LA1 4YG, UK
| |
Collapse
|
220
|
Bapat PS, Nobile CJ. Photodynamic Therapy Is Effective Against Candida auris Biofilms. Front Cell Infect Microbiol 2021; 11:713092. [PMID: 34540717 PMCID: PMC8446617 DOI: 10.3389/fcimb.2021.713092] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/13/2021] [Indexed: 12/23/2022] Open
Abstract
Fungal infections are increasing in prevalence worldwide. The paucity of available antifungal drug classes, combined with the increased occurrence of multidrug resistance in fungi, has led to new clinical challenges in the treatment of fungal infections. Candida auris is a recently emerged multidrug resistant human fungal pathogen that has become a worldwide public health threat. C. auris clinical isolates are often resistant to one or more antifungal drug classes, and thus, there is a high unmet medical need for the development of new therapeutic strategies effective against C. auris. Additionally, C. auris possesses several virulence traits, including the ability to form biofilms, further contributing to its drug resistance, and complicating the treatment of C. auris infections. Here we assessed red, green, and blue visible lights alone and in combination with photosensitizing compounds for their efficacies against C. auris biofilms. We found that (1) blue light inhibited and disrupted C. auris biofilms on its own and that the addition of photosensitizing compounds improved its antibiofilm potential; (2) red light inhibited and disrupted C. auris biofilms, but only in combination with photosensitizing compounds; and (3) green light inhibited C. auris biofilms in combination with photosensitizing compounds, but had no effects on disrupting C. auris biofilms. Taken together, our findings suggest that photodynamic therapy could be an effective non-drug therapeutic strategy against multidrug resistant C. auris biofilm infections.
Collapse
Affiliation(s)
- Priyanka S Bapat
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, CA, United States.,Quantitative and Systems Biology Graduate Program, University of California Merced, Merced, CA, United States
| | - Clarissa J Nobile
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, CA, United States.,Health Sciences Research Institute, University of California Merced, Merced, CA, United States
| |
Collapse
|
221
|
Thieme L, Hartung A, Tramm K, Graf J, Spott R, Makarewicz O, Pletz MW. Adaptation of the Start-Growth-Time Method for High-Throughput Biofilm Quantification. Front Microbiol 2021; 12:631248. [PMID: 34512560 PMCID: PMC8428173 DOI: 10.3389/fmicb.2021.631248] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 07/29/2021] [Indexed: 11/29/2022] Open
Abstract
Colony forming unit (CFU) determination by agar plating is still regarded as the gold standard for biofilm quantification despite being time- and resource-consuming. Here, we propose an adaption of the high-throughput Start-Growth-Time (SGT) method from planktonic to biofilm analysis, which indirectly quantifies CFU/mL numbers by evaluating regrowth curves of detached biofilms. For validation, the effect of dalbavancin, rifampicin and gentamicin against mature biofilms of Staphylococcus aureus and Enterococcus faecium was measured by accessing different features of the viability status of the cell, i.e., the cultivability (conventional agar plating), growth behavior (SGT) and metabolic activity (resazurin assay). SGT correlated well with the resazurin assay for all tested antibiotics, but only for gentamicin and rifampicin with conventional agar plating. Dalbavancin treatment-derived growth curves showed a compared to untreated controls significantly slower increase with reduced cell doubling times and reduced metabolic rate, but no change in CFU numbers was observed by conventional agar plating. Here, unspecific binding of dalbavancin to the biofilm interfered with the SGT methodology since the renewed release of dalbavancin during detachment of the biofilms led to an unintended antimicrobial effect. The application of the SGT method for anti-biofilm testing is therefore not suited for antibiotics which stick to the biofilm and/or to the bacterial cell wall. Importantly, the same applies for the well-established resazurin method for anti-biofilm testing. However, for antibiotics which do not bind to the biofilm as seen for gentamicin and rifampicin, the SGT method presents a much less labor-intensive method suited for high-throughput screening of anti-biofilm compounds.
Collapse
Affiliation(s)
- Lara Thieme
- Institute for Infectious Diseases and Infection Control, Jena University Hospital/Friedrich-Schiller-University, Jena, Germany.,Leibniz Center for Photonics in Infection Research, Jena University Hospital/Friedrich-Schiller-University, Jena, Germany
| | - Anita Hartung
- Institute for Infectious Diseases and Infection Control, Jena University Hospital/Friedrich-Schiller-University, Jena, Germany
| | - Kristina Tramm
- Institute for Infectious Diseases and Infection Control, Jena University Hospital/Friedrich-Schiller-University, Jena, Germany
| | - Julia Graf
- Institute for Infectious Diseases and Infection Control, Jena University Hospital/Friedrich-Schiller-University, Jena, Germany
| | - Riccardo Spott
- Institute for Infectious Diseases and Infection Control, Jena University Hospital/Friedrich-Schiller-University, Jena, Germany
| | - Oliwia Makarewicz
- Institute for Infectious Diseases and Infection Control, Jena University Hospital/Friedrich-Schiller-University, Jena, Germany
| | - Mathias W Pletz
- Institute for Infectious Diseases and Infection Control, Jena University Hospital/Friedrich-Schiller-University, Jena, Germany
| |
Collapse
|
222
|
Pang Z, Zhu Q. Traditional Chinese Medicine is an Alternative Therapeutic Option for Treatment of Pseudomonas aeruginosa Infections. Front Pharmacol 2021; 12:737252. [PMID: 34512364 PMCID: PMC8429605 DOI: 10.3389/fphar.2021.737252] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/18/2021] [Indexed: 12/17/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen causing life-threatening infections in cystic fibrosis patients and immunocompromised individuals, and it is a leading cause of nosocomial infections associated with significant morbidity and mortality. Treatment of P. aeruginosa infections is challenging due to the antibiotic resistance to most of the conventional antibiotics. Development of alternative therapeutic options is urgently demanded for the patients who have antibiotic-resistant infections. Traditional Chinese medicine (TCM) has a clinical history of thousands of years for prevention and treatment of infectious diseases in China, taking advantages of improving clinical outcomes, producing less side effects, inhibiting pathogen, and modulating host immunity. Recent research has revealed a variety of natural products derived from TCM showing significant antimicrobial effects on antibiotic-resistant strains of P. aeruginosa alone or combined with antibiotics in vitro or in animal models, suggesting that TCM is a promising complementary and alternative therapeutic approach for treatment of chronic P. aeruginosa infections. This review summarizes the recent findings attempting to dissect the mechanisms of TCM combating P. aeruginosa infections and highlights the molecular targets of TCM on P. aeruginosa and host.
Collapse
Affiliation(s)
- Zheng Pang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China.,Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China.,Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qingjun Zhu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China.,Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China.,Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
223
|
Qiu G, Wu H, Huang M, Ma T, Schneider A, Oates TW, Weir MD, Xu HHK, Zhao L. Novel calcium phosphate cement with biofilm-inhibition and platelet lysate delivery to enhance osteogenesis of encapsulated human periodontal ligament stem cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112306. [PMID: 34474857 DOI: 10.1016/j.msec.2021.112306] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 01/09/2023]
Abstract
Osteomyelitis is caused by Staphylococcus aureus (S. aureus), with associated progressive bone loss. This study developed for the first time a calcium phosphate cement (CPC) for delivery of doxycycline (DOX) and human platelet lysate (hPL) to fight against S. aureus infection and enhance the osteogenesis of human periodontal ligament stem cells (hPDLSCs). Chitosan-containing CPC scaffolds were fabricated in the absence (CPCC) or presence of DOX (CPCC+DOX). In addition, hPL was encapsulated in alginate microbeads and incorporated into CPCC+DOX (CPCC+DOX+ hPL). Flexural strength of CPCC+DOX + hPL was (5.56 ± 0.55) MPa, lower than (8.26 ± 1.6) MPa of CPCC+DOX (p < 0.05), but exceeding the reported strength of cancellous bone. CPCC+DOX and CPCC+DOX + hPL exhibited strong antibacterial activity against S. aureus, reducing biofilm CFU by 4 orders of magnitude. The hPDLSCs encapsulated in microbeads were co-cultured with the CPCs. The hPDLSCs were able to be released from the microbeads and showed a high proliferation rate, increasing by about 8 folds at 14 days for all groups. The hPL was released from the scaffold and promoted the osteogenic differentiation of hPDLSCs. ALP activity was 28.07 ± 5.15 mU/mg for CPCC+DOX + hPL, higher than 17.36 ± 2.37 mU/mg and 1.34 ± 0.37 mU/mg of CPCC+DOX and CPCC, respectively (p < 0.05). At 7 days, osteogenic genes (ALP, RUNX2, COL-1, and OPN) in CPCC+DOX + hPL were 3-10 folds those of control. The amount of hPDLSC-synthesized bone mineral with CPCC+DOX + hPL was 3.8 folds that of CPCC (p < 0.05). In summary, the novel CPC + DOX + hPL-hPDLSCs scaffold exhibited strong antibacterial activity, excellent cytocompatibility and hPDLSC osteogenic differentiation, showing a promising approach for treatment and prevention of bone infection and enhancement of bone regeneration.
Collapse
Affiliation(s)
- Gengtao Qiu
- Department of Trauma and Joint Surgery, Shunde Hospital, Southern Medical University, Foshan, Guangdong, China; Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA; Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hansen Wu
- General Administration Office, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mingguang Huang
- Department of Trauma and Joint Surgery, Shunde Hospital, Southern Medical University, Foshan, Guangdong, China
| | - Tao Ma
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Abraham Schneider
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD 21201, USA; Member, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Thomas W Oates
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Michael D Weir
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA.
| | - Hockin H K Xu
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA; Member, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Liang Zhao
- Department of Trauma and Joint Surgery, Shunde Hospital, Southern Medical University, Foshan, Guangdong, China; Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
224
|
Li D, Chen T, Zhang Y, Xu Y, Niu H. Synergistical Starvation and Chemo-Dynamic Therapy for Combating Multidrug-Resistant Bacteria and Accelerating Diabetic Wound Healing. Adv Healthc Mater 2021; 10:e2100716. [PMID: 34212536 DOI: 10.1002/adhm.202100716] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/10/2021] [Indexed: 12/18/2022]
Abstract
The application of the antibiotic drug has dramatically decreased the infection and promoted the development of surgery, but drug-resistant bacteria appeared along with the abuse of antibiotics. Especially, wound in diabetic patients provides more glucose for bacteria resulting in poor wound healing. Therefore, it is imminent to explore advanced agents for combating multidrug-resistant bacteria and accelerating diabetic wound healing. Herein, metal-organic frameworks based nanoreactors loaded with glucose oxidase (GOx) and peroxidase-like bovine hemoglobin (BHb) are designed to construct an effective cascaded catalytic antibacterial system. Therein, GOx can cost the glucose, and release H2 O2 simultaneously, which can then be transformed into hydroxyl radicals by BHb. As a result, the as-prepared nanoreactors can play the roles of both starving and killing toward the multidrug-resistant bacteria. Furthermore, the produced gluconic acid can reduce the pH of working condition, which is beneficial for both the enhancement of peroxidase activity and the inhibition of the bacteria growth. More importantly, the constructed nanoreactors can be degraded and excreted from the body in the form of feces, which render the as-proposed nanoreactors qualified as effective and safe materials for both combating multidrug-resistant bacteria in vitro and accelerating the diabetic wound healing in vivo of the mouse model.
Collapse
Affiliation(s)
- Danxia Li
- Department of Urology the Affiliated Hospital of Qingdao University Qingdao 266003 China
| | - Tao Chen
- Institute of Biomedical Engineering College of Life Sciences Qingdao University Qingdao 266071 China
| | - Yanfang Zhang
- Institute of Biomedical Engineering College of Life Sciences Qingdao University Qingdao 266071 China
| | - Yuanhong Xu
- Department of Urology the Affiliated Hospital of Qingdao University Qingdao 266003 China
- Institute of Biomedical Engineering College of Life Sciences Qingdao University Qingdao 266071 China
| | - Haitao Niu
- Department of Urology the Affiliated Hospital of Qingdao University Qingdao 266003 China
| |
Collapse
|
225
|
Alkylimidazolium ionic liquids for biofilm control: Experimental studies on controlling multispecies biofilms in natural waters. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.116859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
226
|
Afonso AC, Oliveira D, Saavedra MJ, Borges A, Simões M. Biofilms in Diabetic Foot Ulcers: Impact, Risk Factors and Control Strategies. Int J Mol Sci 2021; 22:8278. [PMID: 34361044 PMCID: PMC8347492 DOI: 10.3390/ijms22158278] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/13/2022] Open
Abstract
Diabetic foot ulcers (DFUs) are a serious complication from diabetes mellitus, with a huge economic, social and psychological impact on the patients' life. One of the main reasons why DFUs are so difficult to heal is related to the presence of biofilms. Biofilms promote wound inflammation and a remarkable lack of response to host defences/treatment options, which can lead to disease progression and chronicity. In fact, appropriate treatment for the elimination of these microbial communities can prevent the disease evolution and, in some cases, even avoid more serious outcomes, such as amputation or death. However, the detection of biofilm-associated DFUs is difficult due to the lack of methods for diagnostics in clinical settings. In this review, the current knowledge on the involvement of biofilms in DFUs is discussed, as well as how the surrounding environment influences biofilm formation and regulation, along with its clinical implications. A special focus is also given to biofilm-associated DFU diagnosis and therapeutic strategies. An overview on promising alternative therapeutics is provided and an algorithm considering biofilm detection and treatment is proposed.
Collapse
Affiliation(s)
- Ana C. Afonso
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal; (A.C.A.); (D.O.); (A.B.)
- CITAB—Centre for the Research and Technology for Agro-Environment and Biological Sciences, University of Trás-os-Montes e Alto Douro, 5001-801 Vila Real, Portugal;
- CEB—Centre of Biological Engineering, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal
| | - Diana Oliveira
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal; (A.C.A.); (D.O.); (A.B.)
- CIQUP, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal
| | - Maria José Saavedra
- CITAB—Centre for the Research and Technology for Agro-Environment and Biological Sciences, University of Trás-os-Montes e Alto Douro, 5001-801 Vila Real, Portugal;
- Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences, University of Trás-os-Montes e Alto Douro, 5001-801 Vila Real, Portugal
| | - Anabela Borges
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal; (A.C.A.); (D.O.); (A.B.)
| | - Manuel Simões
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal; (A.C.A.); (D.O.); (A.B.)
| |
Collapse
|
227
|
Picchi SC, de Souza E Silva M, Saldanha LL, Ferreira H, Takita MA, Caldana C, de Souza AA. GC-TOF/MS-based metabolomics analysis to investigate the changes driven by N-Acetylcysteine in the plant-pathogen Xanthomonas citri subsp. citri. Sci Rep 2021; 11:15558. [PMID: 34330957 PMCID: PMC8324833 DOI: 10.1038/s41598-021-95113-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 07/09/2021] [Indexed: 11/09/2022] Open
Abstract
N-Acetylcysteine (NAC) is an antioxidant, anti-adhesive, and antimicrobial compound. Even though there is much information regarding the role of NAC as an antioxidant and anti-adhesive agent, little is known about its antimicrobial activity. In order to assess its mode of action in bacterial cells, we investigated the metabolic responses triggered by NAC at neutral pH. As a model organism, we chose the Gram-negative plant pathogen Xanthomonas citri subsp. citri (X. citri), the causal agent of citrus canker disease, due to the potential use of NAC as a sustainable molecule against phytopathogens dissemination in citrus cultivated areas. In presence of NAC, cell proliferation was affected after 4 h, but damages to the cell membrane were observed only after 24 h. Targeted metabolite profiling analysis using GC-MS/TOF unravelled that NAC seems to be metabolized by the cells affecting cysteine metabolism. Intriguingly, glutamine, a marker for nitrogen status, was not detected among the cells treated with NAC. The absence of glutamine was followed by a decrease in the levels of the majority of the proteinogenic amino acids, suggesting that the reduced availability of amino acids affect protein synthesis and consequently cell proliferation.
Collapse
Affiliation(s)
- Simone Cristina Picchi
- Centro de Citricultura "Sylvio Moreira" - Instituto Agronômico de Campinas, Cordeirópolis, São Paulo, 13490-970, Brazil
| | - Mariana de Souza E Silva
- Centro de Citricultura "Sylvio Moreira" - Instituto Agronômico de Campinas, Cordeirópolis, São Paulo, 13490-970, Brazil
| | - Luiz Leonardo Saldanha
- Departamento de Bioquímica e Microbiologia, Instituto de Biociências, Universidade Estadual Paulista, Rio Claro, São Paulo, 13506-900, Brazil
| | - Henrique Ferreira
- Departamento de Bioquímica e Microbiologia, Instituto de Biociências, Universidade Estadual Paulista, Rio Claro, São Paulo, 13506-900, Brazil
| | - Marco Aurélio Takita
- Centro de Citricultura "Sylvio Moreira" - Instituto Agronômico de Campinas, Cordeirópolis, São Paulo, 13490-970, Brazil
| | - Camila Caldana
- Laboratório Nacional de Ciência e Tecnologia do Bioetanol - Centro Nacional de Pesquisa em Energia e Materiais, Campinas, São Paulo, 13083-100, Brazil.,Max-Planck-Institut Für Molekulare Pflanzenphysiologie, Wissenschaftspark Golm, Am Mühlenberg 1, 14476, Potsdam, Germany
| | - Alessandra Alves de Souza
- Centro de Citricultura "Sylvio Moreira" - Instituto Agronômico de Campinas, Cordeirópolis, São Paulo, 13490-970, Brazil.
| |
Collapse
|
228
|
Trebino MA, Shingare RD, MacMillan JB, Yildiz FH. Strategies and Approaches for Discovery of Small Molecule Disruptors of Biofilm Physiology. Molecules 2021; 26:molecules26154582. [PMID: 34361735 PMCID: PMC8348372 DOI: 10.3390/molecules26154582] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 12/02/2022] Open
Abstract
Biofilms, the predominant growth mode of microorganisms, pose a significant risk to human health. The protective biofilm matrix, typically composed of exopolysaccharides, proteins, nucleic acids, and lipids, combined with biofilm-grown bacteria’s heterogenous physiology, leads to enhanced fitness and tolerance to traditional methods for treatment. There is a need to identify biofilm inhibitors using diverse approaches and targeting different stages of biofilm formation. This review discusses discovery strategies that successfully identified a wide range of inhibitors and the processes used to characterize their inhibition mechanism and further improvement. Additionally, we examine the structure–activity relationship (SAR) for some of these inhibitors to optimize inhibitor activity.
Collapse
Affiliation(s)
- Michael A. Trebino
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, CA 95064, USA;
| | - Rahul D. Shingare
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA 95064, USA;
| | - John B. MacMillan
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA 95064, USA;
- Correspondence: (J.B.M.); (F.H.Y.)
| | - Fitnat H. Yildiz
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, CA 95064, USA;
- Correspondence: (J.B.M.); (F.H.Y.)
| |
Collapse
|
229
|
Von Glinski A, Frieler S, Elia CJ, Ansari D, Pierre C, Ishak B, Blecher R, Qutteineh B, Strot S, Oskouian RJ, Chapman JR. Surgical Management of Charcot Spinal Arthropathy in the Face of Possible Infection. Int J Spine Surg 2021; 15:752-762. [PMID: 34315758 DOI: 10.14444/8097] [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] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND The design is a retrospective cohort study. Charcot spinal arthropathy (CSA) is a rare and poorly understood progressive destructive spine condition that usually affects patients with preexisting spinal cord injury. The complexity of this condition, especially when additionally burdened by superimposed infection in the CSA zone, can potentially lead to suboptimal management such as protracted antibiotic therapy, predisposition to hardware failure, and pseudarthrosis. While in noninfected CSA primary stabilization is the major goal, staged surgical management has not been stratified based upon presence of a superinfected CSA. We compare clinical and radiological outcomes of surgical treatment in CSA patients with and without concurrent spinal infections. METHODS Our single-institution database was reviewed for all patients diagnosed with CSA and surgically treated, who were subsequently divided into 2 cohorts: spinal arthropathy with superimposed infection and those without. Those were comparatively studied for complications and reoperation rate. RESULTS Fifteen patients with CSA underwent surgical intervention; mean follow up of 15.3 months (range, 0-43). Eleven patients received stabilization with a quadruple-rod thoracolumbopelvic construct, while 4 patients with superinfected CSA underwent a staged procedure. Patients treated with a staged approach experienced fewer intraoperative complications (0% versus 18%) and fewer revision surgeries (25% versus 36%). Both cohorts had the same eventual healing. CONCLUSIONS Surgical management in CSA patients with primary emphasis on stability and modified surgical treatment based on presence of an active infection in the zone of neuropathic destruction will lead to similar eventual successful results with relatively few and manageable complications in this challenging patient population. LEVEL OF EVIDENCE 4. CLINICAL RELEVANCE The proposed treatment algorithm including the use of a quadruple-rod construct with lumbopelivic fixation and a staged approach in patients with superinfected CSA represents a reasonable option in the surgical treatment of CSA.
Collapse
Affiliation(s)
- Alexander Von Glinski
- Swedish Neuroscience Institute, Swedish Medical Center, Seattle, Washington.,Seattle Science Foundation, Seattle, Washington.,Department of Trauma Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany.,Hansjörg Wyss Hip and Pelvic Center, Swedish Hospital, Seattle, Washington
| | - Sven Frieler
- Swedish Neuroscience Institute, Swedish Medical Center, Seattle, Washington.,Seattle Science Foundation, Seattle, Washington.,Department of Trauma Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany.,Hansjörg Wyss Hip and Pelvic Center, Swedish Hospital, Seattle, Washington
| | - Christopher J Elia
- Swedish Neuroscience Institute, Swedish Medical Center, Seattle, Washington.,Seattle Science Foundation, Seattle, Washington.,Department of Neurosurgery, Riverside University Health Systems, Moreno Valley, California
| | - Darius Ansari
- Swedish Neuroscience Institute, Swedish Medical Center, Seattle, Washington.,Seattle Science Foundation, Seattle, Washington
| | - Clifford Pierre
- Swedish Neuroscience Institute, Swedish Medical Center, Seattle, Washington.,Seattle Science Foundation, Seattle, Washington
| | - Basem Ishak
- Swedish Neuroscience Institute, Swedish Medical Center, Seattle, Washington.,Seattle Science Foundation, Seattle, Washington.,Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Ronen Blecher
- Swedish Neuroscience Institute, Swedish Medical Center, Seattle, Washington
| | - Bilal Qutteineh
- Swedish Neuroscience Institute, Swedish Medical Center, Seattle, Washington
| | - Sarah Strot
- Swedish Neuroscience Institute, Swedish Medical Center, Seattle, Washington
| | - Rod J Oskouian
- Swedish Neuroscience Institute, Swedish Medical Center, Seattle, Washington.,Seattle Science Foundation, Seattle, Washington
| | - Jens R Chapman
- Swedish Neuroscience Institute, Swedish Medical Center, Seattle, Washington
| |
Collapse
|
230
|
Novel Nitro-Heteroaromatic Antimicrobial Agents for the Control and Eradication of Biofilm-Forming Bacteria. Antibiotics (Basel) 2021; 10:antibiotics10070855. [PMID: 34356776 PMCID: PMC8300661 DOI: 10.3390/antibiotics10070855] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/09/2021] [Accepted: 07/10/2021] [Indexed: 12/20/2022] Open
Abstract
The synthesis and biological activity of several novel nitrothiazole, nitrobenzothiazole, and nitrofuran containing antimicrobial agents for the eradication of biofilm-forming Gram-negative and Gram-positive pathogens is described. Nitazoxanide (NTZ), nitrofurantoin, and furazolidone are commercial antimicrobials which were used as models to show how structural modification improved activity toward planktonic bacteria via minimum inhibitory concentration (MIC) assays and biofilms via minimum biofilm eradication concentration (MBEC) assays. Structure–activity relationship (SAR) studies illustrate the ways in which improvements have been made to the aforementioned antimicrobial agents. It is of particular interest in this regard that the introduction of a chloro substituent at the 5-position of NTZ (analog 1b) resulted in marked activity enhancement, as did the replacement of the 2-acetoxy substituent in the latter compound with a basic amine group (analog 7b). It is also of importance that analog 4a, which is a simple methacrylamide, displayed noteworthy activity against S. epidermidis biofilms. These lead compounds identified to have high activity towards biofilms provide promise as starting points in future pro-drug studies.
Collapse
|
231
|
Tomlinson BR, Malof ME, Shaw LN. A global transcriptomic analysis of Staphylococcus aureus biofilm formation across diverse clonal lineages. Microb Genom 2021; 7. [PMID: 34227933 PMCID: PMC8477394 DOI: 10.1099/mgen.0.000598] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
A key characteristic of Staphylococcus aureus infections, and one that also varies phenotypically between clones, is that of biofilm formation, which aids in bacterial persistence through increased adherence and immune evasion. Though there is a general understanding of the process of biofilm formation - adhesion, proliferation, maturation and dispersal - the tightly orchestrated molecular events behind each stage, and what drives variation between S. aureus strains, has yet to be unravelled. Herein we measure biofilm progression and dispersal in real-time across the five major S. aureus CDC-types (USA100-USA500) revealing adherence patterns that differ markedly amongst strains. To gain insight into this, we performed transcriptomic profiling on these isolates at multiple timepoints, compared to planktonically growing counterparts. Our findings support a model in which eDNA release, followed by increased positive surface charge, perhaps drives initial abiotic attachment. This is seemingly followed by cooperative repression of autolysis and activation of poly-N-acetylglucosamine (PNAG) production, which may indicate a developmental shift in structuring the biofilm matrix. As biofilms mature, diminished translational capacity was apparent, with 53 % of all ribosomal proteins downregulated, followed by upregulation of anaerobic respiration enzymes. These findings are noteworthy because reduced cellular activity and an altered metabolic state have been previously shown to contribute to higher antibiotic tolerance and bacterial persistence. In sum, this work is, to our knowledge, the first study to investigate transcriptional regulation during the early, establishing phase of biofilm formation, and to compare global transcriptional regulation both temporally and across multiple clonal lineages.
Collapse
Affiliation(s)
- Brooke R Tomlinson
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, ISA 2015, Tampa, FL, USA
| | - Morgan E Malof
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, ISA 2015, Tampa, FL, USA
| | - Lindsey N Shaw
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, ISA 2015, Tampa, FL, USA
| |
Collapse
|
232
|
Tomlinson BR, Malof ME, Shaw LN. A global transcriptomic analysis of Staphylococcus aureus biofilm formation across diverse clonal lineages. Microb Genom 2021. [PMID: 34227933 DOI: 10.1099/mgen0000598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
A key characteristic of Staphylococcus aureus infections, and one that also varies phenotypically between clones, is that of biofilm formation, which aids in bacterial persistence through increased adherence and immune evasion. Though there is a general understanding of the process of biofilm formation - adhesion, proliferation, maturation and dispersal - the tightly orchestrated molecular events behind each stage, and what drives variation between S. aureus strains, has yet to be unravelled. Herein we measure biofilm progression and dispersal in real-time across the five major S. aureus CDC-types (USA100-USA500) revealing adherence patterns that differ markedly amongst strains. To gain insight into this, we performed transcriptomic profiling on these isolates at multiple timepoints, compared to planktonically growing counterparts. Our findings support a model in which eDNA release, followed by increased positive surface charge, perhaps drives initial abiotic attachment. This is seemingly followed by cooperative repression of autolysis and activation of poly-N-acetylglucosamine (PNAG) production, which may indicate a developmental shift in structuring the biofilm matrix. As biofilms mature, diminished translational capacity was apparent, with 53 % of all ribosomal proteins downregulated, followed by upregulation of anaerobic respiration enzymes. These findings are noteworthy because reduced cellular activity and an altered metabolic state have been previously shown to contribute to higher antibiotic tolerance and bacterial persistence. In sum, this work is, to our knowledge, the first study to investigate transcriptional regulation during the early, establishing phase of biofilm formation, and to compare global transcriptional regulation both temporally and across multiple clonal lineages.
Collapse
Affiliation(s)
- Brooke R Tomlinson
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, ISA 2015, Tampa, FL, USA
| | - Morgan E Malof
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, ISA 2015, Tampa, FL, USA
| | - Lindsey N Shaw
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, ISA 2015, Tampa, FL, USA
| |
Collapse
|
233
|
Silver and Copper Nanoparticles Inhibit Biofilm Formation by Mastitis Pathogens. Animals (Basel) 2021; 11:ani11071884. [PMID: 34202806 PMCID: PMC8300152 DOI: 10.3390/ani11071884] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/20/2021] [Accepted: 06/21/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Bovine mastitis is a common disease in cows. It is caused by many pathogen species, which can form three-dimensional structures composed of bacterial cells, known as biofilms. These structures are almost impermeable to antimicrobials, making treatment difficult. We looked at the influence of metal nanometre-scale particles on biofilm formation by several pathogen species. We analysed the properties of these nanoparticles, determined the concentration needed to inhibit the growth of pathogens and to damage their membranes, and finally, checked how nanoparticles influence biofilm formation. We show that metal nanoparticles (silver and copper nanoparticles and their mixture) limit the formation of biofilm very effectively. These results mean that nanoparticles can be used to cure cattle suffering from mastitis, which will lead to higher milk production and less financial loss. Abstract Bovine mastitis is a common bovine disease, frequently affecting whole herds of cattle. It is often caused by resistant microbes that can create a biofilm structure. The rapidly developing scientific discipline known as nanobiotechnology may help treat this illness, thanks to the extraordinary properties of nanoparticles. The aim of the study was to investigate the inhibition of biofilms created by mastitis pathogens after treatment with silver and copper nanoparticles, both individually and in combination. We defined the physicochemical properties and minimal inhibitory concentration of the nanoparticles and observed their interaction with the cell membrane, as well as the extent of biofilm reduction. The results show that the silver–copper complex was the most active of all nanomaterials tested (biofilm was reduced by nearly 100% at a concentration of 200 ppm for each microorganism species tested). However, silver nanoparticles were also effective individually (biofilm was also reduced by nearly 100% at a concentration of 200 ppm, but at concentrations of 50 and 100 ppm, the extent of reduction was lower than for the complex). Nanoparticles can be used in new alternative therapies to treat bovine mastitis.
Collapse
|
234
|
Bandy A, Tantry B. ESBL Activity, MDR, and Carbapenem Resistance among Predominant Enterobacterales Isolated in 2019. Antibiotics (Basel) 2021; 10:744. [PMID: 34205425 PMCID: PMC8234840 DOI: 10.3390/antibiotics10060744] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/14/2022] Open
Abstract
Antimicrobial-resistance in Enterobacterales is a serious concern in Saudi Arabia. The present study retrospectively analyzed the antibiograms of Enterobacterales identified from 1 January 2019 to 31 December 2019 from a referral hospital in the Aljouf region of Saudi Arabia. The revised document of the Centers for Disease Control (CDC) CR-2015 and Magiorakos et al.'s document were used to define carbapenem resistance and classify resistant bacteria, respectively. The association of carbapenem resistance, MDR, and ESBL with various sociodemographic characteristics was assessed by the chi-square test and odds ratios. In total, 617 Enterobacterales were identified. The predominant (n = 533 (86.4%)) isolates consisted of 232 (37.6%), 200 (32.4%), and 101 (16.4%) Escherichia coli, Klebsiella pneumoniae, and Proteus mirabilis, respectively. In general, 432 (81.0%) and 128 (24.0%) isolates were of MDR and ESBL, respectively. The MDR strains were recovered in higher frequency from intensive care units (OR = 3.24 (1.78-5.91); p < 0.01). E. coli and K. pneumoniae resistance rates to imipenem (2.55 (1.21-5.37); p < 0.01) and meropenem (2.18 (1.01-4.67); p < 0.04), respectively, were significantly higher in winter. The data emphasize that MDR isolates among Enterobacterales are highly prevalent. The studied Enterobacterales exhibited seasonal variation in antimicrobial resistance rates towards carbapenems and ESBL activity.
Collapse
Affiliation(s)
- Altaf Bandy
- Family & Community Medicine, College of Medicine, Jouf University, 74311 Sakaka, Aljouf, Saudi Arabia
| | - Bilal Tantry
- Ex-faculty, Department of microbiology, College of Medicine, Jouf University, 74311 Sakaka, Aljouf, Saudi Arabia;
| |
Collapse
|
235
|
Munteanu AC, Uivarosi V. Ruthenium Complexes in the Fight against Pathogenic Microorganisms. An Extensive Review. Pharmaceutics 2021; 13:874. [PMID: 34199283 PMCID: PMC8232020 DOI: 10.3390/pharmaceutics13060874] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/07/2021] [Accepted: 06/09/2021] [Indexed: 12/13/2022] Open
Abstract
The widespread use of antibiotics has resulted in the emergence of drug-resistant populations of microorganisms. Clearly, one can see the need to develop new, more effective, antimicrobial agents that go beyond the explored 'chemical space'. In this regard, their unique modes of action (e.g., reactive oxygen species (ROS) generation, redox activation, ligand exchange, depletion of substrates involved in vital cellular processes) render metal complexes as promising drug candidates. Several Ru (II/III) complexes have been included in, or are currently undergoing, clinical trials as anticancer agents. Based on the in-depth knowledge of their chemical properties and biological behavior, the interest in developing new ruthenium compounds as antibiotic, antifungal, antiparasitic, or antiviral drugs has risen. This review will discuss the advantages and disadvantages of Ru (II/III) frameworks as antimicrobial agents. Some aspects regarding the relationship between their chemical structure and mechanism of action, cellular localization, and/or metabolism of the ruthenium complexes in bacterial and eukaryotic cells are discussed as well. Regarding the antiviral activity, in light of current events related to the Covid-19 pandemic, the Ru (II/III) compounds used against SARS-CoV-2 (e.g., BOLD-100) are also reviewed herein.
Collapse
Affiliation(s)
- Alexandra-Cristina Munteanu
- Department of General and Inorganic Chemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Valentina Uivarosi
- Department of General and Inorganic Chemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania
| |
Collapse
|
236
|
Algburi A, Alazzawi SA, Al-Ezzy AIA, Weeks R, Chistyakov V, Chikindas ML. Potential Probiotics Bacillus subtilis KATMIRA1933 and Bacillus amyloliquefaciens B-1895 Co-Aggregate with Clinical Isolates of Proteus mirabilis and Prevent Biofilm Formation. Probiotics Antimicrob Proteins 2021; 12:1471-1483. [PMID: 31989448 DOI: 10.1007/s12602-020-09631-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A urinary tract infection (UTI) is a multi-factorial disease including cystitis, pyelonephritis, and pyelitis. After Escherichia coli, Proteus mirabilis is the most common UTI-associated opportunistic pathogen. Antibiotic resistance of bacteria and infection recurrence can be connected to biofilm formation by P. mirabilis. In this study, human and sheep isolates of P. mirabilis were investigated for antibiotic sensitivity using an antibiotic disk test. Co-aggregation of the tested potential probiotic bacilli, Bacillus amyloliquefaciens B-1895 and Bacillus subtilis KATMIRA1933, with the isolated pathogen was also evaluated. Then, the anti-biofilm activity of naturally derived metabolites, such as subtilin and subtilosin, in the bacilli-free supernatants was assessed against biofilms of P. mirabilis isolates. The isolated pathogens were sensitive to 30 μg of amikacin and 5 μg of ciprofloxacin but resistant to other tested antibiotics. After 24 h, auto-aggregation of B. amyloliquefaciens B-1895 was at 89.5% and higher than auto-aggregation of B. subtilis KATMIRA1933 (59.5%). B. amyloliquefaciens B-1895 strongly co-aggregated with P. mirabilis isolates from human UTIs. Cell-free supernatants of B. amyloliquefaciens B-1895 and B. subtilis KATMIRA1933 showed higher antimicrobial activity against biofilms of P. mirabilis isolated from humans as compared with biofilms of sheep isolates. According to our knowledge, this is the first report evaluating the anti-biofilm activity of probiotic spore-forming bacilli against clinical and animal UTI isolates of P. mirabilis. Further studies are recommended to investigate the anti-biofilm activity and the mode of action for the antimicrobial substances produced by these bacilli, subtilosin and subtilin.
Collapse
Affiliation(s)
- Ammar Algburi
- Department of Biotechnology, Science College, University of Diyala, Baqubah, Iraq.
| | - Sarah A Alazzawi
- Department of Microbiology, Veterinary College, University of Diyala, Baqubah, Iraq
| | - Ali Ibrahim Ali Al-Ezzy
- Department of Pathology, College of Veterinary Medicine, University of Diyala, Baqubah, Iraq
| | - Richard Weeks
- Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, Rutgers State University, New Brunswick, NJ, USA
| | | | - Michael L Chikindas
- Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, Rutgers State University, New Brunswick, NJ, USA.,Don State Technical University, Rostov-on-Don, Russia
| |
Collapse
|
237
|
Goel N, Fatima SW, Kumar S, Sinha R, Khare SK. Antimicrobial resistance in biofilms: Exploring marine actinobacteria as a potential source of antibiotics and biofilm inhibitors. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2021; 30:e00613. [PMID: 33996521 PMCID: PMC8105627 DOI: 10.1016/j.btre.2021.e00613] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 02/19/2021] [Accepted: 03/21/2021] [Indexed: 12/12/2022]
Abstract
Antimicrobial resistance (AMR) is one of the serious global public health threats that require immediate action. With the emergence of new resistance mechanisms in infection-causing microorganisms such as bacteria, fungi, and viruses, AMR threatens the effective prevention and treatment of diseases caused by them. This has resulted in prolonged illness, disability, and death. It has been predicted that AMR will lead to over ten million deaths by 2050. The rapid spread of multidrug-resistant bacteria is also causing old antibiotics to become ineffective. Among the diverse factors contributing to AMR, intrinsic biofilm development has been highlighted as an essential contributing facet. Moreover, biofilm-derived antibiotic tolerance leads to serious recurrent chronic infections. Therefore, the discovery of novel bioactive molecules is a potential solution that can help combat AMR. To achieve this, sustained mining of novel antimicrobial leads from actinobacteria, particularly marine actinobacteria, can be a promising strategy. Given their vast diversity and different habitats, the extraordinary capacity of actinobacteria can be tapped to synthesize new antibiotics or bioactive molecules for biofilm inhibition. Advanced screening strategies and novel approaches in the field of modern biochemical and molecular biology can be used to detect such new compounds. In view of this, the present review focuses on understanding some of the recent strategies to inhibit biofilm formation and explores the potential role of marine actinobacteria as sources of novel antibiotics and biofilm inhibitor molecules.
Collapse
Affiliation(s)
- Nikky Goel
- Department of Chemistry, Indian Institute of Technology Delhi, India
| | | | - Sumit Kumar
- Department of Chemistry, Indian Institute of Technology Delhi, India
| | | | - Sunil K. Khare
- Department of Chemistry, Indian Institute of Technology Delhi, India
| |
Collapse
|
238
|
Nordgaard M, Mortensen RMR, Kirk NK, Gallegos‐Monterrosa R, Kovács ÁT. Deletion of Rap-Phr systems in Bacillus subtilis influences in vitro biofilm formation and plant root colonization. Microbiologyopen 2021; 10:e1212. [PMID: 34180604 PMCID: PMC8236291 DOI: 10.1002/mbo3.1212] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/27/2021] [Accepted: 06/01/2021] [Indexed: 12/19/2022] Open
Abstract
Natural isolates of the soil-dwelling bacterium Bacillus subtilis form robust biofilms under laboratory conditions and colonize plant roots. B. subtilis biofilm gene expression displays phenotypic heterogeneity that is influenced by a family of Rap-Phr regulatory systems. Most Rap-Phr systems in B. subtilis have been studied independently, in different genetic backgrounds and under distinct conditions, hampering true comparison of the Rap-Phr systems' impact on bacterial cell differentiation. Here, we investigated each of the 12 Rap-Phr systems of B.subtilis NCIB 3610 for their effect on biofilm formation. By studying single ∆rap-phr mutants, we show that despite redundancy between the cell-cell communication systems, deletion of each of the 12 Rap-Phr systems influences matrix gene expression. These Rap-Phr systems therefore enable fine-tuning of the timing and level of matrix production in response to specific conditions. Furthermore, some of the ∆rap-phr mutants demonstrated altered biofilm formation in vitro and colonization of Arabidopsis thaliana roots, but not necessarily similarly in both processes, indicating that the pathways regulating matrix gene expression and other factors important for biofilm formation may be differently regulated under these distinct conditions.
Collapse
Affiliation(s)
- Mathilde Nordgaard
- Bacterial Interactions and Evolution GroupDTU BioengineeringTechnical University of DenmarkLyngbyDenmark
| | | | - Nikolaj Kaae Kirk
- Bacterial Interactions and Evolution GroupDTU BioengineeringTechnical University of DenmarkLyngbyDenmark
| | | | - Ákos T. Kovács
- Bacterial Interactions and Evolution GroupDTU BioengineeringTechnical University of DenmarkLyngbyDenmark
| |
Collapse
|
239
|
Liu S, Ulugun B, DeFlorio W, Arcot Y, Yegin Y, Salazar KS, Castillo A, Taylor TM, Cisneros-Zevallos L, Akbulut M. Development of durable and superhydrophobic nanodiamond coating on aluminum surfaces for improved hygiene of food contact surfaces. J FOOD ENG 2021. [DOI: 10.1016/j.jfoodeng.2021.110487] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
240
|
Blancas B, Lanzagorta MDL, Jiménez‐Garcia LF, Lara R, Molinari JL, Fernández AM. Study of the ultrastructure of Enterococcus faecalis and Streptococcus mutans incubated with salivary antimicrobial peptides. Clin Exp Dent Res 2021; 7:365-375. [PMID: 33951334 PMCID: PMC8204031 DOI: 10.1002/cre2.430] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/02/2021] [Accepted: 03/23/2021] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVES Enterococcus faecalis has been associated with root canal infections, while Streptococcus mutans has a central role in the etiology of dental caries. One of the main reasons of endodontic failure has been associated to the presence of E. faecalis and the formation of biofilms. S. mutans inhabits the oral cavity, specifically the dental plaque, which is a multispecies biofilm formed on the hard surfaces of the tooth. The biofilm formation is the main factor determining the pathogenicity of numerous bacteria. Natural antimicrobial peptides in the saliva protect against pathogenic bacteria and biofilms. The aim of this study was to assess the ultrastructural damage induced by salivary peptides in bacteria involved in biofilms has not been previously studied. MATERIAL AND METHODS Enterococcus faecalis and S. mutans incubated with cystatin C, chromogranin A, or histatin 5 were morphologically analyzed and counted. The ultrastructural damage was evaluated by transmission electron microscopy (TEM). RESULTS A decrease in bacterial numbers was observed after incubation with cystatin C, chromogranin A, or histatin 5, compared to the control group (P < 0.001). Ultrastructural damage in E. faecalis and S. mutans incubated with salivary peptides was found in the cell wall, plasma membrane with a decreased distance between the bilayers, a granular pattern in the cytoplasm, and pyknotic nucleoids. CONCLUSIONS This study demonstrated that salivary peptides exert antibacterial activity and induce morphological damage on E. faecalis and S. mutans. Knowledge on the ultrastructural damage inflicted by salivary antimicrobial peptides on two important bacteria causing dental caries and root canal infections could aid the design of new therapeutic approaches to facilitate the elimination of these bacteria.
Collapse
Affiliation(s)
- Blanca Blancas
- Departamento de Microbiología y Parasitología, Facultad de MedicinaCol. Universidad Nacional Autónoma de MéxicoMexico CityMexico
| | | | - Luis Felipe Jiménez‐Garcia
- Departamento de Biología Celular, Facultad de CienciasUNAM, Col. Universidad Nacional Autónoma de MéxicoMexico CityMexico
| | - Reyna Lara
- Departamento de Biología Celular, Facultad de CienciasUNAM, Col. Universidad Nacional Autónoma de MéxicoMexico CityMexico
| | - José Luis Molinari
- Departamento de Bioquímica y Biología EstructuralInstituto de Fisiología Celular, Col. Universidad Nacional Autónoma de MéxicoMexico CityMexico
| | - Ana María Fernández
- Departamento de Microbiología y Parasitología, Facultad de MedicinaCol. Universidad Nacional Autónoma de MéxicoMexico CityMexico
- Instituto de Estudios Avanzados en Odontologia Dr. Yury Kuttler, Maestria en EndodonciaMexico CityMexico
- Centro de Investigación en Ciencias de la Salud (CICSA), FCSUniversidad Anáhuac México Campus NorteHuixquilucanMexico
| |
Collapse
|
241
|
Duration of amoxicillin-clavulanate for protracted bacterial bronchitis in children (DACS): a multi-centre, double blind, randomised controlled trial. THE LANCET RESPIRATORY MEDICINE 2021; 9:1121-1129. [PMID: 34048716 DOI: 10.1016/s2213-2600(21)00104-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/04/2021] [Accepted: 02/17/2021] [Indexed: 01/26/2023]
Abstract
BACKGROUND Protracted bacterial bronchitis (PBB) is a leading cause of chronic wet cough in children. The current standard treatment in European and American guidelines is 2 weeks of antibiotics, but the optimal duration of therapy is unknown. We describe the first randomised controlled trial to assess the duration of antibiotic treatment in children with chronic wet cough and suspected PBB. We hypothesise that 4 weeks of amoxicillin-clavulanate is superior to 2 weeks for improving clinical outcomes. METHODS Our parallel, double-blind, placebo-controlled, randomised controlled trial was completed in four Australian hospitals. Children aged 2 months to 19 years with chronic (>4 weeks duration) wet cough, and suspected PBB were randomly assigned (1:1) using permuted block randomisation (stratified by age and site) to 4 weeks of amoxicillin-clavulanate (25-35 mg/kg twice daily oral suspension; 4-week group) or 2 weeks of amoxicillin-clavulanate followed by 2 weeks of placebo (2-week group). The children, caregivers, all the study coordinators, and investigators were masked to treatment assignment until data analysis was completed. The primary outcome was clinical cure (cough resolution) by day 28. Secondary outcomes were recurrence of PBB at 6 months, time to next exacerbation, change in Parent-proxy Cough-Specific Quality-of-Life (PC-QoL) score from baseline to day 28 and from day 28 to 7 months, adverse events, nasal swab bacteriology, and antimicrobial resistance. Analyses followed the intention-to-treat principle. This trial is complete and registered with Australian/New Zealand Registry, ACTRN12616001725459. FINDINGS Between March 8, 2017, and Sept 30, 2019, 106 children were randomly assigned (52 in the 4-week group, median age 2·2 years [IQR 1·3-4·1]; 54 in the 2-week group, median age 1·7 years [1·2-3·8]) with 90 children completing the 4-week treatment. By day 28, the primary endpoint of clinical cure in the 4-week group (32 [62%] of 52 patients) was not significantly different to the 2-week group (38 [70%] of 54 patients; adjusted relative risk 0·87 [95% CI 0·60 to 1·28]; p=0·49). Time to next wet cough exacerbation was significantly longer in the 4-week group than the 2-week group (median 150 days [IQR 38-181] vs 36 days [15-181]; adjusted hazard ratio 0·47 [0·25 to 0·90]; p=0·02). The rate of recurrence of PBB at 6 months was 17 (53%) of 32 patients in the 4-week group vs 28 (74%) of 38 patients in the 2-week group, but the difference between the groups was not significant (adjusted odds ratio 0·39 [0·14 to 1·04]; p=0·07). PC-QoL significantly improved from baseline to day 28 in both groups, but there was no significant difference between them (mean difference in change -0·2 [95% CI -1·0 to 0·6]; p=0·64). From day 28 to 7 months, median PC-QoL remained stable in both groups with no difference in change between them. Data on respiratory pathogens and antimicrobial resistance (paired swabs available for 48 children) were similar between groups. Adverse events occurred in 13 (25%) children in the 2-week group and ten (19%) in the 4-week group (p=0·57). INTERPRETATION A 4-week course of amoxicillin-clavulanate for treating children with chronic wet cough and suspected PBB confers little advantage compared with a 2-week course in achieving clinical cure by 28 days. However, as a 4-week duration led to a longer cough-free period, identifying children who would benefit from a longer antibiotic course is a priority. FUNDING Queensland Children's Hospital Foundation.
Collapse
|
242
|
Jayathilaka EHTT, Rajapaksha DC, Nikapitiya C, De Zoysa M, Whang I. Antimicrobial and Anti-Biofilm Peptide Octominin for Controlling Multidrug-Resistant Acinetobacter baumannii. Int J Mol Sci 2021; 22:ijms22105353. [PMID: 34069596 PMCID: PMC8161146 DOI: 10.3390/ijms22105353] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/12/2021] [Accepted: 05/14/2021] [Indexed: 12/13/2022] Open
Abstract
Acinetobacter baumannii is a serious nosocomial pathogen with multiple drug resistance (MDR), the control of which has become challenging due to the currently used antibiotics. Our main objective in this study is to determine the antibacterial and antibiofilm activities of the antimicrobial peptide, Octominin, against MDR A. baumannii and derive its possible modes of actions. Octominin showed significant bactericidal effects at a low minimum inhibitory concentration (MIC) and the minimum bactericidal concentration (MBC) of 5 and 10 µg/mL, respectively. Time-kill kinetic analysis and bacterial viability tests revealed that Octominin showed a concentration-dependent antibacterial activity. Field-emission scanning electron microscopy (FE-SEM) analysis revealed that Octominin treatment altered the morphology and membrane structure of A. baumannii. Propidium iodide (PI) and reactive oxygen species (ROS) generation assays showed that Octominin increased the membrane permeability and ROS generation in A. baumannii, thereby causing bacterial cell death. Further, a lipopolysaccharides (LPS) binding assay showed an Octominin concentration-dependent LPS neutralization ability. Biofilm formation inhibition and eradication assays further revealed that Octominin inhibited biofilm formation and showed a high biofilm eradication activity against A. baumannii. Furthermore, up to a concentration of 100 µg/mL, Octominin caused no hemolysis and cell viability changes in mammalian cells. An in vivo study in zebrafish showed that the Octominin-treated group had a significantly higher relative percentage survival (54.1%) than the untreated group (16.6%). Additionally, a reduced bacterial load and fewer alterations in histological analysis confirmed the successful control of A. baumannii by Octominin in vivo. Collectively, these data suggest that Octominin exhibits significant antibacterial and antibiofilm activities against the multidrug-resistant A. baumannii, and this AMP can be developed further as a potent AMP for the control of antibiotic resistance.
Collapse
Affiliation(s)
- E. H. T. Thulshan Jayathilaka
- College of Veterinary Medicine, Chungnam National University, Yuseong-gu, Daejeon 34134, Korea; (E.H.T.T.J.); (D.C.R.); (C.N.)
| | - Dinusha C. Rajapaksha
- College of Veterinary Medicine, Chungnam National University, Yuseong-gu, Daejeon 34134, Korea; (E.H.T.T.J.); (D.C.R.); (C.N.)
| | - Chamilani Nikapitiya
- College of Veterinary Medicine, Chungnam National University, Yuseong-gu, Daejeon 34134, Korea; (E.H.T.T.J.); (D.C.R.); (C.N.)
| | - Mahanama De Zoysa
- College of Veterinary Medicine, Chungnam National University, Yuseong-gu, Daejeon 34134, Korea; (E.H.T.T.J.); (D.C.R.); (C.N.)
- Correspondence: (M.D.Z.); (I.W.)
| | - Ilson Whang
- National Marine Biodiversity Institute of Korea (MABIK), 75, Jangsan-ro 101 beon-gil, Janghang-eup, Seochun-gun, Chungchungnam-do 33662, Korea
- Correspondence: (M.D.Z.); (I.W.)
| |
Collapse
|
243
|
Escobar IE, Possamai Rossatto FC, Kim SM, Kang MH, Kim W, Mylonakis E. Repurposing Kinase Inhibitor Bay 11-7085 to Combat Staphylococcus aureus and Candida albicans Biofilms. Front Pharmacol 2021; 12:675300. [PMID: 34025434 PMCID: PMC8133364 DOI: 10.3389/fphar.2021.675300] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/19/2021] [Indexed: 12/03/2022] Open
Abstract
Staphylococcus aureus and Candida spp. are commonly linked with topical biofilm-associated infections such as those found on chronic wounds. These biofilms are notoriously difficult to treat, highlighting the grave need to discover and study new broad-spectrum agents to combat associated infections. Here we report that the kinase inhibitor Bay 11-7085 exhibited bactericidal activity against multidrug-resistant S. aureus with a minimum inhibitory concentration (MIC) of 4 μg/ml. In addition, S. aureus strain MW2 did not acquire resistance to antibiotic pressure. Furthermore, Bay 11-7085 exhibited potency against Candida albicans and the emerging pathogen Candida auris with a MIC of 0.5–1 μg/ml. Bay 11-7085 partially inhibited and eradicated biofilm formation of various pathogens, such as VRSA (vancomycin-resistant S. aureus), as well as antifungal-resistant Candida spp. isolates. Notably, Bay 11-7085 partially inhibited initial cell attachment and formation of a VRSA-C. albicans polymicrobial biofilm in vitro. In contrast to C. albicans, inhibition of VRSA biofilm was linked to initial cell attachment independent of its bactericidal activity. Finally, Bay 11-7085 was effective in vivo at increasing the lifespan of C. elegans during an S. aureus and a C. albicans infection. Our work proposes kinase inhibitor Bay 11-7085 as a potential compound capable of combating biofilms associated with primary multidrug-resistant bacteria and yeast pathogens associated with wound infections.
Collapse
Affiliation(s)
- Iliana E Escobar
- Infectious Diseases Division, Department of Medicine, Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Fernanda Cristina Possamai Rossatto
- Infectious Diseases Division, Department of Medicine, Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States.,Laboratory of Biofilms and Alternative Models, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | - Soo Min Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Min Hee Kang
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Wooseong Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Eleftherios Mylonakis
- Infectious Diseases Division, Department of Medicine, Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| |
Collapse
|
244
|
Qiu L, Wu J, Du W, Nafees M, Yin Y, Ji R, Banwart SA, Guo H. Response of soil bacterial communities to sulfadiazine present in manure: Protection and adaptation mechanisms of extracellular polymeric substances. JOURNAL OF HAZARDOUS MATERIALS 2021; 408:124887. [PMID: 33387717 DOI: 10.1016/j.jhazmat.2020.124887] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 06/12/2023]
Abstract
Extracellular polymeric substances (EPS) play a dominant role in protective biofilms. However, studies exploring the underlying protective mechanism of EPS have mainly focused on activated sludge, whereas their positive roles in protecting soil microbes from environmental stress have not been elucidated. In this study, we revealed the response of soil bacterial communities to various dosages of sulfadiazine (SDZ) present in manure, with a special emphasis on the role of EPS. Sequencing analysis showed that the bacterial community demonstrated stronger symbiotic relationships and weaker competitive interaction patterns to cope with disturbance induced by SDZ. EPS was mainly composed of tyrosine-like and tryptophan-like substances, and moreover, carboxyl, hydroxyl and ether groups were the main functional groups. An adaptation mechanism, namely the enhanced secretion of tryptophan-like substances, could help alleviate the SDZ stress effectively in the biofilms occurring in soil that experienced long-term manure application. Furthermore, the existence of EPS weakened the accumulation of antibiotic resistance genes (ARGs) in soil. Our results for the first time systematically uncover the joint action of biofilm tolerance and ARGs in resisting SDZ stress, which enhances understanding of the protective role of EPS and the underlying mechanisms governing biofilm functions in soil environments.
Collapse
Affiliation(s)
- Linlin Qiu
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Jingjing Wu
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Wenchao Du
- School of Environment, Nanjing Normal University, Nanjing 210023, China
| | - Muhammad Nafees
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Ying Yin
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Rong Ji
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Steven A Banwart
- School of Earth and Environment, University of Leeds, Leeds LS2 9JT, UK; Global Food and Environment Institute, University of Leeds, Leeds LS2 9JT, UK
| | - Hongyan Guo
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu 210023, China.
| |
Collapse
|
245
|
Bhandari S, Khadayat K, Poudel S, Shrestha S, Shrestha R, Devkota P, Khanal S, Marasini BP. Phytochemical analysis of medicinal plants of Nepal and their antibacterial and antibiofilm activities against uropathogenic Escherichia coli. BMC Complement Med Ther 2021; 21:116. [PMID: 33836728 PMCID: PMC8033659 DOI: 10.1186/s12906-021-03293-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 03/30/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND A biofilm is an extracellular polymeric substance (EPS) composed of polysaccharides, proteins, nucleic acids, and lipids that impede antibiotics and immune cells, thus providing a shielded environment for bacterial growth. Due to biofilm formation, some microbes can show up to 1000 fold increased resistance towards the antibiotics than the normal planktonic forms. The study was conducted to screen the crude extracts of medicinal plants used in Nepal for their in vitro antibiofilm activities. METHODS Total phenolic and total flavonoid contents were determined by using a Folin-Ciocalteau reagent and aluminium trichloride method, respectively. Resazurin assay was used to determine the minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC). The initial antibiofilm activities and their inhibitory concentration (IC50) values were determined by the microtiter based modified crystal violet staining method. RESULTS Out of 25 different plant extracts were used for the study, methanolic extracts of 20 plants showed a biofilm inhibition activity against five different strong biofilm producing Escherichia coli strains. Calotropis gigantea exhibited inhibition against all five different E. coli strains with IC50 values ranging from 299.7 ± 20.5 to 427.4 ± 2.7 μg/mL. Apart from that, Eclipta prostrata also showed biofilm formation inhibition, followed by Eupatorium adenophorum, Moringa oleifera, Ocimum tenuifolium, Oxalis lantifolia, Prunus persica, and Urtica parviflora. The extracts of C. gigantea, E. prostrata, Mangifera indica, O. tenuifolium, P. persica, and U. parviflora exhibited a moderate to poor MIC value ranging from 625 to 2500 μg/mL. The highest amount of phenolic content (TPC) was found in Acacia catechu followed by Morus alba, which was 38.9 and 25.1 mg gallic acid equivalents, respectively. The highest amount of flavonoid content was found in A. catechu followed by M. indica, which was 27.1 and 20.8 mg quercetin equivalents, respectively. CONCLUSION Extracts of C. gigantea, E. prostrata, P. persica, U. parviflora, and O. tenuifolium showed antibacterial as well as antibiofilm activity against pathogenic and strong biofilm producing E. coli. Thus, extracts or the pure compound from these medicinal plants could be used as antibiotics in the future.
Collapse
Affiliation(s)
- Sudip Bhandari
- Department of Biotechnology, National College, Tribhuvan University, Naya Bazar, Kathmandu, Nepal
| | - Karan Khadayat
- Department of Biotechnology, National College, Tribhuvan University, Naya Bazar, Kathmandu, Nepal
| | - Sami Poudel
- Department of Biotechnology, National College, Tribhuvan University, Naya Bazar, Kathmandu, Nepal
| | - Sunil Shrestha
- Department of Biotechnology, National College, Tribhuvan University, Naya Bazar, Kathmandu, Nepal
| | - Raju Shrestha
- Department of Microbiology, National College, Tribhuvan University, Naya Bazar, Kathmandu, Nepal
| | - Poonam Devkota
- Department of Biotechnology, National College, Tribhuvan University, Naya Bazar, Kathmandu, Nepal
| | - Santosh Khanal
- Department of Biotechnology, National College, Tribhuvan University, Naya Bazar, Kathmandu, Nepal
| | - Bishnu P Marasini
- Department of Biotechnology, National College, Tribhuvan University, Naya Bazar, Kathmandu, Nepal.
| |
Collapse
|
246
|
Senobar Tahaei SA, Stájer A, Barrak I, Ostorházi E, Szabó D, Gajdács M. Correlation Between Biofilm-Formation and the Antibiotic Resistant Phenotype in Staphylococcus aureus Isolates: A Laboratory-Based Study in Hungary and a Review of the Literature. Infect Drug Resist 2021. [PMID: 33790586 DOI: 10.2147/idrs303992] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
INTRODUCTION Staphylococcus aureus (S. aureus) is an important causative pathogen in human infections. The production of biofilms by bacteria is an important factor, leading to treatment failures. There has been significant interest in assessing the possible relationship between the multidrug-resistant (MDR) status and the biofilm-producer phenotype in bacteria. The aim of our present study was to assess the biofilm-production rates in clinical methicillin-susceptible S. aureus [MSSA] and methicillin-resistant S. aureus [MRSA] isolates from Hungarian hospitals and the correlation between resistance characteristics and their biofilm-forming capacity. METHODS A total of three hundred (n=300) S. aureus isolates (corresponding to MSSA and MRSA isolates in equal measure) were included in this study. Identification of the isolates was carried out using the VITEK 2 ID/AST system and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS). Antimicrobial susceptibility testing was performed using the Kirby-Bauer disk diffusion method and E-tests, confirmation of MRSA status was carried out using PBP2a agglutination assay. Biofilm-production was assessed using the crystal violet (CV) tube-adherence method and the Congo red agar (CRA) plate method. RESULTS There were significant differences among MSSA and MRSA isolates regarding susceptibility-levels to commonly used antibiotics (in case of erythromycin, clindamycin and ciprofloxacin: p<0.001, gentamicin: p=0.023, sulfamethoxazole/trimethoprim: p=0.027, rifampin: p=0.037). In the CV tube adherence-assay, 37% (n=56) of MSSA and 39% (n=58) of MRSA isolates were positive for biofilm-production, while during the use of CRA plates, 41% (n=61) of MSSA and 44% (n=66) of MRSA were positive; no associations were found between methicillin-resistance and biofilm-production. On the other hand, erythromycin, clindamycin and rifampin resistance was associated with biofilm-positivity (p=0.004, p<0.001 and p<0.001, respectively). Biofilm-positive isolates were most common from catheter-associated infections. DISCUSSION Our study emphasizes the need for additional experiments to assess the role biofilms have in the pathogenesis of implant-associated and chronic S. aureus infections.
Collapse
Affiliation(s)
- Seyyed Askhan Senobar Tahaei
- Department of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Szeged, 6720, Hungary
| | - Anette Stájer
- Department of Periodontology, Faculty of Dentistry, University of Szeged, Szeged, 6720, Hungary
| | - Ibrahim Barrak
- Department of Periodontology, Faculty of Dentistry, University of Szeged, Szeged, 6720, Hungary
| | - Eszter Ostorházi
- Institute of Medical Microbiology, Faculty of Medicine, Semmelweis University, Budapest, 1089, Hungary
| | - Dóra Szabó
- Institute of Medical Microbiology, Faculty of Medicine, Semmelweis University, Budapest, 1089, Hungary
| | - Márió Gajdács
- Department of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Szeged, 6720, Hungary.,Institute of Medical Microbiology, Faculty of Medicine, Semmelweis University, Budapest, 1089, Hungary
| |
Collapse
|
247
|
Pervin Z, Hassan MM. Synergistic therapeutic actions of antimicrobial peptides to treat multidrug-resistant bacterial infection. REVIEWS IN MEDICAL MICROBIOLOGY 2021; 32:83-89. [DOI: 10.1097/mrm.0000000000000239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
248
|
Identification of potential therapeutic antimicrobial peptides against Acinetobacter baumannii in a mouse model of pneumonia. Sci Rep 2021; 11:7318. [PMID: 33795739 PMCID: PMC8016998 DOI: 10.1038/s41598-021-86844-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 03/18/2021] [Indexed: 12/25/2022] Open
Abstract
Acinetobacter baumannii-induced nosocomial pneumonia has become a serious clinical problem because of high antibiotic resistance rates. Antimicrobial peptides (AMP) are an ideal alternative strategy due to their broad-spectrum of antimicrobial activity and low incidence of bacterial resistance. However, their application is limited by toxicity and stability in vivo. The present study used a mouse model to directly identify potential AMPs effective for treatment of A. baumannii-induced pneumonia. Fifty-eight AMPs were screened and two identified (SMAP-29 and TP4) to have prophylactic effects which prevented the death of mice with pneumonia. Furthermore, two TP4 derivatives (dN4 and dC4) were found to have therapeutic activity in pneumonia mouse models by peritoneal or intravenous administration. Both dN4 and dC4 also inhibited and/or eliminated A. baumannii biofilms at higher doses. Taken together, these data suggest the AMP derivatives dN4 and dC4 represent a potential treatment strategy for A. baumannii-induced pneumonia.
Collapse
|
249
|
Green KD, Punetha A, Chandrika NT, Hou C, Garneau-Tsodikova S, Tsodikov OV. Development of Single-Stranded DNA Bisintercalating Inhibitors of Primase DnaG as Antibiotics. ChemMedChem 2021; 16:1986-1995. [PMID: 33711198 DOI: 10.1002/cmdc.202100001] [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: 01/01/2021] [Revised: 03/02/2021] [Indexed: 11/07/2022]
Abstract
Many essential enzymes in bacteria remain promising potential targets of antibacterial agents. In this study, we discovered that dequalinium, a topical antibacterial agent, is an inhibitor of Staphylococcus aureus primase DnaG (SaDnaG) with low-micromolar minimum inhibitory concentrations against several S. aureus strains, including methicillin-resistant bacteria. Mechanistic studies of dequalinium and a series of nine of its synthesized analogues revealed that these compounds are single-stranded DNA bisintercalators that penetrate a bacterium by compromising its membrane. The best compound of this series likely interacts with DnaG directly, inhibits both staphylococcal cell growth and biofilm formation, and displays no significant hemolytic activity or toxicity to mammalian cells. This compound is an excellent lead for further development of a novel anti-staphylococcal therapeutic.
Collapse
Affiliation(s)
- Keith D Green
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536-0596, USA
| | - Ankita Punetha
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536-0596, USA
| | | | - Caixia Hou
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536-0596, USA
| | | | - Oleg V Tsodikov
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536-0596, USA
| |
Collapse
|
250
|
Senobar Tahaei SA, Stájer A, Barrak I, Ostorházi E, Szabó D, Gajdács M. Correlation Between Biofilm-Formation and the Antibiotic Resistant Phenotype in Staphylococcus aureus Isolates: A Laboratory-Based Study in Hungary and a Review of the Literature. Infect Drug Resist 2021; 14:1155-1168. [PMID: 33790586 PMCID: PMC8001189 DOI: 10.2147/idr.s303992] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/11/2021] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Staphylococcus aureus (S. aureus) is an important causative pathogen in human infections. The production of biofilms by bacteria is an important factor, leading to treatment failures. There has been significant interest in assessing the possible relationship between the multidrug-resistant (MDR) status and the biofilm-producer phenotype in bacteria. The aim of our present study was to assess the biofilm-production rates in clinical methicillin-susceptible S. aureus [MSSA] and methicillin-resistant S. aureus [MRSA] isolates from Hungarian hospitals and the correlation between resistance characteristics and their biofilm-forming capacity. METHODS A total of three hundred (n=300) S. aureus isolates (corresponding to MSSA and MRSA isolates in equal measure) were included in this study. Identification of the isolates was carried out using the VITEK 2 ID/AST system and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS). Antimicrobial susceptibility testing was performed using the Kirby-Bauer disk diffusion method and E-tests, confirmation of MRSA status was carried out using PBP2a agglutination assay. Biofilm-production was assessed using the crystal violet (CV) tube-adherence method and the Congo red agar (CRA) plate method. RESULTS There were significant differences among MSSA and MRSA isolates regarding susceptibility-levels to commonly used antibiotics (in case of erythromycin, clindamycin and ciprofloxacin: p<0.001, gentamicin: p=0.023, sulfamethoxazole/trimethoprim: p=0.027, rifampin: p=0.037). In the CV tube adherence-assay, 37% (n=56) of MSSA and 39% (n=58) of MRSA isolates were positive for biofilm-production, while during the use of CRA plates, 41% (n=61) of MSSA and 44% (n=66) of MRSA were positive; no associations were found between methicillin-resistance and biofilm-production. On the other hand, erythromycin, clindamycin and rifampin resistance was associated with biofilm-positivity (p=0.004, p<0.001 and p<0.001, respectively). Biofilm-positive isolates were most common from catheter-associated infections. DISCUSSION Our study emphasizes the need for additional experiments to assess the role biofilms have in the pathogenesis of implant-associated and chronic S. aureus infections.
Collapse
Affiliation(s)
- Seyyed Askhan Senobar Tahaei
- Department of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Szeged, 6720, Hungary
| | - Anette Stájer
- Department of Periodontology, Faculty of Dentistry, University of Szeged, Szeged, 6720, Hungary
| | - Ibrahim Barrak
- Department of Periodontology, Faculty of Dentistry, University of Szeged, Szeged, 6720, Hungary
| | - Eszter Ostorházi
- Institute of Medical Microbiology, Faculty of Medicine, Semmelweis University, Budapest, 1089, Hungary
| | - Dóra Szabó
- Institute of Medical Microbiology, Faculty of Medicine, Semmelweis University, Budapest, 1089, Hungary
| | - Márió Gajdács
- Department of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Szeged, 6720, Hungary
- Institute of Medical Microbiology, Faculty of Medicine, Semmelweis University, Budapest, 1089, Hungary
| |
Collapse
|