1
|
Rajkhowa S, Hussain SZ, Agarwal M, Zaheen A, Al-Hussain SA, Zaki MEA. Advancing Antibiotic-Resistant Microbe Combat: Nanocarrier-Based Systems in Combination Therapy Targeting Quorum Sensing. Pharmaceutics 2024; 16:1160. [PMID: 39339197 PMCID: PMC11434747 DOI: 10.3390/pharmaceutics16091160] [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: 07/18/2024] [Revised: 08/17/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
The increase in antibiotic-resistant bacteria presents a significant risk to worldwide public health, emphasizing the necessity of novel approaches to address infections. Quorum sensing, an essential method of communication among bacteria, controls activities like the formation of biofilms, the production of virulence factors, and the synthesis of secondary metabolites according to the number of individuals in the population. Quorum quenching, which interferes with these processes, emerges as a vital approach to diminish bacterial virulence and prevent biofilm formation. Nanocarriers, characterized by their small size, high surface-area-to-volume ratio, and modifiable surface chemistry, offer a versatile platform for the disruption of bacterial communication by targeting various stages within the quorum sensing pathway. These features allow nanocarriers to infiltrate biofilms, disrupt cell membranes, and inhibit bacterial proliferation, presenting a promising alternative to traditional antibiotics. Integrating nanocarrier-based systems into combination therapies provides a multi-pronged approach to infection control, enhancing both the efficacy and specificity of treatment regimens. Nonetheless, challenges related to the stability, safety, and clinical effectiveness of nanomaterial-based antimicrobial treatments remain. Continued research and development are essential to overcoming these obstacles and fully harnessing the potential of nano-antimicrobial therapies. This review emphasizes the importance of quorum sensing in bacterial behavior and highlights the transformative potential of nanotechnology in advancing antimicrobial treatments, offering innovative solutions to combat antibiotic-resistant pathogens.
Collapse
Affiliation(s)
- Sanchaita Rajkhowa
- Centre for Biotechnology and Bioinformatics, Dibrugarh University, Dibrugarh 786004, Assam, India; (S.Z.H.); (M.A.); (A.Z.)
| | - Safrina Zeenat Hussain
- Centre for Biotechnology and Bioinformatics, Dibrugarh University, Dibrugarh 786004, Assam, India; (S.Z.H.); (M.A.); (A.Z.)
| | - Manisha Agarwal
- Centre for Biotechnology and Bioinformatics, Dibrugarh University, Dibrugarh 786004, Assam, India; (S.Z.H.); (M.A.); (A.Z.)
| | - Alaiha Zaheen
- Centre for Biotechnology and Bioinformatics, Dibrugarh University, Dibrugarh 786004, Assam, India; (S.Z.H.); (M.A.); (A.Z.)
| | - Sami A. Al-Hussain
- Department of Chemistry, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia;
| | - Magdi E. A. Zaki
- Department of Chemistry, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia;
| |
Collapse
|
2
|
Everard ML, Priftis K, Koumbourlis AC, Shields MD. Time to re-set our thinking about airways disease: lessons from history, the resurgence of chronic bronchitis / PBB and modern concepts in microbiology. Front Pediatr 2024; 12:1391290. [PMID: 38910961 PMCID: PMC11190372 DOI: 10.3389/fped.2024.1391290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 05/06/2024] [Indexed: 06/25/2024] Open
Abstract
In contrast to significant declines in deaths due to lung cancer and cardiac disease in Westernised countries, the mortality due to 'chronic obstructive pulmonary disease' (COPD) has minimally changed in recent decades while 'the incidence of bronchiectasis' is on the rise. The current focus on producing guidelines for these two airway 'diseases' has hindered progress in both treatment and prevention. The elephant in the room is that neither COPD nor bronchiectasis is a disease but rather a consequence of progressive untreated airway inflammation. To make this case, it is important to review the evolution of our understanding of airway disease and how a pathological appearance (bronchiectasis) and an arbitrary physiological marker of impaired airways (COPD) came to be labelled as 'diseases'. Valuable insights into the natural history of airway disease can be obtained from the pre-antibiotic era. The dramatic impacts of antibiotics on the prevalence of significant airway disease, especially in childhood and early adult life, have largely been forgotten and will be revisited as will the misinterpretation of trials undertaken in those with chronic (bacterial) bronchitis. In the past decades, paediatricians have observed a progressive increase in what is termed 'persistent bacterial bronchitis' (PBB). This condition shares all the same characteristics as 'chronic bronchitis', which is prevalent in young children during the pre-antibiotic era. Additionally, the radiological appearance of bronchiectasis is once again becoming more common in children and, more recently, in adults. Adult physicians remain sceptical about the existence of PBB; however, in one study aimed at assessing the efficacy of antibiotics in adults with persistent symptoms, researchers discovered that the majority of patients exhibiting symptoms of PBB were already on long-term macrolides. In recent decades, there has been a growing recognition of the importance of the respiratory microbiome and an understanding of the ability of bacteria to persist in potentially hostile environments through strategies such as biofilms, intracellular communities, and persister bacteria. This is a challenging field that will likely require new approaches to diagnosis and treatment; however, it needs to be embraced if real progress is to be made.
Collapse
Affiliation(s)
- Mark L Everard
- Division of Paediatrics & Child Health, University of Western Australia, Perth, WA, Australia
| | - Kostas Priftis
- Allergology and Pulmonology Unit, 3rd Paediatric Department, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastassios C Koumbourlis
- Division of Pulmonary & Sleep Medicine, George Washington University School of Medicine & Health Sciences, Washington, DC, United States
| | - Michael D Shields
- Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
3
|
Aonofriesei F. Surfactants' Interplay with Biofilm Development in Staphylococcus and Candida. Pharmaceutics 2024; 16:657. [PMID: 38794319 PMCID: PMC11125353 DOI: 10.3390/pharmaceutics16050657] [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: 04/12/2024] [Revised: 05/09/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
The capacity of micro-organisms to form biofilms is a pervasive trait in the microbial realm. For pathogens, biofilm formation serves as a virulence factor facilitating successful host colonization. Simultaneously, infections stemming from biofilm-forming micro-organisms pose significant treatment challenges due to their heightened resistance to antimicrobial agents. Hence, the quest for active compounds capable of impeding microbial biofilm development stands as a pivotal pursuit in biomedical research. This study presents findings concerning the impact of three surfactants, namely, polysorbate 20 (T20), polysorbate 80 (T80), and sodium dodecyl sulfate (SDS), on the initial stage of biofilm development in both Staphylococcus aureus and Candida dubliniensis. In contrast to previous investigations, we conducted a comparative assessment of the biofilm development capacity of these two taxonomically distant groups, predicated on their shared ability to reduce TTC. The common metabolic trait shared by S. aureus and C. dubliniensis in reducing TTC to formazan facilitated a simultaneous evaluation of biofilm development under the influence of surfactants across both groups. Our results revealed that surfactants could impede the development of biofilms in both species by disrupting the initial cell attachment step. The observed effect was contingent upon the concentration and type of compound, with a higher inhibition observed in culture media supplemented with SDS. At maximum concentrations (5%), T20 and T80 significantly curtailed the formation and viability of S. aureus and C. dubliniensis biofilms. Specifically, T20 inhibited biofilm development by 75.36% in S. aureus and 71.18% in C. dubliniensis, while T80 exhibited a slightly lower inhibitory effect, with values ranging between 66.68% (C. dubliniensis) and 65.54% (S. aureus) compared to the controls. Incorporating these two non-toxic surfactants into pharmaceutical formulations could potentially enhance the inhibitory efficacy of selected antimicrobial agents, particularly in external topical applications.
Collapse
Affiliation(s)
- Florin Aonofriesei
- Department of Natural Sciences, Faculty of Natural and Agricultural Sciences, Ovidius University of Constanta, 1, University Street, 900470 Constanța, Romania
| |
Collapse
|
4
|
Iungin O, Shydlovska O, Moshynets O, Vasylenko V, Sidorenko M, Mickevičius S, Potters G. Metal-based nanoparticles: an alternative treatment for biofilm infection in hard-to-heal wounds. J Wound Care 2024; 33:xcix-cx. [PMID: 38588056 DOI: 10.12968/jowc.2024.33.sup4a.xcix] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Metal-based nanoparticles (MNPs) are promoted as effective compounds in the treatment of bacterial infections and as possible alternatives to antibiotics. These MNPs are known to affect a broad spectrum of microorganisms using a multitude of strategies, including the induction of reactive oxygen species and interaction with the inner structures of the bacterial cells. The aim of this review was to summarise the latest studies about the effect of metal-based nanoparticles on pathogenic bacterial biofilm formed in wounds, using the examples of Gram-positive bacterium Staphylococcus aureus and Gram-negative bacterium Pseudomonas aeruginosa, as well as provide an overview of possible clinical applications.
Collapse
Affiliation(s)
- Olga Iungin
- 1 Kyiv National University of Technologies and Design (KNUTD), Kyiv, Ukraine
- 2 Institute of Molecular Biology and Genetics of the National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Olga Shydlovska
- 1 Kyiv National University of Technologies and Design (KNUTD), Kyiv, Ukraine
| | - Olena Moshynets
- 2 Institute of Molecular Biology and Genetics of the National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Volodymyr Vasylenko
- 3 Vytautas Magnus University, Faculty of Natural Science, Akademija, Lithuania
| | - Marina Sidorenko
- 3 Vytautas Magnus University, Faculty of Natural Science, Akademija, Lithuania
| | - Saulius Mickevičius
- 3 Vytautas Magnus University, Faculty of Natural Science, Akademija, Lithuania
| | - Geert Potters
- 4 Antwerp Maritime Academy, Antwerp, Belgium
- 5 University of Antwerp, Antwerp, Belgium
| |
Collapse
|
5
|
Parmar D, Rosado-Rosa JM, Shrout JD, Sweedler JV. Metabolic insights from mass spectrometry imaging of biofilms: A perspective from model microorganisms. Methods 2024; 224:21-34. [PMID: 38295894 PMCID: PMC11149699 DOI: 10.1016/j.ymeth.2024.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 12/17/2023] [Accepted: 01/16/2024] [Indexed: 02/05/2024] Open
Abstract
Biofilms are dense aggregates of bacterial colonies embedded inside a self-produced polymeric matrix. Biofilms have received increasing attention in medical, industrial, and environmental settings due to their enhanced survival. Their characterization using microscopy techniques has revealed the presence of structural and cellular heterogeneity in many bacterial systems. However, these techniques provide limited chemical detail and lack information about the molecules important for bacterial communication and virulence. Mass spectrometry imaging (MSI) bridges the gap by generating spatial chemical information with unmatched chemical detail, making it an irreplaceable analytical platform in the multi-modal imaging of biofilms. In the last two decades, over 30 species of biofilm-forming bacteria have been studied using MSI in different environments. The literature conveys both analytical advancements and an improved understanding of the effects of environmental variables such as host surface characteristics, antibiotics, and other species of microorganisms on biofilms. This review summarizes the insights from frequently studied model microorganisms. We share a detailed list of organism-wide metabolites, commonly observed mass spectral adducts, culture conditions, strains of bacteria, substrate, broad problem definition, and details of the MS instrumentation, such as ionization sources and matrix, to facilitate future studies. We also compared the spatial characteristics of the secretome under different study designs to highlight changes because of various environmental influences. In addition, we highlight the current limitations of MSI in relation to biofilm characterization to enable cross-comparison between experiments. Overall, MSI has emerged to become an important approach for the spatial/chemical characterization of bacterial biofilms and its use will continue to grow as MSI becomes more accessible.
Collapse
Affiliation(s)
- Dharmeshkumar Parmar
- Department of Chemistry and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States
| | - Joenisse M Rosado-Rosa
- Department of Chemistry and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States
| | - Joshua D Shrout
- Department of Civil and Environmental Engineering and Earth Sciences, University of Notre Dame, Notre Dame, IN 46556, United States; Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Jonathan V Sweedler
- Department of Chemistry and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States.
| |
Collapse
|
6
|
Bilgin M, Dosler S, Otuk G. Antibiotic adjuvant activities of quorum sensing signal molecules DSF and BDSF against mature biofilms of Staphylococci. J Chemother 2024; 36:11-23. [PMID: 37873740 DOI: 10.1080/1120009x.2023.2270743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 10/09/2023] [Indexed: 10/25/2023]
Abstract
Among promising antibiofilm compounds, quorum-sensing (QS) molecules that regulate biological processes such as biofilm formation and intra- or interspecies communication appear to be good candidates. The invitro antibiotic-adjuvant effects of QS molecules diffusible signal factor (DSF) and B. cenocepacia producing-DSF (BDSF) were investigated against mature Staphylococcal biofilms. Broth microdilution methods were used for the determinations of MIC, MBC, MBIC, and MBEC, and bactericidal activities were determined by TKC method. The lowest MICs were obtained with ciprofloxacin and gentamicin, and MBECs with ciprofloxacin. DSF and BDSF at 0.5 µM decreased the MICs as 2-8, and 2-32 fold, respectively. In TKC studies, -cidal activities were achieved by BDSF + gentamycin, or ciprofloxacin, and DSF + daptomycin, vancomycin, meropenem or gentamycin combinations. Synergistic effects were generally obtained with BDSF + gentamicin combinations, followed by DSF + daptomycin against most S. aureus; while BDSF + gentamicin or ciprofloxacin, and DSF + vancomycin or meropenem were synergist against some S. epidermidis biofilms. Also, the antagonist effects were observed with BDSF + meropenem or ciprofloxacin against each MSSE and MSSA. It is estimated that these QS molecules, although it was strain dependent, generally enhanced the antibiotic activity, and would be a new and effective treatment strategy for biofilm control, either alone or as an antibiotic adjuvant.
Collapse
Affiliation(s)
- Merve Bilgin
- Department of Pharmaceutical Microbiology, Istanbul, Istanbul University, Institute of Graduate Studies in Health Sciences, Istanbul, Turkiye
- Department of Pharmaceutical Microbiology, Istanbul Health & Technology University, Faculty of Pharmacy, Istanbul, Turkiye
| | - Sibel Dosler
- Department of Pharmaceutical Microbiology, Istanbul, Istanbul University, Faculty of Pharmacy, Istanbul, Turkiye
| | - Gulten Otuk
- Department of Pharmaceutical Microbiology, Istanbul, Istanbul University, Faculty of Pharmacy, Istanbul, Turkiye
| |
Collapse
|
7
|
Schapira AJ, Dramé M, Olive C, Marion-Sanchez K. Bacterial viability in dry-surface biofilms in healthcare facilities: a systematic review. J Hosp Infect 2024; 144:94-110. [PMID: 38029859 DOI: 10.1016/j.jhin.2023.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Bacteria are known to live inside architectural structures called biofilms. Though standard biofilms have been studied extensively for more than 50 years, little is known about dry-surface biofilms (DSBs). Since 2012, DSBs have been described in several scientific papers, but basic knowledge about the viability and culturability of bacteria remains limited. AIM To conduct a systematic review to determine whether bacteria inside DSBs are viable, culturable, and enumerable. METHODS Eligible articles had to deal with DSBs containing at least one bacterial species involved in healthcare-associated infections, which developed in actual healthcare environments (in-situ) or with the help of any biofilm model (in-vitro). FINDINGS Twenty-four articles were included in the review. Whereas most of them isolated viable bacteria (87% in situ; 100% in vitro), no in-situ study quantified culturable bacteria in the biofilm per unit area. Conversely, 100% of in-vitro studies cultured the bacteria from controls and 94.4% supplied an enumeration of them. Culturable bacteria also grew after 78% of the cleaning, disinfection, or sterilization protocols tested. Microscopic observations after staining the samples with live/dead fluorescent probes (Baclight®) showed large amounts of viable cells in culture-negative samples. CONCLUSION Our study questions the efficacy of current methods for microbiological monitoring of surfaces, since these methods are only based on bacterial culturability. To improve both surface monitoring and cleaning and disinfection protocols, it is necessary to integrate the concept of DSBs which appears to contain a significant amount of viable but non-culturable bacteria.
Collapse
Affiliation(s)
- A-J Schapira
- CHU Martinique, Department of Bacteriology, Hygiene and Environment Laboratory, Fort-de-France Cedex, Martinique; Paris Cité University, Faculty of Health, Paris, France
| | - M Dramé
- CHU Martinique, Department of Clinical Research and Innovation, Fort-de-France Cedex, Martinique; University of the French West Indies, Faculty of Medicine, EpiCliV Research Unit, Martinique
| | - C Olive
- CHU Martinique, Department of Bacteriology, Hygiene and Environment Laboratory, Fort-de-France Cedex, Martinique
| | - K Marion-Sanchez
- CHU Martinique, Department of Bacteriology, Hygiene and Environment Laboratory, Fort-de-France Cedex, Martinique; PCCEI, University of Montpellier, University of the Antilles, INSERM, EFS, Montpellier, France.
| |
Collapse
|
8
|
Kaushik A, Kest H, Sood M, Steussy BW, Thieman C, Gupta S. Biofilm Producing Methicillin-Resistant Staphylococcus aureus (MRSA) Infections in Humans: Clinical Implications and Management. Pathogens 2024; 13:76. [PMID: 38251383 PMCID: PMC10819455 DOI: 10.3390/pathogens13010076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/10/2024] [Accepted: 01/13/2024] [Indexed: 01/23/2024] Open
Abstract
Since its initial description in the 1960s, methicillin-resistant Staphylococcus aureus (MRSA) has developed multiple mechanisms for antimicrobial resistance and evading the immune system, including biofilm production. MRSA is now a widespread pathogen, causing a spectrum of infections ranging from superficial skin issues to severe conditions like osteoarticular infections and endocarditis, leading to high morbidity and mortality. Biofilm production is a key aspect of MRSA's ability to invade, spread, and resist antimicrobial treatments. Environmental factors, such as suboptimal antibiotics, pH, temperature, and tissue oxygen levels, enhance biofilm formation. Biofilms are intricate bacterial structures with dense organisms embedded in polysaccharides, promoting their resilience. The process involves stages of attachment, expansion, maturation, and eventually disassembly or dispersion. MRSA's biofilm formation has a complex molecular foundation, involving genes like icaADBC, fnbA, fnbB, clfA, clfB, atl, agr, sarA, sarZ, sigB, sarX, psm, icaR, and srtA. Recognizing pivotal genes for biofilm formation has led to potential therapeutic strategies targeting elemental and enzymatic properties to combat MRSA biofilms. This review provides a practical approach for healthcare practitioners, addressing biofilm pathogenesis, disease spectrum, and management guidelines, including advances in treatment. Effective management involves appropriate antimicrobial therapy, surgical interventions, foreign body removal, and robust infection control practices to curtail spread within healthcare environments.
Collapse
Affiliation(s)
- Ashlesha Kaushik
- Division of Pediatric Infectious Diseases, St. Luke’s Regional Medical Center, Unity Point Health, 2720 Stone Park Blvd, Sioux City, IA 51104, USA
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Master of Science, Healthcare Quality and Safety, Harvard Medical School, Boston, MA 02115, USA
| | - Helen Kest
- Division of Pediatric Infectious Diseases, St. Joseph’s Children’s Hospital, 703 Main Street, Paterson, NJ 07503, USA;
| | - Mangla Sood
- Department of Pediatrics, Indira Gandhi Medical College, Shimla 171006, India;
| | - Bryan W. Steussy
- Division of Microbiology, St. Luke’s Regional Medical Center, Unity Point Health, 2720 Stone Park Blvd, Sioux City, IA 51104, USA;
| | - Corey Thieman
- Division of Pharmacology, St. Luke’s Regional Medical Center, Unity Point Health, 2720 Stone Park Blvd, Sioux City, IA 51104, USA;
| | - Sandeep Gupta
- Division of Pulmonary and Critical Care, St. Luke’s Regional Medical Center, Unity Point Health, 2720 Stone Park Blvd, Sioux City, IA 51104, USA;
| |
Collapse
|
9
|
Can Karanlık C, Karanlık G, Özdemir S, Tollu G, Erdoğmuş A. Synthesis and characterization of novel BODIPYs and their antioxidant, antimicrobial, photodynamic antimicrobial, antibiofilm and DNA interaction activities. Photochem Photobiol 2024; 100:101-114. [PMID: 37317040 DOI: 10.1111/php.13825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/29/2023] [Accepted: 05/29/2023] [Indexed: 06/16/2023]
Abstract
In the current study, we synthesized and characterized new BODIPY derivatives (1-4) having pyridine or thienyl-pyridine substituents at meso- position and 4-dibenzothienyl or benzo[b]thien-2-yl moieties at 2-,6- positions. We investigated fluorescence properties and the ability to form singlet oxygen. In addition, various biological activities of BODIPYs such as DPPH scavenging, DNA binding/cleavage ability, cell viability inhibition, antimicrobial activity, antimicrobial photodynamic therapy (aPDT) and biofilm inhibition properties were performed. BODIPY derivatives BDPY-3 (3) and BDPY-4 (4) have high fluorescence quantum yields as 0.50 and 0.61 and 1 O2 quantum yields were calculated as 0.83 for BDPY-1 (1), 0.12 for BDPY-2 (2), 0.11 for BDPY-3 and 0.23 for BDPY-4. BODIPY derivatives BDPY-2, BDPY-3 and BDPY-4 displayed 92.54 ± 5.41%, 94.20 ± 5.50%, and 95.03 ± 5.54% antioxidant ability, respectively. BODIPY compounds showed excellent DNA chemical nuclease activity. BDPY-2, BDPY-3 and BDPY-4 also exhibited 100% APDT activity against E. coli at all tested concentrations. In addition to these, they demonstrated a highly effective biofilm inhibition activity against Staphyloccous aureus and Pseudomans aeruginosa. BDPY-4 showed the most effective antioxidant and DNA cleavage activity, while BDPY-3 exhibited the most effective antimicrobial and antibiofilm activity.
Collapse
Affiliation(s)
| | - Gürkan Karanlık
- Department of Chemistry, Yildiz Technical University, Istanbul, Turkey
| | - Sadin Özdemir
- Food Processing Programme, Technical Science Vocational School, Mersin University, Mersin, Turkey
| | - Gülşah Tollu
- Department of Laboratory and Veterinary Health, Technical Science Vocational School, Mersin University, Mersin, Turkey
| | - Ali Erdoğmuş
- Department of Chemistry, Yildiz Technical University, Istanbul, Turkey
- Health Biotechnology Joint Research and Application Center of Excellence, Istanbul, Turkey
| |
Collapse
|
10
|
Wang H, Liu Y, Bai C, Leung SSY. Translating bacteriophage-derived depolymerases into antibacterial therapeutics: Challenges and prospects. Acta Pharm Sin B 2024; 14:155-169. [PMID: 38239242 PMCID: PMC10792971 DOI: 10.1016/j.apsb.2023.08.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/12/2023] [Accepted: 07/22/2023] [Indexed: 01/22/2024] Open
Abstract
Predatory bacteriophages have evolved a vast array of depolymerases for bacteria capture and deprotection. These depolymerases are enzymes responsible for degrading diverse bacterial surface carbohydrates. They are exploited as antibiofilm agents and antimicrobial adjuvants while rarely inducing bacterial resistance, making them an invaluable asset in the era of antibiotic resistance. Numerous depolymerases have been investigated preclinically, with evidence indicating that depolymerases with appropriate dose regimens can safely and effectively combat different multidrug-resistant pathogens in animal infection models. Additionally, some formulation approaches have been developed for improved stability and activity of depolymerases. However, depolymerase formulation is limited to liquid dosage form and remains in its infancy, posing a significant hurdle to their clinical translation, compounded by challenges in their applicability and manufacturing. Future development must address these obstacles for clinical utility. Here, after unravelling the history, diversity, and therapeutic use of depolymerases, we summarized the preclinical efficacy and existing formulation findings of recombinant depolymerases. Finally, the challenges and perspectives of depolymerases as therapeutics for humans were assessed to provide insights for their further development.
Collapse
Affiliation(s)
- Honglan Wang
- School of Pharmacy, the Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yannan Liu
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Changqing Bai
- Department of Respiratory, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Guangdong 518055, China
| | | |
Collapse
|
11
|
Lee-Lopez C, Islam MS, Meléndez AB, Yukl ET. Influence of the Heme Nitric Oxide/Oxygen Binding Protein (H-NOX) on Cell Cycle Regulation in Caulobacter crescentus. Mol Cell Proteomics 2023; 22:100679. [PMID: 37979947 PMCID: PMC10746521 DOI: 10.1016/j.mcpro.2023.100679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 10/29/2023] [Indexed: 11/20/2023] Open
Abstract
The ability of an organism to respond to environmental changes is paramount to survival across a range of conditions. The bacterial heme nitric oxide/oxygen binding proteins (H-NOX) are a family of biofilm-regulating gas sensors that enable bacteria to respond accordingly to the cytotoxic molecule nitric oxide. By interacting with downstream signaling partners, H-NOX regulates the production of the bacterial secondary messenger cyclic diguanylate monophosphate (c-di-GMP) to influence biofilm formation. The aquatic organism Caulobacter crescentus has the propensity to attach to surfaces as part of its transition into the stalked S-phase of its life cycle. This behavior is heavily influenced by intracellular c-di-GMP and thus poses H-NOX as a potential influencer of C. crescentus surface attachment and cell cycle. By generating a strain of C. crescentus lacking hnox, our laboratory has demonstrated that this strain exhibits a considerable growth deficit, an increase in biofilm formation, and an elevation in c-di-GMP. Furthermore, in our comprehensive proteome study of 2779 proteins, 236 proteins were identified that exhibited differential expression in Δhnox C. crescentus, with 132 being downregulated and 104 being upregulated, as determined by a fold change of ≥1.5 or ≤0.66 and a p value ≤0.05. Our systematic analysis unveiled several regulated candidates including GcrA, PopA, RsaA, FtsL, DipM, FlgC, and CpaE that are associated with the regulation of the cellular division process, surface proteins, flagellum, and pili assembly. Further examination of Gene Ontology and pathways indicated that the key differences could be attributed to several metabolic processes. Taken together, our data indicate a role for the HNOX protein in C. crescentus cell cycle progression.
Collapse
Affiliation(s)
- Cameron Lee-Lopez
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico, USA
| | - Md Shariful Islam
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico, USA; Department of Mathematics and Physics, North South University, Dhaka, Bangladesh
| | - Ady B Meléndez
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico, USA
| | - Erik T Yukl
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico, USA.
| |
Collapse
|
12
|
Detomasi TC, Batka AE, Valastyan JS, Hydorn MA, Craik CS, Bassler BL, Marletta MA. Proteases influence colony aggregation behavior in Vibrio cholerae. J Biol Chem 2023; 299:105386. [PMID: 37898401 PMCID: PMC10709122 DOI: 10.1016/j.jbc.2023.105386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/03/2023] [Accepted: 10/16/2023] [Indexed: 10/30/2023] Open
Abstract
Aggregation behavior provides bacteria protection from harsh environments and threats to survival. Two uncharacterized proteases, LapX and Lap, are important for Vibrio cholerae liquid-based aggregation. Here, we determined that LapX is a serine protease with a preference for cleavage after glutamate and glutamine residues in the P1 position, which processes a physiologically based peptide substrate with a catalytic efficiency of 180 ± 80 M-1s-1. The activity with a LapX substrate identified by a multiplex substrate profiling by mass spectrometry screen was 590 ± 20 M-1s-1. Lap shares high sequence identity with an aminopeptidase (termed VpAP) from Vibrio proteolyticus and contains an inhibitory bacterial prepeptidase C-terminal domain that, when eliminated, increases catalytic efficiency on leucine p-nitroanilide nearly four-fold from 5.4 ± 4.1 × 104 M-1s-1 to 20.3 ± 4.3 × 104 M-1s-1. We demonstrate that LapX processes Lap to its mature form and thus amplifies Lap activity. The increase is approximately eighteen-fold for full-length Lap (95.7 ± 5.6 × 104 M-1s-1) and six-fold for Lap lacking the prepeptidase C-terminal domain (11.3 ± 1.9 × 105 M-1s-1). In addition, substrate profiling reveals preferences for these two proteases that could inform in vivo function. Furthermore, purified LapX and Lap restore the timing of the V. cholerae aggregation program to a mutant lacking the lapX and lap genes. Both proteases must be present to restore WT timing, and thus they appear to act sequentially: LapX acts on Lap, and Lap acts on the substrate involved in aggregation.
Collapse
Affiliation(s)
- Tyler C Detomasi
- Department of Chemistry, University of California, Berkeley, Berkeley, California, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
| | - Allison E Batka
- Department of Chemistry, University of California, Berkeley, Berkeley, California, USA
| | - Julie S Valastyan
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA; The Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Molly A Hydorn
- Department of Chemistry, University of California, Berkeley, Berkeley, California, USA; Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Charles S Craik
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
| | - Bonnie L Bassler
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA; The Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Michael A Marletta
- Department of Chemistry, University of California, Berkeley, Berkeley, California, USA; California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, California, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA.
| |
Collapse
|
13
|
Teuber A, Caniglia G, Barth H, Kranz C, Mizaikoff B. Thin-Film Waveguide Laser Spectroscopy: A Novel Platform for Bacterial Analysis. Anal Chem 2023; 95:16600-16608. [PMID: 37883708 DOI: 10.1021/acs.analchem.3c02782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Bacterial sensing based on quantum cascade laser spectroscopy coupled with diamond or gallium arsenide thin-film waveguides is a novel analytical tool for gaining high-resolution infrared spectroscopic information of planktonic and sessile bacteria, as shown in the present study for Escherichia coli. During observation periods of up to 24 h, diamond and gallium arsenide thin-film waveguide laser spectroscopy was compared to information obtained via conventional Fourier transform infrared spectroscopy. The proliferation behavior of E. coli at those surfaces was complementarily investigated using atomic force microscopy and scanning electron microscopy.
Collapse
Affiliation(s)
- Andrea Teuber
- Institute of Analytical and Bioanalytical Chemistry, University of Ulm, 89081 Ulm, Germany
| | - Giada Caniglia
- Institute of Analytical and Bioanalytical Chemistry, University of Ulm, 89081 Ulm, Germany
| | - Holger Barth
- Institute of Experimental and Clinical Pharmacology and Toxicology and Pharmacology of Natural Products, University of Ulm, 89081 Ulm, Germany
| | - Christine Kranz
- Institute of Analytical and Bioanalytical Chemistry, University of Ulm, 89081 Ulm, Germany
| | - Boris Mizaikoff
- Institute of Analytical and Bioanalytical Chemistry, University of Ulm, 89081 Ulm, Germany
- Hahn-Schickard, 89077 Ulm, Germany
| |
Collapse
|
14
|
Aoyama N, Kanematsu H, Barry DM, Miura H, Ogawa A, Kogo T, Kawai R, Hagio T, Hirai N, Kato T, Yoshitake M, Ichino R. AC Electromagnetic Field Controls the Biofilms on the Glass Surface by Escherichia coli & Staphylococcus epidermidis Inhibition Effect. MATERIALS (BASEL, SWITZERLAND) 2023; 16:7051. [PMID: 37959648 PMCID: PMC10649311 DOI: 10.3390/ma16217051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 11/15/2023]
Abstract
Biofilms, mainly comprised of bacteria, form on materials' surfaces due to bacterial activity. They are generally composed of water, extracellular polymeric substances (polysaccharides, proteins, nucleic acids, and lipids), and bacteria. Some bacteria that form biofilms cause periodontal disease, corrosion of the metal materials that make up drains, and slippage. Inside of a biofilm is an environment conducive to the growth and propagation of bacteria. Problems with biofilms include the inability of disinfectants and antibiotics to act on them. Therefore, we have investigated the potential application of alternating electromagnetic fields for biofilm control. We obtained exciting results using various materials' specimens and frequency conditions. Through these studies, we gradually understood that the combination of the type of bacteria, the kind of material, and the application of an electromagnetic field with various low frequencies (4 kHz-12 kHz) changes the circumstances of the onset of the biofilm suppression effect. In this study, relatively high frequencies (20 and 30 kHz) were applied to biofilms caused by Escherichia coli (E. coli) and Staphylococcus epidermidis (S. epidermidis), and quantitative evaluation was performed using staining methods. The sample surfaces were analyzed by Raman spectroscopy using a Laser Raman spectrometer to confirm the presence of biofilms on the surface.
Collapse
Affiliation(s)
- Natsu Aoyama
- Department of Materials Science and Engineering, National Institute of Technology (KOSEN), Suzuka College, (Currently Asahi Kasei Co.), Suzuka 510-0294, Japan; (N.A.); (T.K.); (R.K.)
| | - Hideyuki Kanematsu
- Research Collaboration Promotion Center, National Institute of Technology (KOSEN), Suzuka College, Suzuka 510-0294, Japan
| | - Dana M. Barry
- Department of Electrical and Computer Engineering, Clarkson University, Potsdam, NY 13699, USA;
| | - Hidekazu Miura
- Faculty of Medical Engineering, Suzuka University of Medical Science, Suzuka 510-0293, Japan;
| | - Akiko Ogawa
- Department of Chemistry and Biochemistry, National Institute of Technology (KOSEN), Suzuka College, Suzuka 510-0294, Japan; (A.O.); (N.H.)
| | - Takeshi Kogo
- Department of Materials Science and Engineering, National Institute of Technology (KOSEN), Suzuka College, (Currently Asahi Kasei Co.), Suzuka 510-0294, Japan; (N.A.); (T.K.); (R.K.)
| | - Risa Kawai
- Department of Materials Science and Engineering, National Institute of Technology (KOSEN), Suzuka College, (Currently Asahi Kasei Co.), Suzuka 510-0294, Japan; (N.A.); (T.K.); (R.K.)
| | - Takeshi Hagio
- Institutes of Innovation for Future Society, Graduate School of Engineering, Nagoya University, Nagoya 464-8601, Japan;
| | - Nobumitsu Hirai
- Department of Chemistry and Biochemistry, National Institute of Technology (KOSEN), Suzuka College, Suzuka 510-0294, Japan; (A.O.); (N.H.)
| | - Takehito Kato
- National Institute of Technology (KOSEN), Oyama College, Oyama 323-0806, Japan;
| | - Michiko Yoshitake
- National Institute for Materials Science (NIMS), Tsukuba 305-0047, Japan;
| | - Ryoichi Ichino
- Graduate School of Engineering Chemical Systems Engineering 2, Graduate School of Engineering, Nagoya University, Nagoya 464-8601, Japan;
| |
Collapse
|
15
|
Maliszewska I, Goldeman W. Increasing photoeradication's efficiency of Acinetobacter baumannii by polyphosphonic chelating agents. Photodiagnosis Photodyn Ther 2023; 43:103672. [PMID: 37364665 DOI: 10.1016/j.pdpdt.2023.103672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/17/2023] [Accepted: 06/22/2023] [Indexed: 06/28/2023]
Abstract
Ethylenediamine-N,N,N',N'-tetrakis(methylenephosphonic acid (EDTMP), nitrilotri(methylphosphonic acid (ATMP) and zoledronic acid were studied to enhance the methylene blue-mediated photodynamic inactivation of Acinetobacter baumannii. Laser light (wavelength 638 nm; standard light output 40 mW) was used in all experiment. Planktonic cultures were irradiated for 10, 20 and 30 min which corresponded to the light dose of 63 Jcm‒ 2, 126 Jcm‒2 and 189 Jcm‒2. Biocidal effect depended on the exposure time and it was shown that MB alone caused the highest reduction in the number of viable cells by 3.10 ± 0.2 log10 units after 30 min of irradiation. A significantly more effective killing effect was achieved when the bacteria were pre-treated with zoledronate, ATMP, or EDTMP (prior to photosensitisation) as the number of viable bacteria was reduced by 4.04±0.2 log10, 3.95±0.2 log10 and 4.01 ± 0.2 log10, respectively. The photo-killing effect caused by MB against biofilm pre-incubated with zoledronate, ATMP, or EDTMP and the number of viable bacteria was reduced by 0.80±0.1 log10, 1.25±0.05 log10 and 0.65±0.05 log10, respectively. Polyphosphonic chelating agents increased the efficiency of photo-destruction of A. baumannii by increasing the amount of bound photosensitizer by planktonic cells and biofilm, and increasing the detachment of live planktonic cells from the biofilm. The presence of glucose in the photosensitizing system significantly affected the bacterial photo-elimination. Pre-incubation of planktonic bacteria with the studied polyphosphonic chelating agents in the presence of glucose, and then exposure to light (with MB) for 30 min caused the lethal effect. This photo-eradication protocol (in the case of biofilms) reduced the number of viable bacteria by 2.05±0.2 log10, 3.2±0.2 log10 and 2.02±0.2 log10 for zoledronic acid, ATMP and EDTMP, respectively.
Collapse
Affiliation(s)
- Irena Maliszewska
- Department of Organic and Medicinal Chemistry, Faculty of Chemistry, Wrocaw University of Science and Technology, Wybrzeże Wyspiańskiego 27 Wrocław 50-370, Poland.
| | - Waldemar Goldeman
- Department of Organic and Medicinal Chemistry, Faculty of Chemistry, Wrocaw University of Science and Technology, Wybrzeże Wyspiańskiego 27 Wrocław 50-370, Poland
| |
Collapse
|
16
|
Perry EK, Tan MW. Bacterial biofilms in the human body: prevalence and impacts on health and disease. Front Cell Infect Microbiol 2023; 13:1237164. [PMID: 37712058 PMCID: PMC10499362 DOI: 10.3389/fcimb.2023.1237164] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/11/2023] [Indexed: 09/16/2023] Open
Abstract
Bacterial biofilms can be found in most environments on our planet, and the human body is no exception. Consisting of microbial cells encased in a matrix of extracellular polymers, biofilms enable bacteria to sequester themselves in favorable niches, while also increasing their ability to resist numerous stresses and survive under hostile circumstances. In recent decades, biofilms have increasingly been recognized as a major contributor to the pathogenesis of chronic infections. However, biofilms also occur in or on certain tissues in healthy individuals, and their constituent species are not restricted to canonical pathogens. In this review, we discuss the evidence for where, when, and what types of biofilms occur in the human body, as well as the diverse ways in which they can impact host health under homeostatic and dysbiotic states.
Collapse
Affiliation(s)
| | - Man-Wah Tan
- Department of Infectious Diseases, Genentech, South San Francisco, CA, United States
| |
Collapse
|
17
|
Mu M, Liu S, DeFlorio W, Hao L, Wang X, Salazar KS, Taylor M, Castillo A, Cisneros-Zevallos L, Oh JK, Min Y, Akbulut M. Influence of Surface Roughness, Nanostructure, and Wetting on Bacterial Adhesion. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:5426-5439. [PMID: 37014907 PMCID: PMC10848269 DOI: 10.1021/acs.langmuir.3c00091] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/22/2023] [Indexed: 05/11/2023]
Abstract
Bacterial fouling is a persistent problem causing the deterioration and failure of functional surfaces for industrial equipment/components; numerous human, animal, and plant infections/diseases; and energy waste due to the inefficiencies at internal and external geometries of transport systems. This work gains new insights into the effect of surface roughness on bacterial fouling by systematically studying bacterial adhesion on model hydrophobic (methyl-terminated) surfaces with roughness scales spanning from ∼2 nm to ∼390 nm. Additionally, a surface energy integration framework is developed to elucidate the role of surface roughness on the energetics of bacteria and substrate interactions. For a given bacteria type and surface chemistry; the extent of bacterial fouling was found to demonstrate up to a 75-fold variation with surface roughness. For the cases showing hydrophobic wetting behavior, both increased effective surface area with increasing roughness and decreased activation energy with increased surface roughness was concluded to enhance the extent of bacterial adhesion. For the cases of superhydrophobic surfaces, the combination of factors including (i) the surpassing of Laplace pressure force of interstitial air over bacterial adhesive force, (ii) the reduced effective substrate area for bacteria wall due to air gaps to have direct/solid contact, and (iii) the reduction of attractive van der Waals force that holds adhering bacteria on the substrate were summarized to weaken the bacterial adhesion. Overall, this study is significant in the context of designing antifouling coatings and systems as well as explaining variations in bacterial contamination and biofilm formation processes on functional surfaces.
Collapse
Affiliation(s)
- Minchen Mu
- Artie
McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843, United States
| | - Shuhao Liu
- Artie
McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843, United States
| | - William DeFlorio
- Artie
McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843, United States
| | - Li Hao
- School
of Chemistry and Chemical Engineering, Zhongkai
University of Agriculture and Engineering, Guangzhou, Guangdong 510225, P. R. China
| | - Xunhao Wang
- Department
of Chemical and Environmental Engineering, University of California, Riverside, California 92521, United States
| | - Karla Solis Salazar
- Department
of Food Science and Technology, Texas A&M
University, College Station, Texas 77843, United States
| | - Matthew Taylor
- Department
of Food Science and Technology, Texas A&M
University, College Station, Texas 77843, United States
| | - Alejandro Castillo
- Department
of Food Science and Technology, Texas A&M
University, College Station, Texas 77843, United States
| | - Luis Cisneros-Zevallos
- Department
of Horticultural Sciences, Texas A&M
University, College Station, Texas 77843, United States
| | - Jun Kyun Oh
- Department
of Polymer Science and Engineering, Dankook
University, 152 Jukjeon-ro, Suji-gu, Yongin-si, Gyeonggi-do 16890, Republic of Korea
| | - Younjin Min
- Department
of Chemical and Environmental Engineering, University of California, Riverside, California 92521, United States
| | - Mustafa Akbulut
- Artie
McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
18
|
Doulgeraki AI, Kamarinou CS, Nychas GJE, Argyri AA, Tassou CC, Moulas G, Chorianopoulos N. Role of Microbial Interactions across Food-Related Bacteria on Biofilm Population and Biofilm Decontamination by a TiO 2-Nanoparticle-Based Surfactant. Pathogens 2023; 12:pathogens12040573. [PMID: 37111459 PMCID: PMC10141041 DOI: 10.3390/pathogens12040573] [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: 01/30/2023] [Revised: 03/28/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Microbial interactions play an important role in initial cell adhesion and the endurance of biofilm toward disinfectant stresses. The present study aimed to evaluate the effect of microbial interactions on biofilm formation and the disinfecting activity of an innovative photocatalytic surfactant based on TiO2 nanoparticles. Listeria monocytogenes, Salmonella Enteritidis, Escherichia coli, Leuconostoc spp., Latilactobacillus sakei, Serratia liquefaciens, Serratia proteomaculans, Citrobacter freundii, Hafnia alvei, Proteus vulgaris, Pseudomonas fragi, and Brochothrix thermosphacta left to form mono- or dual-species biofilms on stainless steel (SS) coupons. The effectiveness of the photocatalytic disinfectant after 2 h of exposure under UV light on biofilm decontamination was evaluated. The effect of one parameter i.e., exposure to UV or disinfectant, was also determined. According to the obtained results, the microbial load of a mature biofilm depended on the different species or dual species that had adhered to the surface, while the presence of other species could affect the biofilm population of a specific microbe (p < 0.05). The disinfectant strengthened the antimicrobial activity of UV, as, in most cases, the remaining biofilm population was below the detection limit of the method. Moreover, the presence of more than one species affected the resistance of the biofilm cells to UV and the disinfectant (p < 0.05). In conclusion, this study confirms that microbial interactions affected biofilm formation and decontamination, and it demonstrates the effectiveness of the surfactant with the photocatalytic TiO2 agent, suggesting that it could be an alternative agent with which to disinfect contaminated surfaces.
Collapse
Affiliation(s)
- Agapi I Doulgeraki
- Institute of Technology of Agricultural Products, Hellenic Agricultural Organization-DIMITRA, S. Venizelou 1, 14123 Lycovrissi, Greece
| | - Christina S Kamarinou
- Institute of Technology of Agricultural Products, Hellenic Agricultural Organization-DIMITRA, S. Venizelou 1, 14123 Lycovrissi, Greece
- Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - George-John E Nychas
- Laboratory of Microbiology and Biotechnology of Foods, Department of Food Science and Human Nutrition, School of Food and Nutritional Sciences, Agricultural University of Athens, 11855 Athens, Greece
| | - Anthoula A Argyri
- Institute of Technology of Agricultural Products, Hellenic Agricultural Organization-DIMITRA, S. Venizelou 1, 14123 Lycovrissi, Greece
| | - Chrysoula C Tassou
- Institute of Technology of Agricultural Products, Hellenic Agricultural Organization-DIMITRA, S. Venizelou 1, 14123 Lycovrissi, Greece
| | | | - Nikos Chorianopoulos
- Laboratory of Microbiology and Biotechnology of Foods, Department of Food Science and Human Nutrition, School of Food and Nutritional Sciences, Agricultural University of Athens, 11855 Athens, Greece
| |
Collapse
|
19
|
Sarquis A, Bajrami D, Mizaikoff B, Ladero V, Alvarez MA, Fernandez M. Characterization of the Biofilms Formed by Histamine-Producing Lentilactobacillus parabuchneri Strains in the Dairy Environment. Foods 2023; 12:foods12071503. [PMID: 37048324 PMCID: PMC10093819 DOI: 10.3390/foods12071503] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/29/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
Lentilactobacillus parabuchneri, a lactic acid bacterium, is largely responsible for the production and accumulation of histamine, a toxic biogenic amine, in cheese. L. parabuchneri strains can form biofilms on the surface of industry equipment. Since they are resistant to cleaning and disinfection, they may act as reservoirs of histamine-producing contaminants in cheese. The aim of this study was to investigate the biofilm-producing capacity of L. parabuchneri strains. Using the crystal violet technique, the strains were first categorized as weak, moderate or strong biofilm producers. Analysis of their biofilm matrices revealed them to be mainly composed of proteins. Two strains of each category were then selected to analyze the influence on the biofilm-forming capacity of temperature, pH, carbon source, NaCl concentration and surface material (i.e., focusing on those used in the dairy industry). In general, low temperature (8 °C), high NaCl concentrations (2–3% w/v) and neutral pH (pH 6) prevented biofilm formation. All strains were found to adhere easily to beech wood. These findings increase knowledge of the biofilm-forming capacity of histamine-producing L. parabuchneri strains and how their formation may be prevented for improving food safety.
Collapse
Affiliation(s)
- Agustina Sarquis
- Departmento de Tecnología y Biotecnología de Productos Lácteos, Instituto de Productos Lácteos de Asturias, IPLA, CSIC, 33300 Villaviciosa, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Diellza Bajrami
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Albert Einstein-Allee 11, 89081 Ulm, Germany
| | - Boris Mizaikoff
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Albert Einstein-Allee 11, 89081 Ulm, Germany
- Hahn-Schickard, Institute for Microanalysis Systems, Sedanstrasse 14, 89077 Ulm, Germany
| | - Victor Ladero
- Departmento de Tecnología y Biotecnología de Productos Lácteos, Instituto de Productos Lácteos de Asturias, IPLA, CSIC, 33300 Villaviciosa, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Miguel A. Alvarez
- Departmento de Tecnología y Biotecnología de Productos Lácteos, Instituto de Productos Lácteos de Asturias, IPLA, CSIC, 33300 Villaviciosa, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Maria Fernandez
- Departmento de Tecnología y Biotecnología de Productos Lácteos, Instituto de Productos Lácteos de Asturias, IPLA, CSIC, 33300 Villaviciosa, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
20
|
Gowen R, Gamal A, Di Martino L, McCormick TS, Ghannoum MA. Modulating the Microbiome for Crohn's Disease Treatment. Gastroenterology 2023; 164:828-840. [PMID: 36702360 PMCID: PMC10152883 DOI: 10.1053/j.gastro.2023.01.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/12/2022] [Accepted: 01/06/2023] [Indexed: 01/28/2023]
Abstract
The central role of the gut microbiota in the regulation of health and disease has been convincingly demonstrated. Polymicrobial interkingdom interactions between bacterial (the bacteriome) and fungal (the mycobiome) communities of the gut have become a prominent focus for development of potential therapeutic approaches. In addition to polymicrobial interactions, the complex gut ecosystem also mediates interactions between the host and the microbiota. These interactions are complex and bidirectional; microbiota composition can be influenced by host immune response, disease-specific therapeutics, antimicrobial drugs, and overall ecosystems. However, the gut microbiota also influences host immune response to a drug or therapy by potentially transforming the drug's structure and altering bioavailability, activity, or toxicity. This is especially true in cases where the gut microbiota has produced a biofilm. The negative ramifications of biofilm formation include alteration of gut permeability, enhanced antimicrobial resistance, and alteration of host immune response effectiveness. Natural modulation of the gut microbiota, using probiotic and prebiotic approaches, may also be used to affect the host microbiome, a type of "natural" modulation of the host microbiota composition. In this review, we discuss potential bidirectional interactions between microbes and host, and we describe the changes in gut microbiota induced by probiotic and prebiotic approaches as well as their potential clinical consequences, including biofilm formation. We outline a systematic approach to designing probiotics capable of altering the host microbiota in disease states, using Crohn's disease as a model chronic disease. Understanding how the effective changes in the microbiome may enhance treatment efficacy may unlock the possibility of modulating the gut microbiome to improve treatment using a natural approach.
Collapse
Affiliation(s)
- Rachael Gowen
- Department of Dermatology, Case Western Reserve University, Cleveland, Ohio; University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Ahmed Gamal
- Department of Dermatology, Case Western Reserve University, Cleveland, Ohio; University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Luca Di Martino
- University Hospitals Cleveland Medical Center, Cleveland, Ohio; Department of Medicine, Case Western Reserve University, Cleveland, Ohio; Case Digestive Health Research Institute, Case Western Reserve University, Cleveland Ohio
| | - Thomas S McCormick
- Department of Dermatology, Case Western Reserve University, Cleveland, Ohio; University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Mahmoud A Ghannoum
- Department of Dermatology, Case Western Reserve University, Cleveland, Ohio; University Hospitals Cleveland Medical Center, Cleveland, Ohio.
| |
Collapse
|
21
|
Lu J, Shu Y, Zhang H, Zhang S, Zhu C, Ding W, Zhang W. The Landscape of Global Ocean Microbiome: From Bacterioplankton to Biofilms. Int J Mol Sci 2023; 24:6491. [PMID: 37047466 PMCID: PMC10095273 DOI: 10.3390/ijms24076491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 04/01/2023] Open
Abstract
The development of metagenomics has opened up a new era in the study of marine microbiota, which play important roles in biogeochemical cycles. In recent years, the global ocean sampling expeditions have spurred this research field toward a deeper understanding of the microbial diversities and functions spanning various lifestyles, planktonic (free-living) or sessile (biofilm-associated). In this review, we deliver a comprehensive summary of marine microbiome datasets generated in global ocean expeditions conducted over the last 20 years, including the Sorcerer II GOS Expedition, the Tara Oceans project, the bioGEOTRACES project, the Micro B3 project, the Bio-GO-SHIP project, and the Marine Biofilms. These datasets have revealed unprecedented insights into the microscopic life in our oceans and led to the publication of world-leading research. We also note the progress of metatranscriptomics and metaproteomics, which are confined to local marine microbiota. Furthermore, approaches to transforming the global ocean microbiome datasets are highlighted, and the state-of-the-art techniques that can be combined with data analyses, which can present fresh perspectives on marine molecular ecology and microbiology, are proposed.
Collapse
Affiliation(s)
- Jie Lu
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266100, China
| | - Yi Shu
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao 266100, China;
| | - Heng Zhang
- College of Marine Life Sciences, Ocean University of China, Qingdao 266100, China
| | - Shangxian Zhang
- Haide College, Ocean University of China, Qingdao 266100, China
| | - Chengrui Zhu
- Haide College, Ocean University of China, Qingdao 266100, China
| | - Wei Ding
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao 266100, China;
- College of Marine Life Sciences, Ocean University of China, Qingdao 266100, China
- Haide College, Ocean University of China, Qingdao 266100, China
| | - Weipeng Zhang
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266100, China
- College of Marine Life Sciences, Ocean University of China, Qingdao 266100, China
- Haide College, Ocean University of China, Qingdao 266100, China
| |
Collapse
|
22
|
Nesse LL, Osland AM, Vestby LK. The Role of Biofilms in the Pathogenesis of Animal Bacterial Infections. Microorganisms 2023; 11:608. [PMID: 36985183 PMCID: PMC10059901 DOI: 10.3390/microorganisms11030608] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/04/2023] Open
Abstract
Biofilms are bacterial aggregates embedded in a self-produced, protective matrix. The biofilm lifestyle offers resilience to external threats such as the immune system, antimicrobials, and other treatments. It is therefore not surprising that biofilms have been observed to be present in a number of bacterial infections. This review describes biofilm-associated bacterial infections in most body systems of husbandry animals, including fish, as well as in sport and companion animals. The biofilms have been observed in the auditory, cardiovascular, central nervous, digestive, integumentary, reproductive, respiratory, urinary, and visual system. A number of potential roles that biofilms can play in disease pathogenesis are also described. Biofilms can induce or regulate local inflammation. For some bacterial species, biofilms appear to facilitate intracellular invasion. Biofilms can also obstruct the healing process by acting as a physical barrier. The long-term protection of bacteria in biofilms can contribute to chronic subclinical infections, Furthermore, a biofilm already present may be used by other pathogens to avoid elimination by the immune system. This review shows the importance of acknowledging the role of biofilms in animal bacterial infections, as this influences both diagnostic procedures and treatment.
Collapse
Affiliation(s)
- Live L. Nesse
- Department of Animal Health, Welfare and Food Safety, Norwegian Veterinary Institute, 1433 Ås, Norway
| | - Ane Mohr Osland
- Department of Analysis and Diagnostics, Norwegian Veterinary Institute, 1433 Ås, Norway
| | - Lene K. Vestby
- Department of Analysis and Diagnostics, Norwegian Veterinary Institute, 1433 Ås, Norway
| |
Collapse
|
23
|
Razvi E, Whitfield GB, Reichhardt C, Dreifus JE, Willis AR, Gluscencova OB, Gloag ES, Awad TS, Rich JD, da Silva DP, Bond W, Le Mauff F, Sheppard DC, Hatton BD, Stoodley P, Reinke AW, Boulianne GL, Wozniak DJ, Harrison JJ, Parsek MR, Howell PL. Glycoside hydrolase processing of the Pel polysaccharide alters biofilm biomechanics and Pseudomonas aeruginosa virulence. NPJ Biofilms Microbiomes 2023; 9:7. [PMID: 36732330 PMCID: PMC9894940 DOI: 10.1038/s41522-023-00375-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 01/16/2023] [Indexed: 02/04/2023] Open
Abstract
Pel exopolysaccharide biosynthetic loci are phylogenetically widespread biofilm matrix determinants in bacteria. In Pseudomonas aeruginosa, Pel is crucial for cell-to-cell interactions and reducing susceptibility to antibiotic and mucolytic treatments. While genes encoding glycoside hydrolases have long been linked to biofilm exopolysaccharide biosynthesis, their physiological role in biofilm development is unclear. Here we demonstrate that the glycoside hydrolase activity of P. aeruginosa PelA decreases adherent biofilm biomass and is responsible for generating the low molecular weight secreted form of the Pel exopolysaccharide. We show that the generation of secreted Pel contributes to the biomechanical properties of the biofilm and decreases the virulence of P. aeruginosa in Caenorhabditis elegans and Drosophila melanogaster. Our results reveal that glycoside hydrolases found in exopolysaccharide biosynthetic systems can help shape the soft matter attributes of a biofilm and propose that secreted matrix components be referred to as matrix associated to better reflect their influence.
Collapse
Affiliation(s)
- Erum Razvi
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Gregory B Whitfield
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Département de Microbiologie, Infectiologie, et Immunologie, Faculté de Médecine Université de Montréal, Montréal, QC, Canada
| | - Courtney Reichhardt
- Department of Microbiology, University of Washington, Seattle, WA, USA
- Department of Chemistry, Washington University, St. Louis, MO, USA
| | - Julia E Dreifus
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Alexandra R Willis
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Oxana B Gluscencova
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Erin S Gloag
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA
- Department of Biomedical Sciences and Pathobiology, VA-MD College of Veterinary Medicine, Virginia Tech, VA, 24061, USA
| | - Tarek S Awad
- Department of Materials Science and Engineering, University of Toronto, Toronto, ON, Canada
| | - Jacquelyn D Rich
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Daniel Passos da Silva
- Department of Microbiology, University of Washington, Seattle, WA, USA
- BioVectra Inc. 11 Aviation, Charlottetown, PE, Canada
| | - Whitney Bond
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - François Le Mauff
- Department of Microbiology and Immunology, Faculty of Medicine, McGill University, Montreal, QC, Canada
- Infectious Disease and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- McGill Interdisciplinary Initiative in Infection and Immunity, Montreal, QC, Canada
| | - Donald C Sheppard
- Department of Microbiology and Immunology, Faculty of Medicine, McGill University, Montreal, QC, Canada
- Infectious Disease and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- McGill Interdisciplinary Initiative in Infection and Immunity, Montreal, QC, Canada
| | - Benjamin D Hatton
- Department of Materials Science and Engineering, University of Toronto, Toronto, ON, Canada
| | - Paul Stoodley
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA
- Department of Orthopedics, The Ohio State University, Columbus, OH, 43210, USA
- National Biofilm Innovation Centre (NBIC) and National Centre for Advanced Tribology at Southampton (nCATS), University of Southampton, Southampton, SO17 1BJ, UK
- Department of Microbiology, The Ohio State University, Columbus, OH, 43210, USA
| | - Aaron W Reinke
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Gabrielle L Boulianne
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Daniel J Wozniak
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA
- Department of Microbiology, The Ohio State University, Columbus, OH, 43210, USA
| | - Joe J Harrison
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Matthew R Parsek
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - P Lynne Howell
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON, Canada.
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
24
|
Sharma A, Rishi P, Singh R. In vitro and in vivo evaluation of DNase I in reinstating antibiotic efficacy against Klebsiella pneumoniae biofilms. Pathog Dis 2023; 81:6986254. [PMID: 36633541 DOI: 10.1093/femspd/ftad001] [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: 11/26/2022] [Revised: 01/06/2023] [Accepted: 01/11/2023] [Indexed: 01/13/2023] Open
Abstract
Klebsiella pneumoniae is an opportunistic pathogen associated with biofilm-based infections, which are intrinsically antibiotic resistant. Extracellular DNA plays a crucial role in biofilm formation and self-defence, with nucleases being proposed as promising agents for biofilm disruption. This study evaluated the in vitro and in vivo efficacy of DNase I in improving the activity of cefotaxime, amikacin, and ciprofloxacin against K. pneumoniae biofilms. K. pneumoniae ATCC 700603 and a clinical isolate from catheter-related bloodstream infection were cultured for biofilm formation on microtiter plates, and the antibiofilm activity of the antibiotics (0.03-64 mg/L), with or without bovine pancreatic DNase I (1-32 mg/L) was determined by XTT dye reduction test and viable counting. The effect of ciprofloxacin (2 mg/L) and DNase I (16 mg/L) was further evaluated in vitro on 1-cm-long silicon catheter segments, and in a mouse model of subcutaneous catheter-associated infection. Combination with DNase I did not improve the biofilm-preventive capacity of the three antibiotics or the biofilm-eradicating capacity of cefotaxime and amikacin. The biofilm-eradicating capacity of ciprofloxacin was increased by 8-fold and 4-fold in K. pneumoniae ATCC 700603 and clinical isolate, respectively, with DNase I. The combination therapy caused 99% reduction in biofilm biomass in the mouse model.
Collapse
Affiliation(s)
- Anayata Sharma
- Department of Microbial Biotechnology, Panjab University, Chandigarh, 160014, India
| | - Praveen Rishi
- Department of Microbiology, Panjab University, Chandigarh, 160014, India
| | - Rachna Singh
- Department of Microbial Biotechnology, Panjab University, Chandigarh, 160014, India
| |
Collapse
|
25
|
Mishra S, Gupta A, Upadhye V, Singh SC, Sinha RP, Häder DP. Therapeutic Strategies against Biofilm Infections. LIFE (BASEL, SWITZERLAND) 2023; 13:life13010172. [PMID: 36676121 PMCID: PMC9866932 DOI: 10.3390/life13010172] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023]
Abstract
A biofilm is an aggregation of surface-associated microbial cells that is confined in an extracellular polymeric substance (EPS) matrix. Infections caused by microbes that form biofilms are linked to a variety of animals, including insects and humans. Antibiotics and other antimicrobials can be used to remove or eradicate biofilms in order to treat infections. However, due to biofilm resistance to antibiotics and antimicrobials, clinical observations and experimental research clearly demonstrates that antibiotic and antimicrobial therapies alone are frequently insufficient to completely eradicate biofilm infections. Therefore, it becomes crucial and urgent for clinicians to properly treat biofilm infections with currently available antimicrobials and analyze the results. Numerous biofilm-fighting strategies have been developed as a result of advancements in nanoparticle synthesis with an emphasis on metal oxide np. This review focuses on several therapeutic strategies that are currently being used and also those that could be developed in the future. These strategies aim to address important structural and functional aspects of microbial biofilms as well as biofilms' mechanisms for drug resistance, including the EPS matrix, quorum sensing (QS), and dormant cell targeting. The NPs have demonstrated significant efficacy against bacterial biofilms in a variety of bacterial species. To overcome resistance, treatments such as nanotechnology, quorum sensing, and photodynamic therapy could be used.
Collapse
Affiliation(s)
- Sonal Mishra
- Laboratory of Photobiology and Molecular Microbiology, Centre of Advanced Study in Botany, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Amit Gupta
- Laboratory of Photobiology and Molecular Microbiology, Centre of Advanced Study in Botany, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Vijay Upadhye
- Department of Microbiology, Parul Institute of Applied Science (PIAS), Center of Research for Development (CR4D), Parul University, Vadodara 391760, Gujarat, India
| | - Suresh C. Singh
- Pathkits Healthcare Pvt. Ltd., Gurugram 122001, Haryana, India
| | - Rajeshwar P. Sinha
- Laboratory of Photobiology and Molecular Microbiology, Centre of Advanced Study in Botany, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Donat-P. Häder
- Department of Botany, Emeritus from Friedrich-Alexander University, 91096 Möhrendorf, Germany
- Correspondence: ; Tel.: +49-913-148-730
| |
Collapse
|
26
|
Gholami L, Shahabi S, Jazaeri M, Hadilou M, Fekrazad R. Clinical applications of antimicrobial photodynamic therapy in dentistry. Front Microbiol 2023; 13:1020995. [PMID: 36687594 PMCID: PMC9850114 DOI: 10.3389/fmicb.2022.1020995] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/30/2022] [Indexed: 01/07/2023] Open
Abstract
Given the emergence of resistant bacterial strains and novel microorganisms that globally threaten human life, moving toward new treatment modalities for microbial infections has become a priority more than ever. Antimicrobial photodynamic therapy (aPDT) has been introduced as a promising and non-invasive local and adjuvant treatment in several oral infectious diseases. Its efficacy for elimination of bacterial, fungal, and viral infections and key pathogens such as Streptococcus mutans, Porphyromonas gingivalis, Candida albicans, and Enterococcus faecalis have been investigated by many invitro and clinical studies. Researchers have also investigated methods of increasing the efficacy of such treatment modalities by amazing developments in the production of natural, nano based, and targeted photosensitizers. As clinical studies have an important role in paving the way towards evidence-based applications in oral infection treatment by this method, the current review aimed to provide an overall view of potential clinical applications in this field and summarize the data of available randomized controlled clinical studies conducted on the applications of aPDT in dentistry and investigate its future horizons in the dental practice. Four databases including PubMed (Medline), Web of Science, Scopus and Embase were searched up to September 2022 to retrieve related clinical studies. There are several clinical studies reporting aPDT as an effective adjunctive treatment modality capable of reducing pathogenic bacterial loads in periodontal and peri-implant, and persistent endodontic infections. Clinical evidence also reveals a therapeutic potential for aPDT in prevention and reduction of cariogenic organisms and treatment of infections with fungal or viral origins, however, the number of randomized clinical studies in these groups are much less. Altogether, various photosensitizers have been used and it is still not possible to recommend specific irradiation parameters due to heterogenicity among studies. Reaching effective clinical protocols and parameters of this treatment is difficult and requires further high quality randomized controlled trials focusing on specific PS and irradiation parameters that have shown to have clinical efficacy and are able to reduce pathogenic bacterial loads with sufficient follow-up periods.
Collapse
Affiliation(s)
- Leila Gholami
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, BC, Canada
| | - Shiva Shahabi
- Dental Implants Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Marzieh Jazaeri
- Dental Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mahdi Hadilou
- Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Fekrazad
- Radiation Sciences Research Center, Laser Research Center in Medical Sciences, AJA University of Medical Sciences, Tehran, Iran,International Network for Photo Medicine and Photo Dynamic Therapy (INPMPDT), Universal Scientific Education and Research Network (USERN), Tehran, Iran,*Correspondence: Reza Fekrazad,
| |
Collapse
|
27
|
Bessho S, Grando KCM, Kyrylchuk K, Miller A, Klein-Szanto AJ, Zhu W, Gallucci S, Tam V, Tükel Ç. Systemic exposure to bacterial amyloid curli alters the gut mucosal immune response and the microbiome, exacerbating Salmonella-induced arthritis. Gut Microbes 2023; 15:2221813. [PMID: 37317012 PMCID: PMC10269392 DOI: 10.1080/19490976.2023.2221813] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 06/16/2023] Open
Abstract
The Salmonella biofilm-associated amyloid protein, curli, is a dominant instigator of systemic inflammation and autoimmune responses following Salmonella infection. Systemic curli injections or infection of mice with Salmonella Typhimurium induce the major features of reactive arthritis, an autoimmune disorder associated with Salmonella infection in humans. In this study, we investigated the link between inflammation and microbiota in exacerbating autoimmunity. We studied C57BL/6 mice from two sources, Taconic Farms and Jackson Labs. Mice from Taconic Farms have been reported to have higher basal levels of the inflammatory cytokine IL - 17 than do mice from Jackson Labs due to the differences in their microbiota. When we systemically injected mice with purified curli, we observed a significant increase in diversity in the microbiota of Jackson Labs mice but not in that of the Taconic mice. In Jackson Labs, mice, the most striking effect was the expansion of Prevotellaceae. Furthermore, there were increases in the relative abundance of the family Akkermansiaceae and decreases in families Clostridiaceae and Muribaculaceae in Jackson Labs mice. Curli treatment led to significantly aggravated immune responses in the Taconic mice compared to Jackson Labs counterparts. Expression and production of IL - 1β, a cytokine known to promote IL - 17 production, as well as expression of Tnfa increased in the gut mucosa of Taconic mice in the first 24 hours after curli injections, which correlated with significant increases in the number of neutrophils and macrophages in the mesenteric lymph nodes. A significant increase in the expression of Ccl3 in colon and cecum of Taconic mice injected with curli was detected. Taconic mice injected with curli also had elevated levels of inflammation in their knees. Overall, our data suggest that autoimmune responses to bacterial ligands, such as curli, are amplified in individuals with a microbiome that promote inflammation.
Collapse
Affiliation(s)
- Shingo Bessho
- Center for Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, USA
| | - Kaitlyn C. M. Grando
- Center for Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, USA
| | - Kathrine Kyrylchuk
- Center for Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, USA
| | - Amanda Miller
- Center for Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, USA
| | | | - Wenhan Zhu
- Department of Pathology Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Stefania Gallucci
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Vincent Tam
- Center for Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, USA
| | - Çagla Tükel
- Center for Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, USA
| |
Collapse
|
28
|
The Emerging Role of Probiotics and their Derivatives against Biofilm-Producing MRSA: A Scoping Review. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4959487. [PMID: 36605101 PMCID: PMC9810406 DOI: 10.1155/2022/4959487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 11/20/2022] [Accepted: 12/01/2022] [Indexed: 12/29/2022]
Abstract
Background Methicillin-resistant Staphylococcus aureus (MRSA) is one of the main bacterial pathogens causing chronic infections, mainly because of its capacity to produce biofilm. Biofilm production is one of the underlying strategies for antibacterial drug resistance. Accordingly, preventing and attenuating biofilm production has become an emerging approach to controlling persistent infections. Therefore, this scoping review is aimed at surveying the published literature describing the usage of probiotics and their derivatives against biofilm-producing MRSA. Methods Updated literature searches were conducted across seven electronic databases including Web of Science, PubMed, Scopus, Cochrane Library, ProQuest, Embase, and Google Scholar to identify all original published articles about probiotics against MRSA. In this regard, studies were summarized and analyzed in the present review. Results In the reviewed studies, various microorganisms and compounds were used as probiotics as follows: Lactobacillus species (8 studies), Enterococcus species (4 studies), Bacillus species (2 studies), Streptomyces species (2 studies), Saccharomyces cerevisiae (1 study), Corynebacterium accolens (1 study), and Lactococcus lactis derived Nisin (3 studies). Based on our comprehensive search, 21 studies with eligibility criteria were included in the present review including 12 studies on clinical strains, 6 studies on ATCC, 2 studies simultaneously on clinical and standard strains, and finally 1 study on food sample. Conclusions Our study showed that there was an increasing trend in the number of publications reporting probiotics against biofilm-producing MRSA. The results of this scoping review could use to guide the undertaking of the subsequent systematic reviews. In summary, probiotics with antimicrobial and antibiofilm properties can use as an embedded agent in food products or as a biopharmaceutical in the prevention and treatment of MRSA infections.
Collapse
|
29
|
Šilha D, Syrová P, Syrová L, Janečková J. Smoothie Drinks: Possible Source of Resistant and Biofilm-Forming Microorganisms. Foods 2022; 11:foods11244039. [PMID: 36553778 PMCID: PMC9778333 DOI: 10.3390/foods11244039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/07/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022] Open
Abstract
Smoothie drinks are currently very popular drinks sold especially in fast food establishments. However, smoothies are a significant source of microorganisms. The aim of this study was to evaluate the microbiological quality of smoothies purchased in Eastern Bohemia. A higher prevalence of mesophilic aerobic bacteria (5.4-7.2 log CFU/mL), yeast (4.4-5.9 log CFU/mL) and coliform bacteria (3.1-6.0 log CFU/mL) was observed in vegetable smoothies, in which even the occurrence of enterococci (1.6-3.3 log CFU/mL) was observed. However, the occurrence of S. aureus, Salmonella spp. and Listeria spp. was not observed in any samples. Nevertheless, antimicrobial resistance was observed in 71.8% of the isolated strains. The highest level of resistance was found in isolates from smoothie drinks with predominantly vegetable contents (green smoothie drinks). Considerable resistance was observed in Gram-negative rods, especially to amoxicillin (82.2%) and amoxicillin with clavulanic acid (55.6%). Among enterococci, only one vancomycin-resistant strain was detected. The vast majority of isolated strains were able to form biofilms at a significant level, which increases the clinical importance of these microorganisms. The highest biofilm production was found in Pseudomonas aeruginosa, Kocuria kristinae and Klebsiella pneumoniae. Overall, significant biofilm production was also noted among isolates of Candida spp.
Collapse
Affiliation(s)
- David Šilha
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentská 573, 532-10 Pardubice, Czech Republic
- Correspondence:
| | - Petra Syrová
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentská 573, 532-10 Pardubice, Czech Republic
| | - Lenka Syrová
- Department of Infectious Diagnostics, Hospital of the Pardubice Region, Jana Evangelisty Purkyně 652, 570-14 Litomysl, Czech Republic
| | - Jana Janečková
- Department of Infectious Diagnostics, Hospital of the Pardubice Region, Jana Evangelisty Purkyně 652, 570-14 Litomysl, Czech Republic
| |
Collapse
|
30
|
Xu W, Ceylan Koydemir H. Non-invasive biomedical sensors for early detection and monitoring of bacterial biofilm growth at the point of care. LAB ON A CHIP 2022; 22:4758-4773. [PMID: 36398687 DOI: 10.1039/d2lc00776b] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Bacterial infections have long been a serious global health issue. Biofilm formation complicates matters even more. The biofilm's extracellular polymeric substances (EPSs) matrix protects bacteria from the host's immune responses, yielding strong adhesion and drug resistance as the biofilm matures. Early bacterial biofilm detection and bacterial biofilm growth monitoring are crucial to treating biofilm-associated infections. Current detection methods are highly sensitive but not portable, are time-consuming, and require expensive equipment and complex operating procedures, limiting their use at the point of care. Therefore, there is an urgent need to develop affordable, on-body, and non-invasive biomedical sensors to continuously monitor and detect early biofilm growth at the point of care through personalized telemedicine. Herein, recent advances in developing non-invasive biomedical sensors for early detection and monitoring bacterial biofilm growth are comprehensively reviewed. First, biofilm's life cycle and its impact on the human body, such as biofilm-associated disease and infected medical devices, are introduced together with the challenges of biofilm treatment. Then, the current methods used in clinical and laboratory settings for biofilm detection and their challenges are discussed. Next, the current state of non-invasive sensors for direct and indirect detection of bacterial biofilms are summarized and highlighted with the detection parameters and their design details. Finally, commercially available products, challenges of current devices, and the further trend in biofilm detection sensors are discussed.
Collapse
Affiliation(s)
- Weiming Xu
- Department of Biomedical Engineering, Texas A&M University, College Station, 77843, Texas, USA.
- Center for Remote Health Technologies and Systems, Texas A&M Engineering Experiment Station, College Station, 77843, TX, USA
| | - Hatice Ceylan Koydemir
- Department of Biomedical Engineering, Texas A&M University, College Station, 77843, Texas, USA.
- Center for Remote Health Technologies and Systems, Texas A&M Engineering Experiment Station, College Station, 77843, TX, USA
| |
Collapse
|
31
|
Levipan HA, Irgang R, Opazo LF, Araya-León H, Avendaño-Herrera R. Collective behavior and virulence arsenal of the fish pathogen Piscirickettsia salmonis in the biofilm realm. Front Cell Infect Microbiol 2022; 12:1067514. [PMID: 36544910 PMCID: PMC9760808 DOI: 10.3389/fcimb.2022.1067514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022] Open
Abstract
Piscirickettsiosis is a fish disease caused by the Gram-negative bacterium Piscirickettsia salmonis. This disease has a high socio-economic impact on the Chilean salmonid aquaculture industry. The bacterium has a cryptic character in the environment and their main reservoirs are yet unknown. Bacterial biofilms represent a ubiquitous mechanism of cell persistence in diverse natural environments and a risk factor for the pathogenesis of several infectious diseases, but their microbiological significance for waterborne veterinary diseases, including piscirickettsiosis, have seldom been evaluated. This study analyzed the in vitro biofilm behavior of P. salmonis LF-89T (genogroup LF-89) and CA5 (genogroup EM-90) using a multi-method approach and elucidated the potential arsenal of virulence of the P. salmonis LF-89T type strain in its biofilm state. P. salmonis exhibited a quick kinetics of biofilm formation that followed a multi-step and highly strain-dependent process. There were no major differences in enzymatic profiles or significant differences in cytotoxicity (as tested on the Chinook salmon embryo cell line) between biofilm-derived bacteria and planktonic equivalents. The potential arsenal of virulence of P. salmonis LF-89T in biofilms, as determined by whole-transcriptome sequencing and differential gene expression analysis, consisted of genes involved in cell adhesion, polysaccharide biosynthesis, transcriptional regulation, and gene mobility, among others. Importantly, the global gene expression profiles of P. salmonis LF-89T were not enriched with virulence-related genes upregulated in biofilm development stages at 24 and 48 h. An enrichment in virulence-related genes exclusively expressed in biofilms was also undetected. These results indicate that early and mature biofilm development stages of P. salmonis LF-89T were transcriptionally no more virulent than their planktonic counterparts, which was supported by cytotoxic trials, which, in turn, revealed that both modes of growth induced important and very similar levels of cytotoxicity on the salmon cell line. Our results suggest that the aforementioned biofilm development stages do not represent hot spots of virulence compared with planktonic counterparts. This study provides the first transcriptomic catalogue to select specific genes that could be useful to prevent or control the (in vitro and/or in vivo) adherence and/or biofilm formation by P. salmonis and gain further insights into piscirickettsiosis pathogenesis.
Collapse
Affiliation(s)
- Héctor A. Levipan
- Laboratorio de Ecopatología y Nanobiomateriales, Departamento de Ciencias y Geografía, Facultad de Ciencias Naturales y Exactas, Universidad de Playa Ancha, Valparaíso, Chile,Centro de Espectroscopía Atómica y Molecular (ATMOS-C), Universidad de Playa Ancha, Valparaíso, Chile,*Correspondence: Héctor A. Levipan, ; ; Ruben Avendaño-Herrera, ;
| | - Rute Irgang
- Laboratorio de Patología de Organismos Acuáticos y Biotecnología Acuícola, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Viña del Mar, Chile,Interdisciplinary Center for Aquaculture Research (INCAR), Universidad Andrés Bello, Viña del Mar, Chile
| | - L. Felipe Opazo
- Institute of Ecology and Biodiversity (IEB), Santiago, Chile,Departamento de Ecología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Henry Araya-León
- Laboratorio de Patología de Organismos Acuáticos y Biotecnología Acuícola, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Viña del Mar, Chile,Interdisciplinary Center for Aquaculture Research (INCAR), Universidad Andrés Bello, Viña del Mar, Chile
| | - Ruben Avendaño-Herrera
- Laboratorio de Patología de Organismos Acuáticos y Biotecnología Acuícola, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Viña del Mar, Chile,Interdisciplinary Center for Aquaculture Research (INCAR), Universidad Andrés Bello, Viña del Mar, Chile,Centro de Investigación Marina Quintay (CIMARQ), Universidad Andrés Bello, Quintay, Chile,*Correspondence: Héctor A. Levipan, ; ; Ruben Avendaño-Herrera, ;
| |
Collapse
|
32
|
Sarwar W, Ali Q, Ahmed S. Microscopic visualization of the antibiofilm potential of essential oils against Staphylococcus aureus and Klebsiella pneumoniae. Microsc Res Tech 2022; 85:3921-3931. [PMID: 36250506 DOI: 10.1002/jemt.24243] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/25/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022]
Abstract
Biofilms are known to pose great risks in clinical settings, drinking water systems, and food industries as they show considerable resistance to various environmental stresses. This study investigates the antibiofilm potential of different essential oils against the test organisms Staphylococcus aureus (ATCC 25923) and Klebsiella pneumoniae (ATCC 13883). Moreover, different stages of biofilm formation were also assessed using light microscopic assays. For determining the antibiofilm activity, a total of five essential oils namely cinnamon (Cinnamomum Verum), tea tree (Melaleuca alternifolia), lavender (Lavandula), peppermint (Mentha piperita), and lemongrass (Cymbopogon citratus) were tested for their ability to inhibit the initial attachment of microbial cells as well as the eradication of mature biofilm using the microtitre plate assay. For both the test strains (S. aureus and K. pneumoniae) the concentration of 30 μl/100 μl of cinnamon oil exhibited the highest antibiofilm activity followed by the activity of peppermint oil at the same concentration. These results were further validated by employing the light microscopy assay for observing the antibiofilm potential of cinnamon and peppermint essential oils.
Collapse
Affiliation(s)
- Warda Sarwar
- Department of Microbiology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Qurban Ali
- Department of Microbiology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Safia Ahmed
- Department of Microbiology, Quaid-i-Azam University, Islamabad, Pakistan
| |
Collapse
|
33
|
Sharaf M, Sewid AH, Hamouda HI, Elharrif MG, El-Demerdash AS, Alharthi A, Hashim N, Hamad AA, Selim S, Alkhalifah DHM, Hozzein WN, Abdalla M, Saber T. Rhamnolipid-Coated Iron Oxide Nanoparticles as a Novel Multitarget Candidate against Major Foodborne E. coli Serotypes and Methicillin-Resistant S. aureus. Microbiol Spectr 2022; 10:e0025022. [PMID: 35852338 PMCID: PMC9430161 DOI: 10.1128/spectrum.00250-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 06/21/2022] [Indexed: 01/28/2023] Open
Abstract
Surface-growing antibiotic-resistant pathogenic bacteria such as Escherichia coli and Staphylococcus aureus are emerging as a global health challenge due to dilemmas in clinical treatment. Furthermore, their pathogenesis, including increasingly serious antimicrobial resistance and biofilm formation, makes them challenging to treat by conventional therapy. Therefore, the development of novel antivirulence strategies will undoubtedly provide a path forward in combatting these resistant bacterial infections. In this regard, we developed novel biosurfactant-coated nanoparticles to combine the antiadhesive/antibiofilm properties of rhamnolipid (RHL)-coated Fe3O4 nanoparticles (NPs) with each of the p-coumaric acid (p-CoA) and gallic acid (GA) antimicrobial drugs by using the most available polymer common coatings (PVA) to expand the range of effective antibacterial drugs, as well as a mechanism for their synergistic effect via a simple method of preparation. Mechanistically, the average size of bare Fe3O4 NPs was ~15 nm, while RHL-coated Fe3O4@PVA@p-CoA/GA was about ~254 nm, with a drop in zeta potential from -18.7 mV to -34.3 mV, which helped increase stability. Our data show that RHL-Fe3O4@PVA@p-CoA/GA biosurfactant NPs can remarkably interfere with bacterial growth and significantly inhibited biofilm formation to more than 50% via downregulating IcaABCD and CsgBAC operons, which are responsible for slime layer formation and curli fimbriae production in S. aureus and E. coli, respectively. The novelty regarding the activity of RHL-Fe3O4@PVA@p-CoA/GA biosurfactant NPs reveals their potential effect as an alternative multitarget antivirulence candidate to minimize infection severity by inhibiting biofilm development. Therefore, they could be used in antibacterial coatings and wound dressings in the future. IMPORTANCE Antimicrobial resistance poses a great threat and challenge to humanity. Therefore, the search for alternative ways to target and eliminate microbes from plant, animal, and marine microorganisms is one of the world's concerns today. Furthermore, the extraordinary capacity of S. aureus and E. coli to resist standard antibacterial drugs is the dilemma of all currently used remedies. Methicillin-resistant S. aureus (MRSA) and vancomycin-resistant S. aureus (VRSA) have become widespread, leading to no remedies being able to treat these threatening pathogens. The most widely recognized serotypes that cause severe foodborne illness are E. coli O157:H7, O26:H11, and O78:H10, and they display increasing antimicrobial resistance rates. Therefore, there is an urgent need for an effective therapy that has dual action to inhibit biofilm formation and decrease bacterial growth. In this study, the synthesized RHL-Fe3O4@PVA@p-CoA/GA biosurfactant NPs have interesting properties, making them excellent candidates for targeted drug delivery by inhibiting bacterial growth and downregulating biofilm-associated IcaABCD and CsgBAC gene loci.
Collapse
Affiliation(s)
- Mohamed Sharaf
- Department of Biochemistry, Faculty of Agriculture, Al-Azhar University, Nasr City, Cairo, Egypt
- Department of Biochemistry and Molecular Biology, College of Marine Life Sciences, Ocean University of China, Qingdao, People’s Republic of China
| | - Alaa H. Sewid
- Department of Microbiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
- Department of Biomedical and Diagnostic Sciences, University of Tennessee, Knoxville, Tennessee, USA
| | - H. I. Hamouda
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
- Processes Design and Development Department, Egyptian Petroleum Research Institute, Nasr City, Cairo, Egypt
| | - Mohamed G. Elharrif
- Department of Basic Medical Sciences, Shaqra University, Shaqraa, Kingdom of Saudi Arabia
| | | | - Afaf Alharthi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Nada Hashim
- General Practitioner, Faculty of Medicine, University of Gezira, Wad Medani, Sudan
| | - Anas Abdullah Hamad
- Department of Medical Laboratory Techniques, Al Maarif University College, Al Anbar, Ramadi, Iraq
| | - Samy Selim
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Dalal Hussien M. Alkhalifah
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Wael N. Hozzein
- Botany and Microbiology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Mohnad Abdalla
- Department of Biotechnology, Faculty of Science and Technology, Shendi University, Shendi, Nher Anile, Sudan
| | - Taisir Saber
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
34
|
Board-Davies EL, Rhys-Williams W, Hynes D, Williams D, Farnell DJJ, Love W. Antibacterial and Antibiofilm Potency of XF Drugs, Impact of Photodynamic Activation and Synergy With Antibiotics. Front Cell Infect Microbiol 2022; 12:904465. [PMID: 35846763 PMCID: PMC9279914 DOI: 10.3389/fcimb.2022.904465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
With increasing incidence of antimicrobial resistance, there is an urgent need for novel and effective antibacterials. Destiny Pharma plc have developed a series of porphyrin-based XF drugs, some with dual mechanisms of antibacterial action. An innate mechanism acts through binding to the outer bacterial membrane and a separate, light-activated, photodynamic (PD) mechanism, acts via the generation of reactive oxygen species. This study aimed to assess the innate and PD associated antibacterial activity of XF drugs against planktonic bacteria, their biofilms and combinational effects with conventional antibiotics. Minimum inhibitory concentrations (MICs) were determined for 3 XF drugs against 114 bacterial isolates. MICs for XF-73 and XF-70 were determined (± PD). DPD-207 was designed to not exhibit PD action due to its structure. XF-drugs (± PD) were further assessed for synergy with conventional antibiotics (using a checkerboard assay) and antibiofilm activity against susceptible strains. XF drugs were innately active against all tested Gram-positive isolates. PD action significantly increased bacterial susceptibility to XF-73 and XF-70 for all Gram-positive isolates. Generally, the XF drugs exhibited higher MICs against Gram-negative isolates, however PD significantly enhanced potency, particularly for XF-70. XF-73 and XF-70 exhibited synergy with ertapenem against a methicillin resistant Staphylococcus aureus (MRSA) strain (± PD) and XF-73 with polymyxin B (± PD) against Pseudomonas aeruginosa. No antagonism was seen between the XF drugs and any of the 5 antibiotics tested. The antibiofilm effect of XF drugs was also observed for all Staphylococcus isolates tested. Generally, PD did not enhance activity for other bacterial isolates tested with the exception of XF-73 against Acinetobacter baumannii biofilms. XF drugs exhibited significant antimicrobial activity against Gram-positive bacteria, with PD enhancement of bacterial susceptibility. Additionally, XF drugs displayed synergy with conventional antibiotics and demonstrated antibiofilm effects.
Collapse
Affiliation(s)
| | | | | | - David Williams
- School of Dentistry, Cardiff University, Cardiff, United Kingdom
- *Correspondence: David Williams,
| | | | | |
Collapse
|
35
|
Ribeiro M, Gomes IB, Saavedra MJ, Simões M. Photodynamic therapy and combinatory treatments for the control of biofilm-associated infections. Lett Appl Microbiol 2022; 75:548-564. [PMID: 35689422 DOI: 10.1111/lam.13762] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 04/26/2022] [Accepted: 05/26/2022] [Indexed: 12/20/2022]
Abstract
The advent of antimicrobial resistance has added considerable impact to infectious diseases both in the number of infections and healthcare costs. Furthermore, the relentless emergence of multidrug-resistant bacteria, particularly in the biofilm state, has made mandatory the discovery of new alternative antimicrobial therapies that are capable to eradicate resistant bacteria and impair the development of new forms of resistance. Amongst the therapeutic strategies for treating biofilms, antimicrobial photodynamic therapy (aPDT) has shown great potential in inactivating several clinically relevant micro-organisms, including antibiotic-resistant 'priority bacteria' declared by the WHO as critical pathogens. Its antimicrobial effect is centred on the basis that harmless low-intensity light stimulates a non-toxic dye named photosensitizer, triggering the production of reactive oxygen species upon photostimulation. In addition, combination therapies of aPDT with other antimicrobial agents (e.g. antibiotics) have also drawn considerable attention, as it is a multi-target strategy. Therefore, the present review highlights the recent advances of aPDT against biofilms, also covering progress on combination therapy.
Collapse
Affiliation(s)
- M Ribeiro
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal.,ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal.,CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - I B Gomes
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal.,ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal
| | - M J Saavedra
- Department of Veterinary Sciences, School of Agriculture and Veterinary Science, UTAD, Vila Real, Portugal.,Centre for the Research and Technology for Agro-Environment and Biological Sciences (CITAB), UTAD, Vila Real, Portugal
| | - M Simões
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal.,ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal
| |
Collapse
|
36
|
Belardinelli JM, Li W, Martin KH, Zeiler MJ, Lian E, Avanzi C, Wiersma CJ, Nguyen TV, Angala B, de Moura VCN, Jones V, Borlee BR, Melander C, Jackson M. 2-Aminoimidazoles Inhibit Mycobacterium abscessus Biofilms in a Zinc-Dependent Manner. Int J Mol Sci 2022; 23:ijms23062950. [PMID: 35328372 PMCID: PMC8951752 DOI: 10.3390/ijms23062950] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 02/28/2022] [Accepted: 03/07/2022] [Indexed: 02/04/2023] Open
Abstract
Biofilm growth is thought to be a significant obstacle to the successful treatment of Mycobacterium abscessus infections. A search for agents capable of inhibiting M. abscessus biofilms led to our interest in 2-aminoimidazoles and related scaffolds, which have proven to display antibiofilm properties against a number of Gram-negative and Gram-positive bacteria, including Mycobacterium tuberculosis and Mycobacterium smegmatis. The screening of a library of 30 compounds led to the identification of a compound, AB-2-29, which inhibits the formation of M. abscessus biofilms with an IC50 (the concentration required to inhibit 50% of biofilm formation) in the range of 12.5 to 25 μM. Interestingly, AB-2-29 appears to chelate zinc, and its antibiofilm activity is potentiated by the addition of zinc to the culture medium. Preliminary mechanistic studies indicate that AB-2-29 acts through a distinct mechanism from those reported to date for 2-aminoimidazole compounds.
Collapse
Affiliation(s)
- Juan M. Belardinelli
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA; (J.M.B.); (W.L.); (E.L.); (C.A.); (C.J.W.); (B.A.); (V.C.N.d.M.); (V.J.)
| | - Wei Li
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA; (J.M.B.); (W.L.); (E.L.); (C.A.); (C.J.W.); (B.A.); (V.C.N.d.M.); (V.J.)
| | - Kevin H. Martin
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA; (K.H.M.); (B.R.B.)
| | - Michael J. Zeiler
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; (M.J.Z.); (C.M.)
| | - Elena Lian
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA; (J.M.B.); (W.L.); (E.L.); (C.A.); (C.J.W.); (B.A.); (V.C.N.d.M.); (V.J.)
| | - Charlotte Avanzi
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA; (J.M.B.); (W.L.); (E.L.); (C.A.); (C.J.W.); (B.A.); (V.C.N.d.M.); (V.J.)
| | - Crystal J. Wiersma
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA; (J.M.B.); (W.L.); (E.L.); (C.A.); (C.J.W.); (B.A.); (V.C.N.d.M.); (V.J.)
| | - Tuan Vu Nguyen
- Department of Chemistry, North Carolina State University, Raleigh, NC 27607, USA;
| | - Bhanupriya Angala
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA; (J.M.B.); (W.L.); (E.L.); (C.A.); (C.J.W.); (B.A.); (V.C.N.d.M.); (V.J.)
| | - Vinicius C. N. de Moura
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA; (J.M.B.); (W.L.); (E.L.); (C.A.); (C.J.W.); (B.A.); (V.C.N.d.M.); (V.J.)
| | - Victoria Jones
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA; (J.M.B.); (W.L.); (E.L.); (C.A.); (C.J.W.); (B.A.); (V.C.N.d.M.); (V.J.)
| | - Bradley R. Borlee
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA; (K.H.M.); (B.R.B.)
| | - Christian Melander
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA; (M.J.Z.); (C.M.)
- Department of Chemistry, North Carolina State University, Raleigh, NC 27607, USA;
| | - Mary Jackson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA; (J.M.B.); (W.L.); (E.L.); (C.A.); (C.J.W.); (B.A.); (V.C.N.d.M.); (V.J.)
- Correspondence: ; Tel.: +1-(970)-491-3582
| |
Collapse
|
37
|
Inactivation of Opportunistic Pathogens Acinetobacter baumannii and Stenotrophomonas maltophilia by Antimicrobial Photodynamic Therapy. Microorganisms 2022; 10:microorganisms10030506. [PMID: 35336082 PMCID: PMC8948924 DOI: 10.3390/microorganisms10030506] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/17/2022] [Accepted: 02/23/2022] [Indexed: 02/04/2023] Open
Abstract
Acinetobacter baumannii and Stenotrophomonas maltophilia are opportunistic pathogens causing hospital infections with limited treatment options due to bacterial multidrug resistance. Here, we report that antimicrobial photodynamic therapy (aPDT) based on the natural photosensitizers riboflavin and chlorophyllin inactivates A. baumannii and S. maltophilia. The riboflavin and chlorophyllin photostability experiments assessed the photomodifications of photosensitizers under the conditions subsequently used to inactivate A. baumannii and S. maltophilia. A. baumannii planktonic cells were more sensitive to riboflavin-aPDT, while biofilm bacteria were more efficiently inactivated by chlorophyllin-aPDT. S. maltophilia planktonic and biofilm cells were more susceptible to chlorophyllin-aPDT compared to riboflavin-aPDT. The results suggest that riboflavin- and chlorophyllin-aPDT can be considered as a potential antimicrobial treatment for A. baumannii and S. maltophilia inactivation.
Collapse
|
38
|
Belardinelli JM, Verma D, Li W, Avanzi C, Wiersma CJ, Williams JT, Johnson BK, Zimmerman M, Whittel N, Angala B, Wang H, Jones V, Dartois V, de Moura VCN, Gonzalez-Juarrero M, Pearce C, Schenkel AR, Malcolm KC, Nick JA, Charman SA, Wells TNC, Podell BK, Vennerstrom JL, Ordway DJ, Abramovitch RB, Jackson M. Therapeutic efficacy of antimalarial drugs targeting DosRS signaling in Mycobacterium abscessus. Sci Transl Med 2022; 14:eabj3860. [PMID: 35196022 DOI: 10.1126/scitranslmed.abj3860] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A search for alternative Mycobacterium abscessus treatments led to our interest in the two-component regulator DosRS, which, in Mycobacterium tuberculosis, is required for the bacterium to establish a state of nonreplicating, drug-tolerant persistence in response to a variety of host stresses. We show here that the genetic disruption of dosRS impairs the adaptation of M. abscessus to hypoxia, resulting in decreased bacterial survival after oxygen depletion, reduced tolerance to a number of antibiotics in vitro and in vivo, and the inhibition of biofilm formation. We determined that three antimalarial drugs or drug candidates, artemisinin, OZ277, and OZ439, can target DosS-mediated hypoxic signaling in M. abscessus and recapitulate the phenotypic effects of genetically disrupting dosS. OZ439 displayed bactericidal activity comparable to standard-of-care antibiotics in chronically infected mice, in addition to potentiating the activity of antibiotics used in combination. The identification of antimalarial drugs as potent inhibitors and adjunct inhibitors of M. abscessus in vivo offers repurposing opportunities that could have an immediate impact in the clinic.
Collapse
Affiliation(s)
- Juan Manuel Belardinelli
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Deepshikha Verma
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Wei Li
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Charlotte Avanzi
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Crystal J Wiersma
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - John T Williams
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | | | - Matthew Zimmerman
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Nicholas Whittel
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Bhanupriya Angala
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Han Wang
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Victoria Jones
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Vinicius C N de Moura
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Mercedes Gonzalez-Juarrero
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Camron Pearce
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Alan R Schenkel
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Kenneth C Malcolm
- Department of Medicine, National Jewish Health, Denver, CO, USA.,Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Jerry A Nick
- Department of Medicine, National Jewish Health, Denver, CO, USA.,Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Susan A Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | | | - Brendan K Podell
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | | | - Diane J Ordway
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Robert B Abramovitch
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Mary Jackson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
39
|
Wollanke B, Gerhards H, Ackermann K. Infectious Uveitis in Horses and New Insights in Its Leptospiral Biofilm-Related Pathogenesis. Microorganisms 2022; 10:387. [PMID: 35208842 PMCID: PMC8875353 DOI: 10.3390/microorganisms10020387] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 12/17/2022] Open
Abstract
Uveitis is a sight-threatening eye disease in equids known worldwide that leads to considerable pain and suffering. By far the most common type of uveitis in Germany and neighboring countries is classical equine recurrent uveitis (ERU), which is caused by chronic intraocular leptospiral infection and is the main cause of infectious uveitis in horses. Other infectious causes are extremely rare and are usually clinically distinguishable from ERU. ERU can be treated very effectively by vitreous cavity lavage (vitrectomy). For proper indications of this demanding surgery, it is necessary to differentiate ERU from other types of uveitis in which vitrectomy is not helpful. This can be conducted on the basis of anamnesis in combination with ophthalmologic findings and by aqueous humor examination. During vitrectomy, vitreous material is obtained. These vitreous samples have historically been used for numerous etiologic studies. In this way, a chronic intraocular leptospiral infection has been shown to be the cause of typical ERU and, among other findings, ERU has also been recognized as a biofilm infection, providing new insights into the pathogenesis of ERU and explaining some thus far unexplainable phenomena of ERU. ERU may not only have transmissible aspects to some types of uveitis in humans but may also serve as a model for a spontaneously occurring biofilm infection. Vitreous material obtained during therapeutically indicated vitrectomy can be used for further studies on in vivo biofilm formation, biofilm composition and possible therapeutic approaches.
Collapse
Affiliation(s)
- Bettina Wollanke
- Equine Clinic, Ludwig-Maximilians-University, 80539 Munich, Germany; (H.G.); (K.A.)
| | | | | |
Collapse
|
40
|
Nadar S, Khan T, Patching SG, Omri A. Development of Antibiofilm Therapeutics Strategies to Overcome Antimicrobial Drug Resistance. Microorganisms 2022; 10:microorganisms10020303. [PMID: 35208758 PMCID: PMC8879831 DOI: 10.3390/microorganisms10020303] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
A biofilm is a community of stable microorganisms encapsulated in an extracellular matrix produced by themselves. Many types of microorganisms that are found on living hosts or in the environment can form biofilms. These include pathogenic bacteria that can serve as a reservoir for persistent infections, and are culpable for leading to a broad spectrum of chronic illnesses and emergence of antibiotic resistance making them difficult to be treated. The absence of biofilm-targeting antibiotics in the drug discovery pipeline indicates an unmet opportunity for designing new biofilm inhibitors as antimicrobial agents using various strategies and targeting distinct stages of biofilm formation. The strategies available to control biofilm formation include targeting the enzymes and proteins specific to the microorganism and those involved in the adhesion pathways leading to formation of resistant biofilms. This review primarily focuses on the recent strategies and advances responsible for identifying a myriad of antibiofilm agents and their mechanism of biofilm inhibition, including extracellular polymeric substance synthesis inhibitors, adhesion inhibitors, quorum sensing inhibitors, efflux pump inhibitors, and cyclic diguanylate inhibitors. Furthermore, we present the structure–activity relationships (SAR) of these agents, including recently discovered biofilm inhibitors, nature-derived bioactive scaffolds, synthetic small molecules, antimicrobial peptides, bioactive compounds isolated from fungi, non-proteinogenic amino acids and antibiotics. We hope to fuel interest and focus research efforts on the development of agents targeting the uniquely complex, physical and chemical heterogeneous biofilms through a multipronged approach and combinatorial therapeutics for a more effective control and management of biofilms across diseases.
Collapse
Affiliation(s)
- Sahaya Nadar
- Department of Pharmaceutical Chemistry, St. John Institute of Pharmacy and Research, Mumbai 400056, India;
| | - Tabassum Khan
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, India;
| | - Simon G. Patching
- School of Biomedical Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK
- Correspondence: or (S.G.P.); (A.O.)
| | - Abdelwahab Omri
- The Novel Drug & Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Correspondence: or (S.G.P.); (A.O.)
| |
Collapse
|
41
|
Farah N, Chin VK, Chong PP, Lim WF, Lim CW, Basir R, Chang SK, Lee TY. Riboflavin as a promising antimicrobial agent? A multi-perspective review. CURRENT RESEARCH IN MICROBIAL SCIENCES 2022; 3:100111. [PMID: 35199072 PMCID: PMC8848291 DOI: 10.1016/j.crmicr.2022.100111] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 02/05/2022] [Accepted: 02/07/2022] [Indexed: 12/29/2022] Open
Abstract
Riboflavin demonstrates antioxidant and photosensitizing properties. Riboflavin is able to induce ROS and modulate immune response. Riboflavin possesses potent antimicrobial activity when used alone or combined with other anti-infectives. The riboflavin biosynthesis pathway serves as an ideal drug target against microbes. UVA combination with riboflavin exhibits remarkable antimicrobial effects.
Riboflavin, or more commonly known as vitamin B2, forms part of the component of vitamin B complex. Riboflavin consisting of two important cofactors, flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD), which are involved in multiple oxidative-reduction processes and energy metabolism. Besides maintaining human health, different sources reported that riboflavin can inhibit or inactivate the growth of different pathogens including bacteria, viruses, fungi and parasites, highlighting the possible role of riboflavin as an antimicrobial agent. Moreover, riboflavin and flavins could produce reactive oxygen species (ROS) when exposed to light, inducing oxidative damage in cells and tissues, and thus are excellent natural photosensitizers. Several studies have illustrated the therapeutic efficacy of photoactivated riboflavin against nosocomial infections and multidrug resistant bacterial infections as well as microbial associated biofilm infections, revealing the potential role of riboflavin as a promising antimicrobial candidate, which could serve as one of the alternatives in fighting the global crisis of the emergence of antimicrobial resistance seen in different pathogenic microbes. Riboflavin could also be involved in modulating host immune responses, which might increase the pathogen clearance from host cells and increase host defense against microbial infections. Thus, the dual effects of riboflavin on both pathogens and host immunity, reflected by its potent bactericidal effect and alleviation of inflammation in host cells further imply that riboflavin could be a potential candidate for therapeutic intervention in resolving microbial infections. Hence, this review aimed to provide some insights on the promising role of riboflavin as an antimicrobial candidate and also a host immune-modulator from a multi-perspective view as well as to discuss the application and challenges on using riboflavin in photodynamic therapy against various pathogens and microbial biofilm-associated infections.
Collapse
Affiliation(s)
- Nuratiqah Farah
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, UPM, 43400, Serdang, Selangor, Malaysia
| | - Voon Kin Chin
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, UPM, 43400, Serdang, Selangor, Malaysia
| | - Pei Pei Chong
- School of Biosciences, Taylor's University, No 1, Jalan Taylor's, 47500 Subang Jaya, Selangor, Malaysia
| | - Wai Feng Lim
- Integrative Pharmacogenomics Institute (iPROMISE), Universiti Teknologi MARA, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia
| | - Chee Woei Lim
- Department of Medicine, Faculty of Medicine and Health Sciences, UPM, 43400, Serdang, Selangor, Malaysia
| | - Rusliza Basir
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, UPM, 43400, Serdang, Selangor, Malaysia
| | - Sui Kiat Chang
- Department of Horticulture, Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, Key Laboratory of Post-Harvest Handling of Fruits, Ministry of Agriculture. South China Botanical Garden, Chinese Academy of Sciences. Guangzhou, 510650 China
| | - Tze Yan Lee
- Perdana University School of Liberal Arts, Science and Technology (PUScLST), Suite 9.2, 9th Floor, Wisma Chase Perdana, Changkat Semantan, Damansara Heights, 50490 Kuala Lumpur, Malaysia
- Corresponding author.
| |
Collapse
|
42
|
Asghar S, Khan IU, Salman S, Khalid SH, Ashfaq R, Vandamme TF. Plant-derived nanotherapeutic systems to counter the overgrowing threat of resistant microbes and biofilms. Adv Drug Deliv Rev 2021; 179:114019. [PMID: 34699940 DOI: 10.1016/j.addr.2021.114019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/03/2021] [Accepted: 10/19/2021] [Indexed: 12/17/2022]
Abstract
Since antiquity, the survival of human civilization has always been threatened by the microbial infections. An alarming surge in the resistant microbial strains against the conventional drugs is quite evident in the preceding years. Furthermore, failure of currently available regimens of antibiotics has been highlighted by the emerging threat of biofilms in the community and hospital settings. Biofilms are complex dynamic composites rich in extracellular polysaccharides and DNA, supporting plethora of symbiotic microbial life forms, that can grow on both living and non-living surfaces. These enforced structures are impervious to the drugs and lead to spread of recurrent and non-treatable infections. There is a strong realization among the scientists and healthcare providers to work out alternative strategies to combat the issue of drug resistance and biofilms. Plants are a traditional but rich source of effective antimicrobials with wider spectrum due to presence of multiple constituents in perfect synergy. Other than the biocompatibility and the safety profile, these phytochemicals have been repeatedly proven to overcome the non-responsiveness of resistant microbes and films via multiple pathways such as blocking the efflux pumps, better penetration across the cell membranes or biofilms, and anti-adhesive properties. However, the unfavorable physicochemical attributes and stability issues of these phytochemicals have hampered their commercialization. These issues of the phytochemicals can be solved by designing suitably constructed nanoscaled structures. Nanosized systems can not only improve the physicochemical features of the encapsulated payloads but can also enhance their pharmacokinetic and therapeutic profile. This review encompasses why and how various types of phytochemicals and their nanosized preparations counter the microbial resistance and the biofouling. We believe that phytochemical in tandem with nanotechnological innovations can be employed to defeat the microbial resistance and biofilms. This review will help in better understanding of the challenges associated with developing such platforms and their future prospects.
Collapse
|
43
|
Belardinelli JM, Li W, Avanzi C, Angala SK, Lian E, Wiersma CJ, Palčeková Z, Martin KH, Angala B, de Moura VCN, Kerns C, Jones V, Gonzalez-Juarrero M, Davidson RM, Nick JA, Borlee BR, Jackson M. Unique Features of Mycobacterium abscessus Biofilms Formed in Synthetic Cystic Fibrosis Medium. Front Microbiol 2021; 12:743126. [PMID: 34777289 PMCID: PMC8586431 DOI: 10.3389/fmicb.2021.743126] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 09/28/2021] [Indexed: 12/04/2022] Open
Abstract
Characterizing Mycobacterium abscessus complex (MABSC) biofilms under host-relevant conditions is essential to the design of informed therapeutic strategies targeted to this persistent, drug-tolerant, population of extracellular bacilli. Using synthetic cystic fibrosis medium (SCFM) which we previously reported to closely mimic the conditions encountered by MABSC in actual cystic fibrosis (CF) sputum and a new model of biofilm formation, we show that MABSC biofilms formed under these conditions are substantially different from previously reported biofilms grown in standard laboratory media in terms of their composition, gene expression profile and stress response. Extracellular DNA (eDNA), mannose-and glucose-containing glycans and phospholipids, rather than proteins and mycolic acids, were revealed as key extracellular matrix (ECM) constituents holding clusters of bacilli together. None of the environmental cues previously reported to impact biofilm development had any significant effect on SCFM-grown biofilms, most likely reflecting the fact that SCFM is a nutrient-rich environment in which MABSC finds a variety of ways of coping with stresses. Finally, molecular determinants were identified that may represent attractive new targets for the development of adjunct therapeutics targeting MABSC biofilms in persons with CF.
Collapse
Affiliation(s)
- Juan M Belardinelli
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Wei Li
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Charlotte Avanzi
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Shiva K Angala
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Elena Lian
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Crystal J Wiersma
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Zuzana Palčeková
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Kevin H Martin
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Bhanupriya Angala
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Vinicius C N de Moura
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Callan Kerns
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Victoria Jones
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Mercedes Gonzalez-Juarrero
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Rebecca M Davidson
- Center for Genes, Environment, and Health, National Jewish Health, Denver, CO, United States
| | - Jerry A Nick
- Department of Medicine, National Jewish Health, Denver, CO, United States.,Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Bradley R Borlee
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Mary Jackson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
44
|
Lin J, Xu L, Yang J, Wang Z, Shen X. Beyond dueling: roles of the type VI secretion system in microbiome modulation, pathogenesis and stress resistance. STRESS BIOLOGY 2021; 1:11. [PMID: 37676535 PMCID: PMC10441901 DOI: 10.1007/s44154-021-00008-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 08/09/2021] [Indexed: 09/08/2023]
Abstract
Bacteria inhabit diverse and dynamic environments, where nutrients may be limited and toxic chemicals can be prevalent. To adapt to these stressful conditions, bacteria have evolved specialized protein secretion systems, such as the type VI secretion system (T6SS) to facilitate their survival. As a molecular syringe, the T6SS expels various effectors into neighboring bacterial cells, eukaryotic cells, or the extracellular environment. These effectors improve the competitive fitness and environmental adaption of bacterial cells. Although primarily recognized as antibacterial weapons, recent studies have demonstrated that T6SSs have functions beyond interspecies competition. Here, we summarize recent research on the role of T6SSs in microbiome modulation, pathogenesis, and stress resistance.
Collapse
Affiliation(s)
- Jinshui Lin
- Shaanxi Key Laboratory of Chinese Jujube, College of Life Sciences, Yan'an University, Yan'an, Shaanxi, 716000, People's Republic of China
| | - Lei Xu
- State Key Laboratory of Crop Stress Biology for Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China
| | - Jianshe Yang
- Shaanxi Key Laboratory of Chinese Jujube, College of Life Sciences, Yan'an University, Yan'an, Shaanxi, 716000, People's Republic of China
| | - Zhuo Wang
- State Key Laboratory of Crop Stress Biology for Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China
| | - Xihui Shen
- State Key Laboratory of Crop Stress Biology for Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China.
| |
Collapse
|
45
|
Kasza K, Gurnani P, Hardie KR, Cámara M, Alexander C. Challenges and solutions in polymer drug delivery for bacterial biofilm treatment: A tissue-by-tissue account. Adv Drug Deliv Rev 2021; 178:113973. [PMID: 34530014 DOI: 10.1016/j.addr.2021.113973] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/12/2021] [Accepted: 09/08/2021] [Indexed: 02/07/2023]
Abstract
To tackle the emerging antibiotic resistance crisis, novel antimicrobial approaches are urgently needed. Bacterial communities (biofilms) are a particular concern in this context. Biofilms are responsible for most human infections and are inherently less susceptible to antibiotic treatments. Biofilms have been linked with several challenging chronic diseases, including implant-associated osteomyelitis and chronic wounds. The specific local environments present in the infected tissues further contribute to the rise in antibiotic resistance by limiting the efficacy of systemic antibiotic therapies and reducing drug concentrations at the infection site, which can lead to reoccurring infections. To overcome the shortcomings of systemic drug delivery, encapsulation within polymeric carriers has been shown to enhance antimicrobial efficacy, permeation and retention at the infection site. In this Review, we present an overview of current strategies for antimicrobial encapsulation within polymeric carriers, comparing challenges and solutions on a tissue-by-tissue basis. We compare challenges and proposed drug delivery solutions from the perspective of the local environments for biofilms found in oral, wound, gastric, urinary tract, bone, pulmonary, vaginal, ocular and middle/inner ear tissues. We will also discuss future challenges and barriers to clinical translation for these therapeutics. The following Review demonstrates there is a significant imbalance between the research focus being placed on different tissue types, with some targets (oral and wound biofims) being extensively more studied than others (vaginal and otitis media biofilms and endocarditis). Furthermore, the importance of the local tissue environment when selecting target therapies is demonstrated, with some materials being optimal choices for certain sites of bacterial infection, while having limited applicability in others.
Collapse
|
46
|
Evaluating Metabolic Pathways and Biofilm Formation in Stenotrophomonas maltophilia. J Bacteriol 2021; 204:e0039821. [PMID: 34633868 DOI: 10.1128/jb.00398-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Stenotrophomonas maltophilia has recently arisen as a prominent nosocomial pathogen because of its high antimicrobial resistance and ability to cause chronic respiratory infections. Often the infections are worsened by biofilm formation which enhances antibiotic tolerance. We have previously found that mutation of the gpmA gene, encoding the glycolytic enzyme phosphoglycerate mutase, impacts the formation of this biofilm on biotic and abiotic surfaces at early timepoints. This finding, indicating an association between carbon source and biofilm formation, led us to hypothesize that metabolism would influence S. maltophilia biofilm formation and planktonic growth. In the present study, we tested the impact of various growth substrates on biofilm levels and growth kinetics to determine metabolic requirements for these processes. We found that S. maltophilia wildtype preferred amino acids versus glucose for planktonic and biofilm growth and that gpmA deletion inhibited growth in amino acids. Furthermore, supplementation of the ΔgpmA strain by glucose or ribose phenotypically complemented growth defects. These results suggest that S. maltophilia shuttles amino acid carbon through gluconeogenesis to an undefined metabolic pathway supporting planktonic and biofilm growth. Further evaluation of these metabolic pathways might reveal novel metabolic activities of this pathogen. Importance Stenotrophomonas maltophilia is a prominent opportunistic pathogen that often forms biofilms during infection. However, the molecular mechanisms of virulence and biofilm formation are poorly understood. The glycolytic enzyme phosphoglycerate mutase appears to play a role in biofilm formation, and we used a mutant in its gene (gpmA) to probe the metabolic circuitry potentially involved in biofilm development. The results of our study indicate that S. maltophilia displays unique metabolic activities, which could be exploited for inhibiting growth and biofilm formation of this pathogen.
Collapse
|
47
|
Barnes AMT, Frank KL, Dunny GM. Enterococcal Endocarditis: Hiding in Plain Sight. Front Cell Infect Microbiol 2021; 11:722482. [PMID: 34527603 PMCID: PMC8435889 DOI: 10.3389/fcimb.2021.722482] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
Enterococcus faecalis is a major opportunistic bacterial pathogen of increasing clinical relevance. A substantial body of experimental evidence suggests that early biofilm formation plays a critical role in these infections, as well as in colonization and persistence in the GI tract as a commensal member of the microbiome in most terrestrial animals. Animal models of experimental endocarditis generally involve inducing mechanical valve damage by cardiac catheterization prior to infection, and it has long been presumed that endocarditis vegetation formation resulting from bacterial attachment to the endocardial endothelium requires some pre-existing tissue damage. Here we review both historical and contemporary animal model studies demonstrating the robust ability of E. faecalis to directly attach and form stable microcolony biofilms encased within a bacterially-derived extracellular matrix on the undamaged endovascular endothelial surface. We also discuss the morphological similarities when these biofilms form on other host tissues, including when E. faecalis colonizes the GI epithelium as a commensal member of the normal vertebrate microbiome - hiding in plain sight where it can serve as a source for systemic infection via translocation. We propose that these phenotypes may allow the organism to persist as an undetected infection in asymptomatic individuals and thus provide an infectious reservoir for later clinical endocarditis.
Collapse
Affiliation(s)
- Aaron M. T. Barnes
- Department of Microbiology and Immunology, University of Minnesota School of Medicine, Minneapolis, MN, United States
- Department of Laboratory Medicine and Pathology, University of Minnesota School of Medicine, Minneapolis, MN, United States
| | - Kristi L. Frank
- Department of Microbiology and Immunology, University of Minnesota School of Medicine, Minneapolis, MN, United States
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Gary M. Dunny
- Department of Microbiology and Immunology, University of Minnesota School of Medicine, Minneapolis, MN, United States
| |
Collapse
|
48
|
Targeting antibiotic tolerance in anaerobic biofilms associated with oral diseases: Human antimicrobial peptides LL-37 and lactoferricin enhance the antibiotic efficacy of amoxicillin, clindamycin and metronidazole. Anaerobe 2021; 71:102439. [PMID: 34454095 DOI: 10.1016/j.anaerobe.2021.102439] [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: 06/02/2021] [Revised: 07/27/2021] [Accepted: 08/22/2021] [Indexed: 11/23/2022]
Abstract
Antimicrobial peptides are receiving increasing attention as potential therapeutic agents for treating biofilm-related infections of the oral cavity. Many bacteria residing in biofilms exhibit an enhanced antibiotic tolerance, which grants intrinsically susceptible microorganisms to survive lethal concentrations of antibiotics. In this study, we examined the effects of two endogenous human antimicrobial peptides, LL-37 and human Lactoferricin, on the antibiotic drug efficacy of amoxicillin, clindamycin and metronidazole in two types of polymicrobial biofilms, which aimed to represent frequent oral diseases: (1) facultative anaerobic (Streptococcus mutans, Streptococcus sanguinis, Actinomyces naeslundii) and (2) obligate anaerobic biofilms (Veillonella parvula, Parvimonas micra, Fusobacterium nucleatum). LL-37 and Lactoferricin enhanced the anti-biofilm effect of amoxicillin and clindamycin in facultative anaerobic biofilms. Metronidazole alone was ineffective against facultative anaerobic biofilms, but the presence of LL-37 and Lactoferricin led to a greater biofilm reduction. Obligate anaerobic biofilms showed an increased drug tolerance to amoxicillin and clindamycin, presumably due to metabolic downshifts of the bacteria residing within the biofilm. However, when combined with LL-37 or Lactoferricin, the reduction of obligate anaerobic biofilms was markedly enhanced for all antibiotics, even for amoxicillin and clindamycin. Furthermore, our results suggest that antimicrobial peptides enhance the dispersion of matured biofilms, which may be one of their mechanisms for targeting biofilms. In summary, our study proves that antimicrobial peptides can serve as an auxiliary treatment strategy for combatting enhanced antibiotic tolerance in bacterial biofilms.
Collapse
|
49
|
Birlutiu RM, Mihalache M, Mihalache P, Cismasiu RS, Birlutiu V. Mid-term follow-up results after implementing a new strategy for the diagnosis and management of periprosthetic joint infections. BMC Infect Dis 2021; 21:807. [PMID: 34384360 PMCID: PMC8361652 DOI: 10.1186/s12879-021-06407-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/20/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Periprosthetic joint infections (PJIs) represent one of the most serious complications associated with joint replacement surgeries, a complication also of modern orthopedic surgery despite the efforts that occurred in this field. Frequently PJIs lead to prolonged morbidity, increased costs and mortality. METHODS We are conducting a single-center observational cohort ongoing study in the Academic Emergency Hospital Sibiu, Romania, study in which sonication of the retrieved and as a rapid method of bacteria detection, molecular identification of bacteria by 16S rRNA beacon-based fluorescent in situ hybridization (bbFISH) are used. RESULTS A total of 61 patients were enrolled in this study. The diagnosis of aseptic loosening was established in 30 cases (49.1%) and the diagnosis of periprosthetic joint infection was established at 31 patients (50.8%). The mean follow-up period in the subgroup of patients diagnosed with periprosthetic joint infections was 36.06 ± 12.59 months (range: 1-54). The 25-months Kaplan-Meier survival rate as the end point, as a consequence of the period of enrollment and a different follow-up period for each type of surgical procedure, was 75% after debridement and implant retention, 91.7% after one-stage exchange, 92.3% after two-stage exchange, and 100% after three-stage exchange. There were no significant differences in survival percentage. CONCLUSIONS Our study has good results similar to previously published data. We cannot recommend one strategy of managing prosthetic joint infections over the other. Definitely, there is a need for prospective randomized controlled trials.
Collapse
Affiliation(s)
- Rares Mircea Birlutiu
- Lucian Blaga University of Sibiu, Faculty of Medicine Sibiu, Str. Lucian Blaga, Nr. 2A, 550169, Sibiu, Romania.
| | - Manuela Mihalache
- Lucian Blaga University of Sibiu, Faculty of Medicine Sibiu, Str. Lucian Blaga, Nr. 2A, 550169, Sibiu, Romania
| | - Patricia Mihalache
- Lucian Blaga University of Sibiu, Bd-ul. Victoriei, Nr.10, 550024, Sibiu, Romania
| | - Razvan Silviu Cismasiu
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania.,FOISOR Clinical Hospital of Orthopedics, Traumatology, and Osteoarticular TB Bucharest, Str. Dionisie Lupu nr. 37, Sector 2, 020021, Bucharest, Romania
| | - Victoria Birlutiu
- Lucian Blaga University of Sibiu, Faculty of Medicine Sibiu, Str. Lucian Blaga, Nr. 2A, 550169, Sibiu, Romania.,Academic Emergency Hospital Sibiu - Infectious Diseases Clinic, B-dul Corneliu Coposu, Nr.2-4, 550245, Sibiu, Romania
| |
Collapse
|
50
|
Rancan F, Jurisch J, Günday C, Türeli E, Blume-Peytavi U, Vogt A, Schaudinn C, Günday-Türeli N. Screening of Surfactants for Improved Delivery of Antimicrobials and Poly-Lactic- co-Glycolic Acid Particles in Wound Tissue. Pharmaceutics 2021; 13:1093. [PMID: 34371785 PMCID: PMC8308990 DOI: 10.3390/pharmaceutics13071093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/07/2021] [Accepted: 07/14/2021] [Indexed: 01/14/2023] Open
Abstract
Topical wound management is often a challenge due to the poor penetration of antimicrobials in wound tissue and across the biofilm matrix where bacteria are embedded. Surfactants have been used for decades to improve the stability of formulations, increase drug solubility, and enhance penetration. In this study, we screened different detergents with respect to their cytotoxicity and their ability to improve the penetration of poly-lactic-co-glycolic acid (PLGA) particles in wound tissue. Among the tested surfactants, Kolliphor SLS and Tween 80 increased the penetration of PLGA particles and had a limited cytotoxicity. Then, these surfactants were used to formulate PLGA particles loaded with the poorly water-soluble antibiotic ciprofloxacin. The antimicrobial efficacy of the formulations was tested in a wound infection model based on human ex vivo skin. We found that even though PLGA particles had the same antimicrobial efficiency than the particle-free drug formulation, thanks to their solubilizing and anti-biofilm properties, the surfactants remarkably improved the antimicrobial activity of ciprofloxacin with respect to the drug formulation in water. We conclude that the use of Tween 80 in antimicrobial formulations might be a safe and efficient option to improve the topical antimicrobial management of chronic wound infections.
Collapse
Affiliation(s)
- Fiorenza Rancan
- Research Center for Hair and Skin Science, Department of Dermatology and Allergy, Charité–Universitätsmedizin Berlin 10117, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (J.J.); (U.B.-P.); (A.V.)
| | - Jana Jurisch
- Research Center for Hair and Skin Science, Department of Dermatology and Allergy, Charité–Universitätsmedizin Berlin 10117, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (J.J.); (U.B.-P.); (A.V.)
| | - Cemre Günday
- MyBiotech, 66802 Überherrn, Germany; (C.G.); (E.T.); (N.G.-T.)
| | - Emre Türeli
- MyBiotech, 66802 Überherrn, Germany; (C.G.); (E.T.); (N.G.-T.)
| | - Ulrike Blume-Peytavi
- Research Center for Hair and Skin Science, Department of Dermatology and Allergy, Charité–Universitätsmedizin Berlin 10117, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (J.J.); (U.B.-P.); (A.V.)
| | - Annika Vogt
- Research Center for Hair and Skin Science, Department of Dermatology and Allergy, Charité–Universitätsmedizin Berlin 10117, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (J.J.); (U.B.-P.); (A.V.)
| | - Christoph Schaudinn
- Advanced Light and Electron Microscopy (Zentrum für Biologische Gefahren und Spezielle Pathogene 4), Robert Koch Institute, 13353 Berlin, Germany;
| | | |
Collapse
|