151
|
Isolation, Characterization, and Antibiofilm Activity of Pigments Synthesized by Rhodococcus sp. SC1. Curr Microbiol 2021; 79:15. [PMID: 34905097 DOI: 10.1007/s00284-021-02694-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 10/12/2021] [Indexed: 10/19/2022]
Abstract
Infections from multi-drug resistant bacteria and biofilms constitute a serious problem worldwide. There is a need for new antibacterial and antibiofilm compounds in the fight against infectious diseases. In recent years, pigment-producing microorganisms have drawn a great deal of attention as a promising source for antibacterial and antibiofilm compounds. Here, we report the antibacterial and antibiofilm activity of pigments synthesized by bacteria isolated from soil. This study aimed to perform an evaluation of the antibacterial, antibiofilm, and characteristic of crude pigments from Rhodococcus sp. SC1 isolates. The total pigment extract exhibited antibacterial activity against Gram-positive and Gram-negative reference bacteria with required minimum inhibitory concentration (MIC) values ranging from 64 to 256 µg/ml. Moreover, it reduced biofilm formation of Gram-negative reference bacteria at sub-MIC concentration. For characterization of the pigments, UV-absorbance, thin layer chromatography, fourier transform infrared spectroscopy, and QTOF-LC/MS analyses were performed. The results of this study showed that pigments of Rhodococcus sp. SC1 isolates can be a candidate for medical applications.
Collapse
|
152
|
Mohamed MA, Nasr M, Elkhatib WF, Eltayeb WN, Elshamy AA, El-Sayyad GS. Nanobiotic formulations as promising advances for combating MRSA resistance: susceptibilities and post-antibiotic effects of clindamycin, doxycycline, and linezolid. RSC Adv 2021; 11:39696-39706. [PMID: 35494109 PMCID: PMC9044563 DOI: 10.1039/d1ra08639a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 12/08/2021] [Indexed: 12/15/2022] Open
Abstract
Antimicrobial activity and post-antibiotic effects (PAEs) are both important parameters in determination of the dosage regimen of antimicrobial agents. In the present study, antimicrobial activity and PAEs of clindamycin, doxycycline, linezolid, and their nanobiotic formulations were evaluated against two methicillin resistant Staphylococcus aureus clinical isolates (MRSA) encoded (MRSA-S1 and MRSA-S2). Nanobiotic formulations increased the susceptibility of MRSA isolates by 4-64 folds as compared to their conventional ones. The PAE values were determined after exposure of MRSA isolates for 1 h to 10× the MICs of the tested antibiotics. The duration of PAEs were recorded after bacterial growth in Mueller Hinton broth (MHB) free from antibiotic has been restored. The PAE values for MRSA-S1 were 2.5 h for the conventional antibiotics. However, the PAEs for nanobiotics were 4 h for both clindamycin and linezolid, while 3 h for doxycycline. For MRSA-S2, linezolid and linezolid nanobiotics PAEs were 3 h. PAEs of clindamycin and clindamycin nanobiotics were 3.75 h and 4 h, respectively. Doxycycline and doxycycline nanobiotics revealed the same PAEs patterns of 3.5 h. The findings of the current study may positively influence the pharmacodynamics of the antibiotics and consequently the dosage regimen of nanobiotics as well as on their clinical outcome.
Collapse
Affiliation(s)
- Mennatallah A Mohamed
- Microbiology Department, Faculty of Pharmacy, Misr International University Cairo 19648 Egypt
| | - Maha Nasr
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Ain Shams University, African Union Organization St. Abbassia Cairo 11566 Egypt
| | - Walid F Elkhatib
- Microbiology and Immunology Department, Faculty of Pharmacy, Ain Shams University, African Union Organization St. Abbassia Cairo 11566 Egypt +20-2-24051107 +20-2-24051120
- Department of Microbiology & Immunology, Faculty of Pharmacy, Galala University New Galala City Suez Egypt
| | - Wafaa N Eltayeb
- Microbiology Department, Faculty of Pharmacy, Misr International University Cairo 19648 Egypt
| | - Aliaa A Elshamy
- Microbiology and Public Health Department, Faculty of Pharmacy and Drug Technology, Heliopolis University for Sustainable Development Cairo Belbes Road Cairo 11788 Egypt
| | - Gharieb S El-Sayyad
- Department of Microbiology & Immunology, Faculty of Pharmacy, Galala University New Galala City Suez Egypt
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA) Cairo Egypt +20-2-22749298 +20-2-22727413
| |
Collapse
|
153
|
Dong X, Overton CM, Tang Y, Darby JP, Sun YP, Yang L. Visible Light-Activated Carbon Dots for Inhibiting Biofilm Formation and Inactivating Biofilm-Associated Bacterial Cells. Front Bioeng Biotechnol 2021; 9:786077. [PMID: 34869296 PMCID: PMC8637124 DOI: 10.3389/fbioe.2021.786077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 10/19/2021] [Indexed: 11/13/2022] Open
Abstract
This study aimed to address the significant problems of bacterial biofilms found in medical fields and many industries. It explores the potential of classic photoactive carbon dots (CDots), with 2,2′-(ethylenedioxy)bis (ethylamine) (EDA) for dot surface functionalization (thus, EDA-CDots) for their inhibitory effect on B. subtilis biofilm formation and the inactivation of B. subtilis cells within established biofilm. The EDA-CDots were synthesized by chemical functionalization of selected small carbon nanoparticles with EDA molecules in amidation reactions. The inhibitory efficacy of CDots with visible light against biofilm formation was dependent significantly on the time point when CDots were added; the earlier the CDots were added, the better the inhibitory effect on the biofilm formation. The evaluation of antibacterial action of light-activated EDA-CDots against planktonic B. subtilis cells versus the cells in biofilm indicate that CDots are highly effective for inactivating planktonic cells but barely inactivate cells in established biofilms. However, when coupling with chelating agents (e.g., EDTA) to target the biofilm architecture by breaking or weakening the EPS protection, much enhanced photoinactivation of biofilm-associated cells by CDots was achieved. The study demonstrates the potential of CDots to prevent the initiation of biofilm formation and to inhibit biofilm growth at an early stage. Strategic combination treatment could enhance the effectiveness of photoinactivation by CDots to biofilm-associated cells.
Collapse
Affiliation(s)
- Xiuli Dong
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC, United States
| | | | - Yongan Tang
- Department of Mathematics and Physics, North Carolina Central University, Durham, NC, United States
| | - Jasmine P Darby
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC, United States
| | - Ya-Ping Sun
- Department of Chemistry, Clemson University, Clemson, SC, United States
| | - Liju Yang
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC, United States
| |
Collapse
|
154
|
Cruz A, Condinho M, Carvalho B, Arraiano CM, Pobre V, Pinto SN. The Two Weapons against Bacterial Biofilms: Detection and Treatment. Antibiotics (Basel) 2021; 10:1482. [PMID: 34943694 PMCID: PMC8698905 DOI: 10.3390/antibiotics10121482] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 11/29/2021] [Accepted: 12/01/2021] [Indexed: 12/12/2022] Open
Abstract
Bacterial biofilms are defined as complex aggregates of bacteria that grow attached to surfaces or are associated with interfaces. Bacteria within biofilms are embedded in a self-produced extracellular matrix made of polysaccharides, nucleic acids, and proteins. It is recognized that bacterial biofilms are responsible for the majority of microbial infections that occur in the human body, and that biofilm-related infections are extremely difficult to treat. This is related with the fact that microbial cells in biofilms exhibit increased resistance levels to antibiotics in comparison with planktonic (free-floating) cells. In the last years, the introduction into the market of novel compounds that can overcome the resistance to antimicrobial agents associated with biofilm infection has slowed down. If this situation is not altered, millions of lives are at risk, and this will also strongly affect the world economy. As such, research into the identification and eradication of biofilms is important for the future of human health. In this sense, this article provides an overview of techniques developed to detect and imaging biofilms as well as recent strategies that can be applied to treat biofilms during the several biofilm formation steps.
Collapse
Affiliation(s)
- Adriana Cruz
- iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, University of Lisbon, Av. Rovisco Pais, 1049-001 Lisboa, Portugal;
- i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, University of Lisbon, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Manuel Condinho
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal; (M.C.); (B.C.); (C.M.A.)
| | - Beatriz Carvalho
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal; (M.C.); (B.C.); (C.M.A.)
| | - Cecília M. Arraiano
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal; (M.C.); (B.C.); (C.M.A.)
| | - Vânia Pobre
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal; (M.C.); (B.C.); (C.M.A.)
| | - Sandra N. Pinto
- iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, University of Lisbon, Av. Rovisco Pais, 1049-001 Lisboa, Portugal;
- i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, University of Lisbon, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| |
Collapse
|
155
|
Ink-jet 3D printing as a strategy for developing bespoke non-eluting biofilm resistant medical devices. Biomaterials 2021; 281:121350. [PMID: 35033903 PMCID: PMC7613459 DOI: 10.1016/j.biomaterials.2021.121350] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 12/27/2021] [Indexed: 11/20/2022]
Abstract
Chronic infection as a result of bacterial biofilm formation on implanted medical devices is a major global healthcare problem requiring new biocompatible, biofilm-resistant materials. Here we demonstrate how bespoke devices can be manufactured through ink-jet-based 3D printing using bacterial biofilm inhibiting formulations without the need for eluting antibiotics or coatings. Candidate monomers were formulated and their processability and reliability demonstrated. Formulations for in vivo evaluation of the 3D printed structures were selected on the basis of their in vitro bacterial biofilm inhibitory properties and lack of mammalian cell cytotoxicity. In vivo in a mouse implant infection model, Pseudomonas aeruginosa biofilm formation on poly-TCDMDA was reduced by ~99% when compared with medical grade silicone. Whole mouse bioluminescence imaging and tissue immunohistochemistry revealed the ability of the printed device to modulate host immune responses as well as preventing biofilm formation on the device and infection of the surrounding tissues. Since 3D printing can be used to manufacture devices for both prototyping and clinical use, the versatility of ink-jet based 3D-printing to create personalised functional medical devices is demonstrated by the biofilm resistance of both a finger joint prosthetic and a prostatic stent printed in poly-TCDMDA towards P. aeruginosa and Staphylococcus aureus.
Collapse
|
156
|
Duncker KE, Holmes ZA, You L. Engineered microbial consortia: strategies and applications. Microb Cell Fact 2021; 20:211. [PMID: 34784924 PMCID: PMC8597270 DOI: 10.1186/s12934-021-01699-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/23/2021] [Indexed: 11/10/2022] Open
Abstract
Many applications of microbial synthetic biology, such as metabolic engineering and biocomputing, are increasing in design complexity. Implementing complex tasks in single populations can be a challenge because large genetic circuits can be burdensome and difficult to optimize. To overcome these limitations, microbial consortia can be engineered to distribute complex tasks among multiple populations. Recent studies have made substantial progress in programming microbial consortia for both basic understanding and potential applications. Microbial consortia have been designed through diverse strategies, including programming mutualistic interactions, using programmed population control to prevent overgrowth of individual populations, and spatial segregation to reduce competition. Here, we highlight the role of microbial consortia in the advances of metabolic engineering, biofilm production for engineered living materials, biocomputing, and biosensing. Additionally, we discuss the challenges for future research in microbial consortia.
Collapse
Affiliation(s)
- Katherine E Duncker
- Department of Biomedical Engineering, Duke University, Durham, NC, 27705, USA
| | - Zachary A Holmes
- Department of Biomedical Engineering, Duke University, Durham, NC, 27705, USA
| | - Lingchong You
- Department of Biomedical Engineering, Duke University, Durham, NC, 27705, USA.
| |
Collapse
|
157
|
Current strategies in inhibiting biofilm formation for combating urinary tract infections: Special focus on peptides, nano-particles and phytochemicals. BIOCATALYSIS AND AGRICULTURAL BIOTECHNOLOGY 2021. [DOI: 10.1016/j.bcab.2021.102209] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
158
|
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
|
159
|
Tao J, Yan S, Zhou C, Liu Q, Zhu H, Wen Z. Total flavonoids from Potentilla kleiniana Wight et Arn inhibits biofilm formation and virulence factors production in methicillin-resistant Staphylococcus aureus (MRSA). JOURNAL OF ETHNOPHARMACOLOGY 2021; 279:114383. [PMID: 34214645 DOI: 10.1016/j.jep.2021.114383] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/22/2021] [Accepted: 06/28/2021] [Indexed: 06/13/2023]
Abstract
ETHANOPHARMACOLOGICAL RELEVANCE Potentilla kleiniana Wight et Arn is a wide-spread wild plant in the mountainous areas in southern China. The whole herb has been used as a traditional herbal medicine to treat fever, arthritis, malaria, insect and snake bites, hepatitis, and traumatic injury. In vitro studies have reported the antibacterial activity use of the plant in traditional medicinal systems. AIM OF THE STUDY The aim of this study was to investigate the inhibitory activity of total flavonoid from Potentilla kleiniana Wight et Arn (TFP) on methicillin-resistant Staphylococcus aureus (MRSA) in planktonic state and biofilm state. MATERIALS AND METHODS Antibacterial activities of TFP on planktonic MRSA were determined by agar diffusion method, microtiter plate assay and time-kill curve assay. Electrical conductivity, membrane permeability, membrane potential and autoaggregation were analyzed to study TFP effects on planktonic MRSA growth. Crystal violet (CV) staining and confocal laser scanning microscopy (CLSM) were analyzed to study TFP effects on aggregation and maturation of MRSA biofilm. After TFP treatment, extracellular polymeric substances (EPS) production were examined. Morphological changes in planktonic and MRSA biofilm following TFP treatment were determined with scanning electron microscopy (SEM) and transmission electron microscopy (TEM). Moreover, α-Toxin protein expression and adhesion-related gene expression were also determined. RESULTS The minimum inhibitory concentration (MIC) of TFP against MRSA was 20 μg/mL. The agar diffusion method and time-kill curve assay results indicated that TFP inhibited planktonic MRSA growth. TFP treatment significantly inhibited planktonic MRSA growth by inhibiting autoaggregation, α-hemolysin activity, α-Toxin protein expression, but increasing electrolyte leakage, membrane permeability and membrane potential and impacting cell structure. Moreover, TFP treatment significantly inhibited aggregation and maturation on MRSA biofilm by decreasing surface hydrophobicity, EPS production and adhesion-related gene expression. CONCLUSION The results of this trial provide scientific experimental data on the traditional use of Potentilla Kleiniana Wight et Arn for traumatic injury treatment and further demonstrate the potential of TFP to be developed as a novel anti-biofilm drug.
Collapse
Affiliation(s)
- Junyu Tao
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518000, China; Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, 518000, China; School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou, 521041, China
| | - Shilun Yan
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518000, China; Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, 518000, China; School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou, 521041, China
| | - Chuyue Zhou
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518000, China
| | - Qiong Liu
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518000, China
| | - Hui Zhu
- School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou, 521041, China
| | - Zhen Wen
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518000, China; School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou, 521041, China.
| |
Collapse
|
160
|
Oliva A, Stefani S, Venditti M, Di Domenico EG. Biofilm-Related Infections in Gram-Positive Bacteria and the Potential Role of the Long-Acting Agent Dalbavancin. Front Microbiol 2021; 12:749685. [PMID: 34745053 PMCID: PMC8569946 DOI: 10.3389/fmicb.2021.749685] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/04/2021] [Indexed: 01/08/2023] Open
Abstract
Infections caused by Gram-positive bacteria are a major public health problem due to their increasing resistance to antibiotics. Staphylococcus and Enterococcus species' resistance and pathogenicity are enhanced by their ability to form biofilm. The biofilm lifestyle represents a significant obstacle to treatment because bacterial cells become highly tolerant to a wide range of antimicrobial compounds normally effective against their planktonic forms. Thus, novel therapeutic strategies targeting biofilms are urgently needed. The lipoglycopeptide dalbavancin is a long-acting agent for treating acute bacterial skin and skin structure infections caused by a broad range of Gram-positive pathogens. Recent studies have shown promising activity of dalbavancin against Gram-positive biofilms, including methicillin-resistant S. aureus (MRSA), methicillin-resistant S. epidermidis (MRSE), and vancomycin-susceptible enterococci. This review outlines the mechanisms regulating biofilm development in Staphylococcus and Enterococcus species and the clinical impact of biofilm-related infections. In addition, it discusses the clinical implications and potential therapeutic perspectives of the long-acting drug dalbavancin against biofilm-forming Gram-positive pathogens.
Collapse
Affiliation(s)
- Alessandra Oliva
- Department of Public Health and Infectious Diseases, “La Sapienza” University of Rome, Rome, Italy
| | - Stefania Stefani
- Laboratory of Molecular Medical Microbiology and Antimicrobial Resistance Research (Mmarl), Department of Biomedical and Biotechnological Sciences (Biometec), University of Catania, Catania, Italy
| | - Mario Venditti
- Department of Public Health and Infectious Diseases, “La Sapienza” University of Rome, Rome, Italy
| | | |
Collapse
|
161
|
Abstract
Microbes are hardly seen as planktonic species and are most commonly found as biofilm communities in cases of chronic infections. Biofilms are regarded as a biological condition, where a large group of microorganisms gets adhered to a biotic or abiotic surface. In this context, Pseudomonas aeruginosa, a Gram-negative nosocomial pathogen is the main causative organism responsible for life-threatening and persistent infections in individuals affected with cystic fibrosis and other lung ailments. The bacteria can form a strong biofilm structure when it adheres to a surface suitable for the development of a biofilm matrix. These bacterial biofilms pose higher natural resistance to conventional antibiotic therapy due to their multiple tolerance mechanisms. This prevailing condition has led to an increasing rate of treatment failures associated with P. aeruginosa biofilm infections. A better understanding of the effect of a diverse group of antibiotics on established biofilms would be necessary to avoid inappropriate treatment strategies. Hence, the search for other alternative strategies as effective biofilm treatment options has become a growing area of research. The current review aims to give an overview of the mechanisms governing biofilm formation and the different strategies employed so far in the control of biofilm infections caused by P. aeruginosa. Moreover, this review can also help researchers to search for new antibiofilm agents to tackle the effect of biofilm infections that are currently imprudent to conventional antibiotics.
Collapse
|
162
|
Lopez AJ, Jones LM, Reynolds L, Diaz RC, George IK, Little W, Fleming D, D'souza A, Rennie MY, Rumbaugh KP, Smith AC. Detection of bacterial fluorescence from in vivo wound biofilms using a point-of-care fluorescence imaging device. Int Wound J 2021; 18:626-638. [PMID: 33565263 PMCID: PMC8450799 DOI: 10.1111/iwj.13564] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 12/15/2022] Open
Abstract
Wound biofilms must be identified to target disruption and bacterial eradication but are challenging to detect with standard clinical assessment. This study tested whether bacterial fluorescence imaging could detect porphyrin-producing bacteria within a biofilm using well-established in vivo models. Mouse wounds were inoculated on Day 0 with planktonic bacteria (n = 39, porphyrin-producing and non-porphyrin-producing species, 107 colony forming units (CFU)/wound) or with polymicrobial biofilms (n = 16, 3 biofilms per mouse, each with 1:1:1 parts Staphylococcus aureus/Escherichia coli/Enterobacter cloacae, 107 CFU/biofilm) that were grown in vitro. Mouse wounds inoculated with biofilm underwent fluorescence imaging up to Day 4 or 5. Wounds were then excised and sent for microbiological analysis. Bacteria-matrix interaction was assessed with scanning electron microscopy (SEM) and histopathology. A total of 48 hours after inoculation with planktonic bacteria or biofilm, red fluorescence was readily detected in wounds; red fluorescence intensified up to Day 4. Red fluorescence from biofilms persisted in excised wound tissue post-wash. SEM and histopathology confirmed bacteria-matrix interaction. This pre-clinical study is the first to demonstrate the fluorescence detection of bacterial biofilm in vivo using a point-of-care wound imaging device. These findings have implications for clinicians targeting biofilm and may facilitate improved visualisation and removal of biofilms.
Collapse
Affiliation(s)
- Andrea J. Lopez
- Department of Honors StudiesTexas Tech UniversityLubbockTexasUSA
| | | | - Landrye Reynolds
- Department of Honors StudiesTexas Tech UniversityLubbockTexasUSA
| | - Rachel C. Diaz
- Department of Honors StudiesTexas Tech UniversityLubbockTexasUSA
| | - Isaiah K. George
- Department of Honors StudiesTexas Tech UniversityLubbockTexasUSA
| | - William Little
- Department of Honors StudiesTexas Tech UniversityLubbockTexasUSA
| | - Derek Fleming
- Department of SurgeryTexas Tech University Health Sciences CenterLubbockTexasUSA
- Division of Clinical Microbiology, Department of Laboratory Medicine and PathologyMayo ClinicRochesterMinnesotaUSA
| | | | | | - Kendra P. Rumbaugh
- Department of SurgeryTexas Tech University Health Sciences CenterLubbockTexasUSA
| | | |
Collapse
|
163
|
Wang J, Zhu J, Meng J, Qiu T, Wang W, Wang R, Liu J. Baicalin inhibits biofilm formation by influencing primary adhesion and aggregation phases in Staphylococcus saprophyticus. Vet Microbiol 2021; 262:109242. [PMID: 34562786 DOI: 10.1016/j.vetmic.2021.109242] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/19/2021] [Indexed: 12/20/2022]
Abstract
The ability to form biofilms on surfaces makes Staphylococcus saprophyticus (S. saprophyticus) becomes the main pathogenic factor in nosocomial infections. Previously, we demonstrated that baicalin (Bac) inhibited azithromycin-resistant S. saprophyticus (ARSS) biofilm formation. This investigation aims to explore the influence of baicalin on primary adhesion and aggregation phases of biofilm formation, and the treatment effect of baicalin and azithromycin on ARSS biofilm-associated infection. Crystal violet (CV) staining and scanning electron microscope (SEM) observations clearly showed that sub-inhibitory concentration baicalin inhibited ARSS biofilm formation when baicalin was added before the adhesion and aggregation phases. Baicalin significantly increased the relative adhesion inhibition rate and decreased the rate of bacteria aggregation in a dose-dependent manner. Moreover, CLSM and cell lysis assays revealed that baicalin inhibited the production of surface proteins and cell autolysis in bacteria adhesion and aggregation phases of biofilm formation. Meanwhile, the relative expressions of adhesion-related and autolysis-related genes were down-regulated by baicalin. In vivo, the combination of baicalin and azithromycin succeeded in eradicating ARSS from the mouse cutaneous infection model and decreasing the pathological injuries, the expressions of cytokines in infected tissue, and the number of inflammatory cells in the blood. Simultaneously, baicalin decreased the bacterial burdens in tubes, the level of TNF-α, and the number of monocytes and neutrophils compared with that of the SS and azithromycin groups. Based on these results, baicalin inhibited the adhesion and aggregation phases of biofilm formation by influenced the production of surface proteins and cell autolysis. Baicalin and azithromycin synergetically treated ARSS biofilm-associated infection.
Collapse
Affiliation(s)
- Jinli Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Traditional Chinese Veterinary Medicine Research Center, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Jinyue Zhu
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Traditional Chinese Veterinary Medicine Research Center, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Jinwu Meng
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Traditional Chinese Veterinary Medicine Research Center, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Tianxin Qiu
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Traditional Chinese Veterinary Medicine Research Center, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Wenjia Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Traditional Chinese Veterinary Medicine Research Center, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Rui Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Traditional Chinese Veterinary Medicine Research Center, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Jiaguo Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Traditional Chinese Veterinary Medicine Research Center, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
164
|
Amissah F, Andey T, Ahlschwede KM. Nanotechnology-based therapies for the prevention and treatment of Streptococcus mutans-derived dental caries. J Oral Biosci 2021; 63:327-336. [PMID: 34536629 DOI: 10.1016/j.job.2021.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 09/03/2021] [Accepted: 09/07/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Dental caries results from long-term acid production when sugar is metabolized by a bacterial biofilm, resulting in a loss of calcium and phosphate from the enamel. Streptococcus mutans is a type of acid-producing bacteria and a virulent contributor to oral biofilms. Conventional treatment options, such as cefazolin and ampicillin, have significant levels of bacterial resistance. Other topical agents, such as fluoride, tend to be washed away by saliva, resulting in low therapeutic efficacy. HIGHLIGHT This review aims to highlight the solubility issues that plague poorly water-soluble therapeutic agents, various novel polymeric, and lipid-based nanotechnology systems that aim to improve the retention of therapeutic agents in the oral cavity. CONCLUSION In this review, different formulation types demonstrated improved therapeutic outcomes by enhancing drug solubility, promoting penetration into the deep layers of the biofilm, facilitating prolonged residence time in the buccal cavity, and reducing the emergence of drug-resistant phenotypes. These formulations have a strong potential to give new life to therapeutic agents that have limited physicochemical characteristics.
Collapse
Affiliation(s)
- Felix Amissah
- Department of Pharmaceutical Sciences, College of Pharmacy, Ferris State University, Big Rapids, MI, USA
| | - Terrick Andey
- Department of Pharmaceutical Sciences, School of Pharmacy, MCPHS University, Worcester, MA, USA
| | - Kristen M Ahlschwede
- Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.
| |
Collapse
|
165
|
Ysebaert C, Castado C, Mortier MC, Rioux S, Feron C, Di Paolo E, Weynants V, Blais N, Devos N, Hermand P. UspA2 is a cross-protective Moraxella catarrhalis vaccine antigen. Vaccine 2021; 39:5641-5649. [PMID: 34446318 DOI: 10.1016/j.vaccine.2021.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 11/29/2022]
Abstract
Moraxella catarrhalis (Mcat) is a key pathogen associated with exacerbations of chronic obstructive pulmonary disease (COPD) in adults and playing a significant role in otitis media in children. A vaccine would help to reduce the morbidity and mortality associated with these diseases. UspA2 is an Mcat surface antigen considered earlier as vaccine candidate before the interest in this molecule vanished due to sequence variability. However, the observation that some conserved domains are the target of bactericidal antibodies prompted us to reconsider UspA2 as a potential vaccine antigen. We first determined its prevalence among the COPD patients from the AERIS study, as the prevalence of UspA2 in a COPD-restricted population had yet to be documented. The gene was found in all Mcat isolates either as UspA2 or UspA2H variant. The percentage of UspA2H variant was higher than in any report so far, reaching 51%. A potential link between the role of UspA2H in biofilm formation and this high prevalence is discussed. To study further UspA2 as a vaccine antigen, recombinant UspA2 molecules were designed and used in animal models and bactericidal assays. We showed that UspA2 is immunogenic and that UspA2 immunization clears Mcat pulmonary challenge in a mouse model. In a serum bactericidal assay, anti-UspA2 antibodies generated in mice, guinea pigs or rabbits were able to kill Mcat strains of various origins, including a subset of isolates from the AERIS study, cross-reacting with UspA2H and even UspA1, a closely related Mcat surface protein. In conclusion, UspA2 is a cross-reactive Mcat antigen presenting the characteristics of a vaccine candidate.
Collapse
|
166
|
Goes A, Vidakovic L, Drescher K, Fuhrmann G. Interaction of myxobacteria-derived outer membrane vesicles with biofilms: antiadhesive and antibacterial effects. NANOSCALE 2021; 13:14287-14296. [PMID: 34477714 DOI: 10.1039/d1nr02583j] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Bacterial biofilms are widespread in nature and in medical settings and display a high tolerance to antibiotics and disinfectants. Extracellular vesicles have been increasingly studied to characterise their origins and assess their potential for use as a versatile drug delivery system; however, it remains unclear whether they also have antibiofilm effects. Outer membrane vesicles are lipid vesicles shed by Gram-negative bacteria and, in the case of myxobacteria, carry natural antimicrobial compounds produced by these microorganisms. In this study, we demonstrate that vesicles derived from the myxobacteria Cystobacter velatus Cbv34 and Cystobacter ferrugineus Cbfe23 are highly effective at inhibiting the formation and disrupting biofilms by different bacterial species.
Collapse
Affiliation(s)
- Adriely Goes
- Helmholtz Centre for Infection Research (HZI), Biogenic Nanotherapeutics Group (BION), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Campus E8.1, 66123 Saarbrücken, Germany.
| | | | | | | |
Collapse
|
167
|
Gupta S, Kumar P, Rathi B, Verma V, Dhanda RS, Devi P, Yadav M. Targeting of Uropathogenic Escherichia coli papG gene using CRISPR-dot nanocomplex reduced virulence of UPEC. Sci Rep 2021; 11:17801. [PMID: 34493749 PMCID: PMC8423837 DOI: 10.1038/s41598-021-97224-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023] Open
Abstract
Urinary tract infections (UTI) are the most common infectious diseases in the world. It is becoming increasingly tough to treat because of emergence of antibiotic resistance. So, there is an exigency to develop novel anti-virulence therapeutics to combat multi-drug resistance pathogenic strains. Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) discovery has revolutionized the gene editing technology for targeted approach. The greatest obstacle for CRISPR/Cas9 is cargo delivery systems and both viral and plasmid methods have disadvantages. Here, we report a highly efficient novel CRISPR based gene editing strategy, CRISPR-dots for targeting virulence factor Fimbrial Adhesion (papG gene), the bacterial adhesion molecule. Carbon quantum dots (CQD) were used as a delivery vehicle for Cas9 and gRNA into CFT073, a UPEC strain. CQDs were covalently conjugated to cas9 and papG-targeted guide RNA (gRNA) forming a nanocomplex CRISPR-dots (Cri-dots) as confirmed by DLS and transmission electron microscopy. Cri-dots-papG significantly targeted papG as demonstrated by decrease in the expression of papG.Further papG deficient UPEC had significantly reduced adherence ability and biofilm forming ability as demonstrated by fluorescence microscopy and scanning electron microscopy. Also, papG deficient UPEC had reduced virulence as shown by significantly increased survival of Caenorhabditis elegans (C. elegans) worms compared to UPEC. Our findings suggest that targeting of papG gene using Cri-dots nanocomplexes significantly reduced the pathogenicity of UPEC. Thus, Cri-dots nanocomplex offer a novel anti-bacterial strategy against multi-drug resistant UPEC.
Collapse
Affiliation(s)
- Surbhi Gupta
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, India
| | - Parveen Kumar
- Department of Urology, University of Alabama at Birmingham, Hugh Kaul Genetics Building, Birmingham, AL, USA
| | - Bhawna Rathi
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, India
| | - Vivek Verma
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, India
| | | | - Pooja Devi
- CSIR-Central Scientific Instruments Organisation, Sector-30C, Chandigarh, India
| | - Manisha Yadav
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, India.
- Department of Clinical Sciences, Lund University, Malmö, Sweden.
| |
Collapse
|
168
|
Oz Y, Nabawy A, Fedeli S, Gupta A, Huang R, Sanyal A, Rotello VM. Biodegradable Poly(lactic acid) Stabilized Nanoemulsions for the Treatment of Multidrug-Resistant Bacterial Biofilms. ACS APPLIED MATERIALS & INTERFACES 2021; 13:40325-40331. [PMID: 34416106 PMCID: PMC8573728 DOI: 10.1021/acsami.1c11265] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Biofilm infections caused by multidrug-resistant (MDR) bacteria are an urgent global health threat. Incorporation of natural essential oils into biodegradable oil-in-water cross-linked polymeric nanoemulsions (X-NEs) provides effective eradication of MDR bacterial biofilms. The X-NE platform combines the degradability of functionalized poly(lactic acid) polymers with the antimicrobial activity of carvacrol (from oregano oil). These X-NEs exhibited effective penetration and killing of biofilms formed by pathogenic bacteria. Biofilm-fibroblast coculture models demonstrate that X-NEs selectively eliminate bacteria without harming mammalian cells, making them promising candidates for antibiofilm therapeutics.
Collapse
Affiliation(s)
- Yavuz Oz
- Department of Chemistry, Bogazici University, Istanbul 34342, Turkey
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Ahmed Nabawy
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Stefano Fedeli
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Aarohi Gupta
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Rui Huang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Amitav Sanyal
- Department of Chemistry, Bogazici University, Istanbul 34342, Turkey
- Center for Life Sciences and Technologies, Bogazici University, Istanbul 34342, Turkey
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| |
Collapse
|
169
|
Kono M, Umar NK, Takeda S, Ohtani M, Murakami D, Sakatani H, Kaneko F, Nanushaj D, Hotomi M. Novel Antimicrobial Treatment Strategy Based on Drug Delivery Systems for Acute Otitis Media. Front Pharmacol 2021; 12:640514. [PMID: 34421583 PMCID: PMC8371970 DOI: 10.3389/fphar.2021.640514] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 07/22/2021] [Indexed: 12/27/2022] Open
Abstract
Despite tremendous success of pneumococcal conjugated vaccine and antimicrobial treatment by amoxicillin, acute otitis media (AOM) still remains as a great medical concern. Failure of antimicrobial treatment includes several factors. The middle ear cavity is surrounded by bone tissue, which makes it difficult to maintain sufficient concentration of antibiotics. Tympanic membrane of AOM patients thickens and actually becomes a barrier for topical therapy. This review discusses novel antimicrobial treatment strategies based on drug delivery systems (DDS) for AOM. To deliver drugs enough to kill the pathogenic bacteria without systemic side effects, the development of new antimicrobial treatment strategy applying innovative drug DDS has been expected. The sustained-release DDS can achieve sufficient time for antimicrobial concentrations to exceed minimum inhibitory concentration (MIC) for time-dependent antibiotics as well as enough maximum concentration for dose-dependent antibiotics to eradicate causative pathogens in the middle ear. The development of trans-tympanic membranes of DDS, such as hydrogels with chemical permeation enhancers (CPEs), is another attractive strategy. Phage is a promising strategy for developing DDS-based therapies. The DDS formulations enable antimicrobial treatment of AOM by a single dose and thus, an attractive future antimicrobial treatment for AOM.
Collapse
Affiliation(s)
- Masamitsu Kono
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Nafisa K Umar
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Saori Takeda
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Makiko Ohtani
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Daichi Murakami
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Hideki Sakatani
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Fumie Kaneko
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan.,Department of Otorhinolaryngology, Tokyo Women's Medical University Medical Center East, Tokyo, Japan
| | - Denisa Nanushaj
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Muneki Hotomi
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
170
|
Li X, Fu YN, Huang L, Liu F, Moriarty TF, Tao L, Wei Y, Wang X. Combating Biofilms by a Self-Adapting Drug Loading Hydrogel. ACS APPLIED BIO MATERIALS 2021; 4:6219-6226. [PMID: 35006889 DOI: 10.1021/acsabm.1c00540] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A bacterial biofilm is one of the main reasons that many diseases are difficult to cure. Herein, a teicoplanin (TPN)-loaded self-adapting chitosan-based hydrogel (CPH) system, called TPN-CPH, was prepared by encapsulating antibacterial TPN into CPH. This TPN-CPH can effectively combat preformed biofilms in vitro of Staphylococcus aureus (S. aureus). It has a good therapeutic effect on full-thickness cutaneous wounds in vivo of mice infected with biofilms. In addition, TPN-CPH can accelerate wound healing by self-adapting the wound and providing a moist environment. The operation process of TPN-CPH is simple, and no external stimulation such as light and heat is needed in the treatment process, making it more convenient for clinical application. Furthermore, this is a challenge to use self-adapting hydrogels to adapt the micro-size channels of biofilms. TPN-CPH provides a chitosan-based self-adapting hydrogel system for loading drugs to kill bacteria in biofilms, and thus it is promising for infection control.
Collapse
Affiliation(s)
- Xia Li
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Ya-Nan Fu
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China.,AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz 7270, Switzerland
| | - Lifei Huang
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Fang Liu
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing 100029, P. R. China
| | | | - Lei Tao
- The Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Yen Wei
- The Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Xing Wang
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| |
Collapse
|
171
|
Dai X, Ma J, Zhang Q, Xu Q, Yang L, Gao F. Simultaneous inhibition of planktonic and biofilm bacteria by self-adapting semiconducting polymer dots. J Mater Chem B 2021; 9:6658-6667. [PMID: 34378630 DOI: 10.1039/d1tb01070k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Biofilm infections present an enormous challenge in today's healthcare settings. Currently, pH-switchable antibacterial agents are being developed to eradicate biofilms. However, most pH-switchable antibacterial agents are less lethal to planktonic bacteria under neutral conditions, and cannot prevent the dispersed bacteria from seeding acute infection again. Herein, this work reports the applications of semiconducting polymer dots (Pdots) with a double adhesion mechanism in imaging and inhibiting bacteria inside (weak acidic conditions) and outside (neutral conditions) biofilms. Clew-like Pdots were prepared by covalently linking phenylboronic acid (PBA) and pH-responsive naphthalimide (NA) ramification in semiconducting polymers. Under neutral conditions, the Pdots combined with bacteria through the formation of boronate esters between PBA and diols. Under weakly acidic conditions, the partial borate bond fractured, and the Pdots adhered onto the bacterial surface through the positively charged NA in Pdots. Furthermore, the Pdots display negligible toxicity to mammalian cells and tissues. More importantly, the Pdots can selectively damage the bacterial membrane and inhibit bacteria in vivo. This work highlights the feasibility of using semiconducting Pdots to image and inhibit bacteria inside and outside biofilms, which represents a highly effective strategy to cope with biofilm infections.
Collapse
Affiliation(s)
- Xiaomei Dai
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Key Laboratory of Chemo/Biosensing, Laboratory of Biosensing and Bioimaging (LOBAB), College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, P. R. China.
| | | | | | | | | | | |
Collapse
|
172
|
Guo R, Li K, Tian B, Wang C, Chen X, Jiang X, He H, Hong W. Elaboration on the architecture of pH-sensitive surface charge-adaptive micelles with enhanced penetration and bactericidal activity in biofilms. J Nanobiotechnology 2021; 19:232. [PMID: 34362397 PMCID: PMC8344171 DOI: 10.1186/s12951-021-00980-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 07/27/2021] [Indexed: 01/18/2023] Open
Abstract
Background Biofilm formation is one of the main reasons for persistent bacterial infections. Recently, pH-sensitive copolymers have fascinated incredible attention to tackle biofilm-related infections. However, the proper incorporation of pH-sensitive segments in the polymer chains, which could significantly affect the biofilms targeting ability, has not been particularly investigated. Herein, we synthesized three types of pH-sensitive copolymers based on poly (β-amino ester) (PAE), poly (lactic-co-glycolic acid) (PLA) and polyethylene glycol (PEG), PAE-PLA-mPEG (A-L-E), PLA-PAE-mPEG (L-A-E) and PLA-PEG-PAE (L-E-A) to address this issue. Results The three copolymers could self-assemble into micelles (MA-L-E, ML-A-E and ML-E-A) in aqueous medium. Compared with MA-L-E and ML-A-E, placing the PAE at the distal PEG end of PLA-PEG to yield PLA-PEG-PAE (ML-E-A) was characterized with proper triggering pH, fully biofilm penetration, and high cell membrane binding affinity. Further loaded with Triclosan (TCS), ML-E-A/TCS could efficiently kill the bacteria either in planktonic or biofilm mode. We reasoned that PAE segments would be preferentially placed near the surface and distant from the hydrophobic PLA segments. This would increase the magnitude of surface charge-switching capability, as the cationic PAE+ would easily disassociate from the inner core without conquering the additional hydrophobic force arising from covalent linkage with PLA segments, and rapidly rise to the outermost layer of the micellar surface due to the relative hydrophilicity. This was significant in that it could enable the micelles immediately change its surface charge where localized acidity occurred, and efficiently bind themselves to the bacterial surface where they became hydrolyzed by bacterial lipases to stimulate release of encapsulated TCS even a relatively short residence time to prevent rapid wash-out. In vivo therapeutic performance of ML-E-A/TCS was evaluated on a classical biofilm infection model, implant-related biofilm infection. The result suggested that ML-E-A/TCS was effective for the treatment of implant-related biofilm infection, which was proved by the efficient clearance of biofilm-contaminated catheters and the recovery of surrounding infected tissues. Conclusions In summary, elaboration on the architecture of pH-sensitive copolymers was the first step to target biofilm. The ML-E-A structure may represent an interesting future direction in the treatment of biofilm-relevant infections associated with acidity. Graphic abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-00980-8.
Collapse
Affiliation(s)
- Rong Guo
- School of Pharmacy, Shandong New Drug Loading and Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, People's Republic of China
| | - Keke Li
- School of Pharmacy, Shandong New Drug Loading and Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, People's Republic of China
| | - Baocheng Tian
- School of Pharmacy, Shandong New Drug Loading and Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, People's Republic of China
| | - Changrong Wang
- School of Pharmacy, Shandong New Drug Loading and Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, People's Republic of China
| | - Xiangjun Chen
- School of Pharmacy, Shandong New Drug Loading and Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, People's Republic of China
| | - Xinyu Jiang
- School of Pharmacy, Shandong New Drug Loading and Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, People's Republic of China
| | - Huayu He
- School of Pharmacy, Shandong New Drug Loading and Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, People's Republic of China
| | - Wei Hong
- School of Pharmacy, Shandong New Drug Loading and Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, 346 Guanhai Road, Yantai, 264003, People's Republic of China.
| |
Collapse
|
173
|
Ding M, Zhao W, Song LJ, Luan SF. Stimuli-responsive nanocarriers for bacterial biofilm treatment. RARE METALS 2021; 41:482-498. [PMID: 34366603 PMCID: PMC8333162 DOI: 10.1007/s12598-021-01802-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 06/01/2023]
Abstract
ABSTRACT Bacterial biofilm infections have been threatening the human's life and health globally for a long time because they typically cause chronic and persistent infections. Traditional antibiotic therapies can hardly eradicate biofilms in many cases, as biofilms always form a robust fortress for pathogens inside, inhibiting the penetration of drugs. To address the issues, many novel drug carriers emerged as promising strategies for biofilm treatment. Among them, stimuli-responsive nanocarriers have attracted much attentions for their intriguing physicochemical properties, such as tunable size, shape and surface chemistry, especially smart drug release characteristic. Based on the microenvironmental difference between biofilm infection sites and normal tissue, many stimuli, such as bacterial products accumulating in biofilms (enzymes, glutathione, etc.), lower pH and higher H2O2 levels, have been employed and proved in favor of "on-demand" drug release for biofilm elimination. Additionally, external stimuli including light, heat, microwave and magnetic fields are also able to control the drug releasing behavior artificially. In this review, we summarized recent advances in stimuli-responsive nanocarriers for combating biofilm infections, and mainly, focusing on the different stimuli that trigger the drug release. 摘要 , , 。 , , 。 , , 。 , -, , , , 。 , , (, ), pHH2O2, ""。 , , , , 。 , , 。.
Collapse
Affiliation(s)
- Meng Ding
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese of Academy, Changchun, 130022 China
- College of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026 China
| | - Wei Zhao
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese of Academy, Changchun, 130022 China
- School of Chemistry and Chemical Engineering, Yantai University, Yantai, 264005 China
| | - Ling-Jie Song
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese of Academy, Changchun, 130022 China
| | - Shi-Fang Luan
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese of Academy, Changchun, 130022 China
- National Engineering Laboratory of Medical Implantable Devices, Key Laboratory for Medical Implantable Devices of Shandong Province, WEGO Holding Company Limited, Weihai, 264210 China
| |
Collapse
|
174
|
Liao Y, Ye Z, Qian M, Wang X, Guo Y, Han G, Song Y, Hou J, Liu Y. Photoactive NO hybrids with pseudo-zero-order release kinetics for antimicrobial applications. Org Biomol Chem 2021; 18:5473-5480. [PMID: 32643744 DOI: 10.1039/d0ob00564a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Bacterial infection is a major threat to the health and life of humans due to the development of drug resistance, which is related to biofilm formation. Nitric oxide (NO) has emerged as an important factor in regulating biofilm formation. In order to harness the potential benefits of NO and develop effective antibacterial agents, we designed and synthesized a new class of NO hybrids in which the active scaffold benzothienoazepine was tagged with a nitroso group and further conjugated with quaternary ammoniums or phosphoniums. The temporal release of NO from these hybrids can be achieved by photoactivation. Interestingly, the NO release follows a pseudo-zero-order kinetics, which is easily determined by measuring the fluorescent benzothienoazepine or NO. Compared to the positive control ciprofloxacin, the NO hybrid with triphenyl phosphonium (TPP) exhibited more effective activity against S. aureus biofilm in darkness. Irradiation of the NO hybrid led to higher inhibition against S. aureus biofilm compared to the parental NO hybrid in darkness or the corresponding NO-released product, indicating the combined effect of NO and the NO-released product. Therefore, this new class of NO hybrids includes very promising antimicrobial agents and this work provides a new way for the design of highly effective antimicrobial agents.
Collapse
Affiliation(s)
- Yongfang Liao
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, P. R. China.
| | - Zizhen Ye
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, P. R. China.
| | - Meng Qian
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, P. R. China.
| | - Xing Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, P. R. China.
| | - Yuda Guo
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, P. R. China.
| | - Guifang Han
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, P. R. China.
| | - Yuguang Song
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, P. R. China.
| | - Jingli Hou
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, P. R. China.
| | - Yangping Liu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, P. R. China.
| |
Collapse
|
175
|
Xu Q, Hu X, Wang Y. Alternatives to Conventional Antibiotic Therapy: Potential Therapeutic Strategies of Combating Antimicrobial-Resistance and Biofilm-Related Infections. Mol Biotechnol 2021; 63:1103-1124. [PMID: 34309796 DOI: 10.1007/s12033-021-00371-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/08/2021] [Indexed: 12/14/2022]
Abstract
Antibiotics have been denoted as the orthodox therapeutic agents for fighting bacteria-related infections in clinical practices for decades. Nevertheless, overuse of antibiotics has led to the upsurge of species with antimicrobial resistance (AMR) or multi-drug resistance. Bacteria can also grow into the biofilm, which accounts for at least two-thirds of infections. Distinct gene expression and self-produced heterogeneous hydrated extracellular polymeric substance matrix architecture of biofilm contribute to their tolerance and externally manifest as antibiotic resistance. In this review, the difficulties in combating biofilm formation and AMR are introduced, and novel alternatives to antibiotics such as metal nanoparticles and quaternary ammonium compounds, chitosan and its derivatives, antimicrobial peptides, stimuli-responsive materials, phage therapy and other therapeutic strategies, from compounds to hydrogel, from inorganic to biological, are discussed. We expect to provide useful information for the readers who are seeking for solutions to the problem of AMR and biofilm-related infections.
Collapse
Affiliation(s)
- Qian Xu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, Sichuan, People's Republic of China
| | - Xuefeng Hu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, Sichuan, People's Republic of China.
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, Sichuan, People's Republic of China.
| |
Collapse
|
176
|
Paul P, Das S, Chatterjee S, Shukla A, Chakraborty P, Sarkar S, Maiti D, Das A, Tribedi P. 1,4-Naphthoquinone disintegrates the pre-existing biofilm of Staphylococcus aureus by accumulating reactive oxygen species. Arch Microbiol 2021; 203:4981-4992. [PMID: 34272991 DOI: 10.1007/s00203-021-02485-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 12/01/2022]
Abstract
Staphylococcus aureus causes several nosocomial and community-acquired infections in human host involving biofilm. Thus, strategies need to be explored to curb biofilm threats by either inhibiting the formation of biofilm or disintegrating the pre-existing biofilm. Towards this direction, we had already revealed the biofilm inhibiting properties of 1,4-naphthoquinone against S. aureus. In this study, we have investigated whether this compound can act on pre-existing biofilm. Hence, biofilm of S. aureus was developed first and challenged further with 1,4-naphthoquinone. Experiments such as crystal violet assay, fluorescence microscopy, and estimation of total biofilm protein were performed to confirm the biofilm disintegration properties of 1,4-naphthoquinone. The disintegration of pre-existing biofilm could be attributed to the generation of reactive oxygen species (ROS). To investigate further, we observed that extracellular DNA (eDNA) was found to play an important role in holding the biofilm network as DNaseI treatment could cause an efficient disintegration of the same. To examine the effect of ROS on the eDNA, we exposed pre-existing biofilm to either 1,4-naphthoquinone or a combination of both 1,4-naphthoquinone and ascorbic acid for different length of time. Post-incubation, ROS generation and the amount of eDNA associated with the biofilm were determined wherein an inversely proportional relationship was observed between them. The result indicated that with the increase of ROS generation, the amount of eDNA associated with biofilm got decreased substantially. Thus, the results indicated that the generation of ROS could degrade the eDNA thereby compromising the integrity of biofilm which lead to the disintegration of pre-existing biofilm.
Collapse
Affiliation(s)
- Payel Paul
- Microbial Ecology Laboratory, Department of Biotechnology, The Neotia University, Sarisha, 743368, West Bengal, India
| | - Sharmistha Das
- Microbial Ecology Laboratory, Department of Biotechnology, The Neotia University, Sarisha, 743368, West Bengal, India
| | - Sudipta Chatterjee
- Microbial Ecology Laboratory, Department of Biotechnology, The Neotia University, Sarisha, 743368, West Bengal, India
| | - Aditya Shukla
- Department of Microbiology, University of Calcutta, 35 Ballygunge Circular Road, Calcutta, 700019, India
| | - Poulomi Chakraborty
- Microbial Ecology Laboratory, Department of Biotechnology, The Neotia University, Sarisha, 743368, West Bengal, India
| | - Sarita Sarkar
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Road Scheme VIIM, Calcutta, 700 054, India
| | - Debasish Maiti
- Department of Human Physiology, Tripura University, Agartala, Tripura, India
| | - Amlan Das
- National Institute of Biomedical Genomics, Kalyani, 741251, West Bengal, India
| | - Prosun Tribedi
- Microbial Ecology Laboratory, Department of Biotechnology, The Neotia University, Sarisha, 743368, West Bengal, India.
| |
Collapse
|
177
|
Astrada A, Nakagami G, Minematsu T, Goto T, Kitamura A, Mugita Y, Sanada H. Concurrent validity of biofilm detection by wound blotting on hard-to-heal wounds. J Wound Care 2021; 30:S4-S13. [PMID: 33856931 DOI: 10.12968/jowc.2021.30.sup4.s4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Wound biofilms delay healing of hard-to-heal wounds. Convenient biofilm identification tools for clinical settings are currently not available, hindering biofilm-based wound management. Wound blotting with biofilm staining is a potential tool for biofilm detection, owing to its convenience. Although predictive validity of wound blotting has been established, it is necessary to confirm its concurrent validity. Furthermore, current staining systems employing ruthenium red have some disadvantages for clinical use. This study aimed to evaluate the usability of alcian blue as a substitute for ruthenium red. METHOD Both in vitro and in vivo clinical samples were used to investigate validity and usability. RESULTS The in vitro study showed that proteins and extracellular DNA in biofilms did not affect staining ability of ruthenium red and alcian blue in the detection of biofilms. In the in vivo study, using a wound biofilm model with Pseudomonas aeruginosa, the staining sensitivity of ruthenium red was 88.9% and 100% for alcian blue, with correlation coefficients of signal intensities with native polyacrylamide gel electrophoresis (PAGE) of r=0.67 (p=0.035) and r=0.67 (p=0.036) for ruthenium red and alcian blue, respectively. Results from clinical samples were r=0.75 (p=0.001) for ruthenium red and r=0.77 (p<0.001) for alcian blue. The sensitivities of wound blotting staining by ruthenium red and alcian blue were very high and had a good correlation with native PAGE analysis. CONCLUSION Because the alcian blue procedure is more convenient than the ruthenium red procedure, wound blotting with alcian blue staining would be a promising tool to guide clinicians in delivering biofilm-based wound management.
Collapse
Affiliation(s)
- Adam Astrada
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Gojiro Nakagami
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Global Nursing Research Center, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeo Minematsu
- Global Nursing Research Center, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Skincare Science, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Taichi Goto
- Department of Pain and Translational Symptom Science, University of Maryland School of Nursing, Baltimore, MD, US
| | - Aya Kitamura
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuko Mugita
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiromi Sanada
- Department of Gerontological Nursing/Wound Care Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Global Nursing Research Center, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
178
|
da Rocha LGDO, Ribeiro VST, de Andrade AP, Gonçalves GA, Kraft L, Cieslinski J, Suss PH, Tuon FF. Evaluation of Staphylococcus aureus and Candida albicans biofilms adherence to PEEK and titanium-alloy prosthetic spine devices. EUROPEAN JOURNAL OF ORTHOPAEDIC SURGERY AND TRAUMATOLOGY 2021; 32:981-989. [PMID: 34236512 DOI: 10.1007/s00590-021-03069-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/28/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Titanium and polyether-ether-ketone (PEEK) interbody cages are commonly used for spine fusion. Few data are known about bacterial and yeast biofilms formation in these implants. The aim of this study was to compare Staphylococcus aureus and Candida albicans biofilm formation in the surface of two different interbody devices used routinely in spine surgery. METHODS Six bodies of proof specimens of PEEK and titanium alloy were used for microbiological tests, scanning electron microscopy, and energy-dispersive X-ray spectroscopy. Experimental biofilm was produced with Staphylococcus aureus and Candida albicans, followed by quantitative analysis of planktonic cells and sessile cells. The comparison between the medians of biofilm quantification between the two models was performed using the Mann-Whitney test and considered the statistical difference for a p < 0.05. RESULTS In the S. aureus model, in both planktonic and sessile cell counts, titanium-alloy samples showed lower values for colony forming units per milliliter (UFC/mL) (p < 0.05). The evaluation through the optic density of planktonic and sessile cells showed lower values in the titanium-alloy samples, however, only statistically significant in planktonic cell count (p < 0.05). The count of planktonic yeast cells in PEEK was similar to titanium-alloy samples, while the count of sessile yeast cells in titanium alloy was lower when compared to PEEK (p < 0.05). CONCLUSION Titanium-alloy models were associated with less staphylococcal and Candida biofilm formation when compared with PEEK.
Collapse
Affiliation(s)
- Luiz Gustavo Dal Oglio da Rocha
- Hospital Universitário Cajuru - HUC, Spine Department. Pontifícia, Universidade Católica do Paraná (PUCPR), Avenida São José, 300, Curitiba, Paraná, 80050-350, Brazil.,School of Medicine, Health Sciences Department, Pontifícia Universidade Católica do Paraná (PUCPR), Rua Imaculada Conceição, 1155, Curitiba, Paraná, 80215-901, Brazil
| | - Victoria Stadler Tasca Ribeiro
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Rua Imaculada Conceição, 1155, Curitiba, Paraná, 80215-901, Brazil.,School of Medicine, Health Sciences Department, Pontifícia Universidade Católica do Paraná (PUCPR), Rua Imaculada Conceição, 1155, Curitiba, Paraná, 80215-901, Brazil
| | - Ana Paula de Andrade
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Rua Imaculada Conceição, 1155, Curitiba, Paraná, 80215-901, Brazil.,School of Medicine, Health Sciences Department, Pontifícia Universidade Católica do Paraná (PUCPR), Rua Imaculada Conceição, 1155, Curitiba, Paraná, 80215-901, Brazil
| | - Geiziane Aparecida Gonçalves
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Rua Imaculada Conceição, 1155, Curitiba, Paraná, 80215-901, Brazil
| | - Letícia Kraft
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Rua Imaculada Conceição, 1155, Curitiba, Paraná, 80215-901, Brazil
| | - Juliette Cieslinski
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Rua Imaculada Conceição, 1155, Curitiba, Paraná, 80215-901, Brazil
| | - Paula Hansen Suss
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Rua Imaculada Conceição, 1155, Curitiba, Paraná, 80215-901, Brazil.,School of Medicine, Health Sciences Department, Pontifícia Universidade Católica do Paraná (PUCPR), Rua Imaculada Conceição, 1155, Curitiba, Paraná, 80215-901, Brazil
| | - Felipe Francisco Tuon
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Rua Imaculada Conceição, 1155, Curitiba, Paraná, 80215-901, Brazil. .,School of Medicine, Health Sciences Department, Pontifícia Universidade Católica do Paraná (PUCPR), Rua Imaculada Conceição, 1155, Curitiba, Paraná, 80215-901, Brazil.
| |
Collapse
|
179
|
Triazolo Based-Thiadiazole Derivatives. Synthesis, Biological Evaluation and Molecular Docking Studies. Antibiotics (Basel) 2021; 10:antibiotics10070804. [PMID: 34356726 PMCID: PMC8300616 DOI: 10.3390/antibiotics10070804] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 11/18/2022] Open
Abstract
The goal of this research is to investigate the antimicrobial activity of nineteen previously synthesized 3,6-disubstituted-1,2,4-triazolo[3,4-b]-1,3,4-thiadiazole derivatives. The compounds were tested against a panel of three Gram-positive and three Gram-negative bacteria, three resistant strains, and six fungi. Minimal inhibitory, bactericidal, and fungicidal concentrations were determined by a microdilution method. All of the compounds showed antibacterial activity that was more potent than both reference drugs, ampicillin and streptomycin, against all bacteria tested. Similarly, they were also more active against resistant bacterial strains. The antifungal activity of the compounds was up to 80-fold higher than ketoconazole and from 3 to 40 times higher than bifonazole, both of which were used as reference drugs. The most active compounds (2, 3, 6, 7, and 19) were tested for their inhibition of P. aeruginosa biofilm formation. Among them, compound 3 showed significantly higher antibiofilm activity and appeared to be equipotent with ampicillin. The prediction of the probable mechanism by docking on antibacterial targets revealed that E. coli MurB is the most suitable enzyme, while docking studies on antifungal targets indicated a probable involvement of CYP51 in the mechanism of antifungal activity. Finally, the toxicity testing in human cells confirmed their low toxicity both in cancerous cell line MCF7 and non-cancerous cell line HK-2.
Collapse
|
180
|
Skóra B, Krajewska U, Nowak A, Dziedzic A, Barylyak A, Kus-Liśkiewicz M. Noncytotoxic silver nanoparticles as a new antimicrobial strategy. Sci Rep 2021; 11:13451. [PMID: 34188097 PMCID: PMC8242066 DOI: 10.1038/s41598-021-92812-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/16/2021] [Indexed: 02/05/2023] Open
Abstract
Drug-resistance of bacteria is an ongoing problem in hospital treatment. The main mechanism of bacterial virulency in human infections is based on their adhesion ability and biofilm formation. Many approaches have been invented to overcome this problem, i.e. treatment with antibacterial biomolecules, which have some limitations e.g. enzymatic degradation and short shelf stability. Silver nanoparticles (AgNPs) may be alternative to these strategies due to their unique and high antibacterial properties. Herein, we report on yeast Saccharomyces cerevisiae extracellular-based synthesis of AgNPs. Transmission electron microscopy (TEM) revealed the morphology and structure of the metallic nanoparticles, which showed a uniform distribution and good colloid stability, measured by hydrodynamic light scattering (DLS). The energy dispersive X-ray spectroscopy (EDS) of NPs confirms the presence of silver and showed that sulfur-rich compounds act as a capping agent being adsorbed on the surface of AgNPs. Antimicrobial tests showed that AgNPs inhibit the bacteria growth, while have no impact on fungi growth. Moreover, tested NPs was characterized by high inhibitory potential of bacteria biofilm formation but also eradication of established biofilms. The cytotoxic effect of the NPs on four mammalian normal and cancer cell lines was tested through the metabolic activity, cell viability and wound-healing assays. Last, but not least, ability to deep penetration of the silver colloid to the root canal was imaged by scanning electron microscopy (SEM) to show its potential as the material for root-end filling.
Collapse
Affiliation(s)
- Bartosz Skóra
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management, St. Sucharskiego 2, 35-225, Rzeszów, Poland
| | - Urszula Krajewska
- Institute of Biology and Biotechnology, College of Natural Sciences, University of Rzeszow, St. Pigonia 1, 35-310, Rzeszów, Poland
| | - Anna Nowak
- Institute of Biology and Biotechnology, College of Natural Sciences, University of Rzeszow, St. Pigonia 1, 35-310, Rzeszów, Poland
| | - Andrzej Dziedzic
- College of Natural Sciences, University of Rzeszow, St. Pigonia 1, 35-310, Rzeszow, Poland
| | - Adriana Barylyak
- Laser Department Center of Imlantation and Prosthetic Dentistry "MM", Department of Therapeutical Dentistry, Lviv National Medical University Ukraine, Lviv, Poland
| | - Małgorzata Kus-Liśkiewicz
- Institute of Biology and Biotechnology, College of Natural Sciences, University of Rzeszow, St. Pigonia 1, 35-310, Rzeszów, Poland.
| |
Collapse
|
181
|
Pertusati F, Pileggi E, Richards J, Wootton M, Van Leemputte T, Persoons L, De Coster D, Villanueva X, Daelemans D, Steenackers H, McGuigan C, Serpi M. Drug repurposing: phosphate prodrugs of anticancer and antiviral FDA-approved nucleosides as novel antimicrobials. J Antimicrob Chemother 2021; 75:2864-2878. [PMID: 32688391 DOI: 10.1093/jac/dkaa268] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 05/09/2020] [Accepted: 05/15/2020] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Following a drug repurposing approach, we aimed to investigate and compare the antibacterial and antibiofilm activities of different classes of phosphate prodrugs (HepDirect, cycloSal, SATE and mix SATE) of antiviral and anticancer FDA-approved nucleoside drugs [zidovudine (AZT), floxouridine (FUDR) and gemcitabine (GEM)] against a variety of pathogenic Gram-positive and -negative bacteria. METHODS Ten prodrugs were synthesized and screened for antibacterial activity against seven Gram-negative and two Gram-positive isolates fully susceptible to traditional antibiotics, alongside six Gram-negative and five Gram-positive isolates with resistance mechanisms. Their ability to prevent and eradicate biofilms of different bacterial pathogens in relation to planktonic growth inhibition was also evaluated, together with their effect on proliferation, viability and apoptosis of different eukaryotic cells. RESULTS The prodrugs showed decreased antibacterial activity compared with the parent nucleosides. cycloSal-GEM-monophosphate (MP) prodrugs 20a and 20b were the most active agents against Gram-positive bacteria (Enterococcus faecalis and Staphylococcus aureus) and retained their activity against antibiotic-resistant isolates. cycloSal-FUDR-MP 21a partially retained good activity against the Gram-positive bacteria E. faecalis, Enterococcus faecium and S. aureus. Most of the prodrugs tested displayed very potent preventive antibiofilm specific activity, but not curative. In terms of cytotoxicity, AZT prodrugs did not affect apoptosis or cell viability at the highest concentration tested, and only weak effects on apoptosis and/or cell viability were observed for GEM and FUDR prodrugs. CONCLUSIONS Among the different prodrug approaches, the cycloSal prodrugs appeared the most effective. In particular, cycloSal (17a) and mix SATE (26) AZT prodrugs combine the lowest cytotoxicity with high and broad antibacterial and antibiofilm activity against Gram-negative bacteria.
Collapse
Affiliation(s)
- Fabrizio Pertusati
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, UK
| | - Elisa Pileggi
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, UK
| | - Jennifer Richards
- Public Health Wales Microbiology Cardiff, University Hospital of Wales, Heath Park, Cardiff CF14 4XW, UK
| | - Mandy Wootton
- Public Health Wales Microbiology Cardiff, University Hospital of Wales, Heath Park, Cardiff CF14 4XW, UK
| | - Thijs Van Leemputte
- Centre of Microbial and Plant Genetics, Kasteelpark Arenberg 20, bus 2460, B-3001 Leuven, Belgium
| | - Leentje Persoons
- Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, Herestraat 49-box 1043, Leuven 3000, Belgium
| | - David De Coster
- Centre of Microbial and Plant Genetics, Kasteelpark Arenberg 20, bus 2460, B-3001 Leuven, Belgium
| | - Xabier Villanueva
- Centre of Microbial and Plant Genetics, Kasteelpark Arenberg 20, bus 2460, B-3001 Leuven, Belgium
| | - Dirk Daelemans
- Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, Herestraat 49-box 1043, Leuven 3000, Belgium
| | - Hans Steenackers
- Centre of Microbial and Plant Genetics, Kasteelpark Arenberg 20, bus 2460, B-3001 Leuven, Belgium
| | - Christopher McGuigan
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, UK
| | - Michaela Serpi
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, UK
| |
Collapse
|
182
|
Birk SE, Serioli L, Cavallo V, Haagensen JAJ, Molin S, Nielsen LH, Zór K, Boisen A. Enhanced Eradication of Mucin-Embedded Bacterial Biofilm by Locally Delivered Antibiotics in Functionalized Microcontainers. Macromol Biosci 2021; 21:e2100150. [PMID: 34117842 DOI: 10.1002/mabi.202100150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/07/2021] [Indexed: 12/16/2022]
Abstract
Bacterial biofilm-related infections are difficult to eradicate and require repeated treatments with high doses of antibiotics. Thus, there is an urgent need for new treatment strategies that minimize the use of antibiotics while enhancing biofilm eradication. Functionalized reservoir-based microdevices, such as, microcontainers (MCs), offer, high drug loading capacity, mucus embedment, and tuneable drug release. Here, MCs are loaded with the antibiotic ciprofloxacin (CIP), and sealed with a lid consisting of chitosan (CHI) and a mucolytic agent, N-acetylcysteine (NAC). It is found that CHI and NAC work synergistically, showing improved mucoadhesive and mucolytic properties. To better mimic the in vivo habitat of Pseudomonas aeruginosa (P. aeruginosa), the biofilm is grown in a mucin-containing medium on a newly developed centrifugal microfluidic system. The CHI/NAC coated MCs improve eradication of biofilm (88.22 ± 2.89%) compared to CHI-coated MCs (72.68 ± 3.73%) or bolus injection (39.86 ± 13.28%). The findings suggest that MCs are significantly more efficient than a bolus treatment. Furthermore, CHI/NAC functionalized MCs kill most of the biomass already after 5 h (80.75 ± 3.50%), mainly due to a fast drug release. This is the first time that CHI/NAC has been combined as a coating to explore mucolytic properties on bacterial biofilms.
Collapse
Affiliation(s)
- Stine Egebro Birk
- The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Ørsteds Plads 345C, Lyngby, 2800 Kgs., Denmark
| | - Laura Serioli
- The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Ørsteds Plads 345C, Lyngby, 2800 Kgs., Denmark.,BioInnovation Institute Foundation, Copenhagen N, 2800, Denmark
| | - Valentina Cavallo
- The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Ørsteds Plads 345C, Lyngby, 2800 Kgs., Denmark
| | - Janus Anders Juul Haagensen
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet 220, Lyngby, 2800, Denmark
| | - Søren Molin
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet 220, Lyngby, 2800, Denmark
| | - Line Hagner Nielsen
- The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Ørsteds Plads 345C, Lyngby, 2800 Kgs., Denmark
| | - Kinga Zór
- The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Ørsteds Plads 345C, Lyngby, 2800 Kgs., Denmark.,BioInnovation Institute Foundation, Copenhagen N, 2800, Denmark
| | - Anja Boisen
- The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Ørsteds Plads 345C, Lyngby, 2800 Kgs., Denmark.,BioInnovation Institute Foundation, Copenhagen N, 2800, Denmark
| |
Collapse
|
183
|
Trans-Cinnamaldehyde Attenuates Enterococcus faecalis Virulence and Inhibits Biofilm Formation. Antibiotics (Basel) 2021; 10:antibiotics10060702. [PMID: 34208134 PMCID: PMC8230787 DOI: 10.3390/antibiotics10060702] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/09/2021] [Accepted: 06/09/2021] [Indexed: 11/21/2022] Open
Abstract
Enterococcus faecalis as an important nosocomial pathogen is critically implicated in the pathogenesis of endocarditis, urinary tract, and persistent root canal infections. Its major virulence attributes (biofilm formation, production of proteases, and hemolytic toxins) enable it to cause extensive host tissue damage. With the alarming increase in enterococcal resistance to antibiotics, novel therapeutics are required to inhibit E. faecalis biofilm formation and virulence. Trans-cinnamaldehyde (TC), the main phytochemical in cinnamon essential oils, has demonstrated promising activity against a wide range of pathogens. Here, we comprehensively investigated the effect of TC on planktonic growth, biofilm formation, proteolytic and hemolytic activities, as well as gene regulation in E. faecalis. Our findings revealed that sub-inhibitory concentrations of TC reduced biofilm formation, biofilm exopolysaccharides, as well as its proteolytic and hemolytic activities. Mechanistic studies revealed significant downregulation of the quorum sensing fsr locus and downstream gelE, which are major virulence regulators in E. faecalis. Taken together, our study highlights the potential of TC to inhibit E. faecalis biofilm formation and its virulence.
Collapse
|
184
|
Bhardwaj S, Bhatia S, Singh S, Franco Jr F. Growing emergence of drug-resistant Pseudomonas aeruginosa and attenuation of its virulence using quorum sensing inhibitors: A critical review. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:699-719. [PMID: 34630947 PMCID: PMC8487598 DOI: 10.22038/ijbms.2021.49151.11254] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 11/24/2020] [Indexed: 12/25/2022]
Abstract
A perilous increase in the number of bacterial infections has led to developing throngs of antibiotics for increasing the quality and expectancy of life. Pseudomonas aeruginosa is becoming resistant to all known conventional antimicrobial agents thereby posing a deadly threat to the human population. Nowadays, targeting virulence traits of infectious agents is an alternative approach to antimicrobials that is gaining much popularity to fight antimicrobial resistance. Quorum sensing (QS) involves interspecies communication via a chemical signaling pathway. Under this mechanism, cells work in a concerted manner, communicate with each other with the help of signaling molecules called auto-inducers (AI). The virulence of these strains is driven by genes, whose expression is regulated by AI, which in turn acts as transcriptional activators. Moreover, the problem of antibiotic-resistance in case of infections caused by P. aeruginosa becomes more alarming among immune-compromised patients, where the infectious agents easily take over the cellular machinery of the host while hidden in the QS mediated biofilms. Inhibition of the QS circuit of P. aeruginosa by targeting various signaling pathways such as LasR, RhlR, Pqs, and QScR transcriptional proteins will help in blocking downstream signal transducers which could result in reducing the bacterial virulence. The anti-virulence agent does not pose an immediate selective pressure on growing bacterium and thus reduces the pathogenicity without harming the target species. Here, we review exclusively, the growing emergence of multi-drug resistant (MDR) P. aeruginosa and the critical literature survey of QS inhibitors with their potential application of blocking P. aeruginosa infections.
Collapse
Affiliation(s)
- Snigdha Bhardwaj
- Department of Pharmaceutical Science, SHALOM Institute of Health and Allied Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences (SHUATS), Naini, Prayagraj, India
| | - Sonam Bhatia
- Department of Pharmaceutical Science, SHALOM Institute of Health and Allied Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences (SHUATS), Naini, Prayagraj, India
| | - Shaminder Singh
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, Faridabad - 121 001, Haryana, India
| | - Francisco Franco Jr
- Department of Chemistry, De La Salle University, Manila, Metro Manila, Philippines
| |
Collapse
|
185
|
Fahmide F, Ehsani P, Atyabi SM. Time-dependent behavior of the Staphylococcus aureus biofilm following exposure to cold atmospheric pressure plasma. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:744-751. [PMID: 34630951 PMCID: PMC8487605 DOI: 10.22038/ijbms.2021.52541.11866] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 05/23/2021] [Indexed: 01/13/2023]
Abstract
Objective(s): Formation of Staphylococcus aureus biofilm leads to persistent infection in tissue or on exter-nal and indwelling devices in patients. Cold atmospheric plasma (CAP) is used for eradication of bacterial biofilms and it has diverse applications in the healthcare system. However, there is not sufficient information on the behavior of biofilms during the CAP exposure period. Materials and Methods: Pre-established S. aureus biofilms were exposed to CAP for 0 to 360 sec, then subjected to washing steps and sonication. Subsequently, biomass, number of colonies, vitality of bacteria, structure of colonies, size of produced particles, and viability of bacteria were evaluated by different assays including crystal violet, colony-forming unit, MTT, scanning electron mi-croscopy, confocal laser scanning microscopy, and dynamic light scattering assays. Results: The results showed that the strength of biomass increased in the first 60 sec, then decreased to less than no-CAP treated controls. Moreover, short CAP exposure (≤60 sec) ehances the fusion of the biofilm extracellular matrix and other components, which results in preservation of bacteria during ultra-sonication and washing steps compared with control biofilms. The S. aureus biofilm structure only breaks down following more CAP exposure (> 90 sec) and demolition. Interestingly, the 60 sec CAP exposure could cause the fusion of biofilm compo-nents, and large particles are detectable. Conclusion: According to this study, an inadequate CAP exposure period prevents absolute eradication of biofilm and enhances the preservation of bacteria in stronger biofilm compartments.
Collapse
Affiliation(s)
- Foad Fahmide
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Parastoo Ehsani
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | | |
Collapse
|
186
|
Gomathy V, Manigandan V, Vignesh N, Thabitha A, Saravanan R. Evaluation of antibacterial, teratogenicity and antibiofilm effect of sulfated chitosans extracted from marine waste against microorganism. J BIOACT COMPAT POL 2021. [DOI: 10.1177/08839115211014225] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Biofilms play a key role in infectious diseases, as they may form on the surface and persist after treatment with various antimicrobial agents. The Staphylococcus aureus, Klebsiella pneumoniae, S. typhimurium, P. aeruginosa, and Escherichia coli most frequently associated with medical devices. Chitosan sulphate from marine litter (SCH-MW) was extracted and the mineral components were determined using atomic absorption spectroscopy (AAS). The degree of deacetylation (DA) of SCH was predicted 50% and 33.3% in crab and shrimp waste respectively. The elucidation of the structure of the SCH-MW was portrayed using FT-IR and 1H-NMR spectroscopy. The molecular mass of SCH-MW was determined with Matrix-Assisted Laser Desorption/Ionization-Time of Flight (MALDI-TOF). The teratogenicity of SCH-MW was characterized by the zebrafish embryo (ZFE) model. Antimicrobial activity of SCH-MW was tested with the agar well diffusion method; the inhibitory effect of SCH-MW on biofilm formation was assessed in 96 flat well polystyrene plates. The result revealed that a low concentration of crab-sulfated chitosan inhibited bacterial growth and significantly reduced the anti-biofilm activity of gram-negative and gram-positive bacteria relatively to shrimp. It is potentially against the biofilm formation of pathogenic bacteria.
Collapse
Affiliation(s)
- Velu Gomathy
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| | - Venkatesan Manigandan
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| | - Narasimman Vignesh
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| | - Aavula Thabitha
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| | - Ramachandran Saravanan
- Native Medicine & Marine Pharmacology Laboratory, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| |
Collapse
|
187
|
Srinivasan R, Santhakumari S, Poonguzhali P, Geetha M, Dyavaiah M, Xiangmin L. Bacterial Biofilm Inhibition: A Focused Review on Recent Therapeutic Strategies for Combating the Biofilm Mediated Infections. Front Microbiol 2021; 12:676458. [PMID: 34054785 PMCID: PMC8149761 DOI: 10.3389/fmicb.2021.676458] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/14/2021] [Indexed: 12/31/2022] Open
Abstract
Biofilm formation is a major concern in various sectors and cause severe problems to public health, medicine, and industry. Bacterial biofilm formation is a major persistent threat, as it increases morbidity and mortality, thereby imposing heavy economic pressure on the healthcare sector. Bacterial biofilms also strengthen biofouling, affecting shipping functions, and the offshore industries in their natural environment. Besides, they accomplish harsh roles in the corrosion of pipelines in industries. At biofilm state, bacterial pathogens are significantly resistant to external attack like antibiotics, chemicals, disinfectants, etc. Within a cell, they are insensitive to drugs and host immune responses. The development of intact biofilms is very critical for the spreading and persistence of bacterial infections in the host. Further, bacteria form biofilms on every probable substratum, and their infections have been found in plants, livestock, and humans. The advent of novel strategies for treating and preventing biofilm formation has gained a great deal of attention. To prevent the development of resistant mutants, a feasible technique that may target adhesive properties without affecting the bacterial vitality is needed. This stimulated research is a rapidly growing field for applicable control measures to prevent biofilm formation. Therefore, this review discusses the current understanding of antibiotic resistance mechanisms in bacterial biofilm and intensely emphasized the novel therapeutic strategies for combating biofilm mediated infections. The forthcoming experimental studies will focus on these recent therapeutic strategies that may lead to the development of effective biofilm inhibitors than conventional treatments.
Collapse
Affiliation(s)
- Ramanathan Srinivasan
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, School of Life Sciences, Fujian Agriculture and Forestry University, Fujian, China.,Key Laboratory of Crop Ecology and Molecular Physiology (Fujian Agriculture and Forestry University), Fujian Province University, Fujian, China
| | - Sivasubramanian Santhakumari
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, India
| | | | - Mani Geetha
- PG Research and Department of Microbiology, St. Joseph's College of Arts and Science (Autonomous), Tamil Nadu, India
| | - Madhu Dyavaiah
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, India
| | - Lin Xiangmin
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, School of Life Sciences, Fujian Agriculture and Forestry University, Fujian, China.,Key Laboratory of Crop Ecology and Molecular Physiology (Fujian Agriculture and Forestry University), Fujian Province University, Fujian, China.,Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fujian, China
| |
Collapse
|
188
|
Buzalewicz I, Ulatowska-Jarża A, Kaczorowska A, Gąsior-Głogowska M, Podbielska H, Karwańska M, Wieliczko A, Matczuk AK, Kowal K, Kopaczyńska M. Bacteria Single-Cell and Photosensitizer Interaction Revealed by Quantitative Phase Imaging. Int J Mol Sci 2021; 22:5068. [PMID: 34064730 PMCID: PMC8151141 DOI: 10.3390/ijms22105068] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/23/2021] [Accepted: 05/06/2021] [Indexed: 01/12/2023] Open
Abstract
Quantifying changes in bacteria cells in the presence of antibacterial treatment is one of the main challenges facing contemporary medicine; it is a challenge that is relevant for tackling issues pertaining to bacterial biofilm formation that substantially decreases susceptibility to biocidal agents. Three-dimensional label-free imaging and quantitative analysis of bacteria-photosensitizer interactions, crucial for antimicrobial photodynamic therapy, is still limited due to the use of conventional imaging techniques. We present a new method for investigating the alterations in living cells and quantitatively analyzing the process of bacteria photodynamic inactivation. Digital holographic tomography (DHT) was used for in situ examination of the response of Escherichia coli and Staphylococcus aureus to the accumulation of the photosensitizers immobilized in the copolymer revealed by the changes in the 3D refractive index distributions of single cells. Obtained results were confirmed by confocal microscopy and statistical analysis. We demonstrated that DHT enables real-time characterization of the subcellular structures, the biophysical processes, and the induced local changes of the intracellular density in a label-free manner and at sub-micrometer spatial resolution.
Collapse
Affiliation(s)
- Igor Buzalewicz
- Department of Biomedical Engineering, Faculty of Fundamental Problems of Technology, Wrocław University of Science and Technology, 27 Wybrzeże S. Wyspiańskiego St., 50-370 Wrocław, Poland; (A.U.-J.); (A.K.); (M.G.-G.); (H.P.); (M.K.)
| | - Agnieszka Ulatowska-Jarża
- Department of Biomedical Engineering, Faculty of Fundamental Problems of Technology, Wrocław University of Science and Technology, 27 Wybrzeże S. Wyspiańskiego St., 50-370 Wrocław, Poland; (A.U.-J.); (A.K.); (M.G.-G.); (H.P.); (M.K.)
| | - Aleksandra Kaczorowska
- Department of Biomedical Engineering, Faculty of Fundamental Problems of Technology, Wrocław University of Science and Technology, 27 Wybrzeże S. Wyspiańskiego St., 50-370 Wrocław, Poland; (A.U.-J.); (A.K.); (M.G.-G.); (H.P.); (M.K.)
| | - Marlena Gąsior-Głogowska
- Department of Biomedical Engineering, Faculty of Fundamental Problems of Technology, Wrocław University of Science and Technology, 27 Wybrzeże S. Wyspiańskiego St., 50-370 Wrocław, Poland; (A.U.-J.); (A.K.); (M.G.-G.); (H.P.); (M.K.)
| | - Halina Podbielska
- Department of Biomedical Engineering, Faculty of Fundamental Problems of Technology, Wrocław University of Science and Technology, 27 Wybrzeże S. Wyspiańskiego St., 50-370 Wrocław, Poland; (A.U.-J.); (A.K.); (M.G.-G.); (H.P.); (M.K.)
| | - Magdalena Karwańska
- Department of Epizootiology and Veterinary Administration with Clinic of Infectious Diseases, Wrocław University of Environmental and Life Sciences, 45 Grunwaldzki Square, 50-366 Wrocław, Poland; (M.K.); (A.W.)
| | - Alina Wieliczko
- Department of Epizootiology and Veterinary Administration with Clinic of Infectious Diseases, Wrocław University of Environmental and Life Sciences, 45 Grunwaldzki Square, 50-366 Wrocław, Poland; (M.K.); (A.W.)
| | - Anna K. Matczuk
- Department of Pathology, Division of Microbiology, Faculty of Veterinary Medicine, Wrocław University of Environmental and Life Sciences, 31 C.K. Norwida St., 51-375 Wrocław, Poland;
| | | | - Marta Kopaczyńska
- Department of Biomedical Engineering, Faculty of Fundamental Problems of Technology, Wrocław University of Science and Technology, 27 Wybrzeże S. Wyspiańskiego St., 50-370 Wrocław, Poland; (A.U.-J.); (A.K.); (M.G.-G.); (H.P.); (M.K.)
| |
Collapse
|
189
|
Martín‐Rodríguez AJ, Villion K, Yilmaz‐Turan S, Vilaplana F, Sjöling Å, Römling U. Regulation of colony morphology and biofilm formation in Shewanella algae. Microb Biotechnol 2021; 14:1183-1200. [PMID: 33764668 PMCID: PMC8085958 DOI: 10.1111/1751-7915.13788] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/17/2022] Open
Abstract
Bacterial colony morphology can reflect different physiological stages such as virulence or biofilm formation. In this work we used transposon mutagenesis to identify genes that alter colony morphology and cause differential Congo Red (CR) and Brilliant Blue G (BBG) binding in Shewanella algae, a marine indigenous bacterium and occasional human pathogen. Microscopic analysis of colonies formed by the wild-type strain S. algae CECT 5071 and three transposon integration mutants representing the diversity of colony morphotypes showed production of biofilm extracellular polymeric substances (EPS) and distinctive morphological alterations. Electrophoretic and chemical analyses of extracted EPS showed differential patterns between strains, although the targets of CR and BBG binding remain to be identified. Galactose and galactosamine were the preponderant sugars in the colony biofilm EPS of S. algae. Surface-associated biofilm formation of transposon integration mutants was not directly correlated with a distinct colony morphotype. The hybrid sensor histidine kinase BarA abrogated surface-associated biofilm formation. Ectopic expression of the kinase and mutants in the phosphorelay cascade partially recovered biofilm formation. Altogether, this work provides the basic analysis to subsequently address the complex and intertwined networks regulating colony morphology and biofilm formation in this poorly understood species.
Collapse
Affiliation(s)
| | - Katia Villion
- Department of Microbiology, Tumor and Cell BiologyKarolinska InstitutetStockholmSweden
| | - Secil Yilmaz‐Turan
- Division of GlycoscienceDepartment of ChemistryKTH Royal Institute of TechnologyAlbaNova University CentreStockholmSweden
| | - Francisco Vilaplana
- Division of GlycoscienceDepartment of ChemistryKTH Royal Institute of TechnologyAlbaNova University CentreStockholmSweden
| | - Åsa Sjöling
- Department of Microbiology, Tumor and Cell BiologyKarolinska InstitutetStockholmSweden
| | - Ute Römling
- Department of Microbiology, Tumor and Cell BiologyKarolinska InstitutetStockholmSweden
| |
Collapse
|
190
|
Haney EF, Trimble MJ, Hancock REW. Microtiter plate assays to assess antibiofilm activity against bacteria. Nat Protoc 2021; 16:2615-2632. [PMID: 33911258 DOI: 10.1038/s41596-021-00515-3] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 02/03/2021] [Indexed: 02/02/2023]
Abstract
Bacterial biofilms demonstrate high broad-spectrum adaptive antibiotic resistance and cause two thirds of all infections, but there is a lack of approved antibiofilm agents. Unlike the standard minimal inhibitory concentration assay to assess antibacterial activity against planktonic cells, there is no standardized method to evaluate biofilm inhibition and/or eradication capacity of novel antibiofilm compounds. The protocol described here outlines simple and reproducible methods for assessing the biofilm inhibition and eradication capacities of novel antibiofilm agents against adherent bacterial biofilms grown in 96-well microtiter plates. It employs two inexpensive dyes: crystal violet to stain adhered biofilm biomass and 2,3,5-triphenyl tetrazolium chloride to quantify metabolism of the biofilm cells. The procedure is accessible to any laboratory with a plate reader, requires minimal technical expertise or training and takes 4 or 5 d to complete. Recommendations for how biofilm inhibition and eradication results should be interpreted and presented are also described.
Collapse
Affiliation(s)
- Evan F Haney
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael J Trimble
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Robert E W Hancock
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
191
|
Harrison ZL, Bumgardner JD, Fujiwara T, Baker DL, Jennings JA. In vitro evaluation of loaded chitosan membranes for pain relief and infection prevention. J Biomed Mater Res B Appl Biomater 2021; 109:1735-1743. [PMID: 33871933 DOI: 10.1002/jbm.b.34831] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/01/2021] [Accepted: 02/22/2021] [Indexed: 01/28/2023]
Abstract
Wounds resulting from surgeries, implantation of medical devices, and musculoskeletal trauma result in pain and can also result in infection of damaged tissue. Up to 80% of these infections are due to biofilm formation either on the surface of implanted devices or on surrounding wounded tissue. Bacteria within a biofilm have intrinsic growth and development characteristics that allow them to withstand up to 1,000 times the minimum inhibitory concentration of antibiotics, demonstrating the need for new therapeutics to prevent and treat these infections. Cis-2-decenoic acid (C2DA) is known to disperse preformed biofilms and can prevent biofilm formation entirely for some strains of bacteria. Additionally, local anesthetics like bupivacaine have been shown to have antimicrobial effects against multiple bacterial strains. This study sought to evaluate hexanoic acid-treated electrospun chitosan membranes (HA-ESCM) as wound dressings that release C2DA and bupivacaine to simultaneously prevent infection and alleviate pain associated with musculoskeletal trauma. Release profiles of both therapeutics were evaluated, and membranes were tested in vitro against Methicillin-resistant Staphylococcus aureus (MRSA) to determine efficacy in preventing biofilm infection and bacterial growth. Results indicate that membranes release both therapeutics for 72 hr, and release profile can be tailored by loading concentration. Membranes were effective in preventing biofilm growth but were toxic to fibroblasts when loaded with 2.5 or 5 mg of bupivacaine.
Collapse
Affiliation(s)
- Zoe L Harrison
- Department of Biomedical Engineering, University of Memphis, Memphis, Tennessee, USA
| | - Joel D Bumgardner
- Department of Biomedical Engineering, University of Memphis, Memphis, Tennessee, USA
| | - Tomoko Fujiwara
- Department of Chemistry, University of Memphis, Memphis, Tennessee, USA
| | - Daniel L Baker
- Department of Chemistry, University of Memphis, Memphis, Tennessee, USA
| | - J Amber Jennings
- Department of Biomedical Engineering, University of Memphis, Memphis, Tennessee, USA
| |
Collapse
|
192
|
Dantas Lopes Dos Santos D, Besegato JF, de Melo PBG, Oshiro Junior JA, Chorilli M, Deng D, Bagnato VS, Rastelli ANDS. Curcumin-loaded Pluronic ® F-127 Micelles as a Drug Delivery System for Curcumin-mediated Photodynamic Therapy for Oral Application. Photochem Photobiol 2021; 97:1072-1088. [PMID: 33872402 DOI: 10.1111/php.13433] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/03/2021] [Accepted: 04/10/2021] [Indexed: 12/21/2022]
Abstract
Antimicrobial photodynamic therapy (aPDT) is promising for oral decontamination. Curcumin has been used as photosensitizer; however, the hydrophobic properties can negatively affect aPDT. This study evaluated the aPDT efficacy using Cur-loaded Pluronic® F-127 micelles against Streptococcus mutans and Candida albicans biofilms. Micelles characterization was performed by zeta potential, dynamic light scattering, transmission electron microscopy, absorption and fluorescence spectroscopy. Cur concentrations, cell viability by CFU mL-1 and confocal microscopy were determined. Data were analyzed by parametric and nonparametric tests under 5%. Cur-loaded Pluronic® F-127 exhibited spherical shape, suitable particle size (≤100 nm), adequate polydispersity index, best stability, lower photodegradation and autoaggregation compared to unloaded-Cur. Both microorganisms were sensitive to Cur-loaded Pluronic® F-127 micelles aPDT, with minimum inhibitory concentration (MIC) of 270 μm and 2.1093 μm for S. mutans and C. albicans suspended culture, respectively. Cur-loaded Pluronic® F-127 aPDT exhibited antibacterial/antifungal effect against the biofilms (~3 log10 reduction; P ≤ 0.05); however, similar to unloaded (P ≥ 0.05). Confocal images confirmed these results. Cur-loaded Pluronic® F-127 micelles exhibited good photo-chemical properties and may be a viable alternative to deliver Cur and to improve aPDT effect during the treatment of dental caries. Moreover, Pluronic® micelles can enhance the solubility, stability, permeability and control the release of Cur.
Collapse
Affiliation(s)
- Diego Dantas Lopes Dos Santos
- Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University - UNESP, Araraquara, São Paulo, Brazil
| | - João Felipe Besegato
- Department of Restorative Dentistry, School of Dentistry, São Paulo State University - UNESP, Araraquara, São Paulo, Brazil
| | - Priscila Borges Gobbo de Melo
- Department of Restorative Dentistry, School of Dentistry, São Paulo State University - UNESP, Araraquara, São Paulo, Brazil
| | - João Augusto Oshiro Junior
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University - UNESP, Araraquara, São Paulo, Brazil
| | - Marlus Chorilli
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University - UNESP, Araraquara, São Paulo, Brazil
| | - Dongmei Deng
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam - ACTA, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Vanderlei Salvador Bagnato
- Department of Physics and Materials Science, Physics Institute of São Carlos - IFSC, University of São Paulo - USP, São Carlos, São Paulo, Brazil
| | | |
Collapse
|
193
|
An AY, Choi KYG, Baghela AS, Hancock REW. An Overview of Biological and Computational Methods for Designing Mechanism-Informed Anti-biofilm Agents. Front Microbiol 2021; 12:640787. [PMID: 33927701 PMCID: PMC8076610 DOI: 10.3389/fmicb.2021.640787] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 03/23/2021] [Indexed: 12/29/2022] Open
Abstract
Bacterial biofilms are complex and highly antibiotic-resistant aggregates of microbes that form on surfaces in the environment and body including medical devices. They are key contributors to the growing antibiotic resistance crisis and account for two-thirds of all infections. Thus, there is a critical need to develop anti-biofilm specific therapeutics. Here we discuss mechanisms of biofilm formation, current anti-biofilm agents, and strategies for developing, discovering, and testing new anti-biofilm agents. Biofilm formation involves many factors and is broadly regulated by the stringent response, quorum sensing, and c-di-GMP signaling, processes that have been targeted by anti-biofilm agents. Developing new anti-biofilm agents requires a comprehensive systems-level understanding of these mechanisms, as well as the discovery of new mechanisms. This can be accomplished through omics approaches such as transcriptomics, metabolomics, and proteomics, which can also be integrated to better understand biofilm biology. Guided by mechanistic understanding, in silico techniques such as virtual screening and machine learning can discover small molecules that can inhibit key biofilm regulators. To increase the likelihood that these candidate agents selected from in silico approaches are efficacious in humans, they must be tested in biologically relevant biofilm models. We discuss the benefits and drawbacks of in vitro and in vivo biofilm models and highlight organoids as a new biofilm model. This review offers a comprehensive guide of current and future biological and computational approaches of anti-biofilm therapeutic discovery for investigators to utilize to combat the antibiotic resistance crisis.
Collapse
Affiliation(s)
| | | | | | - Robert E. W. Hancock
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
194
|
Multisubstituted pyrimidines effectively inhibit bacterial growth and biofilm formation of Staphylococcus aureus. Sci Rep 2021; 11:7931. [PMID: 33846401 PMCID: PMC8041844 DOI: 10.1038/s41598-021-86852-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 03/22/2021] [Indexed: 11/25/2022] Open
Abstract
Biofilms are multicellular communities of microorganisms that generally attach to surfaces in a self-produced matrix. Unlike planktonic cells, biofilms can withstand conventional antibiotics, causing significant challenges in the healthcare system. Currently, new chemical entities are urgently needed to develop novel anti-biofilm agents. In this study, we designed and synthesized a set of 2,4,5,6-tetrasubstituted pyrimidines and assessed their antibacterial activity against planktonic cells and biofilms formed by Staphylococcus aureus. Compounds 9e, 10d, and 10e displayed potent activity for inhibiting the onset of biofilm formation as well as for killing pre-formed biofilms of S. aureus ATCC 25923 and Newman strains, with half-maximal inhibitory concentration (IC50) values ranging from 11.6 to 62.0 µM. These pyrimidines, at 100 µM, not only decreased the number of viable bacteria within the pre-formed biofilm by 2–3 log10 but also reduced the amount of total biomass by 30–50%. Furthermore, these compounds were effective against planktonic cells with minimum inhibitory concentration (MIC) values lower than 60 µM for both staphylococcal strains. Compound 10d inhibited the growth of S. aureus ATCC 25923 in a concentration-dependent manner and displayed a bactericidal anti-staphylococcal activity. Taken together, our study highlights the value of multisubstituted pyrimidines to develop novel anti-biofilm agents.
Collapse
|
195
|
Bhandari S, Khadayat K, Poudel S, Shrestha S, Shrestha R, Devkota P, Khanal S, Marasini BP. Phytochemical analysis of medicinal plants of Nepal and their antibacterial and antibiofilm activities against uropathogenic Escherichia coli. BMC Complement Med Ther 2021; 21:116. [PMID: 33836728 PMCID: PMC8033659 DOI: 10.1186/s12906-021-03293-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 03/30/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND A biofilm is an extracellular polymeric substance (EPS) composed of polysaccharides, proteins, nucleic acids, and lipids that impede antibiotics and immune cells, thus providing a shielded environment for bacterial growth. Due to biofilm formation, some microbes can show up to 1000 fold increased resistance towards the antibiotics than the normal planktonic forms. The study was conducted to screen the crude extracts of medicinal plants used in Nepal for their in vitro antibiofilm activities. METHODS Total phenolic and total flavonoid contents were determined by using a Folin-Ciocalteau reagent and aluminium trichloride method, respectively. Resazurin assay was used to determine the minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC). The initial antibiofilm activities and their inhibitory concentration (IC50) values were determined by the microtiter based modified crystal violet staining method. RESULTS Out of 25 different plant extracts were used for the study, methanolic extracts of 20 plants showed a biofilm inhibition activity against five different strong biofilm producing Escherichia coli strains. Calotropis gigantea exhibited inhibition against all five different E. coli strains with IC50 values ranging from 299.7 ± 20.5 to 427.4 ± 2.7 μg/mL. Apart from that, Eclipta prostrata also showed biofilm formation inhibition, followed by Eupatorium adenophorum, Moringa oleifera, Ocimum tenuifolium, Oxalis lantifolia, Prunus persica, and Urtica parviflora. The extracts of C. gigantea, E. prostrata, Mangifera indica, O. tenuifolium, P. persica, and U. parviflora exhibited a moderate to poor MIC value ranging from 625 to 2500 μg/mL. The highest amount of phenolic content (TPC) was found in Acacia catechu followed by Morus alba, which was 38.9 and 25.1 mg gallic acid equivalents, respectively. The highest amount of flavonoid content was found in A. catechu followed by M. indica, which was 27.1 and 20.8 mg quercetin equivalents, respectively. CONCLUSION Extracts of C. gigantea, E. prostrata, P. persica, U. parviflora, and O. tenuifolium showed antibacterial as well as antibiofilm activity against pathogenic and strong biofilm producing E. coli. Thus, extracts or the pure compound from these medicinal plants could be used as antibiotics in the future.
Collapse
Affiliation(s)
- Sudip Bhandari
- Department of Biotechnology, National College, Tribhuvan University, Naya Bazar, Kathmandu, Nepal
| | - Karan Khadayat
- Department of Biotechnology, National College, Tribhuvan University, Naya Bazar, Kathmandu, Nepal
| | - Sami Poudel
- Department of Biotechnology, National College, Tribhuvan University, Naya Bazar, Kathmandu, Nepal
| | - Sunil Shrestha
- Department of Biotechnology, National College, Tribhuvan University, Naya Bazar, Kathmandu, Nepal
| | - Raju Shrestha
- Department of Microbiology, National College, Tribhuvan University, Naya Bazar, Kathmandu, Nepal
| | - Poonam Devkota
- Department of Biotechnology, National College, Tribhuvan University, Naya Bazar, Kathmandu, Nepal
| | - Santosh Khanal
- Department of Biotechnology, National College, Tribhuvan University, Naya Bazar, Kathmandu, Nepal
| | - Bishnu P Marasini
- Department of Biotechnology, National College, Tribhuvan University, Naya Bazar, Kathmandu, Nepal.
| |
Collapse
|
196
|
Pervin Z, Hassan MM. Synergistic therapeutic actions of antimicrobial peptides to treat multidrug-resistant bacterial infection. REVIEWS IN MEDICAL MICROBIOLOGY 2021; 32:83-89. [DOI: 10.1097/mrm.0000000000000239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
197
|
Rao H, Choo S, Rajeswari Mahalingam SR, Adisuri DS, Madhavan P, Md. Akim A, Chong PP. Approaches for Mitigating Microbial Biofilm-Related Drug Resistance: A Focus on Micro- and Nanotechnologies. Molecules 2021; 26:1870. [PMID: 33810292 PMCID: PMC8036581 DOI: 10.3390/molecules26071870] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/03/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023] Open
Abstract
Biofilms play an essential role in chronic and healthcare-associated infections and are more resistant to antimicrobials compared to their planktonic counterparts due to their (1) physiological state, (2) cell density, (3) quorum sensing abilities, (4) presence of extracellular matrix, (5) upregulation of drug efflux pumps, (6) point mutation and overexpression of resistance genes, and (7) presence of persister cells. The genes involved and their implications in antimicrobial resistance are well defined for bacterial biofilms but are understudied in fungal biofilms. Potential therapeutics for biofilm mitigation that have been reported include (1) antimicrobial photodynamic therapy, (2) antimicrobial lock therapy, (3) antimicrobial peptides, (4) electrical methods, and (5) antimicrobial coatings. These approaches exhibit promising characteristics for addressing the impending crisis of antimicrobial resistance (AMR). Recently, advances in the micro- and nanotechnology field have propelled the development of novel biomaterials and approaches to combat biofilms either independently, in combination or as antimicrobial delivery systems. In this review, we will summarize the general principles of clinically important microbial biofilm formation with a focus on fungal biofilms. We will delve into the details of some novel micro- and nanotechnology approaches that have been developed to combat biofilms and the possibility of utilizing them in a clinical setting.
Collapse
Affiliation(s)
- Harinash Rao
- School of Medicine, Taylor’s University, Subang Jaya, Selangor 47500, Malaysia; (H.R.); (D.S.A.); (P.M.)
| | - Sulin Choo
- School of Biosciences, Taylor’s University, Subang Jaya, Selangor 47500, Malaysia;
| | | | - Diajeng Sekar Adisuri
- School of Medicine, Taylor’s University, Subang Jaya, Selangor 47500, Malaysia; (H.R.); (D.S.A.); (P.M.)
| | - Priya Madhavan
- School of Medicine, Taylor’s University, Subang Jaya, Selangor 47500, Malaysia; (H.R.); (D.S.A.); (P.M.)
| | - Abdah Md. Akim
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor 43400, Malaysia
| | - Pei Pei Chong
- School of Biosciences, Taylor’s University, Subang Jaya, Selangor 47500, Malaysia;
| |
Collapse
|
198
|
Design of a novel antimicrobial peptide 1018M targeted ppGpp to inhibit MRSA biofilm formation. AMB Express 2021; 11:49. [PMID: 33770266 PMCID: PMC7997937 DOI: 10.1186/s13568-021-01208-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 03/15/2021] [Indexed: 12/11/2022] Open
Abstract
Methicillin-resistant staphylococcus aureus (MRSA) and its biofilm infection were considered as one of the main international health issues. There are still many challenges for treatment using traditional antibiotics. In this study, a mutant peptide of innate defense regulator (IDR-)1018 named 1018M was designed based on molecular docking and amino acid substitution technology. The antibacterial/biofilm activity and mechanisms against MRSA of 1018M were investigated for the first time. The minimum inhibitory concentration (MIC) of 1018M was reduced 1 time (MIC = 2 μg/mL) compared to IDR-1018. After treatment with 32 μg/mL 1018M for 24 h, the percentage of biofilm decreased by 78.9%, which was more effective than the parental peptide. The results of mechanisms exploration showed that 1018M was more potent than IDR-1018 at destructing bacterial cell wall, permeating cell membrane (20.4%–50.1% vs 1.45%–10.6%) and binding to stringent response signaling molecule ppGpp (increased 27.9%). Additionally, the peptides could also exert their activity by disrupting genomic DNA, regulating the expression of ppGpp metabolism and biofilm forming related genes (RSH, relP, relQ, rsbU, sigB, spA, codY, agrA and icaD). Moreover, the higher temperature, pH and pepsase stabilities provide 1018M better processing, storage and internal environmental tolerance. These data indicated that 1018M may be a potential candidate peptide for the treatment of MRSA and its biofilm infections.
Collapse
|
199
|
Thorat ND, Dworniczek E, Brennan G, Chodaczek G, Mouras R, Gascón Pérez V, Silien C, Tofail SAM, Bauer J. Photo-responsive functional gold nanocapsules for inactivation of community-acquired, highly virulent, multidrug-resistant MRSA. J Mater Chem B 2021; 9:846-856. [PMID: 33367418 DOI: 10.1039/d0tb02047h] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The indiscriminate and sporadic use of antibiotics has contributed to the emergence of drug resistance phenomenon in bacteria including but not limited to Staphylococcus aureus. These drug-resistant bacteria have been threatening safety in hospitals and adversely affecting human health. Here we report a strategy to design photo-stimulated theranostic nanoprobes against methicillin-resistant Staphylococcus aureus (MRSA) "superbug" USA300. The nanocapsule probe is based on gold nanorods (GNRs) coated with pegylated thiol, mPEG-SH, which has been further modified by adding successively a natural antibacterial compound such as curcumin, and a cell targeting deoxyribonucleic acid (DNA) aptamer. We have used this novel gold nanocapsules for near-infrared (NIR) photophysical stimulation against pathogenic bacteria. We have found that the novel nanocapsule blocks biofilm formation and kills bacteria by photothermal action that causes disruption of the bacterial cell wall and membrane. In this approach, multiple drug-resistant Staphylococcus aureus has been captured by these nanocapsules through DNA aptamer targeting. All of the trapped bacteria could be killed in 30 minutes during the NIR stimulation due to the combination of photothermal effect, the generation of reactive oxygen species (ROS) and a loss of transmembrane potential (Δψ). Importantly we did not notice any resistance developed against the photothermal treatment. This is remarkable from an anti-biofilm activity point of view. Importantly, these multifunctional nanocapsules have also shown a surface enhanced Raman spectroscopy (SERS) effect, which could be used to evaluate the success of the inactivation effect during treatment. These results indicate that nanocapsule-based photo treatment can be an alternative antibacterial strategy without contributing to antibiotic resistance, and thus can be used for both environmental and therapeutic applications.
Collapse
Affiliation(s)
- Nanasaheb D Thorat
- Department of Biomedical Engineering, Faculty of Fundamental Problems of Technology, Wroclaw University of Science and Technology, wybrzeże Stanisława Wyspiańskiego 27, Wrocław 50-370, Poland.
| | | | | | | | | | | | | | | | | |
Collapse
|
200
|
Xu M, Li L, Hu Q. The recent progress in photothermal-triggered bacterial eradication. Biomater Sci 2021; 9:1995-2008. [PMID: 33564803 DOI: 10.1039/d0bm02057e] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Increasing evidence suggested that bacterial infection diseases posed a great threat to human health and became the leading cause of mortality. However, the abuse of antibiotics and their residues in the environment result in the emergence and prevalence of drug-resistant bacteria. Photothermal therapy (PTT) has received considerable attention owing to its noninvasiveness, and proved to be promising in preventing bacterial infection diseases. In this review, we first surveyed the recent progress of PTT-based responsive targeting strategies for bacterial killing. We then highlighted the PTT-based smart designs of bio-films, hydrogels and synergistic methods for treating bacterial infections. Existing challenges and perspectives are also discussed to inspire the future development of a PTT-based platform for the efficient therapy of bacterial infections.
Collapse
Affiliation(s)
- Minjie Xu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China.
| | | | | |
Collapse
|