1
|
Patra S, Saha S, Singh R, Tomar N, Gulati P. Biofilm battleground: Unveiling the hidden challenges, current approaches and future perspectives in combating biofilm associated bacterial infections. Microb Pathog 2025; 198:107155. [PMID: 39586337 DOI: 10.1016/j.micpath.2024.107155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/09/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024]
Abstract
A biofilm is a complex aggregation of microorganisms, either of the same or different species, that adhere to a surface and are encased in an extracellular polymeric substances (EPS) matrix. Quorum sensing (QS) and biofilm formation are closely linked, as QS genes regulate the development, maturation, and breakdown of biofilms. Inhibiting QS can be utilized as an effective approach to combat the impacts of biofilm infection. The impact of biofilms includes chronic infections, industrial biofouling, infrastructure corrosion, and environmental contamination as well. Therefore, a deep understanding of biofilms is crucial for enhancing public health, advancing industrial processes, safeguarding the environment, and deepening our knowledge of microbial life as well. This review aims to offer a comprehensive examination of challenges posed by bacterial biofilms, contemporary approaches and strategies for effectively eliminating biofilms, including the inhibition of quorum sensing pathways, while also focusing on emerging technologies and techniques for biofilm treatment. In addition, future research is projected to target the challenges associated with the bacterial biofilms, striving to develop new approaches and improve existing strategies for their effective control and eradication.
Collapse
Affiliation(s)
- Sandeep Patra
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Sumana Saha
- Gujarat Biotechnology University, Gandhinagar, Gujarat, India
| | - Randhir Singh
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Nandini Tomar
- Department of Biotechnology, South Asian University, New Delhi, India
| | - Pallavi Gulati
- Ram Lal Anand College, University of Delhi, New Delhi, India.
| |
Collapse
|
2
|
Alrata L, Abdulsattar D, Madrigal S, Pyeatte SR, Zaghloul M, Abu-Amer W, Arif B, Alhamad T, Remedi M, Lin Y, Zayed MA. Alginate Formulation for Wound Healing Applications. Adv Wound Care (New Rochelle) 2024. [PMID: 39531216 DOI: 10.1089/wound.2024.0081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Significance: Alginate, sourced from seaweed, holds significant importance in industrial and biomedical domains due to its versatile properties. Its chemical composition, primarily comprising β-D-mannuronic acid and α-L-guluronic acid, governs its physical and biological attributes. This polysaccharide, extracted from brown algae and bacteria, offers diverse compositions impacting key factors such as molecular weight, flexibility, solubility, and stability. Recent Advances: Commercial extraction methods yield soluble sodium alginate essential for various biomedical applications. Extraction processes involve chemical treatments converting insoluble alginic acid salts into soluble forms. While biosynthesis pathways in bacteria and algae share similarities, differences in enzyme utilization and product characteristics are noted. Critical Issues: Despite its widespread applicability, challenges persist regarding alginate's stability, biodegradability, and bioactivity. Further understanding of its interactions in complex biological environments and the optimization of extraction and synthesis processes are imperative. Additionally, concerns regarding immune responses to alginate-based implants necessitate thorough investigation. Future Directions: Future research endeavors aim to enhance alginate's stability and bioactivity, facilitating its broader utilization in regenerative medicine and therapeutic interventions. Novel approaches focusing on tailored hydrogel formations, advanced drug delivery systems, and optimized cellular encapsulation techniques hold promise. Continued exploration of alginate's potential in tissue engineering and wound healing, alongside efforts to address critical issues, will drive advancements in biomedical applications.
Collapse
Affiliation(s)
- Louai Alrata
- Department of Surgery, Section of Vascular Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- CardioVascular Research Innovation in Surgery and Engineering Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Dahlia Abdulsattar
- Department of Surgery, Section of Vascular Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- CardioVascular Research Innovation in Surgery and Engineering Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sabrina Madrigal
- Department of Surgery, Section of Vascular Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- CardioVascular Research Innovation in Surgery and Engineering Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sophia R Pyeatte
- Department of Surgery, Section of Vascular Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- CardioVascular Research Innovation in Surgery and Engineering Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Mohamed Zaghloul
- Department of Surgery, Section of Vascular Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- CardioVascular Research Innovation in Surgery and Engineering Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Wahid Abu-Amer
- Department of Surgery, Section of Vascular Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- CardioVascular Research Innovation in Surgery and Engineering Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Batool Arif
- Department of Surgery, Section of Vascular Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- CardioVascular Research Innovation in Surgery and Engineering Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tarek Alhamad
- Department of Medicine, Division of Nephrology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Maria Remedi
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yiing Lin
- Department of Surgery, Section of Transplant Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Mohamed A Zayed
- Department of Surgery, Section of Vascular Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- CardioVascular Research Innovation in Surgery and Engineering Center, Washington University School of Medicine, St. Louis, Missouri, USA
- Division of Molecular Cell Biology, Washington University School of Medicine, St. Louis, Missouri, USA
- Division of Surgical Sciences, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University McKelvey School of Engineering, St. Louis, Missouri, USA
| |
Collapse
|
3
|
Alqasmi M. Therapeutic Interventions for Pseudomonas Infections in Cystic Fibrosis Patients: A Review of Phase IV Trials. J Clin Med 2024; 13:6530. [PMID: 39518670 PMCID: PMC11547045 DOI: 10.3390/jcm13216530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Pseudomonas aeruginosa (Pa) poses a significant threat to individuals with cystic fibrosis (CF), as this bacterium is highly adaptable and resistant to antibiotics. While early-stage Pa infections can often be eradicated with aggressive antibiotic therapy, chronic infections are nearly impossible to eliminate and require treatments that focus on long-term bacterial suppression. Without such suppression, these persistent infections can severely damage the lungs, leading to serious complications and a reduced life expectancy for CF patients. Evidence for a specific treatment regimen for managing Pa infections in CF patients remains limited. This narrative review provides a detailed analysis of antimicrobial therapies assessed in completed phase IV trials, focusing on their safety and efficacy, especially with prolonged use. Key antibiotics, including tobramycin, colistin, meropenem, aztreonam, ceftolozane/tazobactam, ciprofloxacin, and azithromycin, are discussed, emphasizing their use, side effects, and delivery methods. Inhaled antibiotics are preferred for their targeted action and minimal side effects, while systemic antibiotics offer potency but carry risks like nephrotoxicity. The review also explores emerging treatments, such as phage therapy and antibiofilm agents, which show promise in managing chronic infections. Nonetheless, further research is necessary to enhance the safety and effectiveness of existing therapies while investigating new approaches for better long-term outcomes.
Collapse
Affiliation(s)
- Mohammed Alqasmi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia
| |
Collapse
|
4
|
Zhang Y, Wang X, Liang Y, Zhang L, Fan J, Yang Y. A Semisynthetic Oligomannuronic Acid-Based Glycoconjugate Vaccine against Pseudomonas aeruginosa. ACS CENTRAL SCIENCE 2024; 10:1515-1523. [PMID: 39220693 PMCID: PMC11363335 DOI: 10.1021/acscentsci.4c00387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/24/2024] [Accepted: 07/01/2024] [Indexed: 09/04/2024]
Abstract
Pseudomonas aeruginosa is one of the leading causes of nosocomial infections and has become increasingly resistant to multiple antibiotics. However, development of novel classes of antibacterial agents against multidrug-resistant P. aeruginosa is extremely difficult. Herein we develop a semisynthetic oligomannuronic acid-based glycoconjugate vaccine that confers broad protection against infections of both mucoid and nonmucoid strains of P. aeruginosa. The well-defined glycoconjugate vaccine formulated with Freund's adjuvant (FA) employing a highly conserved antigen elicited a strong and specific immune response and protected mice against both mucoid and nonmucoid strains of P. aeruginosa. The resulting antibodies recognized different strains of P. aeruginosa and mediated the opsonic killing of the bacteria at varied levels depending on the amount of alginate expressed on the surface of the strains. Vaccination with the glycoconjugate vaccine plus FA significantly promoted the pulmonary and blood clearance of the mucoid PAC1 strain of P. aeruginosa and considerably improved the survival rates of mice against the nonmucoid PAO1 strain of P. aeruginosa. Thus, the semisynthetic glycoconjugate is a promising vaccine that may provide broad protection against both types of P. aeruginosa.
Collapse
Affiliation(s)
- Yiyue Zhang
- Shanghai
Frontiers Science Center of Optogenetic Techniques for Cell Metabolism,
Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Xiaotong Wang
- Shanghai
Frontiers Science Center of Optogenetic Techniques for Cell Metabolism,
Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Youling Liang
- Shanghai
Frontiers Science Center of Optogenetic Techniques for Cell Metabolism,
Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Liangliang Zhang
- Shanghai
Frontiers Science Center of Optogenetic Techniques for Cell Metabolism,
Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Jiahao Fan
- Engineering
Research Center of Pharmaceutical Process Chemistry, Ministry of Education, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - You Yang
- Shanghai
Frontiers Science Center of Optogenetic Techniques for Cell Metabolism,
Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
- Engineering
Research Center of Pharmaceutical Process Chemistry, Ministry of Education, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| |
Collapse
|
5
|
Liu J, Wang Z, Zeng Y, Wang W, Tang S, Jia A. 1H-Pyrrole-2,5-dicarboxylic acid, a quorum sensing inhibitor from one endophytic fungus in Areca catechu L., acts as antibiotic accelerant against Pseudomonas aeruginosa. Front Cell Infect Microbiol 2024; 14:1413728. [PMID: 39015339 PMCID: PMC11250523 DOI: 10.3389/fcimb.2024.1413728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 05/27/2024] [Indexed: 07/18/2024] Open
Abstract
Pseudomonas aeruginosa has already been stipulated as a "critical" pathogen, emphasizing the urgent need for researching and developing novel antibacterial agents due to multidrug resistance. Bacterial biofilm formation facilitates cystic fibrosis development and restricts the antibacterial potential of many current antibiotics. The capacity of P. aeruginosa to form biofilms and resist antibiotics is closely correlated with quorum sensing (QS). Bacterial QS is being contemplated as a promising target for developing novel antibacterial agents. QS inhibitors are a promising strategy for treating chronic infections. This study reported that the active compound PT22 (1H-pyrrole-2,5-dicarboxylic acid) isolated from Perenniporia tephropora FF2, one endophytic fungus from Areca catechu L., presents QS inhibitory activity against P. aeruginosa. Combined with gentamycin or piperacillin, PT22 functions as a novel antibiotic accelerant against P. aeruginosa. PT22 (0.50 mg/mL, 0.75 mg/mL, and 1.00 mg/mL) reduces the production of QS-related virulence factors, such as pyocyanin and rhamnolipid, and inhibits biofilm formation of P. aeruginosa PAO1 instead of affecting its growth. The architectural disruption of the biofilms was confirmed by visualization through scanning electron microscopy (SEM) and confocal laser scanning microscopy (CLSM). Real-time quantitative PCR (RT-qPCR) indicated that PT22 significantly attenuated the expression of QS-related genes followed by docking analysis of molecules against QS activator proteins. PT22 dramatically increased the survival rate of Galleria mellonella. PT22 combined with gentamycin or piperacillin presents significant inhibition of biofilm formation and eradication of mature biofilm compared to monotherapy, which was also confirmed by visualization through SEM and CLSM. After being treated with PT22 combined with gentamycin or piperacillin, the survival rates of G. mellonella were significantly increased compared to those of monotherapy. PT22 significantly enhanced the susceptibility of gentamycin and piperacillin against P. aeruginosa PAO1. Our results suggest that PT22 from P. tephropora FF2 as a potent QS inhibitor is a candidate antibiotic accelerant to combat the antibiotic resistance of P. aeruginosa.
Collapse
Affiliation(s)
- Junsheng Liu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Zhennan Wang
- Modern Industrial College of Traditional Chinese Medicine and Health, Lishui University, Lishui, China
| | - Yuexiang Zeng
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Wei Wang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Shi Tang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Aiqun Jia
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
6
|
Zhu Y, Wang X, Lu S, Zheng J, Liang Y, Zhang L, Fang P, Xu P, Yu B, Yang Y. Microwave-assisted synthesis of highly sulfated mannuronate glycans as potential inhibitors against SARS-CoV-2. Org Biomol Chem 2024; 22:3986-3994. [PMID: 38695061 DOI: 10.1039/d4ob00466c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Algae-based marine carbohydrate drugs are typically decorated with negative ion groups such as carboxylate and sulfate groups. However, the precise synthesis of highly sulfated alginates is challenging, thus impeding their structure-activity relationship studies. Herein we achieve a microwave-assisted synthesis of a range of highly sulfated mannuronate glycans with up to 17 sulfation sites by overcoming the incomplete sulfation due to the electrostatic repulsion of crowded polyanionic groups. Although the partially sulfated tetrasaccharide had the highest affinity for the receptor binding domain (RBD) of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant, the fully sulfated octasaccharide showed the most potent interference with the binding of the RBD to angiotensin-converting enzyme 2 (ACE2) and Vero E6 cells, indicating that the sulfated oligosaccharides might inhibit the RBD binding to ACE2 in a length-dependent manner.
Collapse
Affiliation(s)
- Yirong Zhu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| | - Xiaotong Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| | - Siqian Lu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| | - Jibin Zheng
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| | - Youling Liang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| | - Liangliang Zhang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| | - Pengfei Fang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Peng Xu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - Biao Yu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
| | - You Yang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.
| |
Collapse
|
7
|
Manna S, Gupta P, Nandi G, Jana S. Recent update on alginate based promising transdermal drug delivery systems. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2023; 34:2291-2318. [PMID: 37368494 DOI: 10.1080/09205063.2023.2230847] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/13/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023]
Abstract
Alongside oral delivery of therapeutics, transdermal delivery systems have gained increased patient acceptability over past few decades. With increasing popularity, novel techniques were employed for transdermal drug targeting which involves microneedle patches, transdermal films and hydrogel based formulations. Hydrogel forming ability along with other rheological behaviour makes natural polysaccharides an attractive option for transdermal use. Being a marine originated anionic polysaccharide, alginates are widely used in pharmaceutical, cosmetics and food industries. Alginate possesses excellent biodegradability, biocompatibility and mucoadhesive properties. Owing to many favourable properties required for transdermal drug delivery systems (TDDS), the application of alginates are increasing in recent times. This review summarizes the source and properties of alginate along with several transdermal delivery techniques including the application of alginate for respective transdermal systems.
Collapse
Affiliation(s)
- Sreejan Manna
- Department of Pharmaceutical Technology, Brainware University, Kolkata, West Bengal, India
| | - Prajna Gupta
- Division of Pharmaceutics, Department of Pharmaceutical Technology, University of North Bengal, Darjeeling, West Bengal, India
| | - Gouranga Nandi
- Division of Pharmaceutics, Department of Pharmaceutical Technology, University of North Bengal, Darjeeling, West Bengal, India
| | - Sougata Jana
- Department of Pharmaceutics, Gupta College of Technological Sciences, Asansol, West Bengal, India
- Department of Health and Family Welfare, Directorate of Health Services, Kolkata, India
| |
Collapse
|
8
|
Rønne ME, Tandrup T, Madsen M, Hunt CJ, Myers PN, Moll JM, Holck J, Brix S, Strube ML, Aachmann FL, Wilkens C, Svensson B. Three alginate lyases provide a new gut Bacteroides ovatus isolate with the ability to grow on alginate. Appl Environ Microbiol 2023; 89:e0118523. [PMID: 37791757 PMCID: PMC10617595 DOI: 10.1128/aem.01185-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 08/03/2023] [Indexed: 10/05/2023] Open
Abstract
Humans consume alginate in the form of seaweed, food hydrocolloids, and encapsulations, making the digestion of this mannuronic acid (M) and guluronic acid (G) polymer of key interest for human health. To increase knowledge on alginate degradation in the gut, a gene catalog from human feces was mined for potential alginate lyases (ALs). The predicted ALs were present in nine species of the Bacteroidetes phylum, of which two required supplementation of an endo-acting AL, expected to mimic cross-feeding in the gut. However, only a new isolate grew on alginate. Whole-genome sequencing of this alginate-utilizing isolate suggested that it is a new Bacteroides ovatus strain harboring a polysaccharide utilization locus (PUL) containing three ALs of families: PL6, PL17, and PL38. The BoPL6 degraded polyG to oligosaccharides of DP 1-3, and BoPL17 released 4,5-unsaturated monouronate from polyM. BoPL38 degraded both alginates, polyM, polyG, and polyMG, in endo-mode; hence, it was assumed to deliver oligosaccharide substrates for BoPL6 and BoPL17, corresponding well with synergistic action on alginate. BoPL17 and BoPL38 crystal structures, determined at 1.61 and 2.11 Å, respectively, showed (α/α)6-barrel + anti-parallel β-sheet and (α/α)7-barrel folds, distinctive for these PL families. BoPL17 had a more open active site than the two homologous structures. BoPL38 was very similar to the structure of an uncharacterized PL38, albeit with a different triad of residues possibly interacting with substrate in the presumed active site tunnel. Altogether, the study provides unique functional and structural insights into alginate-degrading lyases of a PUL in a human gut bacterium.IMPORTANCEHuman ingestion of sustainable biopolymers calls for insight into their utilization in our gut. Seaweed is one such resource with alginate, a major cell wall component, used as a food hydrocolloid and for encapsulation of pharmaceuticals and probiotics. Knowledge is sparse on the molecular basis for alginate utilization in the gut. We identified a new Bacteroides ovatus strain from human feces that grew on alginate and encoded three alginate lyases in a gene cluster. BoPL6 and BoPL17 show complementary specificity toward guluronate (G) and mannuronate (M) residues, releasing unsaturated oligosaccharides and monouronic acids. BoPL38 produces oligosaccharides degraded by BoPL6 and BoPL17 from both alginates, G-, M-, and MG-substrates. Enzymatic and structural characterization discloses the mode of action and synergistic degradation of alginate by these alginate lyases. Other bacteria were cross-feeding on alginate oligosaccharides produced by an endo-acting alginate lyase. Hence, there is an interdependent community in our guts that can utilize alginate.
Collapse
Affiliation(s)
- Mette E. Rønne
- Department of Biotechnology and Biomedicine, Enzyme and Protein Chemistry, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Tobias Tandrup
- Department of Biotechnology and Biomedicine, Enzyme and Protein Chemistry, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Mikkel Madsen
- Department of Biotechnology and Biomedicine, Enzyme and Protein Chemistry, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Cameron J. Hunt
- Department of Biotechnology and Biomedicine, Enzyme Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Pernille N. Myers
- Department of Biotechnology and Biomedicine, Disease Systems Immunology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Janne M. Moll
- Department of Biotechnology and Biomedicine, Disease Systems Immunology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Jesper Holck
- Department of Biotechnology and Biomedicine, Enzyme Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Susanne Brix
- Department of Biotechnology and Biomedicine, Disease Systems Immunology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Mikael L. Strube
- Department of Biotechnology and Biomedicine, Bacterial Ecophysiology and Biotechnology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Finn L. Aachmann
- Department of Biotechnology and Food Science, Norwegian Biopolymer Laboratory (NOBIPOL), NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Casper Wilkens
- Department of Biotechnology and Biomedicine, Enzyme Technology, Technical University of Denmark, Kgs. Lyngby, Denmark
- Department of Biotechnology and Biomedicine, Structural Enzymology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Birte Svensson
- Department of Biotechnology and Biomedicine, Enzyme and Protein Chemistry, Technical University of Denmark, Kgs. Lyngby, Denmark
| |
Collapse
|
9
|
Sánchez-Jiménez A, Llamas MA, Marcos-Torres FJ. Transcriptional Regulators Controlling Virulence in Pseudomonas aeruginosa. Int J Mol Sci 2023; 24:11895. [PMID: 37569271 PMCID: PMC10418997 DOI: 10.3390/ijms241511895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/21/2023] [Accepted: 07/22/2023] [Indexed: 08/13/2023] Open
Abstract
Pseudomonas aeruginosa is a pathogen capable of colonizing virtually every human tissue. The host colonization competence and versatility of this pathogen are powered by a wide array of virulence factors necessary in different steps of the infection process. This includes factors involved in bacterial motility and attachment, biofilm formation, the production and secretion of extracellular invasive enzymes and exotoxins, the production of toxic secondary metabolites, and the acquisition of iron. Expression of these virulence factors during infection is tightly regulated, which allows their production only when they are needed. This process optimizes host colonization and virulence. In this work, we review the intricate network of transcriptional regulators that control the expression of virulence factors in P. aeruginosa, including one- and two-component systems and σ factors. Because inhibition of virulence holds promise as a target for new antimicrobials, blocking the regulators that trigger the production of virulence determinants in P. aeruginosa is a promising strategy to fight this clinically relevant pathogen.
Collapse
Affiliation(s)
| | - María A. Llamas
- Department of Biotechnology and Environmental Protection, Estación Experimental del Zaidín-Consejo Superior de Investigaciones Científicas, 18008 Granada, Spain;
| | - Francisco Javier Marcos-Torres
- Department of Biotechnology and Environmental Protection, Estación Experimental del Zaidín-Consejo Superior de Investigaciones Científicas, 18008 Granada, Spain;
| |
Collapse
|
10
|
Blanco-Romero E, Durán D, Garrido-Sanz D, Redondo-Nieto M, Martín M, Rivilla R. Adaption of Pseudomonas ogarae F113 to the Rhizosphere Environment-The AmrZ-FleQ Hub. Microorganisms 2023; 11:microorganisms11041037. [PMID: 37110460 PMCID: PMC10146422 DOI: 10.3390/microorganisms11041037] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/10/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Motility and biofilm formation are two crucial traits in the process of rhizosphere colonization by pseudomonads. The regulation of both traits requires a complex signaling network that is coordinated by the AmrZ-FleQ hub. In this review, we describe the role of this hub in the adaption to the rhizosphere. The study of the direct regulon of AmrZ and the phenotypic analyses of an amrZ mutant in Pseudomonas ogarae F113 has shown that this protein plays a crucial role in the regulation of several cellular functions, including motility, biofilm formation, iron homeostasis, and bis-(3'-5')-cyclic dimeric guanosine monophosphate (c-di-GMP) turnover, controlling the synthesis of extracellular matrix components. On the other hand, FleQ is the master regulator of flagellar synthesis in P. ogarae F113 and other pseudomonads, but its implication in the regulation of multiple traits related with environmental adaption has been shown. Genomic scale studies (ChIP-Seq and RNA-Seq) have shown that in P. ogarae F113, AmrZ and FleQ are general transcription factors that regulate multiple traits. It has also been shown that there is a common regulon shared by the two transcription factors. Moreover, these studies have shown that AmrZ and FleQ form a regulatory hub that inversely regulate traits such as motility, extracellular matrix component production, and iron homeostasis. The messenger molecule c-di-GMP plays an essential role in this hub since its production is regulated by AmrZ and it is sensed by FleQ and required for its regulatory role. This regulatory hub is functional both in culture and in the rhizosphere, indicating that the AmrZ-FleQ hub is a main player of P. ogarae F113 adaption to the rhizosphere environment.
Collapse
Affiliation(s)
- Esther Blanco-Romero
- Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, Darwin 2, 28049 Madrid, Spain
| | - David Durán
- Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, Darwin 2, 28049 Madrid, Spain
| | - Daniel Garrido-Sanz
- Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, Darwin 2, 28049 Madrid, Spain
- Department of Fundamental Microbiology, University of Lausanne, 1015 Lausanne, Switzerland
| | - Miguel Redondo-Nieto
- Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, Darwin 2, 28049 Madrid, Spain
| | - Marta Martín
- Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, Darwin 2, 28049 Madrid, Spain
| | - Rafael Rivilla
- Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, Darwin 2, 28049 Madrid, Spain
| |
Collapse
|
11
|
Elmassry MM, Colmer-Hamood JA, Kopel J, San Francisco MJ, Hamood AN. Anti- Pseudomonas aeruginosa Vaccines and Therapies: An Assessment of Clinical Trials. Microorganisms 2023; 11:916. [PMID: 37110338 PMCID: PMC10144840 DOI: 10.3390/microorganisms11040916] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Pseudomonas aeruginosa is a Gram-negative opportunistic pathogen that causes high morbidity and mortality in cystic fibrosis (CF) and immunocompromised patients, including patients with ventilator-associated pneumonia (VAP), severely burned patients, and patients with surgical wounds. Due to the intrinsic and extrinsic antibiotic resistance mechanisms, the ability to produce several cell-associated and extracellular virulence factors, and the capacity to adapt to several environmental conditions, eradicating P. aeruginosa within infected patients is difficult. Pseudomonas aeruginosa is one of the six multi-drug-resistant pathogens (ESKAPE) considered by the World Health Organization (WHO) as an entire group for which the development of novel antibiotics is urgently needed. In the United States (US) and within the last several years, P. aeruginosa caused 27% of deaths and approximately USD 767 million annually in health-care costs. Several P. aeruginosa therapies, including new antimicrobial agents, derivatives of existing antibiotics, novel antimicrobial agents such as bacteriophages and their chelators, potential vaccines targeting specific virulence factors, and immunotherapies have been developed. Within the last 2-3 decades, the efficacy of these different treatments was tested in clinical and preclinical trials. Despite these trials, no P. aeruginosa treatment is currently approved or available. In this review, we examined several of these clinicals, specifically those designed to combat P. aeruginosa infections in CF patients, patients with P. aeruginosa VAP, and P. aeruginosa-infected burn patients.
Collapse
Affiliation(s)
- Moamen M. Elmassry
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Jane A. Colmer-Hamood
- Department of Medical Education, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Jonathan Kopel
- Department of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Michael J. San Francisco
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Honors College, Texas Tech University, Lubbock, TX 79409, USA
| | - Abdul N. Hamood
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
12
|
Gonçalves ASC, Leitão MM, Simões M, Borges A. The action of phytochemicals in biofilm control. Nat Prod Rep 2023; 40:595-627. [PMID: 36537821 DOI: 10.1039/d2np00053a] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Covering: 2009 to 2021Antimicrobial resistance is now rising to dangerously high levels in all parts of the world, threatening the treatment of an ever-increasing range of infectious diseases. This has becoming a serious public health problem, especially due to the emergence of multidrug-resistance among clinically important bacterial species and their ability to form biofilms. In addition, current anti-infective therapies have low efficacy in the treatment of biofilm-related infections, leading to recurrence, chronicity, and increased morbidity and mortality. Therefore, it is necessary to search for innovative strategies/antibacterial agents capable of overcoming the limitations of conventional antibiotics. Natural compounds, in particular those obtained from plants, have been exhibiting promising properties in this field. Plant secondary metabolites (phytochemicals) can act as antibiofilm agents through different mechanisms of action from the available antibiotics (inhibition of quorum-sensing, motility, adhesion, and reactive oxygen species production, among others). The combination of different phytochemicals and antibiotics have revealed synergistic or additive effects in biofilm control. This review aims to bring together the most relevant reports on the antibiofilm properties of phytochemicals, as well as insights into their structure and mechanistic action against bacterial pathogens, spanning December 2008 to December 2021.
Collapse
Affiliation(s)
- Ariana S C Gonçalves
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| | - Miguel M Leitão
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| | - Manuel Simões
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| | - Anabela Borges
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| |
Collapse
|
13
|
Agles AA, Bourg IC. Structure-Thermodynamic Relationship of a Polysaccharide Gel (Alginate) as a Function of Water Content and Counterion Type (Na vs Ca). J Phys Chem B 2023; 127:1828-1841. [PMID: 36791328 PMCID: PMC10159261 DOI: 10.1021/acs.jpcb.2c07129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/31/2023] [Indexed: 02/17/2023]
Abstract
Biofilms are the predominant mode of microbial life on Earth, and so a deep understanding of microbial communities─and their impacts on environmental processes─requires a firm understanding of biofilm properties. Because of the importance of biofilms to their microbial inhabitants, microbes have evolved different ways of engineering and reconfiguring the matrix of extracellular polymeric substances (EPS) that constitute the main non-living component of biofilms. This ability makes it difficult to distinguish between the biotic and abiotic origins of biofilm properties. An important route toward establishing this distinction has been the study of simplified models of the EPS matrix. This study builds on such efforts by using atomistic simulations to predict the nanoscale (≤10 nm scale) structure of a model EPS matrix and the sensitivity of this structure to interpolymer interactions and water content. To accomplish this, we use replica exchange molecular dynamics (REMD) simulations to generate all-atom configurations of ten 3.4 kDa alginate polymers at a range of water contents and Ca-Na ratios. Simulated systems are solvated with explicitly modeled water molecules, which allows us to capture the discrete structure of the hydrating water and to examine the thermodynamic stability of water in the gels as they are progressively dehydrated. Our primary findings are that (i) the structure of the hydrogels is highly sensitive to the identity of the charge-compensating cations, (ii) the thermodynamics of water within the gels (specific enthalpy and free energy) are, surprisingly, only weakly sensitive to cation identity, and (iii) predictions of the differential enthalpy and free energy of hydration include a short-ranged enthalpic term that promotes hydration and a longer-ranged (presumably entropic) term that promotes dehydration, where short and long ranges refer to distances shorter or longer than ∼0.6 nm between alginate strands.
Collapse
Affiliation(s)
- Avery A. Agles
- Department
of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Ian C. Bourg
- Department
of Civil and Environmental Engineering and High Meadows Environmental
Institute, Princeton University, Princeton, New Jersey 08544, United States
| |
Collapse
|
14
|
Rossetto V, Moore-Machacek A, Woods DF, Galvão HM, Shanahan RM, Hickey A, O'Leary N, O'Gara F, McGlacken GP, Reen FJ. Structural modification of the Pseudomonas aeruginosa alkylquinoline cell-cell communication signal, HHQ, leads to benzofuranoquinolines with anti-virulence behaviour in ESKAPE pathogens. MICROBIOLOGY (READING, ENGLAND) 2023; 169. [PMID: 36862576 DOI: 10.1099/mic.0.001303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Microbial populations have evolved intricate networks of negotiation and communication through which they can coexist in natural and host ecosystems. The nature of these systems can be complex and they are, for the most part, poorly understood at the polymicrobial level. The Pseudomonas Quinolone Signal (PQS) and its precursor 4-hydroxy-2-heptylquinoline (HHQ) are signal molecules produced by the important nosocomial pathogen
Pseudomonas aeruginosa
. They are known to modulate the behaviour of co-colonizing bacterial and fungal pathogens such as Bacillus atropheaus, Candida albicans and Aspergillus fumigatus. While the structural basis for alkyl-quinolone signalling within
P. aeruginosa
has been studied extensively, less is known about how structural derivatives of these molecules can influence multicellular behaviour and population-level decision-making in other co-colonizing organisms. In this study, we investigated a suite of small molecules derived initially from the HHQ framework, for anti-virulence activity against ESKAPE pathogens, at the species and strain levels. Somewhat surprisingly, with appropriate substitution, loss of the alkyl chain (present in HHQ and PQS) did not result in a loss of activity, presenting a more easily accessible synthetic framework for investigation. Virulence profiling uncovered significant levels of inter-strain variation among the responses of clinical and environmental isolates to small-molecule challenge. While several lead compounds were identified in this study, further work is needed to appreciate the extent of strain-level tolerance to small-molecule anti-infectives among pathogenic organisms.
Collapse
Affiliation(s)
- Veronica Rossetto
- Faculty of Science and Technology, Universidade do Algarve, Algarve, Portugal.,School of Microbiology, University College Cork, Cork, Ireland
| | | | - David F Woods
- School of Microbiology, University College Cork, Cork, Ireland
| | - Helena M Galvão
- Faculty of Science and Technology, Universidade do Algarve, Algarve, Portugal
| | - Rachel M Shanahan
- School of Chemistry and Analytical and Biological Chemistry Research Facility, University College Cork, Cork, Ireland
| | - Aobha Hickey
- School of Chemistry and Analytical and Biological Chemistry Research Facility, University College Cork, Cork, Ireland
| | - Niall O'Leary
- School of Microbiology, University College Cork, Cork, Ireland
| | - Fergal O'Gara
- School of Microbiology, University College Cork, Cork, Ireland.,Biomerit Research Centre, School of Microbiology, University College Cork, Cork, Ireland.,Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Gerard P McGlacken
- School of Chemistry and Analytical and Biological Chemistry Research Facility, University College Cork, Cork, Ireland.,Synthesis and Solid State Pharmaceutical Centre, University College Cork, Cork, Ireland
| | - F Jerry Reen
- School of Microbiology, University College Cork, Cork, Ireland.,Synthesis and Solid State Pharmaceutical Centre, University College Cork, Cork, Ireland
| |
Collapse
|
15
|
Toyos-Rodríguez C, Valero-Calvo D, de la Escosura-Muñiz A. Advances in the screening of antimicrobial compounds using electrochemical biosensors: is there room for nanomaterials? Anal Bioanal Chem 2023; 415:1107-1121. [PMID: 36445455 PMCID: PMC9707421 DOI: 10.1007/s00216-022-04449-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/11/2022] [Accepted: 11/17/2022] [Indexed: 11/30/2022]
Abstract
The abusive use of antimicrobial compounds and the associated appearance of antimicrobial resistant strains are a major threat to human health. An improved antimicrobial administration involves a faster diagnosis and detection of resistances. Antimicrobial susceptibility testing (AST) are the reference techniques for this purpose, relying mainly in the use of culture techniques. The long time required for analysis and the lack of reproducibility of these techniques have fostered the development of high-throughput AST methods, including electrochemical biosensors. In this review, recent electrochemical methods used in AST have been revised, with particular attention on those used for the evaluation of new drug candidates. The role of nanomaterials in these biosensing platforms has also been questioned, inferring that it is of minor importance compared to other applications.
Collapse
Affiliation(s)
- Celia Toyos-Rodríguez
- NanoBioAnalysis Group, Department of Physical and Analytical Chemistry, University of Oviedo, Julián Clavería 8, 33006, Oviedo, Spain
- Biotechnology Institute of Asturias, University of Oviedo, Santiago Gascon Building, 33006, Oviedo, Spain
| | - David Valero-Calvo
- NanoBioAnalysis Group, Department of Physical and Analytical Chemistry, University of Oviedo, Julián Clavería 8, 33006, Oviedo, Spain
- Biotechnology Institute of Asturias, University of Oviedo, Santiago Gascon Building, 33006, Oviedo, Spain
| | - Alfredo de la Escosura-Muñiz
- NanoBioAnalysis Group, Department of Physical and Analytical Chemistry, University of Oviedo, Julián Clavería 8, 33006, Oviedo, Spain.
- Biotechnology Institute of Asturias, University of Oviedo, Santiago Gascon Building, 33006, Oviedo, Spain.
| |
Collapse
|
16
|
Zhang L, Zhang Y, Hua Q, Xu T, Liu J, Zhu Y, Yang Y. Promoter-Controlled Synthesis and Antigenic Evaluation of Mannuronic Acid Alginate Glycans of Pseudomonas aeruginosa. Org Lett 2022; 24:8381-8386. [DOI: 10.1021/acs.orglett.2c03439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Liangliang Zhang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yiyue Zhang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Qingting Hua
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Tong Xu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Junru Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yirong Zhu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - You Yang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| |
Collapse
|
17
|
Louis M, Tahrioui A, Verdon J, David A, Rodrigues S, Barreau M, Manac’h M, Thiroux A, Luton B, Dupont C, Calvé ML, Bazire A, Crépin A, Clabaut M, Portier E, Taupin L, Defontaine F, Clamens T, Bouffartigues E, Cornelis P, Feuilloley M, Caillon J, Dufour A, Berjeaud JM, Lesouhaitier O, Chevalier S. Effect of Phthalates and Their Substitutes on the Physiology of Pseudomonas aeruginosa. Microorganisms 2022; 10:microorganisms10091788. [PMID: 36144390 PMCID: PMC9502294 DOI: 10.3390/microorganisms10091788] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/26/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Phthalates are used in a variety of applications—for example, as plasticizers in polyvinylchloride products to improve their flexibility—and can be easily released into the environment. In addition to being major persistent organic environmental pollutants, some phthalates are responsible for the carcinogenicity, teratogenicity, and endocrine disruption that are notably affecting steroidogenesis in mammals. Numerous studies have thus focused on deciphering their effects on mammals and eukaryotic cells. While multicellular organisms such as humans are known to display various microbiota, including all of the microorganisms that may be commensal, symbiotic, or pathogenic, few studies have aimed at investigating the relationships between phthalates and bacteria, notably regarding their effects on opportunistic pathogens and the severity of the associated pathologies. Herein, the effects of phthalates and their substitutes were investigated on the human pathogen, Pseudomonas aeruginosa, in terms of physiology, virulence, susceptibility to antibiotics, and ability to form biofilms. We show in particular that most of these compounds increased biofilm formation, while some of them enhanced the bacterial membrane fluidity and altered the bacterial morphology.
Collapse
Affiliation(s)
- Mélissande Louis
- Unité de Recherche Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- SéSAD, Fédération de Recherche “Sécurité Sanitaire, Bien Être, Aliment Durable”, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
| | - Ali Tahrioui
- Unité de Recherche Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- SéSAD, Fédération de Recherche “Sécurité Sanitaire, Bien Être, Aliment Durable”, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
| | - Julien Verdon
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
- CNRS UMR7267 Ecologie et Biologie des Interactions (EBI), Université de Poitiers, F-86000 Poitiers, France
| | - Audrey David
- Unité de Recherche Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- SéSAD, Fédération de Recherche “Sécurité Sanitaire, Bien Être, Aliment Durable”, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
| | - Sophie Rodrigues
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
- Université de Bretagne-Sud, EA 3884, LBCM, IUEM, F-56100 Lorient, France
| | - Magalie Barreau
- Unité de Recherche Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- SéSAD, Fédération de Recherche “Sécurité Sanitaire, Bien Être, Aliment Durable”, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
| | - Maëliss Manac’h
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
- Université de Bretagne-Sud, EA 3884, LBCM, IUEM, F-56100 Lorient, France
| | - Audrey Thiroux
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
- CNRS UMR7267 Ecologie et Biologie des Interactions (EBI), Université de Poitiers, F-86000 Poitiers, France
| | - Baptiste Luton
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
- Université de Bretagne-Sud, EA 3884, LBCM, IUEM, F-56100 Lorient, France
| | - Charly Dupont
- Unité de Recherche Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- SéSAD, Fédération de Recherche “Sécurité Sanitaire, Bien Être, Aliment Durable”, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
| | - Marie Le Calvé
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
- Université de Bretagne-Sud, EA 3884, LBCM, IUEM, F-56100 Lorient, France
| | - Alexis Bazire
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
- Université de Bretagne-Sud, EA 3884, LBCM, IUEM, F-56100 Lorient, France
| | - Alexandre Crépin
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
- CNRS UMR7267 Ecologie et Biologie des Interactions (EBI), Université de Poitiers, F-86000 Poitiers, France
| | - Maximilien Clabaut
- Unité de Recherche Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- SéSAD, Fédération de Recherche “Sécurité Sanitaire, Bien Être, Aliment Durable”, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- CNRS UMR7267 Ecologie et Biologie des Interactions (EBI), Université de Poitiers, F-86000 Poitiers, France
| | - Emilie Portier
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
- Université de Bretagne-Sud, EA 3884, LBCM, IUEM, F-56100 Lorient, France
| | - Laure Taupin
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
- Université de Bretagne-Sud, EA 3884, LBCM, IUEM, F-56100 Lorient, France
| | - Florian Defontaine
- Unité de Recherche Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- SéSAD, Fédération de Recherche “Sécurité Sanitaire, Bien Être, Aliment Durable”, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
| | - Thomas Clamens
- Unité de Recherche Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- SéSAD, Fédération de Recherche “Sécurité Sanitaire, Bien Être, Aliment Durable”, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
| | - Emeline Bouffartigues
- Unité de Recherche Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- SéSAD, Fédération de Recherche “Sécurité Sanitaire, Bien Être, Aliment Durable”, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
| | - Pierre Cornelis
- Unité de Recherche Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- SéSAD, Fédération de Recherche “Sécurité Sanitaire, Bien Être, Aliment Durable”, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
| | - Marc Feuilloley
- Unité de Recherche Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- SéSAD, Fédération de Recherche “Sécurité Sanitaire, Bien Être, Aliment Durable”, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
| | - Jocelyne Caillon
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
- EA3826 Thérapeutiques Cliniques et Expérimentales des Infections, Faculté de Médecine, Université de Nantes, F-44000 Nantes, France
| | - Alain Dufour
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
- Université de Bretagne-Sud, EA 3884, LBCM, IUEM, F-56100 Lorient, France
| | - Jean-Marc Berjeaud
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
- CNRS UMR7267 Ecologie et Biologie des Interactions (EBI), Université de Poitiers, F-86000 Poitiers, France
| | - Olivier Lesouhaitier
- Unité de Recherche Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- SéSAD, Fédération de Recherche “Sécurité Sanitaire, Bien Être, Aliment Durable”, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
| | - Sylvie Chevalier
- Unité de Recherche Communication Bactérienne et Stratégies Anti-Infectieuses, CBSA UR4312, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- SéSAD, Fédération de Recherche “Sécurité Sanitaire, Bien Être, Aliment Durable”, Université de Rouen-Normandie, Normandie Université, F-27000 Évreux, France
- IMPERIAL Project Consortium, ANSES, F-94706 Maisons-Alfort, France
- Correspondence: ; Tel.: +33-2-32-29-15-60
| |
Collapse
|
18
|
Sivakumar R, Gunasekaran P, Rajendhran J. Extracytoplasmic sigma factor AlgU contributes to fitness of Pseudomonas aeruginosa PGPR2 during corn root colonization. Mol Genet Genomics 2022; 297:1537-1552. [PMID: 35980488 DOI: 10.1007/s00438-022-01938-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 07/29/2022] [Indexed: 11/28/2022]
Abstract
In bacteria, sigma factors are crucial in determining the plasticity of core RNA polymerase (RNAP) while promoter recognition during transcription initiation. This process is modulated through an intricate regulatory network in response to environmental cues. Previously, an extracytoplasmic function (ECF) sigma factor, AlgU, was identified to positively influence the fitness of Pseudomonas aeruginosa PGPR2 during corn root colonization. In this study, we report that the inactivation of the algU gene encoded by PGPR2_23995 hampers the root colonization ability of PGPR2. An insertion mutant in the algU gene was constructed by allele exchange mutagenesis. The mutant strains displayed threefold decreased root colonization efficiency compared with the wild-type strain when inoculated individually and in the competition assay. The mutant strain was more sensitive to osmotic and antibiotic stresses and showed higher resistance to oxidative stress. On the other hand, the mutant strain showed increased biofilm formation on the abiotic surface, and the expression of the pelB and pslA genes involved in the biofilm matrix formation were up-regulated. In contrast, the expression of algD, responsible for alginate production, was significantly down-regulated in the mutant strain, which is directly regulated by the AlgU sigma factor. The mutant strain also displayed altered motility. The expression of RNA binding protein RsmA was also impeded in the mutant strain. Further, the transcript levels of genes associated with the type III secretion system (T3SS) were analyzed, which revealed a significant down-regulation in the mutant strain. These results collectively provide evidence for the regulatory role of the AlgU sigma factor in modulating gene expression during root colonization.
Collapse
Affiliation(s)
- Ramamoorthy Sivakumar
- Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai, 625 021, India
| | | | - Jeyaprakash Rajendhran
- Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai, 625 021, India.
| |
Collapse
|
19
|
Preclinical Evaluation of Recombinant Microbial Glycoside Hydrolases as Antibiofilm Agents in Acute Pulmonary Pseudomonas aeruginosa Infection. Antimicrob Agents Chemother 2022; 66:e0005222. [PMID: 35862738 PMCID: PMC9380554 DOI: 10.1128/aac.00052-22] [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: 01/09/2023] Open
Abstract
The bacterium Pseudomonas aeruginosa can colonize the airways of patients with chronic lung disease. Within the lung, P. aeruginosa forms biofilms that can enhance resistance to antibiotics and immune defenses. P. aeruginosa biofilm formation is dependent on the secretion of matrix exopolysaccharides, including Pel and Psl. In this study, recombinant glycoside hydrolases (GHs) that degrade Pel and Psl were evaluated alone and in combination with antibiotics in a mouse model of P. aeruginosa infection. Intratracheal GH administration was well tolerated by mice. Pharmacokinetic analysis revealed that, although GHs have short half-lives, administration of two GHs in combination resulted in increased GH persistence. Combining GH prophylaxis and treatment with the antibiotic ciprofloxacin resulted in greater reduction in pulmonary bacterial burden than that with either agent alone. This study lays the foundation for further exploration of GH therapy in bacterial infections.
Collapse
|
20
|
McLean C, Brown K, Windmill J, Dennany L. Innovations In Point-Of-Care Electrochemical Detection Of Pyocyanin. J Electroanal Chem (Lausanne) 2022. [DOI: 10.1016/j.jelechem.2022.116649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
21
|
Kolodkin-Gal I, Cohen-Cymberknoh M, Zamir G, Tsesis I, Rosen E. Targeting Persistent Biofilm Infections: Reconsidering the Topography of the Infection Site during Model Selection. Microorganisms 2022; 10:microorganisms10061164. [PMID: 35744683 PMCID: PMC9231179 DOI: 10.3390/microorganisms10061164] [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: 05/06/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 12/17/2022] Open
Abstract
The physiology of an organism in the environment reflects its interactions with the diverse physical, chemical, and biological properties of the surface. These principles come into consideration during model selection to study biofilm–host interactions. Biofilms are communities formed by beneficial and pathogenic bacteria, where cells are held together by a structured extracellular matrix. When biofilms are associated with a host, chemical gradients and their origins become highly relevant. Conventional biofilm laboratory models such as multiwall biofilm models and agar plate models poorly mimic these gradients. In contrast, ex vivo models possess the partial capacity to mimic the conditions of tissue-associated biofilm and a biofilm associated with a mineralized surface enriched in inorganic components, such as the human dentin. This review will highlight the progress achieved using these settings for two models of persistent infections: the infection of the lung tissue by Pseudomonas aeruginosa and the infection of the root canal by Enterococcus faecalis. For both models, we conclude that the limitations of the conventional in vitro systems necessitate a complimentary experimentation with clinically relevant ex vivo models during therapeutics development.
Collapse
Affiliation(s)
- Ilana Kolodkin-Gal
- Department of Plant Pathology and Microbiology, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
- Correspondence: (I.K.-G.); (I.T.); (E.R.)
| | - Malena Cohen-Cymberknoh
- Pediatric Pulmonary Unit and Cystic Fibrosis Center, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel;
| | - Gideon Zamir
- Department of Experimental Surgery, Hadassah Hebrew University Medical School, Jerusalem 9112001, Israel;
| | - Igor Tsesis
- Department of Endodontics, Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Correspondence: (I.K.-G.); (I.T.); (E.R.)
| | - Eyal Rosen
- Department of Endodontics, Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
- Correspondence: (I.K.-G.); (I.T.); (E.R.)
| |
Collapse
|
22
|
Ma LZ, Wang D, Liu Y, Zhang Z, Wozniak DJ. Regulation of Biofilm Exopolysaccharide Biosynthesis and Degradation in Pseudomonas aeruginosa. Annu Rev Microbiol 2022; 76:413-433. [DOI: 10.1146/annurev-micro-041320-111355] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Microbial communities enmeshed in a matrix of macromolecules, termed as biofilms, are the natural setting of bacteria. Exopolysaccharide is a critical matrix component of biofilms. Here, we focus on biofilm matrix exopolysaccharides in Pseudomonas aeruginosa. This opportunistic pathogen can adapt to a wide range of environments and can form biofilms or aggregates in a variety of surfaces or environments, such as the lungs of people with cystic fibrosis, catheters, wounds, and contact lenses. The ability to synthesize multiple exopolysaccharides is one of the advantages that facilitate bacterial survival in different environments. P. aeruginosa can produce several exopolysaccharides, including alginate, Psl, Pel, and lipopolysaccharide. In this review, we highlight the roles of each exopolysaccharide in P. aeruginosa biofilm development and how bacteria coordinate the biosynthesis of multiple exopolysaccharides and bacterial motility. In addition, we present advances in antibiofilm strategies targeting matrix exopolysaccharides, with a focus on glycoside hydrolases. Expected final online publication date for the Annual Review of Microbiology, Volume 76 is September 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Luyan Z. Ma
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Di Wang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Yiwei Liu
- Department of Microbial Infection and Immunity and Department of Microbiology, Ohio State University, Columbus, Ohio, USA
| | - Zhenyu Zhang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Daniel J. Wozniak
- Department of Microbial Infection and Immunity and Department of Microbiology, Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
23
|
Contribution of Membrane Vesicle to Reprogramming of Bacterial Membrane Fluidity in Pseudomonas aeruginosa. mSphere 2022; 7:e0018722. [PMID: 35603537 PMCID: PMC9241526 DOI: 10.1128/msphere.00187-22] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen capable of resisting environmental insults by applying various strategies, including regulating membrane fluidity and producing membrane vesicles (MVs). This study examined the difference in membrane fluidity between planktonic and biofilm modes of growth in P. aeruginosa and whether the ability to alter membrane rigidity in P. aeruginosa could be transferred via MVs. To this end, planktonic and biofilm P. aeruginosa were compared with respect to the lipid composition of their membranes and their MVs and the expression of genes contributing to alteration of membrane fluidity. Additionally, viscosity maps of the bacterial membrane in planktonic and biofilm lifestyles and under the effect of incubation with bacterial MVs were obtained. Further, the growth rate and biofilm formation capability of P. aeruginosa in the presence of MVs were compared. Results showed that the membrane of the biofilm bacteria is significantly less fluid than the membrane of the planktonic bacteria and is enriched with saturated fatty acids. Moreover, the enzymes involved in altering the structure of existing lipids and favoring membrane rigidification are overexpressed in the biofilm bacteria. MVs of biofilm P. aeruginosa elicit membrane rigidification and delay the bacterial growth in the planktonic lifestyle; conversely, they enhance biofilm development in P. aeruginosa. Overall, the study describes the interplay between the planktonic and biofilm bacteria by shedding light on the role of MVs in altering membrane fluidity. IMPORTANCE Membrane rigidification is a survival strategy in Pseudomonas aeruginosa exposed to stress. Despite various studies dedicated to the mechanism behind this phenomenon, not much attention has been paid to the contribution of the bacterial membrane vesicles (MVs) in this regard. This study revealed that P. aeruginosa rigidifies its membrane in the biofilm mode of growth. Additionally, the capability of decreasing membrane fluidity is transferable to the bacterial population via the bacterial MVs, resulting in reprogramming of bacterial membrane fluidity. Given the importance of membrane rigidification for decreasing the pathogen’s susceptibility to antimicrobials, elucidation of the conditions leading to such biophysicochemical modulation of the P. aeruginosa membrane should be considered for the purpose of developing therapeutic approaches against this resistant pathogen.
Collapse
|
24
|
UVA as environmental signal for alginate production in Pseudomonas aeruginosa: role of this polysaccharide in the protection of planktonic cells and biofilms against lethal UVA doses. Photochem Photobiol Sci 2022; 21:1459-1472. [PMID: 35551642 DOI: 10.1007/s43630-022-00236-w] [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] [Received: 01/20/2022] [Accepted: 04/19/2022] [Indexed: 10/18/2022]
Abstract
Pseudomonas aeruginosa is an extremely versatile microorganism that survives in a wide variety of niches. It is capable to respond rapidly to changes in the environment by producing secondary metabolites and virulence factors, including alginate. Alginate is an extracellular polysaccharide that protects the bacteria from antibiotics and oxidative agents, and enhances cell adhesion to solid surfaces in the process of biofilm formation. In the present study, we analyzed the role of alginate in the response of P. aeruginosa to lethal doses of ultraviolet-A (UVA) radiation, the major fraction of solar UV radiation reaching the Earth's surface. We also studied the role of alginate in the context of the adaptive responses generated when P. aeruginosa is exposed to sublethal doses of UVA radiation. The survival studies demonstrated that alginate has a key role in the resistance of P. aeruginosa to the oxidative stress generated by lethal UVA doses, both in planktonic cells and in static biofilms. In addition, the presence of alginate proved to be essential in the occurrence of adaptive responses such as induction of biofilm formation and cross-protection against hydrogen peroxide and sodium hypochlorite, both generated by exposure to low UVA doses. Finally, we demonstrated that the increase of biofilm formation is accompanied by an increase in alginate concentration in the biofilm matrix, possibly through the ppGpp-dependent induction of genes related to alginate regulation (algR and algU) and biosynthesis (algD operon). Given the importance of alginate in biofilm formation and its protective roles, better understanding of the mechanisms associated to its functions and synthesis is relevant, given the normal exposure of P. aeruginosa to UVA radiation and other types of oxidative stresses.
Collapse
|
25
|
Zhang J, Wu H, Wang D, Wang L, Cui Y, Zhang C, Zhao K, Ma L. Intracellular glycosyl hydrolase PslG shapes bacterial cell fate, signaling, and the biofilm development of Pseudomonas aeruginosa. eLife 2022; 11:e72778. [PMID: 35438634 PMCID: PMC9075953 DOI: 10.7554/elife.72778] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 04/17/2022] [Indexed: 11/22/2022] Open
Abstract
Biofilm formation is one of most important causes leading to persistent infections. Exopolysaccharides are usually a main component of biofilm matrix. Genes encoding glycosyl hydrolases are often found in gene clusters that are involved in the exopolysaccharide synthesis. It remains elusive about the functions of intracellular glycosyl hydrolase and why a polysaccharide synthesis gene cluster requires a glycosyl hydrolase-encoding gene. Here, we systematically studied the physiologically relevant role of intracellular PslG, a glycosyl hydrolase whose encoding gene is co-transcribed with 15 psl genes, which is responsible for the synthesis of exopolysaccharide PSL, a key biofilm matrix polysaccharide in opportunistic pathogen Pseudomonas aeruginosa. We showed that lack of PslG or its hydrolytic activity in this opportunistic pathogen enhances the signaling function of PSL, changes the relative level of cyclic-di-GMP within daughter cells during cell division and shapes the localization of PSL on bacterial periphery, thus results in long chains of bacterial cells, fast-forming biofilm microcolonies. Our results reveal the important roles of intracellular PslG on the cell fate and biofilm development.
Collapse
Affiliation(s)
- Jingchao Zhang
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin UniversityTianjinChina
| | - Huijun Wu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Di Wang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of SciencesBeijingChina
| | - Lanxin Wang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yifan Cui
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Chenxi Zhang
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin UniversityTianjinChina
| | - Kun Zhao
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin UniversityTianjinChina
| | - Luyan Ma
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
26
|
Rojas LJ, Yasmin M, Benjamino J, Marshall SM, DeRonde KJ, Krishnan NP, Perez F, Colin AA, Cardenas M, Martinez O, Pérez-Cardona A, Rhoads DD, Jacobs MR, LiPuma JJ, Konstan MW, Vila AJ, Smania A, Mack AR, Scott JG, Adams MD, Abbo LM, Bonomo RA. Genomic heterogeneity underlies multidrug resistance in Pseudomonas aeruginosa: A population-level analysis beyond susceptibility testing. PLoS One 2022; 17:e0265129. [PMID: 35358221 PMCID: PMC8970513 DOI: 10.1371/journal.pone.0265129] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 02/23/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Pseudomonas aeruginosa is a persistent and difficult-to-treat pathogen in many patients, especially those with Cystic Fibrosis (CF). Herein, we describe a longitudinal analysis of a series of multidrug resistant (MDR) P. aeruginosa isolates recovered in a 17-month period, from a young female CF patient who underwent double lung transplantation. Our goal was to understand the genetic basis of the observed resistance phenotypes, establish the genomic population diversity, and define the nature of sequence evolution over time. METHODS Twenty-two sequential P. aeruginosa isolates were obtained within a 17-month period, before and after a double-lung transplant. At the end of the study period, antimicrobial susceptibility testing, whole genome sequencing (WGS), phylogenetic analyses and RNAseq were performed in order to understand the genetic basis of the observed resistance phenotypes, establish the genomic population diversity, and define the nature of sequence changes over time. RESULTS The majority of isolates were resistant to almost all tested antibiotics. A phylogenetic reconstruction revealed 3 major clades representing a genotypically and phenotypically heterogeneous population. The pattern of mutation accumulation and variation of gene expression suggested that a group of closely related strains was present in the patient prior to transplantation and continued to change throughout the course of treatment. A trend toward accumulation of mutations over time was observed. Different mutations in the DNA mismatch repair gene mutL consistent with a hypermutator phenotype were observed in two clades. RNAseq performed on 12 representative isolates revealed substantial differences in the expression of genes associated with antibiotic resistance and virulence traits. CONCLUSIONS The overwhelming current practice in the clinical laboratories setting relies on obtaining a pure culture and reporting the antibiogram from a few isolated colonies to inform therapy decisions. Our analyses revealed significant underlying genomic heterogeneity and unpredictable evolutionary patterns that were independent of prior antibiotic treatment, highlighting the need for comprehensive sampling and population-level analysis when gathering microbiological data in the context of CF P. aeruginosa chronic infection. Our findings challenge the applicability of antimicrobial stewardship programs based on single-isolate resistance profiles for the selection of antibiotic regimens in chronic infections such as CF.
Collapse
Affiliation(s)
- Laura J. Rojas
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- Research Service, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio, United States of America
- CWRU-Cleveland VAMC Center for Antimicrobial Resistance and Epidemiology (Case VA CARES), Cleveland, Ohio, United States of America
| | - Mohamad Yasmin
- Research Service, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio, United States of America
| | - Jacquelynn Benjamino
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, United States of America
| | - Steven M. Marshall
- Research Service, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio, United States of America
| | - Kailynn J. DeRonde
- Jackson Memorial Hospital, Jackson Health System, Miami, Florida, United States of America
| | - Nikhil P. Krishnan
- Center for Proteomics and Bioinformatics, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- Departments of Translational Hematology and Oncology Research and Radiation Oncology, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Federico Perez
- Medical Service, Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio, United States of America
- CONICET, Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), Córdoba, Argentina
- Division of Infectious Diseases and HIV Medicine, Cleveland, Ohio, United States of America
- GRECC Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio, United States of America
| | - Andrew A. Colin
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Monica Cardenas
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Octavio Martinez
- Jackson Memorial Hospital, Jackson Health System, Miami, Florida, United States of America
- Division of Pulmonology, Department of Pathology University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Armando Pérez-Cardona
- Jackson Memorial Hospital, Jackson Health System, Miami, Florida, United States of America
| | - Daniel D. Rhoads
- Department of Laboratory Medicine and Infection Biology Program, Cleveland Clinic, Cleveland, Ohio, United States of America
- Department of Pathology, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University Cleveland, Ohio, United States of America
| | - Michael R. Jacobs
- Department of Pathology, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University Cleveland, Ohio, United States of America
| | - John J. LiPuma
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Michael W. Konstan
- Department of Pediatrics, Case Western Reserve University School of Medicine and Rainbow Babies and Children’s Hospital, Cleveland, Ohio, United States of America
| | - Alejandro J. Vila
- Instituto de Biología Molecular y Celular de Rosario (IBR, CONICET-UNR), Rosario, Argentina
| | - Andrea Smania
- CONICET, Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), Córdoba, Argentina
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Química Biológica, Córdoba, Argentina
| | - Andrew R. Mack
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- Research Service, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio, United States of America
| | - Jacob G. Scott
- Center for Proteomics and Bioinformatics, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- Departments of Translational Hematology and Oncology Research and Radiation Oncology, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Mark D. Adams
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, United States of America
| | - Lilian M. Abbo
- Jackson Memorial Hospital, Jackson Health System, Miami, Florida, United States of America
- Division of Infectious Diseases Department of Medicine University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Robert A. Bonomo
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- Research Service, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio, United States of America
- CWRU-Cleveland VAMC Center for Antimicrobial Resistance and Epidemiology (Case VA CARES), Cleveland, Ohio, United States of America
- Center for Proteomics and Bioinformatics, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- Medical Service, Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio, United States of America
- Division of Infectious Diseases and HIV Medicine, Cleveland, Ohio, United States of America
- GRECC Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio, United States of America
- Department of Pharmacology, Cleveland, Ohio, United States of America
- Department of Biochemistry Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| |
Collapse
|
27
|
Babaei N, Rasti B, Zamani H. Does ibuprofen affect the expression of alginate genes in pathogenic Pseudomonas aeruginosa strains? Folia Microbiol (Praha) 2022; 67:617-623. [PMID: 35325409 DOI: 10.1007/s12223-022-00962-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 03/07/2022] [Indexed: 11/25/2022]
Abstract
Conversion to mucoid form is a crucial step in the pathogenesis of P. aeruginosa in burns and cystic fibrosis (CF) patients. Alginate is considered the major component of biofilm and is highly associated with the formation of mucoid biofilm in this species. Nonsteroid anti-inflammatory drugs (NSAIDs), including ibuprofen, have shown promising antibacterial and antibiofilm potential for bacterial pathogens. In this study, we aimed to evaluate the effect of ibuprofen on the expression of alginate synthetase (alg8), GDP-mannose dehydrogenase (algD), and alginate lyase (algL) genes in multiple drug-resistant (MDR) P. aeruginosa strains. The biofilm formation potential and the expression of alg8, algD, and algL among the bacteria treated with ibuprofen (at sub-inhibitory concentration) were investigated using the crystal violet staining and real-time PCR assays, respectively. The minimum inhibitory concentration of ibuprofen for the studied strains was determined 1024-2048 µg/mL. We observed that ibuprofen was able to reduce bacterial biofilm by 51-77%. Also, the expression of alg8, algD, and algL decreased by 32, 52, and 48%, respectively. The reduction of the genes responsible for alginate synthesis indicates promising antivirulece potential of ibuprofen to combat P. aeruginosa infection, especially in burns and CF patients. Our findings suggest that ibuprofen could be used to reduce the pathogenicity of P. aeruginosa that could be used in combination with antibiotics to treat drug-resistant infections.
Collapse
Affiliation(s)
- Nastaran Babaei
- Department of Microbiology, Faculty of Basic Sciences, Lahijan Branch, Islamic Azad University (IAU), Lahijan, Guilan, Iran
| | - Behnam Rasti
- Department of Microbiology, Faculty of Basic Sciences, Lahijan Branch, Islamic Azad University (IAU), Lahijan, Guilan, Iran
| | - Hojjatolah Zamani
- Department of Biology, Faculty of Science, University of Guilan, Guilan, Iran.
| |
Collapse
|
28
|
Kiel A, Kaltschmidt BP, Asghari E, Hütten A, Kaltschmidt B, Kaltschmidt C. Bacterial Biofilm Formation on Nano-Copper Added PLA Suited for 3D Printed Face Masks. Microorganisms 2022; 10:439. [PMID: 35208893 PMCID: PMC8875673 DOI: 10.3390/microorganisms10020439] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/10/2022] [Accepted: 02/13/2022] [Indexed: 02/06/2023] Open
Abstract
The COVID-19 Pandemic leads to an increased worldwide demand for personal protection equipment in the medical field, such as face masks. New approaches to satisfy this demand have been developed, and one example is the use of 3D printing face masks. The reusable 3D printed mask may also have a positive effect on the environment due to decreased littering. However, the microbial load on the 3D printed objects is often disregarded. Here we analyze the biofilm formation of Pseudomonas aeruginosa, Staphylococcus aureus, and Escherichia coli on suspected antimicrobial Plactive™ PLA 3D printing filaments and non-antimicrobial Giantarm™ PLA. To characterize the biofilm-forming potential scanning electron microscopy (SEM), Confocal scanning electron microscopy (CLSM) and colony-forming unit assays (CFU) were performed. Attached cells could be observed on all tested 3D printing materials. Gram-negative strains P. aeruginosa and E. coli reveal a strong uniform growth independent of the tested 3D filament (for P. aeruginosa even with stressed induced growth reaction by Plactive™). Only Gram-positive S. aureus shows strong growth reduction on Plactive™. These results suggest that the postulated antimicrobial Plactive™ PLA does not affect Gram-negative bacteria species. These results indicate that reusable masks, while better for our environment, may pose another health risk.
Collapse
Affiliation(s)
- Annika Kiel
- Department of Cell Biology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany; (A.K.); (E.A.); (B.K.)
| | - Bernhard Peter Kaltschmidt
- Department of Thin Films and Physics of Nanostructures, Center of Spinelectronic Materials and Devices, Faculty of Physics, Bielefeld University, 33615 Bielefeld, Germany; (B.P.K.); (A.H.)
| | - Ehsan Asghari
- Department of Cell Biology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany; (A.K.); (E.A.); (B.K.)
| | - Andreas Hütten
- Department of Thin Films and Physics of Nanostructures, Center of Spinelectronic Materials and Devices, Faculty of Physics, Bielefeld University, 33615 Bielefeld, Germany; (B.P.K.); (A.H.)
| | - Barbara Kaltschmidt
- Department of Cell Biology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany; (A.K.); (E.A.); (B.K.)
| | - Christian Kaltschmidt
- Department of Cell Biology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany; (A.K.); (E.A.); (B.K.)
| |
Collapse
|
29
|
Tao J, Yan S, Wang H, Zhao L, Zhu H, Wen Z. Antimicrobial and antibiofilm effects of total flavonoids from Potentilla kleiniana Wight et Arn on Pseudomonas aeruginosa and its potential application to stainless steel surfaces. Lebensm Wiss Technol 2022. [DOI: 10.1016/j.lwt.2021.112631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
30
|
Tan L, Zhang C, Liu F, Chen P, Wei X, Li H, Yi G, Xu Y, Zheng X. Three-compartment septic tanks as sustainable on-site treatment facilities? Watch out for the potential dissemination of human-associated pathogens and antibiotic resistance. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2021; 300:113709. [PMID: 34547574 DOI: 10.1016/j.jenvman.2021.113709] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 09/05/2021] [Accepted: 09/06/2021] [Indexed: 06/13/2023]
Abstract
Improved sanitation is critical important to reduce the spread of human deposited pathogens and antibiotic resistance genes (ARGs). In the China's rural "Toilet Revolution", three-compartment septic tanks (SPTs) are widely used as household domestic sewage treatment facilities. The effluents of SPTs are encouraged to be used as fertilizer in agriculture. However, whether SPT could eliminate fecal pathogens and ARGs is still unrevealed which is crucial in risk assessment of SPT effluent utilization. Herein, we employed metagenomic sequencing to investigate the pathogens and ARGs in rural household SPTs from Tianjin, China. We found that rural household SPT effluents conserved pathogens comparable to that of the influents. A total of 441 ARGs conferring resistance to 26 antibiotic classes were observed in rural household SPTs, with the relative abundance ranging from 709 to 1800 ppm. Results of metagenomic assembly indicated that some ARG-MGE-carrying contigs were carried by pathogens, which may pose risk to human and animal health after being introduced to the environment. This study raises the question of SPTs as sustainable on-site treatment facilities for rural domestic sewage and underscores the need for more attention to the propagation and dissemination of antibiotic-resistant pathogens from SPT to the environments, animals, and humans.
Collapse
Affiliation(s)
- Lu Tan
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin, 300191, China
| | - Chunxue Zhang
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin, 300191, China
| | - Fang Liu
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin, 300191, China
| | - Peizhen Chen
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin, 300191, China
| | - Xiaocheng Wei
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin, 300191, China
| | - Houyu Li
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin, 300191, China
| | - Gao Yi
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin, 300191, China
| | - Yan Xu
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin, 300191, China.
| | - Xiangqun Zheng
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin, 300191, China.
| |
Collapse
|
31
|
Thorn CR, Howell PL, Wozniak DJ, Prestidge CA, Thomas N. Enhancing the therapeutic use of biofilm-dispersing enzymes with smart drug delivery systems. Adv Drug Deliv Rev 2021; 179:113916. [PMID: 34371086 DOI: 10.1016/j.addr.2021.113916] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/27/2021] [Accepted: 08/02/2021] [Indexed: 12/18/2022]
Abstract
Biofilm-dispersing enzymes degrade the extracellular polymeric matrix surrounding bacterial biofilms, disperse the microbial community and increase their susceptibility to antibiotics and immune cells. Challenges for the clinical translation of biofilm-dispersing enzymes involve their susceptibility to denaturation, degradation, and clearance upon administration in vivo. Drug delivery systems aim to overcome these limitations through encapsulation, stabilization and protection from the exterior environment, thereby maintaining the enzymatic activity. Smart drug delivery systems offer target specificity, releasing payloads at the site of infection while minimizing unnecessary systemic exposure. This review highlights critical advances of biofilm-dispersing enzymes as a novel therapeutic approach for biofilm-associated infections. We explore how smart, bio-responsive delivery systems overcome the limiting factors of biofilm-dispersing enzymes and summarize the key systems designed. This review will guide future developments, focusing on utilizing selective and specific therapies in a targeted fashion to meet the unmet therapeutic needs of biofilm infections.
Collapse
Affiliation(s)
- Chelsea R Thorn
- University of South Australia, Clinical and Health Sciences, North Tce, Adelaide, SA 5000, Australia; The Basil Hetzel Institute for Translational Health Research, Woodville, SA 5011, Australia; ARC Centre for Excellence in Bio-Nano Science and Technology, Adelaide, SA 5000, Australia; Biofilm Test Facility, Cancer Research Institute, University of South Australia, North Tce, Adelaide, SA 5000, Australia
| | - P Lynne Howell
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Daniel J Wozniak
- Departments of Microbial Infection and Immunity, Ohio State University, Columbus, OH 43210, USA
| | - Clive A Prestidge
- University of South Australia, Clinical and Health Sciences, North Tce, Adelaide, SA 5000, Australia; ARC Centre for Excellence in Bio-Nano Science and Technology, Adelaide, SA 5000, Australia
| | - Nicky Thomas
- University of South Australia, Clinical and Health Sciences, North Tce, Adelaide, SA 5000, Australia; The Basil Hetzel Institute for Translational Health Research, Woodville, SA 5011, Australia; ARC Centre for Excellence in Bio-Nano Science and Technology, Adelaide, SA 5000, Australia; Biofilm Test Facility, Cancer Research Institute, University of South Australia, North Tce, Adelaide, SA 5000, Australia.
| |
Collapse
|
32
|
Rosiak P, Latanska I, Paul P, Sujka W, Kolesinska B. Modification of Alginates to Modulate Their Physic-Chemical Properties and Obtain Biomaterials with Different Functional Properties. Molecules 2021; 26:7264. [PMID: 34885846 PMCID: PMC8659150 DOI: 10.3390/molecules26237264] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 11/27/2021] [Accepted: 11/28/2021] [Indexed: 01/02/2023] Open
Abstract
Modified alginates have a wide range of applications, including in the manufacture of dressings and scaffolds used for regenerative medicine, in systems for selective drug delivery, and as hydrogel materials. This literature review discusses the methods used to modify alginates and obtain materials with new or improved functional properties. It discusses the diverse biological and functional activity of alginates. It presents methods of modification that utilize both natural and synthetic peptides, and describes their influence on the biological properties of the alginates. The success of functionalization depends on the reaction conditions being sufficient to guarantee the desired transformations and provide modified alginates with new desirable properties, but mild enough to prevent degradation of the alginates. This review is a literature description of efficient methods of alginate functionalization using biologically active ligands. Particular attention was paid to methods of alginate functionalization with peptides, because the combination of the properties of alginates and peptides leads to the obtaining of conjugates with properties resulting from both components as well as a completely new, different functionality.
Collapse
Affiliation(s)
- Piotr Rosiak
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland; (P.R.); (P.P.)
| | - Ilona Latanska
- Tricomed S.A., Swietojanska 5/9, 93-493 Lodz, Poland; (I.L.); (W.S.)
| | - Paulina Paul
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland; (P.R.); (P.P.)
| | - Witold Sujka
- Tricomed S.A., Swietojanska 5/9, 93-493 Lodz, Poland; (I.L.); (W.S.)
| | - Beata Kolesinska
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland; (P.R.); (P.P.)
| |
Collapse
|
33
|
Yahiaoui S, Voos K, Haupenthal J, Wichelhaus TA, Frank D, Weizel L, Rotter M, Brunst S, Kramer JS, Proschak E, Ducho C, Hirsch AKH. N-Aryl mercaptoacetamides as potential multi-target inhibitors of metallo-β-lactamases (MBLs) and the virulence factor LasB from Pseudomonas aeruginosa. RSC Med Chem 2021; 12:1698-1708. [PMID: 34778771 PMCID: PMC8528214 DOI: 10.1039/d1md00187f] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/23/2021] [Indexed: 12/18/2022] Open
Abstract
Increasing antimicrobial resistance is evolving to be one of the major threats to public health. To reduce the selection pressure and thus to avoid a fast development of resistance, novel approaches aim to target bacterial virulence instead of growth. Another strategy is to restore the activity of antibiotics already in clinical use. This can be achieved by the inhibition of resistance factors such as metallo-β-lactamases (MBLs). Since MBLs can cleave almost all β-lactam antibiotics, including the “last resort” carbapenems, their inhibition is of utmost importance. Here, we report on the synthesis and in vitro evaluation of N-aryl mercaptoacetamides as inhibitors of both clinically relevant MBLs and the virulence factor LasB from Pseudomonas aeruginosa. All tested N-aryl mercaptoacetamides showed low micromolar to submicromolar activities on the tested enzymes IMP-7, NDM-1 and VIM-1. The two most promising compounds were further examined in NDM-1 expressing Klebsiella pneumoniae isolates, where they restored the full activity of imipenem. Together with their LasB-inhibitory activity in the micromolar range, this class of compounds can now serve as a starting point for a multi-target inhibitor approach against both bacterial resistance and virulence, which is unprecedented in antibacterial drug discovery. Simultaneous inhibition of metallo-β-lactamases (MBLs) and virulence factors such as LasB from Pseudomonas aeruginosa offers a new approach to combat antibiotic-resistant pathogens.![]()
Collapse
Affiliation(s)
- Samir Yahiaoui
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI) Campus E8 1 66123 Saarbrücken Germany
| | - Katrin Voos
- Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University Campus C2 3 66123 Saarbrücken Germany
| | - Jörg Haupenthal
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI) Campus E8 1 66123 Saarbrücken Germany
| | - Thomas A Wichelhaus
- Institute of Medical Microbiology and Infection Control, University Hospital Frankfurt Paul-Ehrlich-Straße 40 60596 Frankfurt Germany
| | - Denia Frank
- Institute of Medical Microbiology and Infection Control, University Hospital Frankfurt Paul-Ehrlich-Straße 40 60596 Frankfurt Germany
| | - Lilia Weizel
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Max-von-Laue-Straße 9 60438 Frankfurt Germany
| | - Marco Rotter
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Max-von-Laue-Straße 9 60438 Frankfurt Germany
| | - Steffen Brunst
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Max-von-Laue-Straße 9 60438 Frankfurt Germany
| | - Jan S Kramer
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Max-von-Laue-Straße 9 60438 Frankfurt Germany
| | - Ewgenij Proschak
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Max-von-Laue-Straße 9 60438 Frankfurt Germany
| | - Christian Ducho
- Department of Pharmacy, Pharmaceutical and Medicinal Chemistry, Saarland University Campus C2 3 66123 Saarbrücken Germany
| | - Anna K H Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI) Campus E8 1 66123 Saarbrücken Germany .,Department of Pharmacy, Saarland University Campus Building E8 1 66123 Saarbrücken Germany
| |
Collapse
|
34
|
Dharshini RS, Manickam R, Curtis WR, Rathinasabapathi P, Ramya M. Genome analysis of alginate synthesizing Pseudomonas aeruginosa strain SW1 isolated from degraded seaweeds. Antonie van Leeuwenhoek 2021; 114:2205-2217. [PMID: 34661815 DOI: 10.1007/s10482-021-01673-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/07/2021] [Indexed: 11/30/2022]
Abstract
Pseudomonas aeruginosa strain SW1 is an aerobic, motile, Gram-negative, and rod-shaped bacterium isolated from degraded seaweeds. Based on the 16S rRNA gene sequence and MALDI TOF analysis, strain SW1 exhibits 100% similarity to P. aeruginosa DSM 50,071, its closest phylogenetic neighbor. The complete genome of strain SW1 consists of a single circular chromosome with 23,258,857 bp (G + C content of 66%), including 6734 protein-coding sequences, 8 rRNA, and 63 tRNA sequences. The genome of the P. aeruginosa SW1 contains at least 27 genes for the biosynthesis of alginate and other exopolysaccharide involved in biofilm formation. KAAS and GO analysis and functional annotation by COG and CAZymes are consistent with the biosynthesis of alginate. In addition, the presence of antimicrobial resistance, multi-efflux operon, and antibiotic inactivation genes indicate a pathogenic potential similar to strain DSM50071. The high-quality genome and associated annotation provide a starting point to exploit the potential for P. aeruginosa to produce alginate.
Collapse
Affiliation(s)
- Rajathirajan Siva Dharshini
- Faculty of Engineering and Technology, Molecular Genetics Laboratory, Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamil Nadu, 603203, India
| | - Ranjani Manickam
- SRM-DBT Platform for Advanced Life Science Technologies, SRMIST, Chengalpattu, Chennai, Tamil Nadu, 603203, India
| | - Wayne R Curtis
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Pasupathi Rathinasabapathi
- Faculty of Engineering and Technology, Molecular Genetics Laboratory, Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamil Nadu, 603203, India
| | - Mohandass Ramya
- Faculty of Engineering and Technology, Molecular Genetics Laboratory, Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamil Nadu, 603203, India.
| |
Collapse
|
35
|
Chadha J, Harjai K, Chhibber S. Revisiting the virulence hallmarks of Pseudomonas aeruginosa: a chronicle through the perspective of quorum sensing. Environ Microbiol 2021; 24:2630-2656. [PMID: 34559444 DOI: 10.1111/1462-2920.15784] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 12/15/2022]
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen and the leading cause of mortality among immunocompromised patients in clinical setups. The hallmarks of virulence in P. aeruginosa encompass six biologically competent attributes that cumulatively drive disease progression in a multistep manner. These multifaceted hallmarks lay the principal foundation for rationalizing the complexities of pseudomonal infections. They include factors for host colonization and bacterial motility, biofilm formation, production of destructive enzymes, toxic secondary metabolites, iron-chelating siderophores and toxins. This arsenal of virulence hallmarks is fostered and stringently regulated by the bacterial signalling system called quorum sensing (QS). The central regulatory functions of QS in controlling the timely expression of these virulence hallmarks for adaptation and survival drive the disease outcome. This review describes the intricate mechanisms of QS in P. aeruginosa and its role in shaping bacterial responses, boosting bacterial fitness. We summarize the virulence hallmarks of P. aeruginosa, relating them with the QS circuitry in clinical infections. We also examine the role of QS in the development of drug resistance and propose a novel antivirulence therapy to combat P. aeruginosa infections. This can prove to be a next-generation therapy that may eventually become refractory to the use of conventional antimicrobial treatments.
Collapse
Affiliation(s)
- Jatin Chadha
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Kusum Harjai
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Sanjay Chhibber
- Department of Microbiology, Panjab University, Chandigarh, India
| |
Collapse
|
36
|
Sivakumar R, Gunasekaran P, Rajendhran J. Inactivation of CbrAB two-component system hampers root colonization in rhizospheric strain of Pseudomonas aeruginosa PGPR2. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2021; 1864:194763. [PMID: 34530138 DOI: 10.1016/j.bbagrm.2021.194763] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 11/24/2022]
Abstract
Two-component systems (TCS) are one of the signal transduction mechanisms, which sense physiological/biological restraints and respond to changing environmental conditions by regulating the gene expression. Previously, by employing a forward genetic screen (INSeq), we identified that cbrA gene is essential for the fitness of Pseudomonas aeruginosa PGPR2 during root colonization. Here, we report the functional characterization of cbrAB TCS in PGPR2 during root colonization. We constructed insertion mutants in cbrA and its cognate response regulator cbrB. Genetic characterization revealed drastic down-regultion of sRNA crcZ gene in both mutant strains which play a critical role in carbon catabolite repression (CCR). The mutant strains displayed 10-fold decreased root colonization efficiency when compared to the wild-type strain. On the other hand, mutant strains formed higher biofilm on the abiotic surface, and the expression of pelB and pslA genes involved in biofilm matrix formation was up-regulated. In contrast, the expression of algD, responsible for alginate production, and its associated sigma factor algU was significantly down-regulated in mutant strains. We further analyzed the transcript levels of rsmA, controlled by the algU sigma factor, and found that the expression of rsmA was hampered in both mutants. The ability of mutant strains to swim and swarm was significantly hindered. Also, the expression of genes associated with type III secretion system (T3SS) was dysregulated in mutant strains. Taken together, regulation of gene expression by CbrAB TCS is intricate, and we confirm its role beyond carbon and nitrogen assimilation.
Collapse
Affiliation(s)
- Ramamoorthy Sivakumar
- Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | | | - Jeyaprakash Rajendhran
- Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai, India.
| |
Collapse
|
37
|
Burns and biofilms: priority pathogens and in vivo models. NPJ Biofilms Microbiomes 2021; 7:73. [PMID: 34504100 PMCID: PMC8429633 DOI: 10.1038/s41522-021-00243-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 08/02/2021] [Indexed: 02/08/2023] Open
Abstract
Burn wounds can create significant damage to human skin, compromising one of the key barriers to infection. The leading cause of death among burn wound patients is infection. Even in the patients that survive, infections can be notoriously difficult to treat and can cause lasting damage, with delayed healing and prolonged hospital stays. Biofilm formation in the burn wound site is a major contributing factor to the failure of burn treatment regimens and mortality as a result of burn wound infection. Bacteria forming a biofilm or a bacterial community encased in a polysaccharide matrix are more resistant to disinfection, the rigors of the host immune system, and critically, more tolerant to antibiotics. Burn wound-associated biofilms are also thought to act as a launchpad for bacteria to establish deeper, systemic infection and ultimately bacteremia and sepsis. In this review, we discuss some of the leading burn wound pathogens and outline how they regulate biofilm formation in the burn wound microenvironment. We also discuss the new and emerging models that are available to study burn wound biofilm formation in vivo.
Collapse
|
38
|
Thorn CR, Raju D, Lacdao I, Gilbert S, Sivarajah P, Howell PL, Prestidge CA, Thomas N. Protective Liquid Crystal Nanoparticles for Targeted Delivery of PslG: A Biofilm Dispersing Enzyme. ACS Infect Dis 2021; 7:2102-2115. [PMID: 33908759 DOI: 10.1021/acsinfecdis.1c00014] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The glycoside hydrolase, PslG, attacks and degrades the dominant Psl polysaccharide in the exopolymeric substance (EPS) matrix of Pseudomonas aeruginosa biofilms and is a promising therapy to potentiate the effect of antibiotics. However, the need for coadministration with an antibiotic and the potential susceptibility of PslG to proteolysis highlights the need for an effective delivery system. Here, we compared liposomes versus lipid liquid crystal nanoparticles (LCNPs) loaded with PslG and tobramycin as potential formulation approaches to (1) protect PslG from proteolysis, (2) trigger the enzyme's release in the presence of bacteria, and (3) improve the total antimicrobial effect in vitro and in vivo in a Caenorhabditis elegans infection model. LCNPs were an effective formulation strategy for PslG and tobramycin that better protected the enzyme against proteolysis, triggered and sustained the release of PslG, improved the antimicrobial effect by 10-100-fold, and increased the survival of C. elegans infected with P. aeruginosa. Digestible LCNPs had the advantage of triggering the enzyme's release in the presence of bacteria. However, compared to nondigestible LCNPs, negligible differences arose between the LCNPs' ability to protect PslG from proteolysis and potentiate the antimicrobial activity in combination with tobramycin. In C. elegans, the improved antimicrobial efficacy was comparable to tobramycin-LCNPs, although the PslG + tobramycin-LCNPs achieved a greater than 10-fold reduction in bacteria compared to the unformulated combination. Herewith, LCNPs are showcased as a promising protective delivery system for novel biofilm dispersing enzymes combined with antibiotics, enabling infection-directed therapy and improved performance.
Collapse
Affiliation(s)
- Chelsea R. Thorn
- Clinical and Health Science, University of South Australia, North Tce, Adelaide, South Australia 5000, Australia
- The Basil Hetzel Institute for Translational Health Research, Woodville, South Australia 5011, Australia
- ARC Centre for Excellence in Bio-Nano Science and Technology, Adelaide, South Australia 5000, Australia
- Biofilm Test Facility, Cancer Research Institute, University of South Australia, North Tce, Adelaide, South Australia 5000, Australia
| | - Deepa Raju
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Ira Lacdao
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Stephanie Gilbert
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Piyanka Sivarajah
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - P. Lynne Howell
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Clive A. Prestidge
- Clinical and Health Science, University of South Australia, North Tce, Adelaide, South Australia 5000, Australia
- ARC Centre for Excellence in Bio-Nano Science and Technology, Adelaide, South Australia 5000, Australia
| | - Nicky Thomas
- Clinical and Health Science, University of South Australia, North Tce, Adelaide, South Australia 5000, Australia
- The Basil Hetzel Institute for Translational Health Research, Woodville, South Australia 5011, Australia
- ARC Centre for Excellence in Bio-Nano Science and Technology, Adelaide, South Australia 5000, Australia
- Biofilm Test Facility, Cancer Research Institute, University of South Australia, North Tce, Adelaide, South Australia 5000, Australia
| |
Collapse
|
39
|
Alsayari A, Muhsinah AB, Almaghaslah D, Annadurai S, Wahab S. Pharmacological Efficacy of Ginseng against Respiratory Tract Infections. Molecules 2021; 26:molecules26134095. [PMID: 34279434 PMCID: PMC8271507 DOI: 10.3390/molecules26134095] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/01/2021] [Accepted: 07/02/2021] [Indexed: 11/18/2022] Open
Abstract
Respiratory tract infections are underestimated, as they are mild and generally not incapacitating. In clinical medicine, however, these infections are considered a prevalent problem. By 2030, the third most comprehensive reason for death worldwide will be chronic obstructive pulmonary disease (COPD), according to the World Health Organization. The current arsenal of anti-inflammatory drugs shows little or no benefits against COPD. For thousands of years, herbal drugs have been used to cure numerous illnesses; they exhibit promising results and enhance physical performance. Ginseng is one such herbal medicine, known to alleviate pro-inflammatory chemokines and cytokines (IL-2, IL-4, IFN-γ, TNF-α, IL-5, IL-6, IL-8) formed by macrophages and epithelial cells. Furthermore, the mechanisms of action of ginsenoside are still not fully understood. Various clinical trials of ginseng have exhibited a reduction of repeated colds and the flu. In this review, ginseng’s structural features, the pathogenicity of microbial infections, and the immunomodulatory, antiviral, and anti-bacterial effects of ginseng were discussed. The focus was on the latest animal studies and human clinical trials that corroborate ginseng’s role as a therapy for treating respiratory tract infections. The article concluded with future directions and significant challenges. This review would be a valuable addition to the knowledge base for researchers in understanding the promising role of ginseng in treating respiratory tract infections. Further analysis needs to be re-focused on clinical trials to study ginseng’s efficacy and safety in treating pathogenic infections and in determining ginseng-drug interactions.
Collapse
Affiliation(s)
- Abdulrhman Alsayari
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia; (A.A.); (A.B.M.); (S.A.)
| | - Abdullatif Bin Muhsinah
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia; (A.A.); (A.B.M.); (S.A.)
| | - Dalia Almaghaslah
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia;
| | - Sivakumar Annadurai
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia; (A.A.); (A.B.M.); (S.A.)
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia; (A.A.); (A.B.M.); (S.A.)
- Correspondence: or
| |
Collapse
|
40
|
Aziz F, Rashid M, Rehman M, Rafique M, Imran M. Facile and Novel Synthesis of Spiky Gold Nanoparticles as an Efficient Antimicrobial Agent against Pseudomonas Aeruginosa. Comb Chem High Throughput Screen 2021; 25:1230-1236. [PMID: 34139978 DOI: 10.2174/1386207324666210617163037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/19/2021] [Accepted: 04/24/2021] [Indexed: 11/22/2022]
Abstract
AIMS The study aims to develop advanced antibacterial agents as nanoparticles instead of antibiotics due to the emergence of antimicrobial resistance. BACKGROUND Pseudomonas aeruginosa is capable of causing many diseases, including severe bacterial pneumonia. There is a need for an efficient antibacterial agent to kill these pathogens. OBJECTIVE The objective of the study is to synthesize advanced antibacterial agents as nanoparticles for biomedical applications that can play a vital role in killing Gram-negative bacteria (Pseudomonas aeruginosa). METHOD A novel fabricated growth of hydrophilic spiky gold nanoparticles (SGNPs) via reduction method is reported. RESULTS The surface plasmon resonance peak of the synthesized SGNPs was tuned under the near-infrared range. The SGNPs have anisotropic and spiky morphology with 68 nm size and -58 mV surface charge. They are pure, possessing adsorption similar to the organic material. Pseudomonas aeruginosa treated with synthesized SGNPs showed 60% bacterial death at the concentration of 100 μM. CONCLUSION This work consists of the novel synthesis of SGNPs via a safe and simple reduction method. The synthesized SGNPs exhibit strong antibacterial activity against the Gram-negative bacteria Pseudomonas aeruginosa measured using a microplate assay test. The result showed that these SGNPs are ideal for biomedical applications.
Collapse
Affiliation(s)
- Farooq Aziz
- Department of Physics, University of Sahiwal, Sahiwal, 57000, Pakistan
| | - Muhammad Rashid
- Department of Physics, Ghazi University, Dera Ghazi Khan, 32200, Pakistan
| | - Mubashar Rehman
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Muhammad Rafique
- Department of Physics, University of Sahiwal, Sahiwal, 57000, Pakistan
| | - Muhammad Imran
- Department of Chemistry, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
| |
Collapse
|
41
|
Mahajan S, Sunsunwal S, Gautam V, Singh M, Ramya TNC. Biofilm inhibitory effect of alginate lyases on mucoid P. aeruginosa from a cystic fibrosis patient. Biochem Biophys Rep 2021; 26:101028. [PMID: 34095554 PMCID: PMC8165544 DOI: 10.1016/j.bbrep.2021.101028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 05/03/2021] [Accepted: 05/17/2021] [Indexed: 01/21/2023] Open
Abstract
Chronic mucoid Pseudomonas aeruginosa infections are a major scourge in cystic fibrosis patients. Mucoid P. aeruginosa displays structured alginate-rich biofilms that are resistant to antibiotics. Here, we have assessed the efficacy of a panel of alginate lyases in combating mucoid P. aeruginosa biofilms in cystic fibrosis. Albeit we could not demonstrate alginate degradation by alginate lyases in sputum, we demonstrate that the endotypic alginate lyases, CaAly (from Cellulophaga algicola) and VspAlyVI (from Vibrio sp. QY101) and the exotypic alginate lyases, FspAlyFRB (from Falsirhodobacterium sp. alg1), and SA1-IV (from Sphingomonas sp. A1), indeed inhibit biofilm formation by a mucoid P. aeruginosa strain isolated from the sputum of a cystic fibrosis patient with comparative effect to that of the glycoside hydrolase PslG, a promising candidate for biofilm treatment. We believe that these enzymes should be explored for in vivo efficacy in future studies. A P. aeruginosa strain was isolated from the sputum of a cystic fibrosis patient. The anti-biofilm efficacy of endotypic and exotypic alginate lyases was assessed. Alginate lyases CaAly, VspAlyVI, FspAlyFRB, and SA1-IV inhibited biofilm formation. Similar anti-biofilm effect was observed for the glycoside hydrolase, PslG.
Collapse
Affiliation(s)
- Sonal Mahajan
- CSIR- Institute of Microbial Technology, Sector 39A, Chandigarh, 160036, India
| | - Sonali Sunsunwal
- CSIR- Institute of Microbial Technology, Sector 39A, Chandigarh, 160036, India
| | - Vikas Gautam
- Department of Medical Microbiology, Post Graduate Institute of Medical Education and Research, Sector 12, Chandigarh, 160012, India
| | - Meenu Singh
- Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Sector 12, Chandigarh, 160012, India
| | - T N C Ramya
- CSIR- Institute of Microbial Technology, Sector 39A, Chandigarh, 160036, India
| |
Collapse
|
42
|
Ali S, Khan N. Delineation of mechanistic approaches employed by plant growth promoting microorganisms for improving drought stress tolerance in plants. Microbiol Res 2021; 249:126771. [PMID: 33930840 DOI: 10.1016/j.micres.2021.126771] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/01/2021] [Accepted: 04/17/2021] [Indexed: 11/24/2022]
Abstract
Drought stress is expected to increase in intensity, frequency, and duration in many parts of the world, with potential negative impacts on plant growth and productivity. The plants have evolved complex physiological and biochemical mechanisms to respond and adjust to water-deficient environments. The physiological and biochemical mechanisms associated with water-stress tolerance and water-use efficiency have been extensively studied. Besides these adaptive and mitigating strategies, the plant growth-promoting rhizobacteria (PGPR) play a significant role in alleviating plant drought stress. These beneficial microorganisms colonize the endo-rhizosphere/rhizosphere of plants and enhance drought tolerance. The common mechanism by which these microorganisms improve drought tolerance included the production of volatile compounds, phytohormones, siderophores, exopolysaccharides, 1-aminocyclopropane-1-carboxylate deaminase (ACC deaminase), accumulation of antioxidant, stress-induced metabolites such as osmotic solutes proline, alternation in leaf and root morphology and regulation of the stress-responsive genes. The PGPR is an easy and efficient alternative approach to genetic manipulation and crop enhancement practices because plant breeding and genetic modification are time-consuming and expensive processes for obtaining stress-tolerant varieties. In this review, we will elaborate on PGPR's mechanistic approaches in enhancing the plant stress tolerance to cope with the drought stress.
Collapse
Affiliation(s)
- Shahid Ali
- Plant Epigenetic and Development, Northeast Forestry University, Harbin, 150040, China
| | - Naeem Khan
- Department of Agronomy, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
43
|
Kart D, Reçber T, Nemutlu E, Sagiroglu M. Sub-Inhibitory Concentrations of Ciprofloxacin Alone and Combinations with Plant-Derived Compounds against P. aeruginosa Biofilms and Their Effects on the Metabolomic Profile of P. aeruginosa Biofilms. Antibiotics (Basel) 2021; 10:antibiotics10040414. [PMID: 33918895 PMCID: PMC8070142 DOI: 10.3390/antibiotics10040414] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION Alternative anti-biofilm agents are needed to combat Pseudomonas aeruginosa infections. The mechanisms behind these new agents also need to be revealed at a molecular level. MATERIALS AND METHODS The anti-biofilm effects of 10 plant-derived compounds on P. aeruginosa biofilms were investigated using minimum biofilm eradication concentration (MBEC) and virulence assays. The effects of ciprofloxacin and compound combinations on P. aeruginosa in mono and triple biofilms were compared. A metabolomic approach and qRT-PCR were applied to the biofilms treated with ciprofloxacin in combination with baicalein, esculin hydrate, curcumin, and cinnamaldehyde at sub-minimal biofilm inhibitory concentration (MBIC) concentrations to highlight the specific metabolic shifts between the biofilms and to determine the quorum sensing gene expressions, respectively. RESULTS The combinations of ciprofloxacin with curcumin, baicalein, esculetin, and cinnamaldehyde showed more reduced MBICs than ciprofloxacin alone. The quorum sensing genes were downregulated in the presence of curcumin and cinnamaldehyde, while upregulated in the presence of baicalein and esculin hydrate rather than for ciprofloxacin alone. The combinations exhibited different killing effects on P. aeruginosa in mono and triple biofilms without affecting its virulence. The findings of the decreased metabolite levels related to pyrimidine and lipopolysaccharide synthesis and to down-regulated alginate and lasI expressions strongly indicate the role of multifactorial mechanisms for curcumin-mediated P. aeruginosa growth inhibition. CONCLUSIONS The use of curcumin, baicalein, esculetin, and cinnamaldehyde with ciprofloxacin will help fight against P. aeruginosa biofilms. To the best of our knowledge, this is the first study of its kind to define the effect of plant-based compounds as possible anti-biofilm agents with low MBICs for the treatment of P. aeruginosa biofilms through metabolomic pathways.
Collapse
Affiliation(s)
- Didem Kart
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Hacettepe University, Ankara 06100, Turkey;
- Correspondence: ; Tel.: +90-533-690-7637
| | - Tuba Reçber
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara 06100, Turkey; (T.R.); (E.N.)
| | - Emirhan Nemutlu
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara 06100, Turkey; (T.R.); (E.N.)
- Bioanalytic and Omics Laboratory, Faculty of Pharmacy, Hacettepe University, Ankara 06100, Turkey
| | - Meral Sagiroglu
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Hacettepe University, Ankara 06100, Turkey;
| |
Collapse
|
44
|
Wang Y. Liposome as a delivery system for the treatment of biofilm-mediated infections. J Appl Microbiol 2021; 131:2626-2639. [PMID: 33650748 DOI: 10.1111/jam.15053] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 02/22/2021] [Accepted: 02/24/2021] [Indexed: 12/11/2022]
Abstract
Biofilm formation by pathogenic microorganisms has been a tremendous challenge for antimicrobial therapies due to various factors. The biofilm matrix sequesters bacterial cells from the exterior environment and therefore prevents antimicrobial agents from reaching the interior. In addition, biofilm surface extracellular polymeric substances can absorb antimicrobial agents and thus reduce their bioavailability. To conquer these protection mechanisms, liposomes have been developed into a drug delivery system for antimicrobial agents against biofilm-mediated infections. The unique characteristics of liposomes, including versatility for cargoes, target-specificity, nonimmunogenicity, low toxicity, and biofilm matrix-/cell membrane-fusogenicity, remarkably improve the effectiveness of antimicrobial agents and minimize recurrence of infections. This review summarizes current development of liposomal carriers for biofilm therapeutics, presents evidence in their practical applications and discusses their potential limitations.
Collapse
Affiliation(s)
- Y Wang
- School of Agriculture and Food Sciences, University of Queensland, St Lucia, Qld, Australia
| |
Collapse
|
45
|
Liu J, Yu M, Ge Y, Tian Y, Hu B, Zhao Y. The RsmA RNA-Binding Proteins in Pseudomonas syringae Exhibit Distinct and Overlapping Roles in Modulating Virulence and Survival Under Different Nutritional Conditions. FRONTIERS IN PLANT SCIENCE 2021; 12:637595. [PMID: 33719314 PMCID: PMC7952654 DOI: 10.3389/fpls.2021.637595] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/27/2021] [Indexed: 06/12/2023]
Abstract
The post-transcriptional regulator RsmA globally controls gene expression in bacteria. Previous studies showed that RsmA2 and RsmA3 played critical roles in regulating type III secretion system (T3SS), motility, syringafactin, and alginate productions in Pseudomonas syringae pv. tomato strain DC3000 (PstDC3000). In this study, we investigated global gene expression profiles of the wild-type PstDC3000, the rsmA3 mutant, and the rsmA2/A3 double mutant in the hrp-inducing minimum medium (HMM) and King's B (KB) medium. By comparing the rsmA2/A3 and rsmA3 mutants to PstDC3000, a total of 1358 and 1074 differentially expressed genes (DEGs) in HMM, and 870 and 1463 DEGs in KB were uncovered, respectively. When comparing the rsmA2/A3 mutant with the rsmA3 mutant, 277 and 741 DEGs in HMM and KB, respectively, were revealed. Transcriptomic analysis revealed that the rsmY, rsmZ, and rsmX1-5 non-coding small RNAs (ncsRNAs) were positively affected by RsmA2 and RsmA3, while RsmA3 positively regulates the expression of the rsmA2 gene and negatively regulates both rsmA1 and rsmA5 gene expression. Comparative transcriptomic analysis showed that RsmA2 and RsmA3 synergistically influenced the expression of genes involved in T3SS and alginate biosynthesis in HMM and chemotaxis in KB. RsmA2 and RsmA3 inversely affected genes involved in syringafactin production in HMM and ribosomal protein biosynthesis in KB. In addition, RsmA2 played a major role in influencing genes involved in sarcosine and thiamine biosynthesis in HMM and in mannitol and phosphate metabolism in KB. On the other hand, genes involved in fatty acid metabolism, cellulose biosynthesis, signal transduction, and stress responses were mainly impacted by RsmA3 in both HMM and KB; whereas RsmA3 played a major role in controlling genes involved in c-di-GMP, phosphate metabolism, chemotaxis, and capsular polysaccharide in HMM. Furthermore, regulation of syringafactin production and oxidative stress by RsmA2 and RsmA3 was experimentally verified. Our results suggested the potential interplay among the RsmA proteins, which exhibit distinct and overlapping roles in modulating virulence and survival in P. syringae under different nutritional conditions.
Collapse
Affiliation(s)
- Jun Liu
- College of Plant Protection and Key Laboratory of Integrated Management of Crop Diseases and Pests, Nanjing Agricultural University, Nanjing, China
- Department of Crop Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Menghao Yu
- Department of Crop Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Yixin Ge
- College of Plant Protection and Key Laboratory of Integrated Management of Crop Diseases and Pests, Nanjing Agricultural University, Nanjing, China
- Department of Crop Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Yanli Tian
- College of Plant Protection and Key Laboratory of Integrated Management of Crop Diseases and Pests, Nanjing Agricultural University, Nanjing, China
| | - Baishi Hu
- College of Plant Protection and Key Laboratory of Integrated Management of Crop Diseases and Pests, Nanjing Agricultural University, Nanjing, China
| | - Youfu Zhao
- Department of Crop Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
46
|
Schulze A, Mitterer F, Pombo JP, Schild S. Biofilms by bacterial human pathogens: Clinical relevance - development, composition and regulation - therapeutical strategies. MICROBIAL CELL (GRAZ, AUSTRIA) 2021; 8:28-56. [PMID: 33553418 PMCID: PMC7841849 DOI: 10.15698/mic2021.02.741] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 12/19/2022]
Abstract
Notably, bacterial biofilm formation is increasingly recognized as a passive virulence factor facilitating many infectious disease processes. In this review we will focus on bacterial biofilms formed by human pathogens and highlight their relevance for diverse diseases. Along biofilm composition and regulation emphasis is laid on the intensively studied biofilms of Vibrio cholerae, Pseudomonas aeruginosa and Staphylococcus spp., which are commonly used as biofilm model organisms and therefore contribute to our general understanding of bacterial biofilm (patho-)physiology. Finally, therapeutical intervention strategies targeting biofilms will be discussed.
Collapse
Affiliation(s)
- Adina Schulze
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50, 8010 Graz, Austria
- A.S. and F.M. contributed equally to this work
| | - Fabian Mitterer
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50, 8010 Graz, Austria
- A.S. and F.M. contributed equally to this work
| | - Joao P. Pombo
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50, 8010 Graz, Austria
| | - Stefan Schild
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50, 8010 Graz, Austria
- BioTechMed Graz, Austria
- Field of Excellence Biohealth – University of Graz, Graz, Austria
| |
Collapse
|
47
|
Ci F, Jiang H, Zhang Z, Mao X. Properties and potential applications of mannuronan C5-epimerase: A biotechnological tool for modifying alginate. Int J Biol Macromol 2021; 168:663-675. [PMID: 33220370 DOI: 10.1016/j.ijbiomac.2020.11.123] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 11/17/2020] [Accepted: 11/17/2020] [Indexed: 11/23/2022]
Abstract
Given the excellent characteristics of alginate, it is an industrially important polysaccharide. Mannuronan C5-epimerase (MC5E) is an alginate-modifying enzyme that catalyzes the conversion of β-D-mannuronate (M) to its C5 epimer α-L-guluronate (G) in alginate. Both the biological activities and physical properties of alginate are determined by M/G ratios and distribution patterns. Therefore, MC5E is regarded as a biotechnological tool for modifying and processing alginate. Various MC5Es derived from brown algae, Pseudomonas and Azotobacter have been isolated and characterized. With the rapid development of structural biology, the crystal structures and catalytic mechanisms of several MC5Es have been elucidated. It is necessary to comprehensively understand the research status of this alginate-modifying enzyme. In this review, the properties and potential applications of MC5Es isolated from different kinds of organisms are summarized and reviewed. Moreover, future research directions of MC5Es as well as strategies to enhance their properties are elucidated, highlighted, and prospected.
Collapse
Affiliation(s)
- Fangfang Ci
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - Hong Jiang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China.
| | - Zhaohui Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - Xiangzhao Mao
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China.
| |
Collapse
|
48
|
Mahajan S, Ramya TNC. Cellulophaga algicola alginate lyase inhibits biofilm formation of a clinical Pseudomonas aeruginosa strain MCC 2081. IUBMB Life 2020; 73:444-462. [PMID: 33350564 DOI: 10.1002/iub.2442] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/14/2020] [Accepted: 12/17/2020] [Indexed: 12/31/2022]
Abstract
Alginate lyases are potential agents for disrupting alginate-rich Pseudomonas biofilms in the infected lungs of cystic fibrosis patients but there is as yet no clinically approved alginate lyase that can be used as a therapeutic. We report here the endolytic alginate lyase activity of a recombinant Cellulophaga algicola alginate lyase domain (CaAly) encoded by a gene that also codes for an N-terminal carbohydrate-binding module, CBM6, and a central F-type lectin domain (CaFLD). CaAly degraded both polyM and polyG alginates with optimal temperature and pH of 37°C and pH 7, respectively, with greater preference for polyG. Recombinant CaFLD bound to fucosylated glycans with a preference for H-type 2 glycan motif, and did not have any apparent effect on the enzyme activity of the co-associated alginate lyase domain in the recombinant protein construct, CaFLD_Aly. We assessed the potential of CaAly and other alginate lyases previously reported in published literature to inhibit biofilm formation by a clinical strain, Pseudomonas aeruginosa MCC 2081. Of all the alginate lyases tested, CaAly displayed most inhibition of in vitro biofilm formation on plastic surfaces. We also assessed its inhibitory ability against P. aeruginosa 2081 biofilms formed over a monolayer of A549 lung epithelial cells. Our study indicated that CaAly is efficacious in inhibition of biofilm formation even on A549 lung epithelial cell line monolayers.
Collapse
Affiliation(s)
- Sonal Mahajan
- Protein Science and Engineering Department, Institute of Microbial Technology, Chandigarh, India
| | | |
Collapse
|
49
|
Biofilms as Promoters of Bacterial Antibiotic Resistance and Tolerance. Antibiotics (Basel) 2020; 10:antibiotics10010003. [PMID: 33374551 PMCID: PMC7822488 DOI: 10.3390/antibiotics10010003] [Citation(s) in RCA: 216] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/15/2020] [Accepted: 12/19/2020] [Indexed: 12/12/2022] Open
Abstract
Multidrug resistant bacteria are a global threat for human and animal health. However, they are only part of the problem of antibiotic failure. Another bacterial strategy that contributes to their capacity to withstand antimicrobials is the formation of biofilms. Biofilms are associations of microorganisms embedded a self-produced extracellular matrix. They create particular environments that confer bacterial tolerance and resistance to antibiotics by different mechanisms that depend upon factors such as biofilm composition, architecture, the stage of biofilm development, and growth conditions. The biofilm structure hinders the penetration of antibiotics and may prevent the accumulation of bactericidal concentrations throughout the entire biofilm. In addition, gradients of dispersion of nutrients and oxygen within the biofilm generate different metabolic states of individual cells and favor the development of antibiotic tolerance and bacterial persistence. Furthermore, antimicrobial resistance may develop within biofilms through a variety of mechanisms. The expression of efflux pumps may be induced in various parts of the biofilm and the mutation frequency is induced, while the presence of extracellular DNA and the close contact between cells favor horizontal gene transfer. A deep understanding of the mechanisms by which biofilms cause tolerance/resistance to antibiotics helps to develop novel strategies to fight these infections.
Collapse
|
50
|
Wang L, Zhang Y, Overkleeft HS, van der Marel GA, Codée JDC. Reagent Controlled Glycosylations for the Assembly of Well-Defined Pel Oligosaccharides. J Org Chem 2020; 85:15872-15884. [PMID: 32375481 PMCID: PMC7754192 DOI: 10.1021/acs.joc.0c00703] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
![]()
A new
additive, methyl(phenyl)formamide (MPF), is introduced for
the glycosylation of 2-azido-2-deoxyglucose building blocks. A linear
α-(1,4)-glucosamine tetrasaccharide was assembled to prove the
utility of MPF. Next, a hexasaccharide fragment of the Pseudomonas
aeruginosa exopolysaccharide Pel was assembled using a [2
+ 2 + 2] strategy modulated by MPF. The used [galactosazide-α-(1,4)-glucosazide]
disaccharide building blocks were synthesized using a 4,6-O-DTBS protected galactosyl azide donor.
Collapse
Affiliation(s)
- Liming Wang
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Yongzhen Zhang
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Herman S Overkleeft
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Gijsbert A van der Marel
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Jeroen D C Codée
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| |
Collapse
|