1
|
Mayattu K, Rajwade J, Ghormade V. Development of erythromycin loaded PLGA nanoparticles for improved drug efficacy and sustained release against bacterial infections and biofilm formation. Microb Pathog 2024; 197:107083. [PMID: 39454804 DOI: 10.1016/j.micpath.2024.107083] [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: 09/10/2024] [Revised: 10/23/2024] [Accepted: 10/23/2024] [Indexed: 10/28/2024]
Abstract
Bacterial infections are a common cause of sepsis, often leading to high patient mortality. Such infections are challenging to treat due to bacterial resistance to many existing drugs. Erythromycin (Ery) is a macrolide antibiotic used against bacterial infections with reported resistance. Recently, synthetic poly-lactide co-glycolic acid (PLGA) polymer nanoparticles (NPs) have displayed improved drug delivery characteristics and biocompatibility. In this study, PLGA-Ery NPs were synthesized by the o/w emulsion diffusion method, having a particle size of 159 ± 23 nm and displayed 71.89 % of encapsulation efficiency. The PLGA-Ery NPs showed 1.5, 2.1 and 1.5-fold improved MIC and antibacterial efficacy against E. coli, S. aureus, and P. aeruginosa, respectively than the pure drug. As illustrated by scanning electron microscopy, PLGA-Ery NPs caused damage to the bacterial cell walls. Furthermore, a surface coating with PLGA-Ery NPs on a glass surface showed efficient inhibition (>90 %) of the biofilm formation by P. aeruginosa, as determined by fluorescence microscopy and MTT assay. This study demonstrates that PLGA-Ery NPs can increase the efficiency of erythromycin and can suppress the growth and biofilm formation of P. aeruginosa. Such polymeric nanoparticles drug nanoformulations have potential as an antimicrobial and as a surface coating for medical devices.
Collapse
Affiliation(s)
- Kamal Mayattu
- Nanobioscience Group, Agharkar Research Institute, GG Agarkar Road, Pune, 411004, India
| | - Jyutika Rajwade
- Nanobioscience Group, Agharkar Research Institute, GG Agarkar Road, Pune, 411004, India
| | - Vandana Ghormade
- Nanobioscience Group, Agharkar Research Institute, GG Agarkar Road, Pune, 411004, India.
| |
Collapse
|
2
|
Sarkar S, Roy A, Mitra R, Kundu S, Banerjee P, Acharya Chowdhury A, Ghosh S. Escaping the ESKAPE pathogens: A review on antibiofilm potential of nanoparticles. Microb Pathog 2024; 194:106842. [PMID: 39117012 DOI: 10.1016/j.micpath.2024.106842] [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: 04/18/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
ESKAPE pathogens, a notorious consortium comprising Enterococcusfaecium, Staphylococcusaureus, Klebsiellapneumoniae, Acinetobacterbaumannii, Pseudomonasaeruginosa, and Enterobacter species, pose formidable challenges in healthcare settings due to their multidrug-resistant nature. The increasing global cases of antimicrobial-resistant ESKAPE pathogens are closely related to their remarkable ability to form biofilms. Thus, understanding the unique mechanisms of antimicrobial resistance of ESKAPE pathogens and the innate resilience of biofilms against traditional antimicrobial agents is important for developing innovative strategies to establish effective control methods against them. This review offers a thorough analysis of biofilm dynamics, with a focus on the general mechanisms of biofilm formation, the significant contribution of persister cells in the resistance mechanisms, and the recurrence of biofilms in comparison to planktonic cells. Additionally, this review highlights the potential strategies of nanoparticles for managing biofilms in the ESKAPE group of pathogens. Nanoparticles, with their unique physicochemical properties, provide promising opportunities for disrupting biofilm structures and improving antimicrobial effectiveness. The review has explored interactions between nanoparticles and biofilms, covering a range of nanoparticle types such as metal, metal-oxide, surface-modified, and functionalized nanoparticles, along with organic nanoparticles and nanomaterials. The additional focus of this review also encompasses green synthesis techniques of nanoparticles that involve plant extract and supernatants from bacterial and fungal cultures as reducing agents. Furthermore, the use of nanocomposites and nano emulsions in biofilm management of ESKAPE is also discussed. To conclude, the review addresses the current obstacles and future outlooks in nanoparticle-based biofilm management, stressing the necessity for further research and development to fully exploit the potential of nanoparticles in addressing biofilm-related challenges.
Collapse
Affiliation(s)
| | - Ankita Roy
- Department of Biosciences, JIS University, Kolkata, India
| | - Rangan Mitra
- Department of Biosciences, JIS University, Kolkata, India
| | - Sweta Kundu
- Department of Biosciences, JIS University, Kolkata, India
| | | | | | - Suparna Ghosh
- Department of Biosciences, JIS University, Kolkata, India.
| |
Collapse
|
3
|
Joshi T, Vijayakumar S, Ghosh S, Mathpal S, Ramaiah S, Anbarasu A. Identifying Novel Therapeutics for the Resistant Mutant "F533L" in PBP3 of Pseudomonas aeruginosa Using ML Techniques. ACS OMEGA 2024; 9:28046-28060. [PMID: 38973840 PMCID: PMC11223260 DOI: 10.1021/acsomega.4c00929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 07/09/2024]
Abstract
Pseudomonas aeruginosa (P. aeruginosa) is a highly infectious and antibiotic-resistant bacterium, which causes acute and chronic nosocomial infections. P. aeruginosa exhibits multidrug resistance due to the emergence of resistant mutants. The bacterium takes advantage of intrinsic and acquired resistance mechanisms to resist almost every antibiotic. To overcome the drug-resistance problem, there is a need to develop effective drugs against antibiotic-resistant mutants. Therefore, in this study, we selected the F533L mutation in PBP3 (penicillin-binding protein 3) because of its important role in β-lactam recognition. To target this mutation, we screened 147 antibacterial compounds from PubChem through a machine-learning model developed based on the decision stump algorithm with 75.75% accuracy and filtered out 55 compounds. Subsequently, out of 55 compounds, 47 compounds were filtered based on their drug-like activity. These 47 compounds were subjected to virtual screening to obtain binding affinity compounds. The binding affinity range of all 47 compounds was -11.3 to -4.6 kcal mol-1. The top 10 compounds were examined according to their binding with the mutation point. A molecular dynamic simulation of the top 8 compounds was conducted to understand the stability of the compounds containing the mutated PBP3. Out of 8 compounds, 3 compounds, namely, macozinone, antibacterial agent 71, and antibacterial agent 123, showed good stability and were validated by RMSD, RMSF, and binding-free analysis. The findings of this study revealed promising antibacterial compounds against the F533L mutant PBP3. Furthermore, developments in these compounds may pave the way for novel therapeutic interventions.
Collapse
Affiliation(s)
- Tushar Joshi
- Medical
and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
- Department
of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Santhiya Vijayakumar
- Medical
and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
- Department
of Integrative Biology, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Soumyadip Ghosh
- Medical
and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
- Department
of Bio-Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Shalini Mathpal
- Medical
and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
- Department
of Bio-Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Sudha Ramaiah
- Medical
and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
- Department
of Bio-Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Anand Anbarasu
- Medical
and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
- Department
of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| |
Collapse
|
4
|
Ammazzalorso A, Granese A, De Filippis B. Recent trends and challenges to overcome Pseudomonas aeruginosa infections. Expert Opin Ther Pat 2024; 34:493-509. [PMID: 38683024 DOI: 10.1080/13543776.2024.2348602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/24/2024] [Indexed: 05/01/2024]
Abstract
INTRODUCTION Pseudomonas aeruginosa (PA) is a Gram-negative bacterium that can cause a wide range of severe infections in immunocompromised patients. The most difficult challenge is due to its ability to rapidly develop multi drug-resistance. New strategies are urgently required to improve the outcome of patients with PA infections. The present patent review highlights the new molecules acting on different targets involved in the antibiotic resistance. AREA COVERED This review offers an insight into new potential PA treatment disclosed in patent literature. From a broad search of documents claiming new PA inhibitors, we selected and summarized molecules that showed in vitro and in vivo activity against PA spp. in the period 2020 and 2023. We collected the search results basing on the targets explored. EXPERT OPINION This review examined the main patented compounds published in the last three years, with regard to the structural novelty and the identification of innovative targets. The main areas of antibiotic resistance have been explored. The compounds are structurally unrelated to earlier antibiotics, characterized by a medium-high molecular weight and the presence of heterocycle rings. Peptides and antibodies have also been reported as potential alternatives to chemical treatment, hereby expanding the therapeutic possibilities in this field.
Collapse
Affiliation(s)
| | - Arianna Granese
- Department of Drug Chemistry and Technology, "Sapienza" University of Rome, Rome, Italy
| | | |
Collapse
|
5
|
Gholami A, Minai-Tehrani D, Farewell A, Eriksson LA. Discovery of novel inhibitors for Pseudomonas aeruginosa lipase enzyme from in silico and in vitro studies. J Biomol Struct Dyn 2024; 42:2197-2210. [PMID: 37098781 DOI: 10.1080/07391102.2023.2203258] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 04/10/2023] [Indexed: 04/27/2023]
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen prone to developing drug-resistance and is a major cause of infection for burn patients and patients suffering from cystic fibrosis or are hospitalized in intensive care units. One of the virulence factors of this bacterium is the lipase enzyme that degrades the extracellular matrix of the host tissue and promotes invasion. Bromhexine is a mucolytic drug and has recently been reported to function as a competitive inhibitor of lipase with an IC50 value of 49 µM. In the present study, an attempt was made to identify stronger inhibitors from the ChEMBL database of bioactive compounds, as compared to the reference compound Bromhexine. Following docking and MD simulations, four hit compounds (N1-N4) were selected that showed promising binding modes and low RMSD values indicative of stable protein-ligand complexes. From subsequent binding pose metadynamics (BPMD) simulations, two of these (N2 and N4) stood out as more potent than Bromhexine, displaying stable interactions with residues in the catalytic site of the enzyme. Biological investigations were performed for all four compounds. Among them, the same two hit compounds were found to be the most effective binders with IC50 values of 22.1 and 27.5 µM, respectively; i.e. roughly twice as efficient as the reference Bromhexine. Taken together, our results show that these hits can be promising new candidates to use as leads for the development of drugs targeting the P. aeruginosa lipase enzyme.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Asma Gholami
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
- Department of Chemistry and Molecular Biology, University of Gothenburg, Göteborg, Sweden
| | - Dariush Minai-Tehrani
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Anne Farewell
- Department of Chemistry and Molecular Biology, University of Gothenburg, Göteborg, Sweden
- Centre for Antibiotic Resistance Research, University of Gothenburg, Göteborg, Sweden
| | - Leif A Eriksson
- Department of Chemistry and Molecular Biology, University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
6
|
Sathe N, Beech P, Croft L, Suphioglu C, Kapat A, Athan E. Pseudomonas aeruginosa: Infections and novel approaches to treatment "Knowing the enemy" the threat of Pseudomonas aeruginosa and exploring novel approaches to treatment. INFECTIOUS MEDICINE 2023; 2:178-194. [PMID: 38073886 PMCID: PMC10699684 DOI: 10.1016/j.imj.2023.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/30/2023] [Accepted: 05/05/2023] [Indexed: 03/09/2024]
Abstract
Pseudomonas aeruginosa is an aerobic Gram-negative rod-shaped bacterium with a comparatively large genome and an impressive genetic capability allowing it to grow in a variety of environments and tolerate a wide range of physical conditions. This biological flexibility enables the P. aeruginosa to cause a broad range of infections in patients with serious underlying medical conditions, and to be a principal cause of health care associated infection worldwide. The clinical manifestations of P. aeruginosa include mostly health care associated infections and community-acquired infections. P. aeruginosa possesses an array of virulence factors that counteract host defence mechanisms. It can directly damage host tissue while utilizing high levels of intrinsic and acquired antimicrobial resistance mechanisms to counter most classes of antibiotics. P. aeruginosa co-regulates multiple resistance mechanisms by perpetually moving targets poses a significant therapeutic challenge. Thus, there is an urgent need for novel approaches in the development of anti-Pseudomonas agents. Here we review the principal infections caused by P. aeruginosa and we discuss novel therapeutic options to tackle antibiotic resistance and treatment of P. aeruginosa infections that may be further developed for clinical practice.
Collapse
Affiliation(s)
- Nikhil Sathe
- Reliance Life Sciences Pvt. Ltd., Dhirubhai Ambani Life Sciences Centre, Thane Belapur Road, Rabale, Navi Mumbai 400701, India
- School of Life and Environmental Sciences, Deakin University, Melbourne Burwood Campus, 221 Burwood Highway, Burwood Victoria 3125, Australia
| | - Peter Beech
- School of Life and Environmental Sciences, Deakin University, Melbourne Burwood Campus, 221 Burwood Highway, Burwood Victoria 3125, Australia
| | - Larry Croft
- School of Life and Environmental Sciences, Deakin University, Melbourne Burwood Campus, 221 Burwood Highway, Burwood Victoria 3125, Australia
| | - Cenk Suphioglu
- NeuroAllergy Research Laboratory, School of Life and Environmental Sciences, Deakin University, Geelong Campus at Waurn Ponds, 75 Pigdons Road, Waurn Ponds Victoria 3216, Australia
| | - Arnab Kapat
- Reliance Life Sciences Pvt. Ltd., Dhirubhai Ambani Life Sciences Centre, Thane Belapur Road, Rabale, Navi Mumbai 400701, India
| | - Eugene Athan
- School of Medicine, Deakin University, PO Box 281 Geelong 3220, Australia
| |
Collapse
|
7
|
Kanak KR, Dass RS, Pan A. Anti-quorum sensing potential of selenium nanoparticles against LasI/R, RhlI/R, and PQS/MvfR in Pseudomonas aeruginosa: a molecular docking approach. Front Mol Biosci 2023; 10:1203672. [PMID: 37635941 PMCID: PMC10449602 DOI: 10.3389/fmolb.2023.1203672] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/25/2023] [Indexed: 08/29/2023] Open
Abstract
Pseudomonas aeruginosa is an infectious pathogen which has the ability to cause primary and secondary contagions in the blood, lungs, and other body parts of immunosuppressed individuals, as well as community-acquired diseases, such as folliculitis, osteomyelitis, pneumonia, and others. This opportunistic bacterium displays drug resistance and regulates its pathogenicity via the quorum sensing (QS) mechanism, which includes the LasI/R, RhlI/R, and PQS/MvfR systems. Targeting the QS systems might be an excellent way to treat P. aeruginosa infections. Although a wide array of antibiotics, namely, newer penicillins, cephalosporins, and combination drugs are being used, the use of selenium nanoparticles (SeNPs) to cure P. aeruginosa infections is extremely rare as their mechanistic interactions are weakly understood, which results in carrying out this study. The present study demonstrates a computational approach of binding the interaction pattern between SeNPs and the QS signaling proteins in P. aeruginosa, utilizing multiple bioinformatics approaches. The computational investigation revealed that SeNPs were acutely 'locked' into the active region of the relevant proteins by the abundant residues in their surroundings. The PatchDock-based molecular docking analysis evidently indicated the strong and significant interaction between SeNPs and the catalytic cleft of LasI synthase (Phe105-Se = 2.7 Å and Thr121-Se = 3.8 Å), RhlI synthase (Leu102-Se = 3.7 Å and Val138-Se = 3.2 Å), transcriptional receptor protein LasR (Lys42-Se = 3.9 Å, Arg122-Se = 3.2 Å, and Glu124-Se = 3.9 Å), RhlR (Tyr43-Se = 2.9 Å, Tyr45-Se = 3.4 Å, and His61-Se = 3.5 Å), and MvfR (Leu208-Se = 3.2 Å and Arg209-Se = 4.0 Å). The production of acyl homoserine lactones (AHLs) was inhibited by the use of SeNPs, thereby preventing QS as well. Obstructing the binding affinity of transcriptional regulatory proteins may cause the suppression of LasR, RhlR, and MvfR systems to become inactive, thereby blocking the activation of QS-regulated virulence factors along with their associated gene expression. Our findings clearly showed that SeNPs have anti-QS properties against the established QS systems of P. aeruginosa, which strongly advocated that SeNPs might be a potent solution to tackle drug resistance and a viable alternative to conventional antibiotics along with being helpful in therapeutic development to cure P. aeruginosa infections.
Collapse
Affiliation(s)
- Kanak Raj Kanak
- Fungal Genetics and Mycotoxicology Laboratory, Department of Microbiology, School of Life Sciences, Pondicherry University (A Central University), Pondicherry, India
| | - Regina Sharmila Dass
- Fungal Genetics and Mycotoxicology Laboratory, Department of Microbiology, School of Life Sciences, Pondicherry University (A Central University), Pondicherry, India
| | - Archana Pan
- Department of Bioinformatics, School of Life Sciences, Pondicherry University (A Central University), Pondicherry, India
| |
Collapse
|
8
|
Kumar L, Bisen M, Harjai K, Chhibber S, Azizov S, Lalhlenmawia H, Kumar D. Advances in Nanotechnology for Biofilm Inhibition. ACS OMEGA 2023; 8:21391-21409. [PMID: 37360468 PMCID: PMC10286099 DOI: 10.1021/acsomega.3c02239] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023]
Abstract
Biofilm-associated infections have emerged as a significant public health challenge due to their persistent nature and increased resistance to conventional treatment methods. The indiscriminate usage of antibiotics has made us susceptible to a range of multidrug-resistant pathogens. These pathogens show reduced susceptibility to antibiotics and increased intracellular survival. However, current methods for treating biofilms, such as smart materials and targeted drug delivery systems, have not been found effective in preventing biofilm formation. To address this challenge, nanotechnology has provided innovative solutions for preventing and treating biofilm formation by clinically relevant pathogens. Recent advances in nanotechnological strategies, including metallic nanoparticles, functionalized metallic nanoparticles, dendrimers, polymeric nanoparticles, cyclodextrin-based delivery, solid lipid nanoparticles, polymer drug conjugates, and liposomes, may provide valuable technological solutions against infectious diseases. Therefore, it is imperative to conduct a comprehensive review to summarize the recent advancements and limitations of advanced nanotechnologies. The present Review encompasses a summary of infectious agents, the mechanisms that lead to biofilm formation, and the impact of pathogens on human health. In a nutshell, this Review offers a comprehensive survey of the advanced nanotechnological solutions for managing infections. A detailed presentation has been made as to how these strategies may improve biofilm control and prevent infections. The key objective of this Review is to summarize the mechanisms, applications, and prospects of advanced nanotechnologies to provide a better understanding of their impact on biofilm formation by clinically relevant pathogens.
Collapse
Affiliation(s)
- Lokender Kumar
- School
of Biotechnology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh 173229, India
- Cancer
Biology Laboratory, Raj Khosla Centre for Cancer Research, Shoolini University, Solan, Himachal Pradesh 173229, India
| | - Monish Bisen
- School
of Biotechnology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh 173229, India
| | - Kusum Harjai
- Department
of Microbiology, Panjab University, Chandigarh 160014, India
| | - Sanjay Chhibber
- Department
of Microbiology, Panjab University, Chandigarh 160014, India
| | - Shavkatjon Azizov
- Laboratory
of Biological Active Macromolecular Systems, Institute of Bioorganic
Chemistry, Academy of Sciences Uzbekistan, Tashkent 100125, Uzbekistan
- Faculty
of Life Sciences, Pharmaceutical Technical
University, Tashkent 100084, Uzbekistan
| | - Hauzel Lalhlenmawia
- Department
of Pharmacy, Regional Institute of Paramedical
and Nursing Sciences, Zemabawk, Aizawl, Mizoram 796017, India
| | - Deepak Kumar
- Department
of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh173229, India
| |
Collapse
|
9
|
Christine E, Olive C, Louisin M, Dramé M, Marion‐Sanchez K. A new spray-based method for the in-vitro development of dry-surface biofilms. Microbiologyopen 2023; 12:e1330. [PMID: 36825879 PMCID: PMC9834607 DOI: 10.1002/mbo3.1330] [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: 06/21/2022] [Accepted: 10/12/2022] [Indexed: 01/13/2023] Open
Abstract
The inanimate environment immediately surrounding the patient in healthcare facilities is a reservoir of microorganisms embedded in dry-surface biofilms (DSB). These biofilms, first highlighted in 2012, are increasingly studied, but currently available in-vitro models only allow for the growth of semi-hydrated biofilms. We developed a new in-vitro method under actual dehydration conditions based on the hypothesis that surface contamination is mainly due to splashes of respiratory secretions. The main objective of this study was to show that the operating conditions we have defined allowed the growth of DSB with a methicillin resistant Staphylococcus aureus strain. The second objective was to show that extended-spectrum beta-lactamase-producing Enterobacteriaceae, that is, Klebsiella pneumoniae and Enterobacter cloacae were also able to grow such biofilms under these conditions. Monobacterial suspensions in sterile artificial saliva (SAS) were sprayed onto polyethylene surfaces. Nutrients and hydration were provided daily by spraying SAS enriched with 20% of Brain Heart Infusion broth. The primary outcome was mean surface coverage measured by image analysis after crystal violet staining. The method applied to S. aureus for 12 days resulted in reproducible and repeatable DSB consisting of isolated and confluent microcolonies embedded in extracellular polymeric substances as shown in scanning electron microscopy images. Similar DSB were obtained with both Enterobacteriaceae applying the same method. No interspecies variation was shown between the three strains in terms of surface coverage. These first trials are the starting point for a 3-year study currently in process.
Collapse
Affiliation(s)
- Esther Christine
- Department of BacteriologyHygiene and Environment Laboratory, CHU MartiniqueCS 90632Fort‐de‐FranceCedexMartinique
| | - Claude Olive
- Department of BacteriologyHygiene and Environment Laboratory, CHU MartiniqueCS 90632Fort‐de‐FranceCedexMartinique
| | - Myriam Louisin
- Department of BacteriologyHygiene and Environment Laboratory, CHU MartiniqueCS 90632Fort‐de‐FranceCedexMartinique
| | - Moustapha Dramé
- Department of Clinical Research and InnovationCHU MartiniqueCS 90632Fort‐de‐FranceCedexMartinique
| | - Karine Marion‐Sanchez
- Department of BacteriologyHygiene and Environment Laboratory, CHU MartiniqueCS 90632Fort‐de‐FranceCedexMartinique
- Department of Hospital HygieneCHU MartiniqueCS 90632Fort‐de‐FranceCedexMartinique
- Pathogenesis and Control of Chronic and Emerging Infections, Université de Montpellier, Université des Antilles, Inserm, Etablissement Français du SangCHU MartiniqueMontpellierFrance
| |
Collapse
|
10
|
Sarveswari HB, Gupta KK, Durai R, Solomon AP. Development of a smart pH-responsive nano-polymer drug, 2-methoxy-4-vinylphenol conjugate against the intestinal pathogen, Vibrio cholerae. Sci Rep 2023; 13:1250. [PMID: 36690664 PMCID: PMC9871008 DOI: 10.1038/s41598-023-28033-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 01/11/2023] [Indexed: 01/24/2023] Open
Abstract
Vibrio cholerae causes cholera, an acute diarrhoeal disease. The virulence in V. cholerae is regulated by the quorum-sensing mechanism and response regulator LuxO positively regulates the expression of virulence determinants adhesion, biofilm formation, and cholera toxin production. Previous in-silico studies revealed that 2-methoxy-4-vinylphenol could bind to the ATP binding site of LuxO and the complex was compact and stable in pHs like intestinal pHs. Here, we have explored the polymeric nano-formulation of 2-methoxy-4-vinylphenol using cellulose acetate phthalate for controlled drug release and their effectiveness in attenuating the expression of V. cholerae virulence. Physico-chemical characterization of the formulation showed particles with a mean size of 91.8 ± 14 nm diameter and surface charge of - 14.7 ± 0.07 mV. The uniform round polymeric nanoparticles formed displayed about 51% burst release of the drug at pH 7 by 3rd h, followed by a controlled linear release in alkaline pH. The polymeric nanoparticles demonstrated a tenfold increase in intestinal membrane permeability ex-vivo. At lower concentrations, the 2-methoxy-4-vinylphenol polymeric nanoparticles were non-cytotoxic to Int 407 cells. In-vitro analysis at pH 6, pH 7, pH 8, and pH 9 revealed that cellulose acetate phthalate-2-methoxy-4-vinylphenol nanoparticles were non-bactericidal at concentrations up to 500 μg/mL. At 31.25 μg/mL, the nanoparticles inhibited about 50% of the biofilm formation of V. cholerae MTCC 3905 and HYR14 strains. At this concentration, the adherence of V. cholerae MTCC 3905 and HYR14 to Int 407 cell lines were also significantly affected. Gene expression analysis revealed that the expression of tcp, qrr, and ct at pH 6, 7, 8, and 9 has reduced. The CAP-2M4VP nanoparticles have demonstrated the potential to effectively reduce the virulence of V. cholerae in-vitro.
Collapse
Affiliation(s)
- Hema Bhagavathi Sarveswari
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, 613401, India
| | - Krishna Kant Gupta
- School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, 613401, India
| | - Ramyadevi Durai
- Pharmaceutical Technology Laboratory, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, 613401, India.
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, 613401, India.
| |
Collapse
|
11
|
A Fucosylated Lactose-Presenting Tetravalent Glycocluster Acting as a Mutual Ligand of Pseudomonas aeruginosa Lectins A (PA-IL) and B (PA-IIL)-Synthesis and Interaction Studies. Int J Mol Sci 2022; 23:ijms232416194. [PMID: 36555839 PMCID: PMC9782601 DOI: 10.3390/ijms232416194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
The Gram-negative bacterium Pseudomonas aeruginosa is an important opportunistic human pathogen associated with cystic fibrosis. P. aeruginosa produces two soluble lectins, the d-galactose-specific lectin PA-IL (LecA) and the l-fucose-specific lectin PA-IIL (LecB), among other virulence factors. These lectins play an important role in the adhesion to host cells and biofilm formation. Moreover, PA-IL is cytotoxic to respiratory cells in the primary culture. Therefore, these lectins are promising therapeutic targets. Specifically, carbohydrate-based compounds could inhibit their activity. In the present work, a 3-O-fucosyl lactose-containing tetravalent glycocluster was synthesized and utilized as a mutual ligand of galactophilic and fucophilic lectins. Pentaerythritol equipped with azido ethylene glycol-linkers was chosen as a multivalent scaffold and the glycocluster was constructed by coupling the scaffold with propargyl 3-O-fucosyl lactoside using an azide-alkyne 1,3-dipolar cycloaddition reaction. The interactions between the glycocluster and PA-IL or PA-IIL were investigated by isothermal titration microcalorimetry and saturation transfer difference NMR spectroscopy. These results may assist in the development of efficient anti-adhesion therapy for the treatment of a P. aeruginosa infection.
Collapse
|
12
|
Schifano NP, Caputo GA. Investigation of the Role of Hydrophobic Amino Acids on the Structure-Activity Relationship in the Antimicrobial Venom Peptide Ponericin L1. J Membr Biol 2022; 255:537-551. [PMID: 34792624 PMCID: PMC9114170 DOI: 10.1007/s00232-021-00204-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 10/13/2021] [Indexed: 10/19/2022]
Abstract
Venom mixtures from insects, reptiles, and mollusks have long been a source of bioactive peptides which often have alternative uses as therapeutics. While these molecules act in numerous capacities, there have been many venom components that act on the target cells through membrane disruptive mechanisms. These peptides have long been of interest as potential antimicrobial peptide platforms, but the inherent cytotoxicity of venom peptides often results in poor therapeutic potential. Despite this, efforts are ongoing to identify and characterize venom peptide which exhibit high antimicrobial activity with low cytotoxicity and modify these to further enhance the efficacy while reducing toxicity. One example is ponericin L1 from Neoponera goeldii which has been demonstrated to have good antimicrobial activity and low in vitro cytotoxicity. The L1 sequence was modified by uniformly replacing the native hydrophobic residues with either Leu, Ile, Phe, Ala, or Val. Spectroscopic and microbiological approaches were employed to investigate how the amino acid sequence changes impacted membrane interaction, secondary structure, and antimicrobial efficacy. The L1 derivatives showed varying degrees of bilayer interaction, in some cases driven by bilayer composition. Several of the variants exhibited enhanced antimicrobial activity compared to the parent strain, while others lost all activity. Interestingly, the variant containing Val lost all antimicrobial activity and ability to interact with bilayers. Taken together the results indicate that peptide secondary structure, amino acid composition, and hydrophobicity all play a role in peptide activity, although this is a delicate balance that can result in non-specific binding or complete loss of activity if specific amino acids are incorporated.
Collapse
Affiliation(s)
- Nicholas P Schifano
- Department of Chemistry & Biochemistry, Rowan University, 201 Mullica Hill Road, Glassboro, NJ, 08028, USA
| | - Gregory A Caputo
- Department of Chemistry & Biochemistry, Rowan University, 201 Mullica Hill Road, Glassboro, NJ, 08028, USA.
- Department of Molecular & Cellular Biosciences, Rowan University, 201 Mullica Hill Road, Glassboro, NJ, 08028, USA.
| |
Collapse
|
13
|
Wojtczak K, Byrne JP. Structural Considerations for Building Synthetic Glycoconjugates as Inhibitors for Pseudomonas aeruginosa Lectins. ChemMedChem 2022; 17:e202200081. [PMID: 35426976 PMCID: PMC9321714 DOI: 10.1002/cmdc.202200081] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/13/2022] [Indexed: 11/16/2022]
Abstract
Pseudomonas aeruginosa is a pathogenic bacterium, responsible for a large portion of nosocomial infections globally and designated as critical priority by the World Health Organisation. Its characteristic carbohydrate-binding proteins LecA and LecB, which play a role in biofilm-formation and lung-infection, can be targeted by glycoconjugates. Here we review the wide range of inhibitors for these proteins (136 references), highlighting structural features and which impact binding affinity and/or therapeutic effects, including carbohydrate selection; linker length and rigidity; and scaffold topology, particularly for multivalent candidates. We also discuss emerging therapeutic strategies, which build on targeting of LecA and LecB, such as anti-biofilm activity, anti-adhesion and drug-delivery, with promising prospects for medicinal chemistry.
Collapse
Affiliation(s)
- Karolina Wojtczak
- School of Biological and Chemical SciencesNational University of Ireland GalwayUniversity RoadGalwayIreland
| | - Joseph P. Byrne
- School of Biological and Chemical SciencesNational University of Ireland GalwayUniversity RoadGalwayIreland
| |
Collapse
|
14
|
Alhajj N, O'Reilly NJ, Cathcart H. Developing ciprofloxacin dry powder for inhalation: A story of challenges and rational design in the treatment of cystic fibrosis lung infection. Int J Pharm 2021; 613:121388. [PMID: 34923051 DOI: 10.1016/j.ijpharm.2021.121388] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 12/08/2021] [Accepted: 12/12/2021] [Indexed: 12/11/2022]
Abstract
Cystic fibrosis (CF) is an inherited multisystem disease affecting the lung which leads to a progressive decline in lung function as a result of malfunctioning mucociliary clearance and subsequent chronic bacterial infections. Pseudomonas aeruginosa is the predominant cause of lung infection in CF patients and is associated with significant morbidity and mortality. Thus, antibiotic therapy remains the cornerstone of the treatment of CF. Pulmonary delivery of antibiotics for lung infections significantly reduces the required dose and the associated systemic side effects while improving therapeutic outcomes. Ciprofloxacin is one of the most widely used antibiotics against P. aeruginosa and the most effective fluoroquinolone. However, in spite of the substantial amount of research aimed at developing ciprofloxacin powder for inhalation, none of these formulations has been commercialized. Here, we present an integrated view of the diverse challenges associated with delivering ciprofloxacin dry particles to the lungs of CF patients and the rationales behind recent formulations of ciprofloxacin dry powder for inhalation. This review will discuss the challenges in developing ciprofloxacin powder for inhalation along with the physiological and pathophysiological challenges such as ciprofloxacin lung permeability, overproduction of viscous mucus and bacterial biofilms. The review will also discuss the current and emerging particle engineering approaches to overcoming these challenges. By doing so, we believe the review will help the reader to understand the current limitations in developing an inhalable ciprofloxacin powder and explore new opportunities of rational design strategies.
Collapse
Affiliation(s)
- Nasser Alhajj
- Pharmaceutical and Molecular Biotechnology Research Centre (PMBRC), Waterford Institute of Technology, Waterford, Ireland.
| | - Niall J O'Reilly
- Pharmaceutical and Molecular Biotechnology Research Centre (PMBRC), Waterford Institute of Technology, Waterford, Ireland; SSPC - The Science Foundation Ireland Research Centre for Pharmaceuticals, Ireland
| | - Helen Cathcart
- Pharmaceutical and Molecular Biotechnology Research Centre (PMBRC), Waterford Institute of Technology, Waterford, Ireland
| |
Collapse
|
15
|
Thambirajoo M, Maarof M, Lokanathan Y, Katas H, Ghazalli NF, Tabata Y, Fauzi MB. Potential of Nanoparticles Integrated with Antibacterial Properties in Preventing Biofilm and Antibiotic Resistance. Antibiotics (Basel) 2021; 10:1338. [PMID: 34827276 PMCID: PMC8615099 DOI: 10.3390/antibiotics10111338] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/24/2021] [Accepted: 10/26/2021] [Indexed: 01/13/2023] Open
Abstract
Nanotechnology has become an emerging technology in the medical field and is widely applicable for various clinical applications. The potential use of nanoparticles as antimicrobial agents is greatly explored and taken into consideration as alternative methods to overcome the challenges faced by healthcare workers and patients in preventing infections caused by pathogenic microorganisms. Among microorganisms, bacterial infections remain a major hurdle and are responsible for high morbidity and mortality globally, especially involving those with medical conditions and elderly populations. Over time, these groups are more vulnerable to developing resistance to antibiotics, as bacterial biofilms are difficult to destroy or eliminate via antibiotics; thus, treatment becomes unsuccessful or ineffective. Mostly, bacterial biofilms and other microbes can be found on medical devices and wounds where they disperse their contents which cause infections. To inhibit biofilm formations and overcome antibiotic resistance, antimicrobial-loaded nanoparticles alone or combined with other substances could enhance the bactericidal activity of nanomaterials. This includes killing the pathogens effectively without harming other cells or causing any adverse effects to living cells. This review summarises the mechanisms of actions employed by the different types of nanoparticles which counteract infectious agents in reducing biofilm formation and improve antibiotic therapy for clinical usage.
Collapse
Affiliation(s)
- Maheswary Thambirajoo
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (M.T.); (M.M.); (Y.L.)
| | - Manira Maarof
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (M.T.); (M.M.); (Y.L.)
| | - Yogeswaran Lokanathan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (M.T.); (M.M.); (Y.L.)
| | - Haliza Katas
- Centre for Drug Delivery Research, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| | - Nur Fatiha Ghazalli
- Biomaterials Unit, School of Dental Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Malaysia;
| | - Yasuhiko Tabata
- Department of Biomaterials, Institute for Frontier Medical Sciences, Kyoto University, 53 Kawara-cho Shogoin, Sakyo-ku, Kyoto 606-8507, Japan;
| | - Mh Busra Fauzi
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (M.T.); (M.M.); (Y.L.)
| |
Collapse
|
16
|
Birk SE, Boisen A, Nielsen LH. Polymeric nano- and microparticulate drug delivery systems for treatment of biofilms. Adv Drug Deliv Rev 2021; 174:30-52. [PMID: 33845040 DOI: 10.1016/j.addr.2021.04.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/23/2021] [Accepted: 04/06/2021] [Indexed: 12/11/2022]
Abstract
Now-a-days healthcare systems face great challenges with antibiotic resistance and low efficacy of antibiotics when combating pathogenic bacteria and bacterial biofilms. Administration of an antibiotic in its free form is often ineffective due to lack of selectivity to the infectious site and breakdown of the antibiotic before it exerts its effect. Therefore, polymeric delivery systems, where the antibiotic is encapsulated into a formulation, have shown great promise, facilitating a high local drug concentration at the site of infection, a controlled drug release and less drug degradation. All this leads to improved therapeutic effects and fewer systemic side effects together with a lower risk of developing antibiotic resistance. Here, we review and provide a comprehensive overview of polymer-based nano- and microparticles as carriers for antimicrobial agents and their effect on eradicating bacterial biofilms. We have a main focus on polymeric particulates containing poly(lactic-co-glycolic acid), chitosan and polycaprolactone, but also strategies involving combinations of these polymers are included. Different production techniques are reviewed and important parameters for biofilm treatment are discussed such as drug loading capacity, control of drug release, influence of particle size and mobility in biofilms. Additionally, we reflect on other promising future strategies for combating biofilms such as lipid-polymer hybrid particles, enzymatic biofilm degradation, targeted/triggered antibiotic delivery and future alternatives to the conventional particles.
Collapse
|
17
|
O'Reilly C, Blasco S, Parekh B, Collins H, Cooke G, Gunnlaugsson T, Byrne JP. Ruthenium-centred btp glycoclusters as inhibitors for Pseudomonas aeruginosa biofilm formation. RSC Adv 2021; 11:16318-16325. [PMID: 35479152 PMCID: PMC9030604 DOI: 10.1039/d0ra05107a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 04/26/2021] [Indexed: 11/21/2022] Open
Abstract
Carbohydrate-decorated clusters (glycoclusters) centred on a Ru(ii) ion were synthesised and tested for their activity against Pseudomonas aeruginosa biofilm formation. These clusters were designed by conjugating a range of carbohydrate motifs (galactose, glucose, mannose and lactose, as well as galactose with a triethylene glycol spacer) to a btp (2,6-bis(1,2,3-triazol-4-yl)pyridine) scaffold. This scaffold, which possesses a C2 symmetry, is an excellent ligand for d-metal ions, and thus the formation of the Ru(ii)-centred glycoclusters 7 and 8Gal was achieved from 5 and 6Gal; each possessing four deprotected carbohydrates. Glycocluster 8Gal, which has a flexible spacer between the btp and galactose moieties, showed significant inhibition of P. aeruginosa bacterial biofilm formation. By contrast, glycocluster 7, which lacked the flexible linker, didn't show significant antimicrobial effects and neither does the ligand 6Gal alone. These results are proposed to arise from carbohydrate–lectin interactions with LecA, which are possible for the flexible metal-centred multivalent glycocluster. Metal-centred glycoclusters present a structurally versatile class of antimicrobial agent for P. aeruginosa, of which this is, to the best of our knowledge, the first example. Ruthenium-centred glycoclusters based on carbohydrate-functionalised bis(triazolyl)pyridine ligands show Pseudomonas aeruginosa biofilm inhibition, with activity that is dependent on ligand structure.![]()
Collapse
Affiliation(s)
- Ciaran O'Reilly
- School of Chemistry, Trinity Biomedical Sciences Institute, Trinity College Dublin Ireland.,School of Medicine, University College Dublin Belfield Dublin 4 Ireland
| | - Salvador Blasco
- School of Chemistry, Trinity Biomedical Sciences Institute, Trinity College Dublin Ireland
| | - Bina Parekh
- School of Medicine, University College Dublin Belfield Dublin 4 Ireland
| | - Helen Collins
- Department of Applied Science, Tallaght Campus, Technological University Dublin Ireland
| | - Gordon Cooke
- School of Medicine, University College Dublin Belfield Dublin 4 Ireland.,Department of Applied Science, Tallaght Campus, Technological University Dublin Ireland
| | | | - Joseph P Byrne
- School of Chemistry, National University of Ireland Galway University Road Galway Ireland
| |
Collapse
|
18
|
Fattah RAFA, Fathy FEZY, Mohamed TAH, Elsayed MS. Effect of chitosan nanoparticles on quorum sensing-controlled virulence factors and expression of LasI and RhlI genes among Pseudomonas aeruginosa clinical isolates. AIMS Microbiol 2021; 7:415-430. [PMID: 35071940 PMCID: PMC8712529 DOI: 10.3934/microbiol.2021025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 10/20/2021] [Indexed: 12/25/2022] Open
Abstract
<abstract>
<p>Antibiotic-resistant strains of <italic>Pseudomonas aeruginosa (P. aeruginosa</italic>) pose a major threat for healthcare-associated and community-acquired infections. <italic>P. aeruginosa</italic> is recognized as an opportunistic pathogen using quorum sensing (QS) system to regulate the expression of virulence factors and biofilm development. Thus, meddling with the QS system would give alternate methods of controlling the pathogenicity. This study aimed to assess the inhibitory impact of chitosan nanoparticles (CS-NPs) on <italic>P. aeruginosa</italic> virulence factors regulated by QS (e.g., motility and biofilm formation) and <italic>LasI</italic> and <italic>RhlI</italic> gene expression. Minimum inhibitory concentration (MIC) of CS-NPs against 30 isolates of <italic>P. aeruginosa</italic> was determined. The CS-NPs at sub-MIC were utilized to assess their inhibitory effect on motility, biofilm formation, and the expression levels of <italic>LasI</italic> and <italic>RhlI</italic> genes. CS-NPs remarkably inhibited the tested virulence factors as compared to the controls grown without the nanoparticles. The mean (±SD) diameter of swimming motility was decreased from 3.93 (±1.5) to 1.63 (±1.02) cm, and the mean of the swarming motility was reduced from 3.5 (±1.6) to 1.9 (±1.07) cm. All isolates became non-biofilm producers, and the mean percentage rate of biofilm inhibition was 84.95% (±6.18). Quantitative real-time PCR affirmed the opposition of QS activity by lowering the expression levels of <italic>LasI</italic> and <italic>RhlI</italic> genes; the expression level was decreased by 90- and 100-folds, respectively. In conclusion, the application of CS-NPs reduces the virulence factors significantly at both genotypic and phenotypic levels. These promising results can breathe hope in the fight against resistant <italic>P. aeruginosa</italic> by repressing its QS-regulated virulence factors.</p>
</abstract>
Collapse
|
19
|
Antiquorum Sensing Activity of Copper Nanoparticle in Pseudomonas aeruginosa: An In Silico Approach. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/s40011-020-01193-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
20
|
Rathnayake K, Patel U, Pham C, McAlpin A, Budisalich T, Jayawardena SN. Targeted Delivery of Antibiotic Therapy to Inhibit Pseudomonas aeruginosa Using Lipid-Coated Mesoporous Silica Core–Shell Nanoassembly. ACS APPLIED BIO MATERIALS 2020; 3:6708-6721. [DOI: 10.1021/acsabm.0c00622] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Kavini Rathnayake
- Department of Chemistry, The University of Alabama in Huntsville, Huntsville, Alabama 35899, United States
| | - Unnati Patel
- Department of Chemistry, The University of Alabama in Huntsville, Huntsville, Alabama 35899, United States
| | - Chi Pham
- Department of Chemistry, The University of Alabama in Huntsville, Huntsville, Alabama 35899, United States
| | - Anna McAlpin
- Department of Chemistry, The University of Alabama in Huntsville, Huntsville, Alabama 35899, United States
| | - Travis Budisalich
- Department of Chemistry, The University of Alabama in Huntsville, Huntsville, Alabama 35899, United States
| | - Surangi N. Jayawardena
- Department of Chemistry, The University of Alabama in Huntsville, Huntsville, Alabama 35899, United States
| |
Collapse
|
21
|
Paunova-Krasteva T, Haladjova E, Petrov P, Forys A, Trzebicka B, Topouzova-Hristova T, R Stoitsova S. Destruction of Pseudomonas aeruginosa pre-formed biofilms by cationic polymer micelles bearing silver nanoparticles. BIOFOULING 2020; 36:679-695. [PMID: 32741293 DOI: 10.1080/08927014.2020.1799354] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 06/30/2020] [Accepted: 07/14/2020] [Indexed: 06/11/2023]
Abstract
Pseudomonas aeruginosa is an opportunistic Gram-negative pathogen often associated with biofilm infections. This study evaluated the capacity for biofilm destruction of a novel combination of cationic polymer micelles formed from poly(2-(dimethylamino)ethyl methacrylate)-b-poly(ε-caprolactone)-b-poly(2-(dimethylamino)ethyl methacrylate) (PDMAEMA-PCL-PDMAEMA) triblock copolymer either alone, or loaded with silver nanoparticles (M_AgNPs). Pre-formed P. aeruginosa biofilms were incubated with either blank micelles, AgNO3, or M_AgNPs. Biofilm biomass (crystal violet assay), metabolic activity (Alamar blue reduction), structure (SEM) and viability (CLSM after Live/Dead staining, or plating for CFU) were checked. The results showed that the micelles alone loosened the biofilm matrix, and caused some alterations in the bacterial surface. AgNO3 killed the bacteria in situ leaving dead biofilm bacteria on the surface. M_AgNPs combined the two types of activities causing significant biofilm reduction, and alteration and death of biofilm bacteria. Therefore, the applied PDMAEMA-based micelles appear to be a successful candidate for the treatment of P. aeruginosa biofilm infections.
Collapse
Affiliation(s)
| | - Emi Haladjova
- Bulgarian Academy of Sciences, Institute of Polymers, Sofia, Bulgaria
| | - Petar Petrov
- Bulgarian Academy of Sciences, Institute of Polymers, Sofia, Bulgaria
| | - Aleksander Forys
- Polish Academy of Sciences, Centre of Polymer and Carbon Materials, Zabrze, Poland
| | - Barbara Trzebicka
- Polish Academy of Sciences, Centre of Polymer and Carbon Materials, Zabrze, Poland
| | | | - Stoyanka R Stoitsova
- Bulgarian Academy of Sciences, The Stephan Angeloff Institute of Microbiology, Sofia, Bulgaria
| |
Collapse
|
22
|
Chirgwin ME, Dedloff MR, Holban AM, Gestal MC. Novel Therapeutic Strategies Applied to Pseudomonas aeruginosa Infections in Cystic Fibrosis. MATERIALS 2019; 12:ma12244093. [PMID: 31817881 PMCID: PMC6947192 DOI: 10.3390/ma12244093] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/25/2019] [Accepted: 12/05/2019] [Indexed: 11/16/2022]
Abstract
Cystic fibrosis (CF) is one of the most prevalent genetic diseases and a total of 1700 different genetic mutations can cause this condition. Patients that suffer this disease have a thickening of the mucus, creating an environment that promotes bacterial infections. Pseudomonas aeruginosa is a ubiquitous bacterium, which is frequently found in the lungs of CF patients. P. aeruginosa is known for its high level of antibiotic resistance as well as its high rate of mutation that allows it to rapidly evolve and adapt to a multitude of conditions. When a CF lung is infected with P. aeruginosa, the decay of the patient is accelerated, but there is little that can be done apart from controlling the infection with antibiotics. Novel strategies to control P. aeruginosa infection are imperative, and nanotechnology provides novel approaches to drug delivery that are more efficient than classic antibiotic treatments. These drug delivery systems are offering new prospects, especially for these patients with special mucus conditions and bacterial characteristics that limit antibiotic use.
Collapse
Affiliation(s)
- Michael E. Chirgwin
- Department of Chemical Engineering, Clarkson University, Potsdam, NY 13699, USA;
| | | | - Alina Maria Holban
- Department of Microbiology, Faculty of Biology, University of Bucharest, 030018 Bucharest, Romania;
- Research Institute of the University of Bucharest (ICUB), 050107 Bucharest, Romania
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politechnica of Bucharest, 1-7 Polizu Street, 011061 Bucharest, Romania
| | - Monica C. Gestal
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
- Correspondence: or
| |
Collapse
|
23
|
Malinovská L, Thai Le S, Herczeg M, Vašková M, Houser J, Fujdiarová E, Komárek J, Hodek P, Borbás A, Wimmerová M, Csávás M. Synthesis of β-d-galactopyranoside-Presenting Glycoclusters, Investigation of Their Interactions with Pseudomonas aeruginosa Lectin A (PA-IL) and Evaluation of Their Anti-Adhesion Potential. Biomolecules 2019; 9:E686. [PMID: 31683947 PMCID: PMC6920806 DOI: 10.3390/biom9110686] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 12/19/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic human pathogen associated with cystic fibrosis. This bacterium produces, among other virulence factors, a soluble d-galactose-specific lectin PA-IL (LecA). PA-IL plays an important role in the adhesion to the host cells and is also cytotoxic. Therefore, this protein is an interesting therapeutic target, suitable for inhibition by carbohydrate-based compounds. In the current study, β-d-galactopyranoside-containing tri- and tetravalent glycoclusters were synthesized. Methyl gallate and pentaerythritol equipped with propargyl groups were chosen as multivalent scaffolds and the galactoclusters were built from the above-mentioned cores by coupling ethylene or tetraethylene glycol-bridges and peracetylated propargyl β-d-galactosides using 1,3-dipolar azide-alkyne cycloaddition. The interaction between galactoside derivatives and PA-IL was investigated by several biophysical methods, including hemagglutination inhibition assay, isothermal titration calorimetry, analytical ultracentrifugation, and surface plasmon resonance. Their ability to inhibit the adhesion of P. aeruginosa to bronchial cells was determined by ex vivo assay. The newly synthesized multivalent galactoclusters proved to be significantly better ligands than simple d-galactose for lectin PA-IL and as a result, two representatives of the dendrimers were able to decrease adhesion of P. aeruginosa to bronchial cells to approximately 32% and 42%, respectively. The results may provide an opportunity to develop anti-adhesion therapy for the treatment of P. aeruginosa infection.
Collapse
Affiliation(s)
- Lenka Malinovská
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic.
| | - Son Thai Le
- Department of Pharmaceutical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary.
- Doctoral School of Pharmaceutical Sciences, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary.
| | - Mihály Herczeg
- Department of Pharmaceutical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary.
- Research Group for Oligosaccharide Chemistry of Hungarian Academy of Sciences, Egyetem tér 1, H-4032 Debrecen, Hungary.
| | - Michaela Vašková
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40 Prague 2, Czech Republic.
| | - Josef Houser
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic.
| | - Eva Fujdiarová
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic.
| | - Jan Komárek
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic.
| | - Petr Hodek
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40 Prague 2, Czech Republic.
| | - Anikó Borbás
- Department of Pharmaceutical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary.
| | - Michaela Wimmerová
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic.
- Department of Biochemistry, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic.
| | - Magdolna Csávás
- Department of Pharmaceutical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary.
| |
Collapse
|