1
|
Zhang Y, Zhao H, Ji SB, Xing HC. Clinical analysis of Klebsiella pneumoniae infection in patients with liver cirrhosis in Beijing. World J Hepatol 2024; 16:1441-1449. [DOI: 10.4254/wjh.v16.i12.1441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/07/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The incidence of Klebsiella pneumoniae (K. pneumoniae) infection in patients with cirrhosis has been increasing over recent years, posing certain difficulties in clinical treatment.
AIM To analyze the clinical features of patients with liver cirrhosis and identify the risk factors to help the early diagnosis and treatment of these diseases.
METHODS Clinical data and laboratory tests were collected from 72 patients with cirrhosis confirmed by secretion or blood culture of K. pneumoniae infection at Beijing Ditan Hospital, Capital Medical University, between May 2016 and October 2018. Data from hospitalized patients with liver cirrhosis and K. pneumoniae infections, including age, sex, antimicrobial use, length of stay, site of infection, distribution of pathogenic bacteria, complications, invasive operations, laboratory indicators, treatment, and clinical regression, were extracted and retrospectively analyzed. Clinical data and biochemical values were included in the multivariate logistic regression analysis.
RESULTS A total of 52 men and 20 women, with an age range from 29 to 85 years and an average age of 57.7 ± 12.54, were analyzed. The incidence of hospital K. pneumoniae infection in patients with cirrhosis was approximately 19.44%. The most common the infection site was the bloodstream, followed by the respiratory tract, abdominal cavity, and biliary tract. Risk factors for infection were old age, long hospital stays, gastrointestinal bleeding, and low serum albumin levels, while prophylactic antibiotics were protective factors. The multivariate analysis suggested that other infections, chronic diseases, and invasive procedures were independent factors.
CONCLUSION In clinical practice, the length of hospital stays should be shortened as much as possible, invasive operations should be reduced, antibiotics should be rationally used, and the patients’ liver function should be timely improved. This is of great significance for reducing the incidence of hospital infection.
Collapse
Affiliation(s)
- Yu Zhang
- Center of Liver Diseases Division 3, Beijing Ditan Hospital, Capital Medical University, Peking University Ditan Teaching Hospital, Beijing 100015, China
| | - Hong Zhao
- Center of Liver Diseases Division 1, Beijing Ditan Hospital, Capital Medical University, Peking University Ditan Teaching Hospital, Beijing 100015, China
| | - Shi-Bo Ji
- Center of Liver Diseases Division 3, Beijing Ditan Hospital, Capital Medical University, Peking University Ditan Teaching Hospital, Beijing 100015, China
| | - Hui-Chun Xing
- Center of Liver Diseases Division 3, Beijing Ditan Hospital, Capital Medical University, Peking University Ditan Teaching Hospital, Beijing 100015, China
| |
Collapse
|
2
|
Yadav B, Jaiswal A, Kumar D, Karad DD, Joshi PR, Kamble DP, Mercier C, Kharat AS. Sub-minimum inhibitory concentrations in ceftazidime exacerbate the formation of Acinetobacter baumannii biofilms. Microb Pathog 2024; 199:107229. [PMID: 39675437 DOI: 10.1016/j.micpath.2024.107229] [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: 10/18/2024] [Revised: 12/04/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
Associated with nosocomial infections, the environmental Gram-negative coccobacillus A. baumannii leads to various kinds of high mortality-rate infections among which pneumonias mainly in immune-compromised people from health-care facilities. A critical component of the current antibiotic resistance problem is the presence of antibiotics sub-minimum inhibitory concentrations (sub-MICs) in a variety of natural settings including drinking water, sewage water, rivers, lakes, and natural sludge. In India, third-generation cephalosporins such as ceftazidime (CAZ) count among the most often prescribed β-lactams to treat infections by A. baumannii. In this study, we showed that CAZ sub-MICs 1/reduce adhesion to lung epithelial cells and slow down the growth of the A. baumannii KSK1 strain, which nevertheless quickly resumes its growth; 2/alter the morphology of A. baumannii KSK1 planktonic cells and induce the formation of bacterial aggregates that resemble biofilms; 3/increase the in vitro formation of biofilms by A. baumannii KSK1 bacterial cells. Our findings underscore the importance of considering sub-MICs in antibiotic therapy and environmental contamination as the antibiotics sub-MICs potentially found in wastewater may contribute to the selection causing antibiotic resistance and persistence of antibiotic-resistant strains.
Collapse
Affiliation(s)
- Bipin Yadav
- Laboratory of Applied Microbiology and Cancer Remedies, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Anjali Jaiswal
- Laboratory of Applied Microbiology and Cancer Remedies, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Durgesh Kumar
- Laboratory of Applied Microbiology and Cancer Remedies, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Dilip D Karad
- Department of Microbiology, Shri Shivaji Mahavidyalaya, Barshi, 413401, India
| | - Prashant R Joshi
- Laboratory of Applied Microbiology and Cancer Remedies, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India; Department of Chemistry, S. B. E. S. College of Sciences, Chattrapati Sambhajinagar, 431001, India
| | - Dhanraj P Kamble
- Department of Chemistry, S. B. E. S. College of Sciences, Chattrapati Sambhajinagar, 431001, India
| | - Corinne Mercier
- Translational Innovation in Medicine and Complexity (TIMC) Laboratory, Translational Microbiology - Evolution - Engineering (TrEE) Team, UMR5525 CNRS/Université Grenoble Alpes/VetAgro'Sup, Grenoble, France.
| | - Arun S Kharat
- Laboratory of Applied Microbiology and Cancer Remedies, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
3
|
Raghoonanadan A, Dweba Y, Aruwa CE, Sabiu S. Metabolomic fingerprinting, molecular modelling and experimental bioprospection of Helianthus annuus seed cultivars as Pseudomonas aeruginosa LasR modulators. Bioorg Chem 2024; 154:108046. [PMID: 39693924 DOI: 10.1016/j.bioorg.2024.108046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 11/26/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024]
Abstract
The Pseudomonas aeruginosa LasR quorum sensing system (QSS) is central to regulating the expression of several pathogenicity factors. Also, while seed- and/or plant-derived products have been investigated as QSS regulators, the impact of Helianthus annuus (Pannar sunflower seed cultivars) extracts and metabolites as LasR modulators remain underexplored. Thus, this study focused on the untargeted metabolomic profiling (Liquid Chromatography-Mass Spectrometry), in vitro and in silico (docking, pharmacokinetics, dynamic simulation) bioprospection of Pannar seed cultivars' extracts and metabolites as LasR modulators. The extracts showed significant QS modulating properties (motility, violacein, biofilm, cell attachment, pyocyanin inhibition) with the PAN 7102 CLP seed cultivar (74.3 %) being the most potent, compared to azithromycin (65 %) and cinnamaldehyde (62 %). Chemometric principal component analysis (PCA) analysis showed distinct metabolite signatures with 52.5 % variance across the six cultivars that was driven by aqueous and ethanolic extracts of PAN 7102, 7160, and 7156 cultivars. The presence of methoxymellein, hydroxytetradecanedioic acid, koninginin G, geoside, pinellic acid and methylpicraquassioside A were reported for the first time. The profiled metabolites were subjected to a 100-ns molecular dynamics simulation following molecular docking. Binding free energy (ΔGbind) calculations revealed obolactone (-48.26 kcal/mol), 1,4-bis(phenylglyoxaloyl)benzene (-45.06 kcal/mol), cyclocanaliculatin (-43.41 kcal/mol), 5-hydroxy-7-methoxy-2-phenylchroman-4-one (-39.18 kcal/mol) and lonchocarpin (-33.78 kcal/mol) as first-time putative leads relative to azithromycin (-32.09 kcal/mol). All lead metabolites also conformed to Lipinski's rule of 5 (Ro5), and their LasR bound complexes were thermodynamically stable and compact given their strong bond interactions. Findings indicate that metabolomic profiling remain key to identifying new compounds from underexplored species. H. annuus lead metabolites and extracts may also play key roles as LasR modulators. Further structural modification of the 5 leads could aid their development into novel, oral therapeutics targeting LasR to mitigate resistant P. aeruginosa infections.
Collapse
Affiliation(s)
- Akshay Raghoonanadan
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban 4000, KwaZulu-Natal, South Africa.
| | - Yamkela Dweba
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban 4000, KwaZulu-Natal, South Africa.
| | - Christiana E Aruwa
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban 4000, KwaZulu-Natal, South Africa.
| | - Saheed Sabiu
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban 4000, KwaZulu-Natal, South Africa.
| |
Collapse
|
4
|
Aggarwal M, Patra A, Awasthi I, George A, Gagneja S, Gupta V, Capalash N, Sharma P. Drug repurposing against antibiotic resistant bacterial pathogens. Eur J Med Chem 2024; 279:116833. [PMID: 39243454 DOI: 10.1016/j.ejmech.2024.116833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/22/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024]
Abstract
The growing prevalence of MDR and XDR bacterial pathogens is posing a critical threat to global health. Traditional antibiotic development paths have encountered significant challenges and are drying up thus necessitating innovative approaches. Drug repurposing, which involves identifying new therapeutic applications for existing drugs, offers a promising alternative to combat resistant pathogens. By leveraging pre-existing safety and efficacy data, drug repurposing accelerates the development of new antimicrobial therapy regimes. This review explores the potential of repurposing existing FDA approved drugs against the ESKAPE and other clinically relevant bacterial pathogens and delves into the identification of suitable drug candidates, their mechanisms of action, and the potential for combination therapies. It also describes clinical trials and patent protection of repurposed drugs, offering perspectives on this evolving realm of therapeutic interventions against drug resistance.
Collapse
Affiliation(s)
- Manya Aggarwal
- Departmen of Microbiology, Panjab University, Chandigarh, India
| | - Anushree Patra
- Departmen of Microbiology, Panjab University, Chandigarh, India
| | - Ishita Awasthi
- Departmen of Microbiology, Panjab University, Chandigarh, India
| | - Annu George
- Departmen of Microbiology, Panjab University, Chandigarh, India
| | - Simran Gagneja
- Departmen of Microbiology, Panjab University, Chandigarh, India
| | - Varsha Gupta
- Department of Microbiology, Government Multi-speciality hospital, Sector 16, Chandigarh, India
| | - Neena Capalash
- Department of Biotechnology, Panjab University, Chandigarh, India
| | - Prince Sharma
- Departmen of Microbiology, Panjab University, Chandigarh, India.
| |
Collapse
|
5
|
Jeong GJ, Khan F, Tabassum N, Kim YM. Motility of Acinetobacter baumannii: regulatory systems and controlling strategies. Appl Microbiol Biotechnol 2024; 108:3. [PMID: 38159120 DOI: 10.1007/s00253-023-12975-6] [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: 10/05/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 01/03/2024]
Abstract
Acinetobacter baumannii is a Gram-negative opportunistic zoonotic pathogenic bacterium that causes nosocomial infections ranging from minor to life-threatening. The clinical importance of this zoonotic pathogen is rapidly increasing due to the development of multiple resistance mechanisms and the synthesis of numerous virulence factors. Although no flagellum-mediated motility exists, it may move through twitching or surface-associated motility. Twitching motility is a coordinated multicellular movement caused by the extension, attachment, and retraction of type IV pili, which are involved in surface adherence and biofilm formation. Surface-associated motility is a kind of movement that does not need appendages and is most likely driven by the release of extra polymeric molecules. This kind of motility is linked to the production of 1,3-diaminopropane, lipooligosaccharide formation, natural competence, and efflux pump proteins. Since A. baumannii's virulence qualities are directly tied to motility, it is possible that its motility may be used as a specialized preventative or therapeutic measure. The current review detailed the signaling mechanism and involvement of various proteins in controlling A. baumannii motility. As a result, we have thoroughly addressed the role of natural and synthetic compounds that impede A. baumannii motility, as well as the underlying action mechanisms. Understanding the regulatory mechanisms behind A. baumannii's motility features will aid in the development of therapeutic drugs to control its infection. KEY POINTS: • Acinetobacter baumannii exhibits multiple resistance mechanisms. • A. baumannii can move owing to twitching and surface-associated motility. • Natural and synthetic compounds can attenuate A. baumannii motility.
Collapse
Affiliation(s)
- Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Fazlurrahman Khan
- Institute of Fisheries Sciences, Pukyong National University, Busan, 48513, Republic of Korea.
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea.
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea.
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Young-Mog Kim
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea.
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea.
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
6
|
Yang S, Meng X, Zhen Y, Baima Q, Wang Y, Jiang X, Xu Z. Strategies and mechanisms targeting Enterococcus faecalis biofilms associated with endodontic infections: a comprehensive review. Front Cell Infect Microbiol 2024; 14:1433313. [PMID: 39091674 PMCID: PMC11291369 DOI: 10.3389/fcimb.2024.1433313] [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: 05/15/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024] Open
Abstract
Enterococcus faecalis is one of the main microorganisms that infects root canals, ranking among the most prevalent microorganisms associated with endodontic treatment failure. Given its pervasive presence in persistent endodontic infections, the successful elimination of Enterococcus faecalis is crucial for effective endodontic treatment and retreatment. Furthermore, Enterococcus faecalis can form biofilms - defense structures that microbes use to fight environmental threats. These biofilms confer resistance against host immune system attacks and antibiotic interventions. Consequently, the presence of biofilms poses a significant challenge in the complete eradication of Enterococcus faecalis and its associated disease. In response, numerous scholars have discovered promising outcomes in addressing Enterococcus faecalis biofilms within root canals and undertaken endeavors to explore more efficacious approaches in combating these biofilms. This study provides a comprehensive review of strategies and mechanisms for the removal of Enterococcus faecalis biofilms.
Collapse
Affiliation(s)
- Shipeng Yang
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Xiuping Meng
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yuqi Zhen
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Quzhen Baima
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yu Wang
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Xinmiao Jiang
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Zhibo Xu
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
7
|
Shein AMS, Hongsing P, Smith OK, Phattharapornjaroen P, Miyanaga K, Cui L, Ishikawa H, Amarasiri M, Monk PN, Kicic A, Chatsuwan T, Pletzer D, Higgins PG, Abe S, Wannigama DL. Current and novel therapies for management of Acinetobacter baumannii-associated pneumonia. Crit Rev Microbiol 2024:1-22. [PMID: 38949254 DOI: 10.1080/1040841x.2024.2369948] [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: 09/25/2023] [Accepted: 06/11/2024] [Indexed: 07/02/2024]
Abstract
Acinetobacter baumannii is a common pathogen associated with hospital-acquired pneumonia showing increased resistance to carbapenem and colistin antibiotics nowadays. Infections with A. baumannii cause high patient fatalities due to their capability to evade current antimicrobial therapies, emphasizing the urgency of developing viable therapeutics to treat A. baumannii-associated pneumonia. In this review, we explore current and novel therapeutic options for overcoming therapeutic failure when dealing with A. baumannii-associated pneumonia. Among them, antibiotic combination therapy administering several drugs simultaneously or alternately, is one promising approach for optimizing therapeutic success. However, it has been associated with inconsistent and inconclusive therapeutic outcomes across different studies. Therefore, it is critical to undertake additional clinical trials to ascertain the clinical effectiveness of different antibiotic combinations. We also discuss the prospective roles of novel antimicrobial therapies including antimicrobial peptides, bacteriophage-based therapy, repurposed drugs, naturally-occurring compounds, nanoparticle-based therapy, anti-virulence strategies, immunotherapy, photodynamic and sonodynamic therapy, for utilizing them as additional alternative therapy while tackling A. baumannii-associated pneumonia. Importantly, these innovative therapies further require pharmacokinetic and pharmacodynamic evaluation for safety, stability, immunogenicity, toxicity, and tolerability before they can be clinically approved as an alternative rescue therapy for A. baumannii-associated pulmonary infections.
Collapse
Affiliation(s)
- Aye Mya Sithu Shein
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Center of Excellence in, Antimicrobial Resistance and Stewardship Research, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Parichart Hongsing
- Mae Fah Luang University Hospital, Chiang Rai, Thailand
- School of Integrative Medicine, Mae Fah Luang University, Chiang Rai, Thailand
| | - O'Rorke Kevin Smith
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Phatthranit Phattharapornjaroen
- Department of Emergency Medicine, Center of Excellence, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Department of Surgery, Sahlgrenska Academy, Institute of Clinical Sciences, Gothenburg University, Gothenburg, Sweden
| | - Kazuhiko Miyanaga
- Division of Bacteriology, School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Longzhu Cui
- Division of Bacteriology, School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Hitoshi Ishikawa
- Yamagata Prefectural University of Health Sciences, Kamiyanagi, Japan
| | - Mohan Amarasiri
- Laboratory of Environmental Hygiene, Department of Health Science, School of Allied Health Sciences, Kitasato University, Kitasato, Sagamihara-Minami, Japan
| | - Peter N Monk
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield Medical School, UK
| | - Anthony Kicic
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
- Centre for Cell Therapy and Regenerative Medicine, Medical School, The University of Western Australia, Nedlands, Western Australia, Australia
- Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, Western Australia, Australia
- School of Population Health, Curtin University, Bentley, Western Australia, Australia
| | - Tanittha Chatsuwan
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Center of Excellence in, Antimicrobial Resistance and Stewardship Research, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Daniel Pletzer
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Paul G Higgins
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
- German Centre for Infection Research, Partner site Bonn-Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Shuichi Abe
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Dhammika Leshan Wannigama
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Center of Excellence in, Antimicrobial Resistance and Stewardship Research, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
- School of Medicine, Faculty of Health and Medical Sciences, The University of Western Australia, Nedlands, Western Australia, Australia
- Biofilms and Antimicrobial Resistance Consortium of ODA receiving countries, The University of Sheffield, Sheffield, UK
- Pathogen Hunter's Research Team, Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
| |
Collapse
|
8
|
Gad AI, El-Ganiny AM, Eissa AG, Noureldin NA, Nazeih SI. Miconazole and phenothiazine hinder the quorum sensing regulated virulence in Pseudomonas aeruginosa. J Antibiot (Tokyo) 2024; 77:454-465. [PMID: 38724627 PMCID: PMC11208154 DOI: 10.1038/s41429-024-00731-5] [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: 08/15/2023] [Revised: 02/20/2024] [Accepted: 04/13/2024] [Indexed: 06/28/2024]
Abstract
Antibiotic resistance is a major health problem worldwide. Pseudomonas aeruginosa is a Gram-negative pathogen with an arsenal of virulence factors and elevated antimicrobial resistance. It is a leading cause of nosocomial infections with high morbidity and mortality. The significant time and effort required to develop new antibiotics can be circumvented using alternative therapeutic strategies, including anti-virulence targets. This study aimed to investigate the anti-virulence activity of the FDA-approved drugs miconazole and phenothiazine against P. aeruginosa. The phenotypic effect of sub-inhibitory concentrations of miconazole and phenothiazine on biofilm, pyocyanin, protease, rhamnolipid and hemolysin activities in PAO1 strain was examined. qRT-PCR was used to assess the effect of drugs on quorum-sensing genes that regulate virulence. Further, the anti-virulence potential of miconazole and phenothiazine was evaluated in silico and in vivo. Miconazole showed significant inhibition of Pseudomonas virulence by reducing biofilm-formation approximately 45-48%, hemolytic-activity by 59%, pyocyanin-production by 47-49%, rhamnolipid-activity by approximately 42-47% and protease activity by 36-40%. While, phenothiazine showed lower anti-virulence activity, it inhibited biofilm (31-35%), pyocyanin (37-39%), protease (32-40%), rhamnolipid (35-40%) and hemolytic activity (47-56%). Similarly, there was significantly reduced expression of RhlR, PqsR, LasI and LasR following treatment with miconazole, but less so with phenothiazine. In-silico analysis revealed that miconazole had higher binding affinity than phenothiazine to LasR, RhlR, and PqsR QS-proteins. Furthermore, there was 100% survival in mice injected with PAO1 treated with miconazole. In conclusion, miconazole and phenothiazine are promising anti-virulence agents for P. aeruginosa.
Collapse
Affiliation(s)
- Amany I Gad
- Microbiology and Immunology Department, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, 11786, Egypt
| | - Amira M El-Ganiny
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| | - Ahmed G Eissa
- Medicinal Chemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Nada A Noureldin
- Medicinal Chemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Shaimaa I Nazeih
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| |
Collapse
|
9
|
Karasiński M, Wnorowska U, Daniluk T, Deptuła P, Łuckiewicz M, Paprocka P, Durnaś B, Skłodowski K, Sawczuk B, Savage PB, Piktel E, Bucki R. Investigating the Effectiveness of Ceragenins against Acinetobacter baumannii to Develop New Antimicrobial and Anti-Adhesive Strategies. Int J Mol Sci 2024; 25:7036. [PMID: 39000144 PMCID: PMC11241064 DOI: 10.3390/ijms25137036] [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: 05/30/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
A growing body of experimental data indicates that ceragenins (CSAs), which mimic the physicochemical properties of the host's cationic antimicrobial peptide, hold promise for the development of a new group of broad-spectrum antimicrobials. Here, using a set of in vivo experiments, we assessed the potential of ceragenins in the eradication of an important etiological agent of nosocomial infections, Acinetobacter baumannii. Assessment of the bactericidal effect of ceragenins CSA-13, CSA-44, and CSA-131 on clinical isolates of A. baumannii (n = 65) and their effectiveness against bacterial cells embedded in the biofilm matrix after biofilm growth on abiotic surfaces showed a strong bactericidal effect of the tested molecules regardless of bacterial growth pattern. AFM assessment of bacterial cell topography, bacterial cell stiffness, and adhesion showed significant membrane breakdown and rheological changes, indicating the ability of ceragenins to target surface structures of A. baumannii cells. In the cell culture of A549 lung epithelial cells, ceragenin CSA-13 had the ability to inhibit bacterial adhesion to host cells, suggesting that it interferes with the mechanism of bacterial cell invasion. These findings highlight the potential of ceragenins as therapeutic agents in the development of antimicrobial strategies against bacterial infections caused by A. baumannii.
Collapse
Affiliation(s)
- Maciej Karasiński
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, 15-222 Białystok, Poland; (M.K.); (U.W.); (T.D.); (K.S.)
| | - Urszula Wnorowska
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, 15-222 Białystok, Poland; (M.K.); (U.W.); (T.D.); (K.S.)
| | - Tamara Daniluk
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, 15-222 Białystok, Poland; (M.K.); (U.W.); (T.D.); (K.S.)
| | - Piotr Deptuła
- Independent Laboratory of Nanomedicine, Medical University of Białystok, 15-222 Białystok, Poland; (P.D.); (M.Ł.); (E.P.)
| | - Milena Łuckiewicz
- Independent Laboratory of Nanomedicine, Medical University of Białystok, 15-222 Białystok, Poland; (P.D.); (M.Ł.); (E.P.)
| | - Paulina Paprocka
- Department of Microbiology and Immunology, Institute of Medical Science, Collegium Medicum, Jan Kochanowski University in Kielce, 25-317 Kielce, Poland; (P.P.); (B.D.)
| | - Bonita Durnaś
- Department of Microbiology and Immunology, Institute of Medical Science, Collegium Medicum, Jan Kochanowski University in Kielce, 25-317 Kielce, Poland; (P.P.); (B.D.)
| | - Karol Skłodowski
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, 15-222 Białystok, Poland; (M.K.); (U.W.); (T.D.); (K.S.)
| | - Beata Sawczuk
- Department of Prosthodontics, Medical University of Bialystok, Sklodowskiej 24a, 15-276 Bialystok, Poland;
| | - Paul B. Savage
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA;
| | - Ewelina Piktel
- Independent Laboratory of Nanomedicine, Medical University of Białystok, 15-222 Białystok, Poland; (P.D.); (M.Ł.); (E.P.)
| | - Robert Bucki
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, 15-222 Białystok, Poland; (M.K.); (U.W.); (T.D.); (K.S.)
| |
Collapse
|
10
|
Arenas S, Rivera N, Méndez Casallas FJ, Galvis B. Assessing Diesel Tolerance of Chromobacterium violaceum: Insights from Growth Kinetics, Substrate Utilization, and Implications for Microbial Adaptation. ACS OMEGA 2024; 9:23741-23752. [PMID: 38854507 PMCID: PMC11154896 DOI: 10.1021/acsomega.4c01698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/30/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024]
Abstract
This study aimed to determine the tolerance of Chromobacterium violaceum ATCC 12472 to diesel. The growth of the strain was evaluated through exposure to various diesel concentrations (1, 2.5, 5, 7.5, and 10% v/v), with continuous monitoring of growth via optical density measurements until the death phase was reached. Employing a logistic model, we analyzed the growth kinetics of C. violaceum and compared them with five other models to comprehend substrate utilization dynamics. Our results indicate that optimal bacterial growth occurred at 2.5% (v/v) or 18,125 mg/L diesel, while both higher and lower concentrations manifested inhibitory and increasingly stressful effects. The Aiba model emerged as the most fitting representation of substrate utilization by C. violaceum. In addition, our findings underscore the remarkable diesel tolerance of C. violaceum ATCC 12472, despite the inherently stressful nature of the medium. This study contributes to the understanding of microbial responses to environmental stressors and highlights the pivotal role of the substrate concentration in influencing microbial growth. These insights have implications for bioremediation strategies and enhance our understanding of bacterial ecological resilience in the presence of hydrocarbon pollutants.
Collapse
Affiliation(s)
- Sebastián Arenas
- Programa
de Ingeniería ambiental y Sanitaria, Universidad de La Salle, Bogotá 110231, Colombia
| | - Nathaly Rivera
- Programa
de Ingeniería ambiental y Sanitaria, Universidad de La Salle, Bogotá 110231, Colombia
| | | | - Boris Galvis
- Escuela
de Ingeniería de los Recursos Naturales y del Ambiente—EIDENAR, Universidad del Valle, Cali 760042, Colombia
| |
Collapse
|
11
|
Saad HM, Elekhnawy E, Shaldam MA, Alqahtani MJ, Altwaijry N, Attallah NGM, Hussein IA, Ibrahim HA, Negm WA, Salem EA. Rosuvastatin and diosmetin inhibited the HSP70/TLR4 /NF-κB p65/NLRP3 signaling pathways and switched macrophage to M2 phenotype in a rat model of acute kidney injury induced by cisplatin. Biomed Pharmacother 2024; 171:116151. [PMID: 38262148 DOI: 10.1016/j.biopha.2024.116151] [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: 10/14/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/25/2024] Open
Abstract
Numerous efforts to manage acute kidney injury (AKI) were unsuccessful because its pathophysiology is still poorly understood. Thus, our research hotspot was to explore the possible renoprotective effects of rosuvastatin (Ros) and diosmetin (D) on macrophage polarization and the role of HSP70/TLR4/MyD88/NF-κB p65/NLRP3/STAT3 signaling in cis-induced AKI and study the activity of D against uropathogenic bacteria. Fifty-four albino male rats were randomized into 9 groups equally: Control, Ros, D20, D40, untreated Cis, and Cis groups cotreated with Ros, D20, D40 and Ros+D40 for 10 days. Our results indicated that Ros and D, in a dose-dependent manner, markedly restored body weight, systolic blood pressure, and renal histological architecture besides significantly upregulated SOD levels, expression of anti-inflammatory CD163 macrophages, arginase1levels, IL-10 levels,STAT3 and PCNA immunoreactivity. Also, they significantly downregulated renal index, serum urea, serum creatinine, serum cystatin c, inflammatory biomarkers (C reactive protein, IL1β & TNF-α), MDA levels, HSP70/TLR4/MyD88/NF-κB p65/NLRP3 expressions, proinflammatory CD68 macrophages and caspase-3 immunoreactivity, resulting in a reversal of cis-induced renal damage. These findings were further confirmed by molecular docking that showed the binding affinity of Ros and D towards TLR4 and NLRP3. Furthermore, D had antibacterial action with a minimum inhibitory concentration ranging from 128 to 256 µg/mL and caused a delay in the growth of the tested isolates, and negatively affected the membrane integrity. In conclusion, Ros and D had antioxidant, anti-inflammatory and antiapoptotic properties and switched macrophage from proinflammatory CD68 to anti-inflammatory CD163. Additionally, the targeting of HSP70/TLR4/MyD88/NF-κB p65/NLRP3/STAT3 signals are effective therapeutic strategy in AKI.
Collapse
Affiliation(s)
- Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matrouh, Egypt.
| | - Engy Elekhnawy
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafr El-Sheikh 33516, Egypt
| | - Moneerah J Alqahtani
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia.
| | - Najla Altwaijry
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia.
| | | | - Ismail A Hussein
- Department of Pharmacognosy and Medicinal Plants, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt
| | - Hanaa A Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tanta, Egypt
| | - Walaa A Negm
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Esraa A Salem
- Department of Medical Physiology, Faculty of Medicine, Menoufia University, Shebeen ElKom 32511, Egypt
| |
Collapse
|
12
|
Vadakkan K, Ngangbam AK, Sathishkumar K, Rumjit NP, Cheruvathur MK. A review of chemical signaling pathways in the quorum sensing circuit of Pseudomonas aeruginosa. Int J Biol Macromol 2024; 254:127861. [PMID: 37939761 DOI: 10.1016/j.ijbiomac.2023.127861] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/26/2023] [Accepted: 11/01/2023] [Indexed: 11/10/2023]
Abstract
Pseudomonas aeruginosa, an increasingly common competitive and biofilm organism in healthcare infection with sophisticated, interlinked and hierarchic quorum systems (Las, Rhl, PQS, and IQS), creates the greatest threats to the medical industry and has rendered prevailing chemotherapy medications ineffective. The rise of multidrug resistance has evolved into a concerning and potentially fatal occurrence for human life. P. aeruginosa biofilm development is assisted by exopolysaccharides, extracellular DNA, proteins, macromolecules, cellular signaling and interaction. Quorum sensing is a communication process between cells that involves autonomous inducers and regulators. Quorum-induced infectious agent biofilms and the synthesis of virulence factors have increased disease transmission, medication resistance, infection episodes, hospitalizations and mortality. Hence, quorum sensing may be a potential therapeutical target for bacterial illness, and developing quorum inhibitors as an anti-virulent tool could be a promising treatment strategy for existing antibiotics. Quorum quenching is a prevalent technique for treating infections caused by microbes because it diminishes microbial pathogenesis and increases microbe biofilm sensitivity to antibiotics, making it a potential candidate for drug development. This paper examines P. aeruginosa quorum sensing, the hierarchy of quorum sensing mechanism, quorum sensing inhibition and quorum sensing inhibitory agents as a drug development strategy to supplement traditional antibiotic strategies.
Collapse
Affiliation(s)
- Kayeen Vadakkan
- Department of Biology, St. Mary's College, Thrissur, Kerala 680020, India; Manipur International University, Imphal, Manipur 795140, India.
| | | | - Kuppusamy Sathishkumar
- Rhizosphere Biology Laboratory, Department of Microbiology, Bharathidasan University, Tiruchirappalli, Tamil Nadu 620024, India; Department of Computational Biology, Institute of Bioinformatics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Thandalam, Chennai 602 105, Tamil Nadu, India
| | | | | |
Collapse
|
13
|
Morgaan HA, Omar HMG, Zakaria AS, Mohamed NM. Repurposing carvacrol, cinnamaldehyde, and eugenol as potential anti-quorum sensing agents against uropathogenic Escherichia coli isolates in Alexandria, Egypt. BMC Microbiol 2023; 23:300. [PMID: 37872476 PMCID: PMC10591344 DOI: 10.1186/s12866-023-03055-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 10/11/2023] [Indexed: 10/25/2023] Open
Abstract
BACKGROUND Urinary tract infections represent one of the most frequent hospital and community-acquired infections with uropathogenic Escherichia coli (UPEC) being the main causative agent. The global increase in the emergence of multidrug-resistant (MDR) UPEC necessitates exploring novel approaches. Repurposing natural products as anti-quorum sensing (QS) agents to impede bacterial virulence is gaining momentum nowadays. Hence, this study investigates the anti-QS potentials of carvacrol, cinnamaldehyde, and eugenol against E. coli isolated from urine cultures of Egyptian patients. RESULTS Antibiotic susceptibility testing was performed for 67 E. coli isolates and 94% of the isolates showed MDR phenotype. The usp gene was detected using PCR and accordingly, 45% of the isolates were categorized as UPEC. Phytochemicals, at their sub-inhibitory concentrations, inhibited the swimming and twitching motilities of UPEC isolates, with eugenol showing the highest inhibitory effect. The agents hindered the biofilm-forming ability of the tested isolates, at two temperature sets, 37 and 30 °C, where eugenol succeeded in significantly inhibiting the biofilm formation by > 50% at both investigated temperatures, as compared with untreated controls. The phytochemicals were shown to downregulate the expression of the QS gene (luxS) and critical genes related to motility, asserting their anti-QS potential. Further, the combinatory activity of the phytoproducts with five antibiotics was assessed by checkerboard assay. The addition of the phytoproducts significantly reduced the minimum inhibitory concentrations of the antibiotics and generated several synergistic or partially synergistic combinations, some of which have not been previously explored. CONCLUSIONS Overall, carvacrol, cinnamaldehyde, and eugenol could be repurposed as potential anti-QS agents, which preferentially reduce the QS-based communication and attenuate the cascades of gene expression, thus decreasing the production of virulence factors in UPEC, and eventually, subsiding their pathogenicity. Furthermore, the synergistic combinations of these agents with antibiotics might provide a new perspective to circumvent the side effects brought about by high antibiotic doses, thereby paving the way for overcoming antibiotic resistance.
Collapse
Affiliation(s)
- Hadeer A Morgaan
- Microbiology and Immunology Department, Faculty of Pharmacy, Alexandria University, El-Khartoom Square, Azarita, Alexandria, Egypt
| | - Hoda M G Omar
- Microbiology and Immunology Department, Faculty of Pharmacy, Alexandria University, El-Khartoom Square, Azarita, Alexandria, Egypt
| | - Azza S Zakaria
- Microbiology and Immunology Department, Faculty of Pharmacy, Alexandria University, El-Khartoom Square, Azarita, Alexandria, Egypt
| | - Nelly M Mohamed
- Microbiology and Immunology Department, Faculty of Pharmacy, Alexandria University, El-Khartoom Square, Azarita, Alexandria, Egypt.
| |
Collapse
|
14
|
Murali M, Ahmed F, Gowtham HG, Aribisala JO, Abdulsalam RA, Shati AA, Alfaifi MY, Sayyed RZ, Sabiu S, Amruthesh KN. Exploration of CviR-mediated quorum sensing inhibitors from Cladosporium spp. against Chromobacterium violaceum through computational studies. Sci Rep 2023; 13:15505. [PMID: 37726386 PMCID: PMC10509224 DOI: 10.1038/s41598-023-42833-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 09/15/2023] [Indexed: 09/21/2023] Open
Abstract
An opportunistic human pathogenic bacterium, Chromobacterium violaceum resists the potency of most antibiotics by exploiting the quorum sensing system within their community to control virulence factor expression. Therefore, blocking the quorum sensing mechanism could help to treat several infectious caused by this organism. The quorum sensing receptor (CviR) of C. violaceum was used as a model target in the current investigation to identify potentially novel quorum sensing inhibitors from Cladosporium spp. through in silico computational approaches. The molecular docking results confirmed the anti-quorum sensing potential of bioactive compounds from Cladosporium spp. through binding to CviR with varying docking scores between - 5.2 and - 9.5 kcal/mol. Relative to the positive control [Azithromycin (- 7.4 kcal/mol)], the top six metabolites of Cladosporium spp. had higher docking scores and were generally greater than - 8.5 kcal/mol. The thermodynamic stability and binding affinity refinement of top-ranked CviR inhibitors were further studied through a 160 ns molecular dynamic (MD) simulation. The Post-MD simulation analysis confirmed the top-ranked compounds' affinity, stability, and biomolecular interactions with CviR at 50 ns, 100 ns, and 160 ns with Coniochaetone K of the Cladosporium spp. having the highest binding free energy (- 30.87 kcal/mol) and best interactions (two consistent hydrogen bond contact) following the 160 ns simulation. The predicted pharmacokinetics properties of top selected compounds point to their drug likeliness, potentiating their chance as a possible drug candidate. Overall, the top-ranked compounds from Cladosporium spp., especially Coniochaetone K, could be identified as potential C. violaceum CviR inhibitors. The development of these compounds as broad-spectrum antibacterial medicines is thus possible in the future following the completion of further preclinical and clinical research.
Collapse
Affiliation(s)
- Mahadevamurthy Murali
- Applied Plant Pathology Laboratory, Department of Studies in Botany, University of Mysore, Manasagangotri, Mysore, 570006, India
| | - Faiyaz Ahmed
- Department of Clinical Nutrition, College of Applied Health Sciences in Ar Rass, Qassim University, 51452, Buraydah, Saudi Arabia
| | | | - Jamiu Olaseni Aribisala
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban, South Africa
| | - Rukayat Abiola Abdulsalam
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban, South Africa
| | - Ali A Shati
- Faculty of Science, Biology Department, King Khalid University, 9004, Abha, Saudi Arabia
| | - Mohammad Y Alfaifi
- Faculty of Science, Biology Department, King Khalid University, 9004, Abha, Saudi Arabia
| | - R Z Sayyed
- Department of Microbiology, PSGVP Mandal's S I Patil Arts, G B Patel Science and STKV Sangh Commerce College, Shahada, 425409, India.
| | - Saheed Sabiu
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban, South Africa.
| | - Kestur Nagaraj Amruthesh
- Applied Plant Pathology Laboratory, Department of Studies in Botany, University of Mysore, Manasagangotri, Mysore, 570006, India.
| |
Collapse
|
15
|
Mosallam FM, Abbas HA, Shaker GH, Gomaa SE. Alleviating the virulence of Pseudomonas aeruginosa and Staphylococcus aureus by ascorbic acid nanoemulsion. Res Microbiol 2023; 174:104084. [PMID: 37247797 DOI: 10.1016/j.resmic.2023.104084] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 05/31/2023]
Abstract
The high incidence of persistent multidrug resistant bacterial infections is a worldwide public health burden. Alternative strategies are required to deal with such issue including the use of drugs with anti-virulence activity. The application of nanotechnology to develop advanced Nano-materials that target quorum sensing regulated virulence factors is an attractive approach. Synthesis of ascorbic acid Nano-emulsion (ASC-NEs) and assessment of its activity in vitro against the virulence factors and its protective ability against pathogenesis as well as the effect against expression of quorum sensing genes of Pseudomonas aeruginosa and Staphylococcus aureus isolates. Ascorbic acid Nano-emulsion was characterized by DLS Zetasizer Technique, Zeta potential; Transmission Electron Microscopy (TEM) and Fourier transform infrared spectroscopy (FT-IR). The antibacterial activity of ASC-NEs was tested by the broth microdilution method and the activity of their sub-MIC against the expression of quorum sensing controlled virulence was investigated using phenotypic experiments and RT-PCR. The protective activity of ASC-NEs against P. aeruginosa as well as S. aureus pathogenesis was tested in vivo. Phenotypically, ASC-NEs had strong virulence inhibitory activity against the tested bacteria. The RT-PCR experiment showed that it exhibited significant QS inhibitory activity. The in vivo results showed that ASC-NEs protected against staphylococcal infection, however, it failed to protect mice against Pseudomonal infection. These results suggest the promising use of nanoformulations against virulence factors in multidrug resistant P. aeruginosa and S. aureus. However, further studies are required concerning the potential toxicity, clearance and phamacokinetics of the nanoformulations.
Collapse
Affiliation(s)
- Farag M Mosallam
- Drug Microbiology Lab., Drug Radiation Research Department, Biotechnology Division, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt.
| | - Hisham A Abbas
- Department of Microbiology and Immunology-Faculty of Pharmacy-Zagazig University, Zagazig, Egypt
| | - Ghada H Shaker
- Department of Microbiology and Immunology-Faculty of Pharmacy-Zagazig University, Zagazig, Egypt
| | - Salwa E Gomaa
- Department of Microbiology and Immunology-Faculty of Pharmacy-Zagazig University, Zagazig, Egypt
| |
Collapse
|
16
|
Venkateswaran P, Vasudevan S, David H, Shaktivel A, Shanmugam K, Neelakantan P, Solomon AP. Revisiting ESKAPE Pathogens: virulence, resistance, and combating strategies focusing on quorum sensing. Front Cell Infect Microbiol 2023; 13:1159798. [PMID: 37457962 PMCID: PMC10339816 DOI: 10.3389/fcimb.2023.1159798] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 06/08/2023] [Indexed: 07/18/2023] Open
Abstract
The human-bacterial association is long-known and well-established in terms of both augmentations of human health and attenuation. However, the growing incidents of nosocomial infections caused by the ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter sp.) call for a much deeper understanding of these organisms. Adopting a holistic approach that includes the science of infection and the recent advancements in preventing and treating infections is imperative in designing novel intervention strategies against ESKAPE pathogens. In this regard, this review captures the ingenious strategies commissioned by these master players, which are teamed up against the defenses of the human team, that are equally, if not more, versatile and potent through an analogy. We have taken a basketball match as our analogy, dividing the human and bacterial species into two teams playing with the ball of health. Through this analogy, we make the concept of infectious biology more accessible.
Collapse
Affiliation(s)
- Parvathy Venkateswaran
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Sahana Vasudevan
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Helma David
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Adityan Shaktivel
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Karthik Shanmugam
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Prasanna Neelakantan
- Division of Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| |
Collapse
|
17
|
Ressler AJ, Frate M, Hontoria A, Ream A, Timms E, Li H, Stettler LD, Bollinger A, Poor JE, Parra MA, Ma H, Seeram NP, Meschwitz SM, Henry GE. Synthesis, anti-ferroptosis, anti-quorum sensing, antibacterial and DNA interaction studies of chromene-hydrazone derivatives. Bioorg Med Chem 2023; 90:117369. [PMID: 37320993 DOI: 10.1016/j.bmc.2023.117369] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/15/2023] [Accepted: 06/05/2023] [Indexed: 06/17/2023]
Abstract
Nineteen chromene-hydrazone derivatives containing a variety of structural modifications on the hydrazone moiety were synthesized. Structure-activity correlations were investigated to determine the influence of structural variations on anti-ferroptosis, anti-quorum sensing, antibacterial, DNA cleavage and DNA binding properties. Ferroptosis inhibitory activity was determined by measuring the ability of the derivatives to reverse erastin-induced ferroptosis. Several of the derivatives were more effective than fisetin at inhibiting ferroptosis, with the thiosemicarbazone derivative being the most effective. Quorum sensing inhibition was evaluated using Vibrio harveyi, and both V. harveyi and Staphylococcus aureus were used to determine antibacterial activity. The semicarbazone and benzensulfonyl hydrazone derivatives showed moderate quorum sensing inhibition with IC50 values of 27 μM and 22 μM, respectively, while a few aryl hydrazone and pyridyl hydrazone derivatives showed bacterial growth inhibition, with MIC values ranging from 3.9 to 125 μM. In addition, the interaction of the hydrazone derivatives with DNA was investigated by gel electrophoresis, UV-Vis spectroscopy and molecular docking. All of the derivatives cleaved plasmid DNA and showed favorable interaction with B-DNA through minor groove binding. Overall, this work highlights a broad range of pharmacological applications for chromene-hydrazone derivatives.
Collapse
Affiliation(s)
- Andrew J Ressler
- Department of Chemistry, Susquehanna University, 514 University Avenue, Selinsgrove, PA 17870, USA
| | - Marissa Frate
- Department of Chemistry, Salve Regina University, Newport, Rhode Island 02840, USA
| | - Ana Hontoria
- Department of Chemistry, Salve Regina University, Newport, Rhode Island 02840, USA
| | - Anna Ream
- Department of Chemistry, Susquehanna University, 514 University Avenue, Selinsgrove, PA 17870, USA
| | - Eliza Timms
- Department of Chemistry, Salve Regina University, Newport, Rhode Island 02840, USA
| | - Huifang Li
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Lauren D Stettler
- Department of Chemistry, Susquehanna University, 514 University Avenue, Selinsgrove, PA 17870, USA
| | - Ashton Bollinger
- Department of Chemistry, Susquehanna University, 514 University Avenue, Selinsgrove, PA 17870, USA
| | - Jenna E Poor
- Department of Chemistry, Susquehanna University, 514 University Avenue, Selinsgrove, PA 17870, USA
| | - Michael A Parra
- Department of Chemistry, Susquehanna University, 514 University Avenue, Selinsgrove, PA 17870, USA
| | - Hang Ma
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Navindra P Seeram
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Susan M Meschwitz
- Department of Chemistry, Salve Regina University, Newport, Rhode Island 02840, USA.
| | - Geneive E Henry
- Department of Chemistry, Susquehanna University, 514 University Avenue, Selinsgrove, PA 17870, USA.
| |
Collapse
|
18
|
Akshaya BS, Premraj K, Iswarya C, Muthusamy S, Ibrahim HIM, Khalil HE, Gunasekaran V, Vickram S, Senthil Kumar V, Palanisamy S, Thirugnanasambantham K. Cinnamaldehyde inhibits Enterococcus faecalis biofilm formation and promotes clearance of its colonization by modulation of phagocytes in vitro. Microb Pathog 2023:106157. [PMID: 37268049 DOI: 10.1016/j.micpath.2023.106157] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 06/04/2023]
Abstract
The nosocomial pathogen, Enterococcus faecalis plays a crucial role in the pathogenesis of variety of infections including endocarditis, urinary tract, and recurrent root canal infections. Primary virulence factors of E. faecalis such as biofilm formation, gelatinase production and suppression of host innate immune response can severely harm host tissue. Thus, novel treatments are needed to prevent E. faecalis biofilm development and pathogenicity due to the worrisome rise in enterococcal resistance to antibiotics. The primary phytochemical in cinnamon essential oils, cinnamaldehyde, has shown promising efficacy against a variety of infections. Here, we looked into how cinnamaldehyde affected the growth of biofilms, the activity of the enzyme gelatinase, and gene expression in E. faecalis. In addition, we looked at the influence of cinnamaldehyde on RAW264.7 macrophages' interaction with biofilm and planktonic E. faecalis in terms of intracellular bacterial clearance, NO generation, and macrophage migration in vitro. According to our research, cinnamaldehyde attenuated the biofilm formation potential of planktonic E. faecalis and gelatinase activity of the biofilm at non-lethal concentrations. The expression of the quorum sensing fsr locus and its downstream gene gelE in biofilms were also found to be significantly downregulated by cinnamaldehyde. Results also demonstrated that cinnamaldehyde treatment increased NO production, intracellular bacterial clearance, and migration of RAW264.7 macrophages in presence of both biofilm and planktonic E. faecalis. Overall these results suggest that cinnamaldehyde has the ability to inhibit E. faecalis biofilm formation and modulate host innate immune response for better clearance of bacterial colonization.
Collapse
Affiliation(s)
- Balasubramanian Sennammal Akshaya
- Pondicherry Centre for Biological Science and Educational Trust, Sundararaja Nagar, Pondicherry, 605004, India; Department of Biotechnology, Vels Institute of Science, Technology and Advanced Studies, Chennai, Tamil Nadu, India
| | - Kumar Premraj
- Pondicherry Centre for Biological Science and Educational Trust, Sundararaja Nagar, Pondicherry, 605004, India
| | - Christian Iswarya
- Pondicherry Centre for Biological Science and Educational Trust, Sundararaja Nagar, Pondicherry, 605004, India
| | - Suganthi Muthusamy
- Department of Biotechnology, Vels Institute of Science, Technology and Advanced Studies, Chennai, Tamil Nadu, India
| | - Hairul-Islam Mohamed Ibrahim
- Pondicherry Centre for Biological Science and Educational Trust, Sundararaja Nagar, Pondicherry, 605004, India; Biological Science College of Science, King Faisal University, Al Ahsa, 31982, Saudi Arabia
| | - Hany Ezzat Khalil
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, 31982, Saudi Arabia; Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Vaishnavi Gunasekaran
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Sundaram Vickram
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, India
| | - Venugopal Senthil Kumar
- Pondicherry Centre for Biological Science and Educational Trust, Sundararaja Nagar, Pondicherry, 605004, India; Tamil Nadu State Council for Science and Technology, DOTE Campus, Chennai, 600025, Tamil Nadu, India
| | - Senthilkumar Palanisamy
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India.
| | - Krishnaraj Thirugnanasambantham
- Pondicherry Centre for Biological Science and Educational Trust, Sundararaja Nagar, Pondicherry, 605004, India; Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, India.
| |
Collapse
|
19
|
Ruggieri F, Compagne N, Antraygues K, Eveque M, Flipo M, Willand N. Antibiotics with novel mode of action as new weapons to fight antimicrobial resistance. Eur J Med Chem 2023; 256:115413. [PMID: 37150058 DOI: 10.1016/j.ejmech.2023.115413] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/09/2023] [Accepted: 04/22/2023] [Indexed: 05/09/2023]
Abstract
Antimicrobial resistance (AMR) is a major public health issue, causing 5 million deaths per year. Without any action plan, AMR will be in a near future the leading cause of death ahead of cancer. AMR comes from the ability of bacteria to rapidly develop and share resistance mechanisms towards current antibiotics, rendering them less effective. To circumvent this issue and avoid the phenomenon of cross-resistance, new antibiotics acting on novel targets or with new modes of action are required. Today, the pipeline of potential new treatments with these characteristics includes promising compounds such as gepotidacin, zoliflodacin, ibezapolstat, MGB-BP-3, CRS-3123, afabicin and TXA-709, which are currently in clinical trials, and lefamulin, which has been recently approved by FDA and EMA. In this review, we report the chemical synthesis, mode of action, structure-activity relationships, in vitro and in vivo activities as well as clinical data of these eight small molecules listed above.
Collapse
Affiliation(s)
- Francesca Ruggieri
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Nina Compagne
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Kevin Antraygues
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Maxime Eveque
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Marion Flipo
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Nicolas Willand
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, F-59000, Lille, France.
| |
Collapse
|
20
|
Kilic T, Bali EB. Biofilm control strategies in the light of biofilm-forming microorganisms. World J Microbiol Biotechnol 2023; 39:131. [PMID: 36959476 DOI: 10.1007/s11274-023-03584-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/15/2023] [Indexed: 03/25/2023]
Abstract
Biofilm is a complex consortium of microorganisms attached to biotic or abiotic surfaces and live in self-produced or acquired extracellular polymeric substances (EPSs). EPSs are mainly formed by lipids, polysaccharides, proteins, and extracellular DNAs. The adherence to the surface of microbial communities is seen in food, medical, dental, industrial, and environmental fields. Biofilm development in food processing areas challenges food hygiene, and human health. In addition, bacterial attachment and biofilm formation on medical implants inside human tissue can cause multiple critical chronic infections. More than 30 years of international research on the mechanisms of biofilm formation have been underway to address concerns about bacterial biofilm infections. Antibiofilm strategies contain cold atmospheric plasma, nanotechnological, phage-based, antimicrobial peptides, and quorum sensing inhibition. In the last years, the studies on environmentally-friendly techniques such as essential oils and bacteriophages have been intensified to reduce microbial growth. However, the mechanisms of the biofilm matrix formation are still unclear. This review aims to discuss the latest antibiofilm therapeutic strategies against biofilm-forming bacteria.
Collapse
Affiliation(s)
- Tugba Kilic
- Department of Medical Services and Techniques, Program of Medical Laboratory Techniques, Vocational School of Health Services, Gazi University, Ankara, 06830, Turkey.
| | - Elif Burcu Bali
- Department of Medical Services and Techniques, Program of Medical Laboratory Techniques, Vocational School of Health Services, Gazi University, Ankara, 06830, Turkey
| |
Collapse
|
21
|
Genome Analysis of Pseudomonas aeruginosa Strains from Chronically Infected Patients with High Levels of Persister Formation. Pathogens 2023; 12:pathogens12030426. [PMID: 36986348 PMCID: PMC10051920 DOI: 10.3390/pathogens12030426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
The appearance of persister cells with low metabolic rates are key factors leading to antibiotic treatment failure. Such persisters are multidrug tolerant and play a key role in the recalcitrance of biofilm-based chronic infections. Here, we present the genomic analyses of three distinct Pseudomonas aeruginosa Egyptian persister-isolates recovered from chronic human infections. To calculate the persister frequencies, viable counts were determined before and after treatment with levofloxacin. The susceptibilities of isolates to different antibiotics were determined using the agar-dilution method. To determine their recalcitrance, the levofloxacin persisters were further challenged with lethal concentrations of meropenem, tobramycin, or colistin. Furthermore, the biofilm formation of the persister strains was estimated phenotypically, and they were reported to be strong biofilm-forming strains. The genotypic characterization of the persisters was performed using whole genome sequencing (WGS) followed by phylogenetic analysis and resistome profiling. Interestingly, out of the thirty-eight clinical isolates, three isolates (8%) demonstrated a persister phenotype. The three levofloxacin-persister isolates were tested for their susceptibility to selected antibiotics; all of the tested isolates were multidrug resistant (MDR). Additionally, the P. aeruginosa persisters were capable of surviving over 24 h and were not eradicated after exposure to 100X-MIC of levofloxacin. WGS for the three persisters revealed a smaller genome size compared to PAO1-genome. Resistome profiling indicated the presence of a broad collection of antibiotic-resistance genes, including genes encoding for antibiotic-modifying enzymes and efflux pump. Phylogenetic analysis indicated that the persister isolates belong to a distinct clade rather than the deposited P. aeruginosa strains in the GenBank. Conclusively, the persister isolates in our study are MDR and form a highly strong biofilm. WGS revealed a smaller genome that belongs to a distinct clade.
Collapse
|
22
|
Gao J, Hu X, Xu C, Guo M, Li S, Yang F, Pan X, Zhou F, Jin Y, Bai F, Cheng Z, Wu Z, Chen S, Huang X, Wu W. Neutrophil-mediated delivery of the combination of colistin and azithromycin for the treatment of bacterial infection. iScience 2022; 25:105035. [PMID: 36117992 PMCID: PMC9474925 DOI: 10.1016/j.isci.2022.105035] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/29/2022] [Accepted: 08/24/2022] [Indexed: 11/21/2022] Open
Abstract
Novel treatment strategies are in urgent need to deal with the rapid development of antibiotic-resistant superbugs. Combination therapies and targeted drug delivery have been exploited to promote treatment efficacies. In this study, we loaded neutrophils with azithromycin and colistin to combine the advantages of antibiotic combinations, targeted delivery, and immunomodulatory effect of azithromycin to treat infections caused by Gram-negative pathogens. Delivery of colistin into neutrophils was mediated by fusogenic liposome, while azithromycin was directly taken up by neutrophils. Neutrophils loaded with the drugs maintained the abilitity to generate reactive oxygen species and migrate. In vitro assays demonstrated enhanced bactericidal activity against multidrug-resistant pathogens and reduced inflammatory cytokine production by the drug-loaded neutrophils. A single intravenous administration of the drug-loaded neutrophils effectively protected mice from Pseudomonas aeruginosa infection in an acute pneumonia model. This study provides a potential effective therapeutic approach for the treatment of bacterial infections. Neutrophils are loaded with colistin and azithromycin in vitro The loaded drugs enhance the bactericidal effect and reduce the inflammatory response Drug-loaded neutrophils conferred effective protection against bacterial infection
Collapse
Affiliation(s)
- Jiacong Gao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xueyan Hu
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China.,Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Congjuan Xu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Mingming Guo
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shouyi Li
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Fan Yang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xiaolei Pan
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Fangyu Zhou
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yongxin Jin
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Fang Bai
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Zhihui Cheng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Zhenzhou Wu
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shuiping Chen
- Department of Laboratory Medicine, 5th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xinglu Huang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China.,Joint Laboratory of Nanozymes, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Weihui Wu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
23
|
Bilosomes as Nanoplatform for Oral Delivery and Modulated In Vivo Antimicrobial Activity of Lycopene. Pharmaceuticals (Basel) 2022; 15:ph15091043. [PMID: 36145264 PMCID: PMC9505130 DOI: 10.3390/ph15091043] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 01/19/2023] Open
Abstract
Owing to the disseminating resistance among pathogenic bacteria, especially Klebsiella pneumoniae, there is a high need for alternate compounds with antibacterial activity. Herein, lycopene was isolated from Lycopersicon esculentum L. Molecular docking approach was employed to explore lycopene binding affinity to selected vital proteins of K. pneumoniae with the binding mechanisms being investigated. This proposed a promising antibacterial activity of lycopene. However, the pharmacological use of lycopene is hampered by its poor solubility and limited oral bioavailability. Accordingly, bilosomes were fabricated for oral lycopene delivery. The computed entrapment efficiency, mean vesicular size, and zeta potential values for the optimized formulation were 93.2 ± 0.6%, 485.8 ± 35.3 nm, and −38.3 ± 4, respectively. In vitro drug release studies revealed controlled lycopene release from constructed bilosomes, with the drug liberation being based on the Higuchi kinetics model. Transmission electron microscopic evaluation of bilosomes revealed spherical nanovesicles free from aggregates. Moreover, the in vitro and in vivo antibacterial activity of lycopene and its constructed formulations against multidrug-resistant K. pneumoniae isolates were explored. The optimized bilosomes exhibited the lowest minimum inhibitory concentrations ranging from 8 to 32 µg/mL. In addition, scanning electron microscopy revealed remarkable deformation and lysis of the bilosomes-treated bacterial cells. Regarding in vivo investigation, a lung infection model in mice was employed. The tested bilosomes reduced the inflammation and congestion in the treated mice’s lung tissues, resulting in normal-sized bronchioles and alveoli with very few congested vessels. In addition, it resulted in a significant reduction in pulmonary fibrosis. In conclusion, this study investigated the potential activity of the naturally isolated lycopene in controlling infections triggered by multidrug-resistant K. pneumoniae isolates. Furthermore, it introduced bilosomes as a promising biocompatible nanocarrier for modulation of oral lycopene delivery and in vivo antimicrobial activity.
Collapse
|
24
|
Rather MA, Saha D, Bhuyan S, Jha AN, Mandal M. Quorum Quenching: A Drug Discovery Approach Against Pseudomonas aeruginosa. Microbiol Res 2022; 264:127173. [PMID: 36037563 DOI: 10.1016/j.micres.2022.127173] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/06/2022] [Accepted: 08/14/2022] [Indexed: 11/19/2022]
Abstract
Pseudomonas aeruginosa, a ubiquitous opportunistic and nosocomial biofilm-forming pathogen with complex, interconnected and hierarchical nature of QS systems (Las, Rhl, PQS, and IQS), is posing the biggest challenge to the healthcare sector and have made current chemotherapies incapable. Conventional antibiotics designed to intercept the biochemical or physiological processes precisely of planktonic microorganisms exert extreme selective pressure and develop resistance against them thereby emphasizing the development of alternative therapeutic approaches. Additionally, quorum sensing induced pathogenic microbial biofilms and production of virulence factors have intensified the pathogenicity, drug resistance, recurrence of infections, hospital visits, morbidity, and mortality many-folds. In this regard, QS could be a potential druggable target and the discovery of QS inhibiting agents as an anti-virulent measure could serve as an alternative therapeutic approach to conventional antibiotics. Quorum quenching (QQ) is a preferred strategy to combat microbial infections since it attenuates the pathogenicity of microbes and enhances the microbial biofilm susceptibility to antibiotics, thus qualifying as a suitable target for drug discovery. This review discusses the QS-induced pathogenicity of P. aeruginosa, the hierarchical QS systems, and QS inhibition as a drug discovery approach to complement classical antibiotic strategy.
Collapse
Affiliation(s)
- Muzamil Ahmad Rather
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur 784028, Assam, India
| | - Debanjan Saha
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur 784028, Assam, India
| | - Shuvam Bhuyan
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur 784028, Assam, India
| | - Anupam Nath Jha
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur 784028, Assam, India
| | - Manabendra Mandal
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur 784028, Assam, India.
| |
Collapse
|
25
|
Abdel-Karim SAAM, El-Ganiny AMA, El-Sayed MA, Abbas HAA. Promising FDA-approved drugs with efflux pump inhibitory activities against clinical isolates of Staphylococcus aureus. PLoS One 2022; 17:e0272417. [PMID: 35905077 PMCID: PMC9337675 DOI: 10.1371/journal.pone.0272417] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/19/2022] [Indexed: 12/04/2022] Open
Abstract
Background and objectives Staphylococcus aureus is an opportunistic pathogen that causes wide range of nosocomial and community-acquired infections which have spread worldwide leading to an urgent need for developing effective anti-staphylococcal agents. Efflux is an important resistance mechanism that bacteria used to fight the antimicrobial action. This study aimed to investigate the efflux mechanism in S. aureus and assess diclofenac, domperidone, glyceryl trinitrate and metformin as potential efflux pump inhibitors that can be used in combination with antibiotics for treating topical infections caused by S. aureus. Materials and methods Efflux was detected qualitatively by the ethidium bromide Cart-Wheel method followed by investigating the presence of efflux genes by polymerase chain reaction. Twenty-six isolates were selected for further investigation of efflux by Cart-Wheel method in absence and presence of tested compounds followed by quantitative efflux assay. Furthermore, antibiotics minimum inhibitory concentrations in absence and presence of tested compounds were determined. The effects of tested drugs on expression levels of efflux genes norA, fexA and tetK were determined by quantitative real time-polymerase chain reaction. Results Efflux was found in 65.3% of isolates, the prevalence of norA, tetK, fexA and msrA genes were 91.7%, 77.8%, 27.8% and 6.9%. Efflux assay revealed that tested drugs had potential efflux inhibitory activities, reduced the antibiotic’s MICs and significantly decreased the relative expression of efflux genes. Conclusion Diclofenac sodium, domperidone and glyceryl trinitrate showed higher efflux inhibitory activities than verapamil and metformin. To our knowledge, this is the first report that shows that diclofenac sodium, glyceryl trinitrate and domperidone have efflux pump inhibitory activities against S. aureus.
Collapse
Affiliation(s)
| | | | - Mona Abdelmonem El-Sayed
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | | |
Collapse
|
26
|
Zeng YX, Liu JS, Wang YJ, Tang S, Wang DY, Deng SM, Jia AQ. Actinomycin D: a novel Pseudomonas aeruginosa quorum sensing inhibitor from the endophyte Streptomyces cyaneochromogenes RC1. World J Microbiol Biotechnol 2022; 38:170. [PMID: 35904625 DOI: 10.1007/s11274-022-03360-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 07/15/2022] [Indexed: 10/16/2022]
Abstract
The infections caused by Pseudomonas aeruginosa are difficult to treat due to its multidrug resistance. A promising strategy for controlling P. aeruginosa infection is targeting the quorum sensing (QS) system. Actinomycin D isolated from the metabolite of endophyte Streptomyces cyaneochromogenes RC1 exhibited good anti-QS activity against P. aeruginosa PAO1. Actinomycin D (50, 100, and 200 μg/mL) significantly inhibited the motility as well as reduced the production of multiple virulence factors including pyocyanin, protease, rhamnolipid, and siderophores. The images of confocal laser scanning microscopy and scanning electron microscopy revealed that the treatment of actinomycin D resulted in a looser and flatter biofilm structure. Real-time quantitative PCR analysis showed that the expression of QS-related genes lasI, rhlI, rhlR, pqsR, pslA, and pilA were downregulated dramatically. The production of QS signaling molecules N-(3-oxododecanoyl)-L-homoserine lactone and N-butanoyl-L-homoserine lactone were also decreased by actinomycin D. These findings suggest that actinomycin D, a potent in vitro anti-virulence agent, is a promising candidate to treat P. aeruginosa infection by interfering with the QS systems.
Collapse
Affiliation(s)
- Yue-Xiang Zeng
- School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China.,State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, 570228, China
| | - Jun-Sheng Liu
- School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China.,State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, 570228, China
| | - Ying-Jie Wang
- School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China
| | - Shi Tang
- School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China.,State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, 570228, China
| | - Da-Yong Wang
- School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China
| | - Shi-Ming Deng
- School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China
| | - Ai-Qun Jia
- School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China. .,State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, 570228, China. .,One Health Institute, Hainan University, Haikou, 570228, China.
| |
Collapse
|
27
|
Anti-Biofilm and Antibacterial Activities of Cycas media R. Br Secondary Metabolites: In Silico, In Vitro, and In Vivo Approaches. Antibiotics (Basel) 2022; 11:antibiotics11080993. [PMID: 35892383 PMCID: PMC9394325 DOI: 10.3390/antibiotics11080993] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/18/2022] [Accepted: 07/22/2022] [Indexed: 11/17/2022] Open
Abstract
Enterococcus species possess many virulence factors that have an essential role in exacerbating the infections caused by them. The current study aimed to evaluate the effect of the secondary metabolites ginkgetin (GINK) and sotetsuflavone (SOTE), isolated from Cycas media R. Br dichloromethane fraction, on Enterococcus faecalis (E. faecalis) isolates for the first time. The antibacterial and antivirulence activities of the isolated compounds were investigated using docking studies and in vitro by determination of the minimum inhibitory concentrations (MICs). Additionally, flow cytometry and scanning electron microscope (SEM) were utilized to assess the effect of SOTE on the tested bacteria. Moreover, crystal violet assay and qRT-PCR were used to test the effect of SOTE on the biofilm-forming ability of E. faecalis isolates. In addition, a systemic infection model was utilized in vivo to investigate the antibacterial activity of SOTE. We found that both GINK and SOTE showed a good affinity for the five proteins enrolled in the virulence of E. faecalis, with SOTE being the highest, suggesting the possible mechanisms for the antivirulence activity of both ligands. In addition, SOTE exhibited a higher antibacterial activity than GINK, as the values of the MICs of SOTE were lower than those of GINK. Thus, we performed the in vitro and in vivo assays on SOTE. However, they did not exhibit any significant variations (p > 0.05) in the membrane depolarization of E. faecalis isolates. Moreover, as evaluated by SEM, SOTE caused distortion and deformation in the treated cells. Regarding its impact on the biofilm formation, it inhibited the biofilm-forming ability of the tested isolates, as determined by crystal violet assay and qRT-PCR. The in vivo experiment revealed that SOTE resulted in a reduction of the inflammation of the liver and spleen with an increase in the survival rate. SOTE also improved the liver-function tests and decreased tumor necrosis factor-alpha using immunostaining and the inflammation markers, interleukins (IL-1β and IL-6), using ELISA. Thus, we can conclude that SOTE could be a promising compound that should be investigated in future preclinical and clinical studies.
Collapse
|
28
|
Inhibitory Effects of Compounds from Plumula nelumbinis on Biofilm and Quorum Sensing Against P. aeruginosa. Curr Microbiol 2022; 79:236. [PMID: 35767197 DOI: 10.1007/s00284-022-02914-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 05/23/2022] [Indexed: 11/03/2022]
Abstract
Quorum sensing (QS), which controls the survival and virulence of Pseudomonas aeruginosa, including the formation of biofilm, is considered to be a new target to overcome pathogens. The aim of this study was to identify new QS inhibitors against P. aeruginosa and provide potential treatments for clinical infections. In this study, 25 compounds were isolated from Plumula nelumbini. Among these compounds, C25 showed the most significant biofilm inhibition activity, reaching 44.63% at 100 μM without inhibiting bacterial growth. Furthermore, C25 showed significant inhibition activity of rhamnolipid, pyocyanin, and elastase. Further mechanistic studies have confirmed that C25 could downregulate key genes in the QS system, including lasI, lasR, lasA, lasB, and pqsR, and Molecular docking studies have shown that C25 can bind to the active sites of the LasR and PqsR receptors. The present study suggests that C25 is a promising QS inhibitor for treating P. aeruginosa infections.
Collapse
|
29
|
Elshaer SL, Shaldam MA, Shaaban MI. Ketoprofen, Piroxicam and Indomethacin Suppressed Quorum Sensing and Virulence Factors in Acinetobacter baumannii. J Appl Microbiol 2022; 133:2182-2197. [PMID: 35503000 DOI: 10.1111/jam.15609] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/23/2022] [Accepted: 04/28/2022] [Indexed: 12/01/2022]
Abstract
AIM Quorum sensing (QS) inhibition is a promising strategy to suppress bacterial virulence, and control infection caused by Gram-negative and Gram-positive bacteria. This study explores the quorum sensing inhibiting activity of the non-steroidal anti-inflammatory drugs (NSAIDs) in Acinetobacter baumannii. METHODS AND RESULTS Ketoprofen, piroxicam, and indomethacin revealed QS inhibition via elimination of violacein production of the reporter strain Chromobacterium violaceum ATCC 12472 without affecting bacterial growth. The minimal inhibitory concentration (MIC) of ketoprofen, piroxicam, and indomethacin was determined against A. baumannii strains ATCC 17978, ATCC 19606, A1, A11, and A27 by the microbroth dilution method. The MICs of ketoprofen against tested isolates were 3.12-6.25 mg mL-1 , piroxicam MICs were 1.25-2.5 mg mL-1 , and indomethacin MICs were 3.12-12.5 mg mL-1 . Those compounds significantly inhibited QS-associated virulence factors such as biofilm formation, and surface motility, as well as, significantly increased bacterial tolerance to oxidative stress without affecting bacterial growth. On the molecular level, the three compounds significantly inhibited the transcription of QS regulatory genes abaI/abaR, and biofilm regulated genes cusD, and pgaB. Molecular docking analysis revealed potent binding affinity of the three compounds with AbaI via hydrogen and/or hydrophobic bonds. CONCLUSION These results indicate that NSAIDs, ketoprofen, piroxicam, and indomethacin, could be potential inhibitors of the QS and could suppress the QS-related virulence factors of A. baumannii. SIGNIFICANCE AND IMPACT Ketoprofen, piroxicam, and indomethacin could provide promising implications and strategies for combating the virulence, and pathogenesis of A. baumannii.
Collapse
Affiliation(s)
- Soha Lotfy Elshaer
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, 35516, Egypt
| | - Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafr El Sheikh 33516, Egypt
| | - Mona I Shaaban
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, 35516, Egypt
| |
Collapse
|
30
|
Escobar-Muciño E, Arenas-Hernández MMP, Luna-Guevara ML. Mechanisms of Inhibition of Quorum Sensing as an Alternative for the Control of E. coli and Salmonella. Microorganisms 2022; 10:884. [PMID: 35630329 PMCID: PMC9143355 DOI: 10.3390/microorganisms10050884] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/08/2022] [Accepted: 04/10/2022] [Indexed: 02/05/2023] Open
Abstract
Quorum sensing (QS) is a process of cell-cell communication for bacteria such as E. coli and Salmonella that cause foodborne diseases, with the production, release, and detection of autoinducer (AI) molecules that participate in the regulation of virulence genes. All of these proteins are useful in coordinating collective behavior, the expression of virulence factors, and the pathogenicity of Gram-negative bacteria. In this work, we review the natural or synthetic inhibitor molecules of QS that inactivate the autoinducer and block QS regulatory proteins in E. coli and Salmonella. Furthermore, we describe mechanisms of QS inhibitors (QSIs) that act as competitive inhibitors, being a useful tool for preventing virulence gene expression through the downregulation of AI-2 production pathways and the disruption of signal uptake. In addition, we showed that QSIs have negative regulatory activity of genes related to bacterial biofilm formation on clinical artifacts, which confirms the therapeutic potential of QSIs in the control of infectious pathogens. Finally, we discuss resistance to QSIs, the design of next-generation QSIs, and how these molecules can be leveraged to provide a new antivirulence therapy to combat diseases caused by E. coli or Salmonella.
Collapse
Affiliation(s)
- Esmeralda Escobar-Muciño
- Posgrado en Microbiología, Centro de Investigación en Ciencias Microbiológicas, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, Puebla C.P. 72570, Pue, Mexico;
| | - Margarita M. P. Arenas-Hernández
- Posgrado en Microbiología, Centro de Investigación en Ciencias Microbiológicas, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, Puebla C.P. 72570, Pue, Mexico;
| | - M. Lorena Luna-Guevara
- Colegío de Ingeniería en Alimentos, Facultad de Ingeniería Química, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, Puebla C.P. 72570, Pue, Mexico
| |
Collapse
|
31
|
She P, Liu Y, Xu L, Li Y, Li Z, Liu S, Hussain Z, Wu Y. SPR741, Double- or Triple-Combined With Erythromycin and Clarithromycin, Combats Drug-Resistant Klebsiella pneumoniae, Its Biofilms, and Persister Cells. Front Cell Infect Microbiol 2022; 12:858606. [PMID: 35372124 PMCID: PMC8971605 DOI: 10.3389/fcimb.2022.858606] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 02/25/2022] [Indexed: 11/13/2022] Open
Abstract
Klebsiella pneumoniae has emerged as a major clinical and public health threat owing to the increasing prevalence of healthcare-associated infections caused by multidrug-resistant or extensively drug-resistant strains. However, increasing antibiotic resistance and the absence of clinically effective antimicrobial agents make combination therapy an urgent need. This study investigated the anti-microbial activity of SPR741, a polymyxin B derivative, in combination with macrolide antibiotics (erythromycin and clarithromycin), against extensively drug-resistant and pandrug-resistant K. pneumoniae. Monotherapy, double, and triple combination therapies were performed to identify the most effective treatment combination using in vitro checkerboard, time-killing kinetics. Furthermore, we evaluated the biofilm eradication and persister cell-killing activity of these combinations using laser confocal microscopy and colony forming unit counting. In addition, a neutropenic mouse thigh infection model was used to assess the therapeutic efficacy and toxicity of the triple antibiotic combination against pandrug-resistant K. pneumoniae in vivo. Our results suggested that SPR741 combined with macrolides exhibited strong synergistic antibacterial activity against extensively drug-resistant and pandrug-resistant K. pneumoniae. These antibiotic combinations could also effectively eradicate highly resistant bacterial biofilms and persister cells in vitro and demonstrate considerable efficacy and low toxicity in vivo. In summary, our findings indicated that SPR741, in combination with macrolide antibiotics (double or triple combination), has the potential to serve as a novel treatment option against drug-resistant K. pneumoniae -related infections.
Collapse
Affiliation(s)
- Pengfei She
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yaqian Liu
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Lanlan Xu
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yimin Li
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zehao Li
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Shasha Liu
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zubair Hussain
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yong Wu
- Department of Laboratory Medicine, The First Hospital of Changsha, Changsha, China
| |
Collapse
|
32
|
Medina Lopez AI, Fregoso DR, Gallegos A, Yoon DJ, Fuentes JJ, Crawford R, Kaba H, Yang H, Isseroff RR. Beta adrenergic receptor antagonist can modify
Pseudomonas aeruginosa
biofilm formation in vitro: Implications for chronic wounds. FASEB J 2022; 36:e22057. [DOI: 10.1096/fj.202100717rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 10/20/2021] [Accepted: 11/08/2021] [Indexed: 11/11/2022]
Affiliation(s)
| | - Daniel R. Fregoso
- Department of Dermatology University of California, Davis Davis California USA
| | - Anthony Gallegos
- Department of Dermatology University of California, Davis Davis California USA
| | - Daniel J. Yoon
- Department of Dermatology University of California, Davis Davis California USA
| | - Jaime J. Fuentes
- Department of Biological Sciences California State University Sacramento Sacramento California USA
| | - Robert Crawford
- Department of Biological Sciences California State University Sacramento Sacramento California USA
| | - Hawa Kaba
- Department of Dermatology University of California, Davis Davis California USA
| | - Hsin‐ya Yang
- Department of Dermatology University of California, Davis Davis California USA
| | - R. Rivkah Isseroff
- Department of Dermatology University of California, Davis Davis California USA
- Dermatology Section VA Northern California Health Care System Mather USA
| |
Collapse
|
33
|
Nadar S, Khan T, Patching SG, Omri A. Development of Antibiofilm Therapeutics Strategies to Overcome Antimicrobial Drug Resistance. Microorganisms 2022; 10:microorganisms10020303. [PMID: 35208758 PMCID: PMC8879831 DOI: 10.3390/microorganisms10020303] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
A biofilm is a community of stable microorganisms encapsulated in an extracellular matrix produced by themselves. Many types of microorganisms that are found on living hosts or in the environment can form biofilms. These include pathogenic bacteria that can serve as a reservoir for persistent infections, and are culpable for leading to a broad spectrum of chronic illnesses and emergence of antibiotic resistance making them difficult to be treated. The absence of biofilm-targeting antibiotics in the drug discovery pipeline indicates an unmet opportunity for designing new biofilm inhibitors as antimicrobial agents using various strategies and targeting distinct stages of biofilm formation. The strategies available to control biofilm formation include targeting the enzymes and proteins specific to the microorganism and those involved in the adhesion pathways leading to formation of resistant biofilms. This review primarily focuses on the recent strategies and advances responsible for identifying a myriad of antibiofilm agents and their mechanism of biofilm inhibition, including extracellular polymeric substance synthesis inhibitors, adhesion inhibitors, quorum sensing inhibitors, efflux pump inhibitors, and cyclic diguanylate inhibitors. Furthermore, we present the structure–activity relationships (SAR) of these agents, including recently discovered biofilm inhibitors, nature-derived bioactive scaffolds, synthetic small molecules, antimicrobial peptides, bioactive compounds isolated from fungi, non-proteinogenic amino acids and antibiotics. We hope to fuel interest and focus research efforts on the development of agents targeting the uniquely complex, physical and chemical heterogeneous biofilms through a multipronged approach and combinatorial therapeutics for a more effective control and management of biofilms across diseases.
Collapse
Affiliation(s)
- Sahaya Nadar
- Department of Pharmaceutical Chemistry, St. John Institute of Pharmacy and Research, Mumbai 400056, India;
| | - Tabassum Khan
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, India;
| | - Simon G. Patching
- School of Biomedical Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK
- Correspondence: or (S.G.P.); (A.O.)
| | - Abdelwahab Omri
- The Novel Drug & Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Correspondence: or (S.G.P.); (A.O.)
| |
Collapse
|
34
|
El-Ganiny AM, Gad AI, El-Sayed MA, Shaldam MA, Abbas HA. The promising anti-virulence activity of candesartan, domperidone, and miconazole on Staphylococcus aureus. Braz J Microbiol 2021; 53:1-18. [PMID: 34773629 DOI: 10.1007/s42770-021-00655-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 10/17/2021] [Indexed: 12/15/2022] Open
Abstract
Staphylococcus aureus is a primary cause of hospital and community-acquired infections. With the emergence of multidrug-resistant S. aureus strains, there is a need for new drugs discovery. Due to the poor supply of new antimicrobials, targeting virulence of S. aureus may generate weaker selection for resistant strains, anti-virulence agents disarm the pathogen instead of killing it. In this study, the ability of the FDA-approved drugs domperidone, candesartan, and miconazole as inhibitors of S. aureus virulence was investigated. The effect of tested drugs was evaluated against biofilm formation, lipase, protease, hemolysin, and staphyloxanthin production by using phenotypic and genotypic methods. At sub-inhibitory concentrations, candesartan, domperidone, and miconazole showed a significant inhibition of hemolysin (75.8-96%), staphyloxanthin (81.2-85%), lipase (50-65%), protease (40-64%), and biofilm formation (71.4-90%). Domperidone and candesartan have similar activity and were more powerful than miconazole against S. aureus virulence. The hemolysins and lipase inhibition were the greatest under the domperidone effect. Candesartan showed a remarkable reduction in staphyloxanthin production. The highest inhibitory effect of proteolytic activity was obtained with domperidone and candesartan. Biofilm was significantly reduced by miconazole. Expression levels of crtM, sigB, sarA, agrA, hla, fnbA, and icaA genes were significantly reduced under candesartan (68.98-82.7%), domperidone (62.6-77.2%), and miconazole (32.96-52.6%) at sub-MIC concentrations. Candesartan showed the highest inhibition activity against crtM, sigB, sarA, agrA, hla, and icaA expression followed by domperidone then miconazole. Domperidone showed the highest downregulation activity against fnbA gene. In conclusion, candesartan, domperidone, and miconazole could serve as anti-virulence agents for attenuation of S. aureus pathogenicity.
Collapse
Affiliation(s)
- Amira M El-Ganiny
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Amany I Gad
- Microbiology and Immunology Department, School of Pharmacy and Pharmaceutical Industries, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt.
| | - Mona A El-Sayed
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Moataz A Shaldam
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Hisham A Abbas
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| |
Collapse
|
35
|
Nolan C, Behrends V. Sub-Inhibitory Antibiotic Exposure and Virulence in Pseudomonas aeruginosa. Antibiotics (Basel) 2021; 10:antibiotics10111393. [PMID: 34827331 PMCID: PMC8615142 DOI: 10.3390/antibiotics10111393] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 12/20/2022] Open
Abstract
Pseudomonas aeruginosa is a prime opportunistic pathogen, one of the most important causes of hospital-acquired infections and the major cause of morbidity and mortality in cystic fibrosis lung infections. One reason for the bacterium's pathogenic success is the large array of virulence factors that it can employ. Another is its high degree of intrinsic and acquired resistance to antibiotics. In this review, we first summarise the current knowledge about the regulation of virulence factor expression and production. We then look at the impact of sub-MIC antibiotic exposure and find that the virulence-antibiotic interaction for P. aeruginosa is antibiotic-specific, multifaceted, and complex. Most studies undertaken to date have been in vitro assays in batch culture systems, involving short-term (<24 h) antibiotic exposure. Therefore, we discuss the importance of long-term, in vivo-mimicking models for future work, particularly highlighting the need to account for bacterial physiology, which by extension governs both virulence factor expression and antibiotic tolerance/resistance.
Collapse
|
36
|
Khadke SK, Lee JH, Kim YG, Raj V, Lee J. Assessment of Antibiofilm Potencies of Nervonic and Oleic Acid against Acinetobacter baumannii Using In Vitro and Computational Approaches. Biomedicines 2021; 9:biomedicines9091133. [PMID: 34572317 PMCID: PMC8466663 DOI: 10.3390/biomedicines9091133] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/25/2021] [Accepted: 08/29/2021] [Indexed: 12/24/2022] Open
Abstract
Acinetobacter baumannii is a nosocomial pathogen, and its biofilms are tolerant to desiccation, nutrient starvation, and antimicrobial treatment on biotic and abiotic surfaces, tissues, and medical devices. Biofilm formation by A. baumannii is triggered by a quorum sensing cascade, and we hypothesized that fatty acids might inhibit its biofilm formation by interfering with quorum sensing. Initially, we investigated the antibiofilm activities of 24 fatty acids against A. baumannii ATCC 17978 and two clinical isolates. Among these fatty acids, two unsaturated fatty acids, nervonic and oleic acid, at 20 μg/mL significantly inhibited A. baumannii biofilm formation without affecting its planktonic cell growth (MICs were >500 μg/mL) and markedly decreased the motility of A. baumannii but had no toxic effect on the nematode Caenorhabditis elegans. Interestingly, molecular dynamic simulations showed that both fatty acids bind to the quorum sensing acyl homoserine lactone synthase (AbaI), and decent conformational stabilities of interactions between the fatty acids and AbaI were exhibited. Our results demonstrate that nervonic and oleic acid inhibit biofilm formation by A. baumannii strains and may be used as lead molecules for the control of persistent A. baumannii infections.
Collapse
Affiliation(s)
| | | | | | | | - Jintae Lee
- Correspondence: ; Tel.: +82-53-810-2533; Fax: +82-53-810-4631
| |
Collapse
|
37
|
Lazar V, Holban AM, Curutiu C, Chifiriuc MC. Modulation of Quorum Sensing and Biofilms in Less Investigated Gram-Negative ESKAPE Pathogens. Front Microbiol 2021; 12:676510. [PMID: 34394026 PMCID: PMC8359898 DOI: 10.3389/fmicb.2021.676510] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 06/30/2021] [Indexed: 01/20/2023] Open
Abstract
Pathogenic bacteria have the ability to sense their versatile environment and adapt by behavioral changes both to the external reservoirs and the infected host, which, in response to microbial colonization, mobilizes equally sophisticated anti-infectious strategies. One of the most important adaptive processes is the ability of pathogenic bacteria to turn from the free, floating, or planktonic state to the adherent one and to develop biofilms on alive and inert substrata; this social lifestyle, based on very complex communication networks, namely, the quorum sensing (QS) and response system, confers them an increased phenotypic or behavioral resistance to different stress factors, including host defense mechanisms and antibiotics. As a consequence, biofilm infections can be difficult to diagnose and treat, requiring complex multidrug therapeutic regimens, which often fail to resolve the infection. One of the most promising avenues for discovering novel and efficient antibiofilm strategies is targeting individual cells and their QS mechanisms. A huge amount of data related to the inhibition of QS and biofilm formation in pathogenic bacteria have been obtained using the well-established gram-positive Staphylococcus aureus and gram-negative Pseudomonas aeruginosa models. The purpose of this paper was to revise the progress on the development of antibiofilm and anti-QS strategies in the less investigated gram-negative ESKAPE pathogens Klebsiella pneumoniae, Acinetobacter baumannii, and Enterobacter sp. and identify promising leads for the therapeutic management of these clinically significant and highly resistant opportunistic pathogens.
Collapse
Affiliation(s)
- Veronica Lazar
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
| | - Alina Maria Holban
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
| | - Carmen Curutiu
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
| | - Mariana Carmen Chifiriuc
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
| |
Collapse
|
38
|
Could the analgesic drugs, paracetamol and indomethacin, function as quorum sensing inhibitors? Microb Pathog 2021; 158:105097. [PMID: 34284088 DOI: 10.1016/j.micpath.2021.105097] [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: 06/29/2020] [Revised: 10/05/2020] [Accepted: 07/15/2021] [Indexed: 11/22/2022]
Abstract
The current failure of antimicrobials in treating life-threatening diseases, the high rate of multidrug resistant pathogens and the slow progress in the development of new antibiotics directed scientists to develop antivirulence drugs that targets quorum sensing (QS). In many microbes, QS acts as a communication system which control pathogenicity of microbes. Analgesics can be beneficial in controlling virulence traits of microbes and hence they may augment the efficacy of antimicrobials. In this study, two analgesics were screened for the inhibition of QS in Chromobacterium violaceum CV026 and their effects on virulence production in Pseudomonas aeruginosa PAO1 strain and clinical isolates of Acinetobacter baumannii were evaluated. The traits investigated were biofilm formation, pyocyanin and rhamnolipid production, twitching, swarming or surface associated motilities, production of protease, phospholipase and gelatinase enzymes and sensitivity to oxidative stress. Relative expression of abaI gene was calculated by performing qRT-PCR. Docking analysis of paracetamol as QSI (quorum sensing inhibitor) of AbaI and AbaR proteins was performed. Paracetamol inhibited QS in CV026, but indomethacin devoids anti-QS activity. Paracetamol inhibited virulence factors of PAO1. It strongly inhibited biofilm formation, and swarming by 66.4% and 57.1%, respectively. While, it moderately to slightly inhibited rhamnolipid, pyocyanin, gelatinase, resistance to oxidative stress, protease and twitching motility by 33.3%, 33.1% 17.5%, 9.1%, 8.7% and 7.7%, respectively. For A. baumannii, paracetamol strongly inhibited biofilm by 39.7-93% and phospholipase enzyme by 8.7-100%, reduced twitching and surface motility by 6.7-82.5% and 7.7-29.4%, respectively, And slightly reduced sensitivity to oxidative stress by 3.3-36.4%. Paracetamol at sub-MIC suppressed the expression of abaI gene by 32% in A. baumannii. Docking studies suggested that paracetamol can bind to AbaR and AbaI proteins and bind more to AbaR, hence it may act by inhibiting AHL signal reception. As a conclusion, paracetamol, beside its analgesic activity, has anti-QS activity and could be used in the eradication of P. aeruginosa and A. baumannii infections in combination with antibiotics.
Collapse
|
39
|
Pimentel C, Le C, Tuttobene MR, Subils T, Papp-Wallace KM, Bonomo RA, Tolmasky ME, Ramirez MS. Interaction of Acinetobacter baumannii with Human Serum Albumin: Does the Host Determine the Outcome? Antibiotics (Basel) 2021; 10:antibiotics10070833. [PMID: 34356754 PMCID: PMC8300715 DOI: 10.3390/antibiotics10070833] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/06/2021] [Accepted: 07/06/2021] [Indexed: 01/16/2023] Open
Abstract
Acinetobacter baumannii has become a serious threat to human health due to its extreme antibiotic resistance, environmental persistence, and capacity to survive within the host. Two A. baumannii strains, A118 and AB5075, commonly used as model systems, and three carbapenem-resistant strains, which are becoming ever more dangerous due to the multiple drugs they can resist, were exposed to 3.5% human serum albumin (HSA) and human serum (HS) to evaluate their response with respect to antimicrobial resistance, biofilm formation, and quorum sensing, all features responsible for increasing survival and persistence in the environment and human body. Expression levels of antibiotic resistance genes were modified differently when examined in different strains. The cmlA gene was upregulated or downregulated in conditions of exposure to 3.5% HSA or HS depending on the strain. Expression levels of pbp1 and pbp3 tended to be increased by the presence of HSA and HS, but the effect was not seen in all strains. A. baumannii A118 growing in the presence of HS did not experience increased expression of these genes. Aminoglycoside-modifying enzymes were also expressed at higher or lower levels in the presence of HSA or HS. Still, the response was not uniform; in some cases, expression was enhanced, and in other cases, it was tapered. While A. baumannii AB5075 became more susceptible to rifampicin in the presence of 3.5% HSA or HS, strain A118 did not show any changes. Expression of arr2, a gene involved in resistance to rifampicin present in A. baumannii AMA16, was expressed at higher levels when HS was present in the culture medium. HSA and HS reduced biofilm formation and production of N-Acyl Homoserine Lactone, a compound intimately associated with quorum sensing. In conclusion, HSA, the main component of HS, stimulates a variety of adaptative responses in infecting A. baumannii strains.
Collapse
Affiliation(s)
- Camila Pimentel
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92831-3599, USA; (C.P.); (C.L.); (M.E.T.)
| | - Casin Le
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92831-3599, USA; (C.P.); (C.L.); (M.E.T.)
| | - Marisel R. Tuttobene
- Área Biología Molecular, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario 2000, Argentina;
| | - Tomas Subils
- Instituto de Procesos Biotecnológicos y Químicos de Rosario (IPROBYQ, CONICET-UNR), Rosario S2002LRK, Argentina;
| | - Krisztina M. Papp-Wallace
- Research Service and GRECC, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH 44106, USA; (K.M.P.-W.); (R.A.B.)
- Departments of Medicine, Pharmacology, Molecular Biology and Microbiology, Biochemistry, Proteomics and Bioinformatics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- CWRU-Cleveland VAMC Center for Antimicrobial Resistance and Epidemiology (Case VA CARES), Cleveland, OH 44106, USA
| | - Robert A. Bonomo
- Research Service and GRECC, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH 44106, USA; (K.M.P.-W.); (R.A.B.)
- Departments of Medicine, Pharmacology, Molecular Biology and Microbiology, Biochemistry, Proteomics and Bioinformatics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- CWRU-Cleveland VAMC Center for Antimicrobial Resistance and Epidemiology (Case VA CARES), Cleveland, OH 44106, USA
| | - Marcelo E. Tolmasky
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92831-3599, USA; (C.P.); (C.L.); (M.E.T.)
| | - Maria Soledad Ramirez
- Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA 92831-3599, USA; (C.P.); (C.L.); (M.E.T.)
- Correspondence: ; Tel.: +1-657-278-4562
| |
Collapse
|
40
|
Zhong S, He S. Quorum Sensing Inhibition or Quenching in Acinetobacter baumannii: The Novel Therapeutic Strategies for New Drug Development. Front Microbiol 2021; 12:558003. [PMID: 33597937 PMCID: PMC7882596 DOI: 10.3389/fmicb.2021.558003] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 01/06/2021] [Indexed: 01/01/2023] Open
Abstract
Acinetobacter baumannii is a Gram-negative opportunistic nosocomial pathogen, which can cause ventilator-related and blood infection in critically ill patients. The resistance of A. baumannii clinical isolates to common antimicrobials and their tolerance to desiccation have emerged as a serious problem to public health. In the process of pathogenesis, bacteria release signals, which regulate virulence and pathogenicity-related genes. Such bacteria coordinate their virulent behavior in a cell density-dependent phenomenon called quorum sensing (QS). In contrast, the two main approaches of QS interference, quorum sensing inhibitors (QSIs) and quorum quenching (QQ) enzymes, have been developed to reduce the virulence of bacteria, thus reducing the pressure to produce bacterial drug resistance. Therefore, QSIs or QQ enzymes, which interfere with these processes, might potentially inhibit bacterial QS and ultimately biofilm formation. In this review, we aim to describe the state-of-art in the QS process in A. baumannii and elaborate on the use of QSIs or QQ enzymes as antimicrobial drugs in various potential sites of the QS pathway.
Collapse
Affiliation(s)
- Shan Zhong
- Department of Acupuncture, Guilin Hospital of Traditional Chinese Medicine, Guilin, China
| | - Songzhe He
- Department of Clinical Laboratory, Affiliated Hospital of Guilin Medical University, Guilin, China.,Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
41
|
She P, Liu Y, Luo Z, Chen L, Zhou L, Hussain Z, Wu Y. PA2146 Gene Knockout Is Associated With Pseudomonas aeruginosa Pathogenicity in Macrophage and Host Immune Response. Front Cell Infect Microbiol 2020; 10:559803. [PMID: 33134189 PMCID: PMC7579411 DOI: 10.3389/fcimb.2020.559803] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/08/2020] [Indexed: 12/22/2022] Open
Abstract
Pseudomonas aeruginosa is a common gram-negative bacterium that usually causes nosocomial infection. The main pathogenicity of P. aeruginosa is caused by its virulence factors. PA2146 is reported to be a potential virulence-regulating gene and is highly expressed in the biofilms of P. aeruginosa. However, the effect of PA2146 mutant (PAO1ΔPA2146) on the macrophage immune response and murine models has not been reported. In the present study, PA2146 knockout was performed by homologous recombination. We found that PAO1ΔPA2146 stimulation significantly increased pyocyanin production but inhibited interleukin-6 secretion by neutrophils compared to PAO1 stimulation. In addition, PAO1ΔPA2146 treatment significantly inhibited cytokine production in macrophages independent of cell killing. In an acute pneumonia murine infection model, treatment with P. aeruginosa infected with PAO1ΔPA2146 inhibited cytokine secretion in the lungs but increased the infiltration of inflammatory cells compared to the wild-type group. The paradoxical results indicate that PA2146 deletion may also increase the production of virulence factors other than pyocyanin, which may not only increase inflammatory cell infiltration in the lungs but also lead to immune cells “shock.” Overall, our findings suggest that PA2146 could serve as a P. aeruginosa virulence-regulating gene that regulates its macrophage and host immune response.
Collapse
Affiliation(s)
- Pengfei She
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yiqing Liu
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhen Luo
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Lihua Chen
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Linying Zhou
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zubair Hussain
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yong Wu
- Department of Clinical Laboratory, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
42
|
Peng Q, Lin F, Ling B. In vitro activity of biofilm inhibitors in combination with antibacterial drugs against extensively drug-resistant Acinetobacter baumannii. Sci Rep 2020; 10:18097. [PMID: 33093606 PMCID: PMC7581519 DOI: 10.1038/s41598-020-75218-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 10/13/2020] [Indexed: 12/15/2022] Open
Abstract
Acinetobacter baumannii is a common pathogen of nosocomial infection, and its ability to form biofilms further contributes to its virulence and multidrug resistance, posing a great threat to global public health. In this study, we investigated the inhibitory effects of five biofilm inhibitors (BFIs) (zinc lactate, stannous fluoride, furanone, azithromycin, and rifampicin) on biofilm formation of nine extensively drug-resistant A. baumannii (XDRAB), and assessed the synergistic antibacterial effects of these BFIs when combined with one of four conventional anti-A. baumannii antibiotics (imipenem, meropenem, tigecycline, and polymyxin B). Each of the five BFIs tested was found to be able to significantly inhibit biofilm formation of all the clinical isolates tested under sub-minimal inhibitory concentrations. Then, we observed synergistic effects (in 22%, 56% and 11% of the isolates) and additive effects (56%, 44% and 44%) when zinc lactate, stannous fluoride and furanone were combined with tigecycline, respectively. When zinc lactate and stannous fluoride were each used with a carbapenem (imipenem or meropenem), in 33% and 56-67% of the isolates, they showed synergistic and additive effects, respectively. Additivity in > 50% of the isolates was detected when rifampicin was combined with imipenem, meropenem, tigecycline, or polymyxin B; and a 100% additivity was noted with azithromycin-polymyxin B combination. However, antagonism and indifference were noted for polymyxin B in its combination with zinc lactate and stannous fluoride, respectively. In conclusion, five BFIs in combination with four antibacterial drugs showed different degrees of in vitro synergistic and additive antibacterial effects against XDRAB.
Collapse
Affiliation(s)
- Qin Peng
- Sichuan Province College Key Laboratory of Structure-Specific Small Molecule Drugs, Chengdu Medical College, Chengdu, 610500, China
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, China
- Department of Pharmacy, Nanchong Central Hospital, Nanchong, 637000, China
| | - Fei Lin
- Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Baodong Ling
- Sichuan Province College Key Laboratory of Structure-Specific Small Molecule Drugs, Chengdu Medical College, Chengdu, 610500, China.
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, China.
| |
Collapse
|
43
|
Diclofenac mitigates virulence of multidrug-resistant Staphylococcus aureus. Arch Microbiol 2020; 202:2751-2760. [PMID: 32737541 DOI: 10.1007/s00203-020-01992-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/17/2020] [Accepted: 07/21/2020] [Indexed: 12/20/2022]
Abstract
Staphylococcus aureus is an opportunistic pathogen that has the ability to cause a wide range of diseases including superficial infection and severe invasive life threatening infections. The pathogenicity of S. aureus is mediated by a group of virulence factors that mediate the colonization and penetration. The antibiotic resistance of S. aureus has evolved due to the abuse of antibiotics rendering the cure of infection very difficult especially with the shortage in new antibiotic production. To combat this shortage, repurposing of FDA-approved drugs against the virulence factors is a new strategy. The analgesic drug Diclofenac was found to have anti-virulence activity against Pseudomonas aeruginosa and Proteus mirabilis. This study aimed to demonstrate the anti-virulence effect of diclofenac against clinical MRSA isolates phenotypically and genotypically using qRT-PCR. In this study, diclofenac showed significant reduction in biofilm formation when compared to controls, the inhibition ranged between 22.67% and 70%. Also, remarkable inhibition of hemolysin activity was found (5.4-66.34%). Additionally, diclofenac has inhibitory activity against the staphyloxanthin production (8-57.2%). The results were confirmed by qRT-PCR that showed significant down-regulation of tested virulence genes. The down-regulation ranged from 43 to 64.05% for SarA, 36.85-64.75% for AgrA, 50-63.2% for hla, 38.55-60.35% for FnbA, 46.75-61.05% for IcaA, 27.55-64% for SigB and 51.05-72.8% for CrtM. In conclusion, diclofenac can be used in combination with antibiotics as anti-virulence agent against MDR-MRSA which will enhance the ability of immune system to eradicate infection.
Collapse
|