1
|
Wu Y, Wen F, Gou S, Ran Q, Chu Y, Ma W, Zhao K. Multifaceted quorum-sensing inhibiting activity of 3-(Benzo[d][1,3]dioxol-4-yl)oxazolidin-2-one mitigates Pseudomonas aeruginosa virulence. Virulence 2025; 16:2479103. [PMID: 40104940 DOI: 10.1080/21505594.2025.2479103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 11/10/2024] [Accepted: 03/03/2025] [Indexed: 03/20/2025] Open
Abstract
As antibiotic resistance escalates into a global health crisis, novel therapeutic approaches against infectious diseases are in urgent need. Pseudomonas aeruginosa, an adaptable opportunistic pathogen, poses substantial challenges in treating a range of infections. The quorum-sensing (QS) system plays a pivotal role in orchestrating the production of a large set of virulence factors in a cell density-dependent manner, and the anti-virulence strategy targeting QS may show huge potential. Here, we present a comprehensive investigation into the potential of the synthesized compound 3-(benzo[d][1,3]dioxol-4-yl)oxazolidin-2-one (OZDO, C10H9NO4) as a QS inhibitor to curb the virulence of P. aeruginosa. By employing an integrated approach encompassing in silico screening, in vitro and in vivo functional identification, we elucidated the multifaceted effects of OZDO. Molecular docking predicted that OZDO interfered with three core regulatory proteins of P. aeruginosa QS system. Notably, OZDO exhibited significant inhibition on the production of pyocyanin, rhamnolipid and extracellular proteases, biofilm formation, and cell motilities of P. aeruginosa. Transcriptomic analysis and quantitative real-time PCR displayed the down-regulation of QS-controlled genes in OZDO-treated PAO1, reaffirming the QS-inhibition activity of OZDO. In vivo assessments using a Caenorhabditis elegans-infection model demonstrated OZDO mitigated P. aeruginosa pathogenicity, particularly against the hypervirulent strain PA14. Moreover, OZDO in combination with polymyxin B and aztreonam presented a promising avenue for innovative anti-infective therapy. Our study sheds light on the multifaceted potential of OZDO as an anti-virulence agent and its significance in combating P. aeruginosa-associated infections.
Collapse
Affiliation(s)
- Yi Wu
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Fulong Wen
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Shiyi Gou
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Qiman Ran
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Yiwen Chu
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Wenbo Ma
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Kelei Zhao
- Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Adhikary R, Sarkar I, Patel D, Gang S, Nath UK, Hazra S. Deciphering antibiotic resistance, quorum sensing, and biofilm forming genes of Micrococcus luteus from hemodialysis tunneled cuffed catheter tips of renal failure patients. Arch Microbiol 2025; 207:114. [PMID: 40186781 DOI: 10.1007/s00203-025-04310-6] [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: 01/29/2025] [Revised: 03/06/2025] [Accepted: 03/16/2025] [Indexed: 04/07/2025]
Abstract
Catheter-related bloodstream infections create a significant challenge in healthcare system, often complicated by antibiotic resistance and biofilm formation of multi-drug resistance and virulent bacterial pathogens. This study focused on biofilm-forming efficiency, and underlying genetic mechanisms in Micrococcus luteus HL_Chru_C3, isolated from a hemodialysis catheter tip. The isolate exhibited resistance to multiple antibiotic classes, including beta-lactams and glycopeptides. Biofilm assays revealed that M. luteus HL_Chru_C3 formed optimum biofilms at high concentration of carbohydrates (500 mM), and pH 5 but there was no significant role of mineral salts. Whole-genome sequencing and bioinformatic analysis using CARD, KAAS, and KEGG databases identified genes associated with antibiotic resistance (ftsI, pbp1a/2, vanY, alr, ddl, murF, mraY, and murG), quorum sensing (genes from the opp family, sec, cylA, ccfA, phnA, phnB, phzC, rpfB, clp, and toxE), and biofilm formation (phnA, phnB, cyaB, vfr, vps, glgC, wecB, wecC, and cysE). The predicted mechanisms of action for these genes, based on homology to other organisms, suggest complex interactions contributing to the observed phenotypes. This study provides an insight into the genetic basis of antibiotic resistance and biofilm formation in M. luteus HL_Chru_C3 isolated from a hemodialysis catheter, highlighting the need for effective infection control strategies to combat CRBSIs.
Collapse
Affiliation(s)
- Rajsekhar Adhikary
- Department of Bioscience and Bioengineering, Indian Institute of Technology-Roorkee, Roorkee, Uttarakhand, India
| | - Indrani Sarkar
- Department of Bioscience and Bioengineering, Indian Institute of Technology-Roorkee, Roorkee, Uttarakhand, India
- Department of Medical Oncology and Haematology, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| | - Dhara Patel
- Department of Medical Laboratory Technology, Bapubhai Desaibhai Patel Institue of Paramedical Sciences (BDIPS), Charotar University of Science and Technology, CHARUSAT Campus, Changa, Anand, Gujarat, 388421, India
| | - Sishir Gang
- Department of Nephrology, Muljibhai Patel Urological Hospital, Nadiad, Gujarat, India
| | - Uttam Kumar Nath
- Department of Medical Oncology and Haematology, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| | - Saugata Hazra
- Department of Bioscience and Bioengineering, Indian Institute of Technology-Roorkee, Roorkee, Uttarakhand, India.
- Centre for Nanotechnology, Indian Institute of Technology-Roorkee, Roorkee, Uttarakhand, India.
| |
Collapse
|
3
|
Ran Q, Yuan Y, Wu Y, Gan X, Deng J, Chu Y, Ji Q, Wang X, Zhao K. Two amino-substituted diphenyl fumaramide derivatives inhibit the virulence regulated by quorum sensing system of Pseudomonas aeruginosa. J Appl Microbiol 2025; 136:lxaf038. [PMID: 39971733 DOI: 10.1093/jambio/lxaf038] [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: 09/09/2024] [Revised: 02/06/2025] [Accepted: 02/18/2025] [Indexed: 02/21/2025]
Abstract
AIM Pseudomonas aeruginosa employs the quorum sensing (QS) system, a sophisticated cell-to-cell communication mechanism, to modulate the synthesis and secretion of a range of virulence factors, which contribute to the establishment of acute or chronic infections in hosts. This study seeks to attenuate the virulence of P. aeruginosa by inhibiting the QS system, thereby reducing its pathogenicity as a promising alternative to traditional antibiotics. METHODS AND RESULTS Two compounds with an amino-substituted diphenyl fumaramide core, N1-(4-bromophenyl)-N4-(4'-oxo-3',4'-dihydro-1'H-spiro [cyclopentane-1,2'-quinazolin]-6'-yl) fumaramide (10D) and N1-(3-chloro-4-fluorophenyl)-N4-(4-oxo-3,4,4',5'-tetrahydro-1H,2'H-spiro [quinazoline-2,3'-thiophen]-6-yl) fumaramide (12A), were identified through in-silico screening. The QS inhibitory potential of both compounds was explored in vitro and in vivo. In in vitro experiments, neither compound exhibited bactericidal effects but significantly inhibited the production of QS-regulated extracellular protease and pyocyanin. Quantitative PCR analysis revealed that QS-activated genes and downstream virulence genes were transcriptionally suppressed by 10D or 12A. Molecular docking and molecular dynamics simulations predicted stable interactions between these compounds and the key QS regulators LasR and PqsR. When combined with polymyxin B, kanamycin, and levofloxacin, 10D and 12A exhibited synergistic antibacterial activity. Furthermore, compounds 10D and 12A significantly improved the survival of mice challenged with P. aeruginosa and effectively reduced the bacterial load in the lungs. CONCLUSION This study indicates that 10D and 12A possess considerable QS inhibitory potential, effectively attenuating the pathogenicity of P. aeruginosa. Moreover, the study offers structural insights and methodological guidance for the advancement of anti-virulence drug development.
Collapse
Affiliation(s)
- Qiman Ran
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu 610106 Sichuan, China
| | - Yang Yuan
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu 610106 Sichuan, China
| | - Yi Wu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu 610106 Sichuan, China
| | - Xiongyao Gan
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu 610106 Sichuan, China
| | - Junfeng Deng
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu 610106 Sichuan, China
| | - Yiwen Chu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu 610106 Sichuan, China
| | - Qinggang Ji
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Xinrong Wang
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu 610106 Sichuan, China
| | - Kelei Zhao
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu 610106 Sichuan, China
| |
Collapse
|
4
|
Zhao Y, Seenivasan B, Li R, Li C, Zhang Y, Ravichandran V, Zhong L, Li A. Exploring daidzein dimethyl ether from Albizzia lebbeck as a novel quorum sensing inhibitor against Pseudomonas aeruginosa: Insights from in vitro and in vivo studies. Bioorg Chem 2025; 156:108168. [PMID: 39864373 DOI: 10.1016/j.bioorg.2025.108168] [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: 10/28/2024] [Revised: 01/03/2025] [Accepted: 01/13/2025] [Indexed: 01/28/2025]
Abstract
Infections of multidrug-resistant pathogens including Pseudomonas aeruginosa, cause a high risk of mortality in immunocompromised patients and underscore the need for novel natural antibacterial drugs. In this study, common phytochemicals prevalent in fruits and vegetables have been demonstrated for their ability to inhibit quorum sensing (QS) in Pseudomonas aeruginosa PAO1 (PA). Ten compounds were screened virtually by molecular docking, among which, daidzein dimethyl ether originally from Albizzia lebbeck showed the most significant inhibitory effect on the formation of biofilm and the accumulation of virulence factors, including elastase, pyocyanin and rhamnolipid in PA. Further, both qRT-PCR analysis of key QS components including LasR in PA and luminescence detection of LasR as a reporter in a heterologous system revealed that daidzein dimethyl ether at 10 µM significantly inhibited the transcription of lasR and its downstream targeting genes. At the same time, MD simulations also showed that daidzein dimethyl ether could reduce the stability of LasR. Furthermore, the protective effect of daidzein dimethyl ether against PA infection was demonstrated using zebrafish infection model. It was found to reduce significantly the inflammation in the PA-infected zebrafish and increase their survival rate by inhibiting prominently the accumulation of reactive oxygen species (ROS) and reducing fish mortality in PA-infected zebrafish larvae. Additionally, open field tests suggested that PA-infected zebrafish were observed with impaired swimming behaviour, but daidzein dimethyl ether-treatment rescued zebrafish from such swimming abnormalities. Histopathological analysis revealed that zebrafish treated with both PA and daidzein dimethyl ether showed obvious integrity in intestine tissues, compared to those with only PA treatment. This study firstly demonstrated the preventive advantages of daidzein dimethyl ether in an animal model against PA infection. It is worthwhile to explore further its potential therapeutic intervention as an antimicrobial agent.
Collapse
Affiliation(s)
- Yiming Zhao
- Helmholtz International Lab for Anti-Infectives, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China
| | - Boopathi Seenivasan
- Helmholtz International Lab for Anti-Infectives, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China
| | - Ruijuan Li
- Helmholtz International Lab for Anti-Infectives, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China
| | - Caiyun Li
- Helmholtz International Lab for Anti-Infectives, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China
| | - Youming Zhang
- Helmholtz International Lab for Anti-Infectives, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China; Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Key Laboratory of Genome Manipulation and Biosynthesis, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China.
| | - Vinothkannan Ravichandran
- Helmholtz International Lab for Anti-Infectives, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China; Center for Drug Discovery and Development (CD3), Amity Institute of Biotechnology, Amity University Maharashtra, Panvel, Mumbai, Maharashtra 410206, India.
| | - Lin Zhong
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Key Laboratory of Genome Manipulation and Biosynthesis, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China.
| | - Aiying Li
- Helmholtz International Lab for Anti-Infectives, State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China.
| |
Collapse
|
5
|
Chigozie VU, Saki M, Esimone CO. Molecular structural arrangement in quorum sensing and bacterial metabolic production. World J Microbiol Biotechnol 2025; 41:71. [PMID: 39939401 DOI: 10.1007/s11274-025-04280-3] [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: 12/14/2024] [Accepted: 01/28/2025] [Indexed: 02/14/2025]
Abstract
Quorum sensing (QS) regulates bacterial behaviors such as biofilm formation, virulence, and metabolite production through signaling molecules like acyl-homoserine lactones (AHLs), peptides, and AI-2. These signals are pivotal in bacterial communication, influencing pathogenicity and industrial applications. This review explores the molecular architecture of QS signals and their role in metabolite production, emphasizing structural modifications that disrupt bacterial communication to control virulence and enhance industrial processes. Key findings highlight the development of synthetic QS analogs, engineered inhibitors, and microbial consortia as innovative tools in biotechnology and medicine. The review underscores the potential of molecular engineering in managing microbial behaviors and optimizing applications like biofuel production, bioplastics, and anti-virulence therapies. Additionally, cross-species signaling mechanisms, particularly involving AI-2, reveal new opportunities for regulating interspecies cooperation and competition. This synthesis aims to bridge molecular insights with practical applications, showcasing how QS-based technologies can drive advancements in microbial biotechnology and therapeutic strategies.
Collapse
Affiliation(s)
- Victor U Chigozie
- Department of Pharmaceutical Microbiology and Biotechnology, David Umahi Federal University of Health Sciences, Ohaozara, Ebonyi State, Nigeria.
- International Institute for Pharmaceutical Research (IIPR), Ohaozara, Ebonyi State, Nigeria.
| | - Morteza Saki
- Department of Microbiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Charles O Esimone
- Department of Pharmaceutical Microbiology and Biotechnology, Nnamdi Azikiwe University, Awka, Nigeria
| |
Collapse
|
6
|
Al-Daghistani HI, Matalqah SM, Shadid KA, Abu-Niaaj LF, Zein S, Abo-Ali RM. Quorum Quenching of P. aeruginosa by Portulaca oleracea Methanolic Extract and Its Phytochemical Profile. Pathogens 2025; 14:163. [PMID: 40005538 PMCID: PMC11858189 DOI: 10.3390/pathogens14020163] [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: 12/27/2024] [Revised: 01/19/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
Quorum sensing (QS) is a molecular communication mechanism among bacterial cells. It is critical in regulating virulence factors, motility, antibiotic resistance, and biofilm formation. Pseudomonas aeruginosa is a Gram-negative opportunistic pathogen linked to healthcare-associated infections, food poisoning, and biofilm formation. Treating infections caused by pathogenic bacteria has become a challenge due to the development of multi-antibiotic resistance upon continuous exposure of bacteria to antibiotics. An alternative strategy to conventional antimicrobials to decrease the bacterial pathogenicity is QS inhibition, also known as quorum quenching. Using plant-derived compounds is an environmentally friendly strategy to block the bacterial QS and inhibit bacterial growth. Portulaca oleracea is a popular plant in different countries and is also used in traditional medicine. It is widely consumed raw in salads and as garnishes, though it can be cooked as a vegetarian dish. This study evaluates the antimicrobial activity of the methanolic extract of P. oleracea and its effectiveness in blocking or attenuating the QS of P. aeruginosa. The agar well diffusion method used for screening the antibacterial activity showed a significant growth inhibition of P. aeruginosa by the extract at 500 mg/mL with a minimum inhibitory concentration of 31.25 mg/mL. A bioindicator bacterium, Chromobacterium violaceum CV026, was used to determine the effect of the methanolic extract on the QS of P. aeruginosa. The results indicated a significant reduction in biofilm formation, pyocyanin production, and LasA staphylolytic activity. The phytochemical analysis by Gas Chromatography-Mass Spectrometry showed that the methanolic extract contained several phenols, alkaloids, esters, and other compounds previously reported to have antibacterial and antioxidant effects. These findings highlight the effectiveness of P. oleracea methanolic extract in attenuating the QS and virulence factors of P. aeruginosa. This study suggests that P. oleracea is an important source of natural antimicrobials and its use would be beneficial in food and pharmaceutical applications.
Collapse
Affiliation(s)
- Hala I. Al-Daghistani
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan;
| | - Sina M. Matalqah
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan; (S.M.M.); (K.A.S.)
| | - Khalid A. Shadid
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan; (S.M.M.); (K.A.S.)
| | - Lubna F. Abu-Niaaj
- Department of Agricultural and Life Sciences, College of Engineering, Science, Technology and Agriculture, Central State University, Wilberforce, OH 45384, USA
| | - Sima Zein
- Department of Pharmaceutical Biotechnology, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan;
| | - Raeda M. Abo-Ali
- Faculty of Nursing, Al-Balqa Applied University, Amman 19117, Jordan;
| |
Collapse
|
7
|
Liu S, Rahman MR, Wu H, Qin W, Wang Y, Su G. Development and application of hydrogels in pathogenic bacteria detection in foods. J Mater Chem B 2025; 13:1229-1251. [PMID: 39690945 DOI: 10.1039/d4tb01341g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Hydrogels are 3D networks of water-swollen hydrophilic polymers. It possesses unique properties (e.g., carrying biorecognition elements and creating a micro-environment) that make it highly suitable for bacteria detection (e.g., expedited and effective bacteria detection) and mitigation of bacterial contamination in specific environments (e.g., food systems). This study first introduces the materials used to create hydrogels for bacteria detection and the mechanisms for detection. We also summarize different hydrogel-based detection methods that rely on external stimuli and biorecognition elements, such as enzymes, temperature, pH, antibodies, and oligonucleotides. Subsequently, a range of widely utilized bacterial detection technologies were discussed where recently hydrogels are being used. These modifications allow for precise, real-time diagnostics across varied food matrices, responding effectively to industry needs for sensitivity, scalability, and portability. After highlighting the utilization of hydrogels and their role in these detection techniques, we outline limitations and advancements in the methods for the detection of foodborne pathogenic bacteria, especially the potential application of hydrogels in the food industry.
Collapse
Affiliation(s)
- Shuxiang Liu
- College of Food Science, Sichuan Agricultural University, Ya'an 625014, China.
| | - Md Rashidur Rahman
- College of Food Science, Sichuan Agricultural University, Ya'an 625014, China.
| | - Hejun Wu
- College of Science, Sichuan Agricultural University, Ya'an, 625000, China.
| | - Wen Qin
- College of Food Science, Sichuan Agricultural University, Ya'an 625014, China.
| | - Yanying Wang
- College of Science, Sichuan Agricultural University, Ya'an, 625000, China.
| | - Gehong Su
- College of Science, Sichuan Agricultural University, Ya'an, 625000, China.
| |
Collapse
|
8
|
Weng J, Guo Y, Gu J, Chen R, Wang X. Inovirus-Encoded Peptides Induce Specific Toxicity in Pseudomonas aeruginosa. Viruses 2025; 17:112. [PMID: 39861901 PMCID: PMC11769263 DOI: 10.3390/v17010112] [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: 12/17/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Pseudomonas aeruginosa is a common opportunistic pathogen associated with nosocomial infections. The primary treatment for infections typically involves antibiotics, which can lead to the emergence of multidrug-resistant strains. Therefore, there is a pressing need for safe and effective alternative methods. Phage therapy stands out as a promising approach. However, filamentous prophages (Pfs) commonly found in P. aeruginosa encode genes with phage defense activity, thereby reducing the efficacy of phage therapy. Through a genomic analysis of the Pf4 prophage, we identified a 102 bp gene co-transcribed with the upstream gene responsible for phage release (zot gene), giving rise to a 33-amino-acid polypeptide that we have named Pf4-encoded toxic polypeptide (PftP4). The overexpression of PftP4 demonstrated cellular toxicity in P. aeruginosa, with subcellular localization indicating its presence in the cell membrane and a subsequent increase in membrane permeability. Notably, PftP4 homologues are found in multiple Pf phages and exhibit specificity in their toxicity towards P. aeruginosa among the tested bacterial strains. Our study reveals that the novel Pf-encoded polypeptide PftP4 has the potential to selectively target and eradicate P. aeruginosa, offering valuable insights for combating P. aeruginosa infections.
Collapse
Affiliation(s)
- Juehua Weng
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 511458, China; (J.W.); (J.G.); (R.C.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yunxue Guo
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 511458, China; (J.W.); (J.G.); (R.C.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiayu Gu
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 511458, China; (J.W.); (J.G.); (R.C.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ran Chen
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 511458, China; (J.W.); (J.G.); (R.C.)
| | - Xiaoxue Wang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 511458, China; (J.W.); (J.G.); (R.C.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Tropical Oceanography, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 511458, China
| |
Collapse
|
9
|
Gonzalez-Prada I, Borges A, Santos-Torres B, Magariños B, Simões M, Concheiro A, Alvarez-Lorenzo C. Antimicrobial cyclodextrin-assisted electrospun fibers loaded with carvacrol, citronellol and cinnamic acid for wound healing. Int J Biol Macromol 2024; 277:134154. [PMID: 39116822 DOI: 10.1016/j.ijbiomac.2024.134154] [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: 05/31/2024] [Revised: 07/14/2024] [Accepted: 07/23/2024] [Indexed: 08/10/2024]
Abstract
This work aimed to explore an alternative to the use of antibiotics for prevention and treatment of wounds infection caused by two common bacterial pathogens Staphylococcus aureus and Pseudomonas aeruginosa. For this purpose, three different essential oil components (EOCs), namely carvacrol, citronellol and cinnamic acid, were loaded into electrospun fibers of poly-ε-caprolactone (PCL) aided by alpha-cyclodextrin (αCD) and hydroxypropyl-β-cyclodextrin (HPβCD). Electrospun-fibers prepared with each EOC and their mixtures were screened for antimicrobial capability and characterized regarding morphological, mechanical, thermal, surface polarity, antibiofilm and antioxidant properties. αCD formed poly(pseudo)rotaxanes with PCL and weakly interacted with EOCs, while HPβCD facilitated EOC encapsulation and formation of homogeneous fibers (500-1000 nm diameter) without beads. PCL/HPβCD fibers with high concentration of EOCs (mainly carvacrol and cinnamic acid) showed strong antibiofilm (>3 log CFU reduction) and antioxidant activity (10-50% DPPH scavenging effects). Different performances were recorded for the EOCs and their mixtures; cinnamic acid migrated to fiber surface and was released faster. Fibers biocompatibility was verified using hemolysis tests and in ovo tissue integration and angiogenesis assays. Overall, HPβCD facilitates complete release of EOCs from the fibers to the aqueous medium, being an environment-friendly and cost-effective strategy for the treatment of infected wounds.
Collapse
Affiliation(s)
- Iago Gonzalez-Prada
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Faculty of Pharmacy, Institute of Materials (iMATUS), and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain
| | - Anabela Borges
- LEPABE - Department of Chemical Engineering, Faculty of Engineering, University of Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Portugal
| | - Beatriz Santos-Torres
- Departamento de Microbiología y Parasitología, Facultad de Biología, CIBUS, Universidade de Santiago de Compostela, Spain
| | - Beatriz Magariños
- Departamento de Microbiología y Parasitología, Facultad de Biología, CIBUS, Universidade de Santiago de Compostela, Spain
| | - Manuel Simões
- LEPABE - Department of Chemical Engineering, Faculty of Engineering, University of Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Portugal
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Faculty of Pharmacy, Institute of Materials (iMATUS), and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Faculty of Pharmacy, Institute of Materials (iMATUS), and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain.
| |
Collapse
|
10
|
Jeong GJ, Khan F, Tabassum N, Jo DM, Jung WK, Kim YM. Roles of Pseudomonas aeruginosa siderophores in interaction with prokaryotic and eukaryotic organisms. Res Microbiol 2024; 175:104211. [PMID: 38734157 DOI: 10.1016/j.resmic.2024.104211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 05/05/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that produces two types of siderophores, pyoverdine and pyochelin, that play pivotal roles in iron scavenging from the environment and host cells. P. aeruginosa siderophores can serve as virulence factors and perform various functions. Several bacterial and fungal species are likely to interact with P. aeruginosa due to its ubiquity in soil and water as well as its potential to cause infections in plants, animals, and humans. Siderophores produced by P. aeruginosa play critical roles in iron scavenging for prokaryotic species (bacteria) and eukaryotic hosts (fungi, animals, insects, invertebrates, and plants) as well. This review provides a comprehensive discussion of the role of P. aeruginosa siderophores in interaction with prokaryotes and eukaryotes as well as their underlying mechanisms of action. The evolutionary relationship between P. aeruginosa siderophore recognition receptors, such as FpvA, FpvB, and FptA, and those of other bacterial species has also been investigated.
Collapse
Affiliation(s)
- Geum-Jae Jeong
- 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
| | - Fazlurrahman Khan
- 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; Institute of Fisheries Science, Pukyong National University. Busan 48513, Republic of Korea; International Graduate Program of Fisheries Science, 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
| | - Du-Min Jo
- National Marine Biodiversity Institute of Korea, Seochun, Chungcheongnam-do, 33662, Republic of Korea
| | - Won-Kyo Jung
- 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; Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence and New-senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan, 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
|
11
|
Niazy AA, Lambarte RNA, Sumague TS, Vigilla MGB, Bin Shwish NM, Kamalan R, Daeab EK, Aljehani NM. FTY720 Reduces the Biomass of Biofilms in Pseudomonas aeruginosa in a Dose-Dependent Manner. Antibiotics (Basel) 2024; 13:621. [PMID: 39061303 PMCID: PMC11273553 DOI: 10.3390/antibiotics13070621] [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: 04/29/2024] [Revised: 06/11/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Pseudomonas aeruginosa, a nosocomial pathogen, has strong biofilm capabilities, representing the main source of infection in the human body. Repurposing existing drugs has been explored as an alternative strategy to combat emerging antibiotic-resistant pathogens. Fingolimod hydrochloride (FTY720), an immunomodulatory drug for multiple sclerosis, has shown promising antimicrobial effects against some ESKAPE pathogens. Therefore, the effects of FTY720 on the biofilm capabilities of Pseudomonas aeruginosa were investigated in this study. It was determined that FTY720 inhibited the growth of P. aeruginosa PAO1 at 100 µM. The significant reduction in PAO1 cell viability was observed to be dose-dependent. Additional cytotoxicity analysis on human cell lines showed that FTY720 significantly reduced viabilities at sub-inhibitory concentrations of 25-50 µM. Microtiter assays and confocal analysis confirmed reductions in biofilm mass and thickness and the cell survivability ratio in the presence of FTY720. Similarly, virulence production and biofilm-related gene expression (rhlA, rhlB, pilA, pilI, fliC, fliD and algR) were determined. The results demonstrate that pigment production was affected and quantitative real-time PCR analysis showed a variable degree of reduced gene expression in response to FTY720 at 12.5-50 µM. These findings suggest that FTY720 could be repurposed as an alternative antibiofilm agent against Pseudomonas aeruginosa.
Collapse
Affiliation(s)
- Abdurahman A. Niazy
- Department of Oral Medicine and Diagnostic Sciences, College of Dentistry, King Saud University, Riyadh 11545, Saudi Arabia
- Molecular and Cell Biology Laboratory, Prince Naif bin AbdulAziz Health Research Center, College of Dentistry, King Saud University Medical City, King Saud University, Riyadh 11545, Saudi Arabia; (R.N.A.L.); (T.S.S.); (N.M.B.S.)
| | - Rhodanne Nicole A. Lambarte
- Molecular and Cell Biology Laboratory, Prince Naif bin AbdulAziz Health Research Center, College of Dentistry, King Saud University Medical City, King Saud University, Riyadh 11545, Saudi Arabia; (R.N.A.L.); (T.S.S.); (N.M.B.S.)
| | - Terrence S. Sumague
- Molecular and Cell Biology Laboratory, Prince Naif bin AbdulAziz Health Research Center, College of Dentistry, King Saud University Medical City, King Saud University, Riyadh 11545, Saudi Arabia; (R.N.A.L.); (T.S.S.); (N.M.B.S.)
| | - Mary Grace B. Vigilla
- Molecular and Cell Biology Laboratory, Prince Naif bin AbdulAziz Health Research Center, College of Dentistry, King Saud University Medical City, King Saud University, Riyadh 11545, Saudi Arabia; (R.N.A.L.); (T.S.S.); (N.M.B.S.)
| | - Najla M. Bin Shwish
- Molecular and Cell Biology Laboratory, Prince Naif bin AbdulAziz Health Research Center, College of Dentistry, King Saud University Medical City, King Saud University, Riyadh 11545, Saudi Arabia; (R.N.A.L.); (T.S.S.); (N.M.B.S.)
| | - Ranan Kamalan
- Research Center, College of Dentistry, King Saud University, Riyadh 11451, Saudi Arabia
| | - Eid Khulaif Daeab
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| | - Nami M. Aljehani
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| |
Collapse
|
12
|
Talaat R, Abu El-Naga MN, El-Bialy HAA, El-Fouly MZ, Abouzeid MA. Quenching of quorum sensing in multi-drug resistant Pseudomonas aeruginosa: insights on halo-bacterial metabolites and gamma irradiation as channels inhibitors. Ann Clin Microbiol Antimicrob 2024; 23:31. [PMID: 38600513 PMCID: PMC11007959 DOI: 10.1186/s12941-024-00684-5] [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: 10/31/2023] [Accepted: 03/03/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Anti-virulence therapy is a promising strategy to treat multi-drug resistant (MDR) pathogens. Pseudomonas aeruginosa is a potent opportunistic pathogen because of an array of virulence factors that are regulated by quorum sensing systems. METHODS The virulence features of four multi-drug resistant P. aeruginosa strains were investigated upon exposure to the sub-lethal dose of gamma rays (1 kGy), and sub-inhibitory concentrations of bioactive metabolites recovered from local halophilic strains in comparison to control. Then, the gene expression of AHL-mediated quorum sensing systems (las/rhl) was quantitatively determined in treated and untreated groups by real-time PCR. RESULTS The bioactive metabolites recovered from halophilic strains previously isolated from saline ecosystems were identified as Halomonas cupida (Halo-Rt1), H. elongate (Halo-Rt2), Vigibacillus natechei (Halo-Rt3), Sediminibacillus terrae (Halo-Rt4) and H. almeriensis (Halo-Rt5). Results revealed that both gamma irradiation and bioactive metabolites significantly reduced the virulence factors of the tested MDR strains. The bioactive metabolites showed a maximum efficiency for inhibiting biofilm formation and rhamnolipids production whereas the gamma irradiation succeeded in decreasing other virulence factors to lower levels in comparison to control. Quantitative-PCR results showed that AHL-mediated quorum sensing systems (las/rhl) in P. aeruginosa strains were downregulated either by halo-bacterial metabolites or gamma irradiation in all treatments except the upregulation of both lasI internal gene and rhlR intact gene in P. aeruginosa NCR-RT3 and both rhlI internal gene and rhlR intact gene in P. aeruginosa U3 by nearly two folds or more upon exposure to gamma irradiation. The most potent result was observed in the expression of lasI internal gene that was downregulated by more than ninety folds in P. aeruginosa NCR-RT2 after treatment with metabolites of S. terrae (Halo-Rt4). Analyzing metabolites recovered from H. cupida (Halo-Rt1) and H. elongate (Halo-Rt2) using LC-ESI-MS/MS revealed many chemical compounds that have quorum quenching properties including glabrol, 5,8-dimethoxyquinoline-2-carbaldehyde, linoleoyl ethanolamide, agelasine, penigequinolones derivatives, berberine, tetracosanoic acid, and liquidambaric lactone in the former halophile and phloretin, lycoctonine, fucoxanthin, and crassicauline A in the latter one. CONCLUSION QS inhibitors can significantly reduce the pathogenicity of MDR P. aeruginosa strains; and thus can be an effective and successful strategy for treating antibiotic resistant traits.
Collapse
Affiliation(s)
- Reham Talaat
- Radiation Microbiology Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Mohamed N Abu El-Naga
- Radiation Microbiology Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Heba Abd Alla El-Bialy
- Radiation Microbiology Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt.
| | - Mohie Z El-Fouly
- Radiation Microbiology Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Mohamed A Abouzeid
- Microbiology Department, Faculty of Science, Ain Shams University, Cairo, Egypt
- Faculty of Science, Galala University, Suez, Egypt
| |
Collapse
|
13
|
Mancuso G, Trinchera M, Midiri A, Zummo S, Vitale G, Biondo C. Novel Antimicrobial Approaches to Combat Bacterial Biofilms Associated with Urinary Tract Infections. Antibiotics (Basel) 2024; 13:154. [PMID: 38391540 PMCID: PMC10886225 DOI: 10.3390/antibiotics13020154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/24/2024] Open
Abstract
Urinary tract infections (UTIs) are prevalent bacterial infections in both community and healthcare settings. They account for approximately 40% of all bacterial infections and require around 15% of all antibiotic prescriptions. Although antibiotics have traditionally been used to treat UTIs for several decades, the significant increase in antibiotic resistance in recent years has made many previously effective treatments ineffective. Biofilm on medical equipment in healthcare settings creates a reservoir of pathogens that can easily be transmitted to patients. Urinary catheter infections are frequently observed in hospitals and are caused by microbes that form a biofilm after a catheter is inserted into the bladder. Managing infections caused by biofilms is challenging due to the emergence of antibiotic resistance. Biofilms enable pathogens to evade the host's innate immune defences, resulting in long-term persistence. The incidence of sepsis caused by UTIs that have spread to the bloodstream is increasing, and drug-resistant infections may be even more prevalent. While the availability of upcoming tests to identify the bacterial cause of infection and its resistance spectrum is critical, it alone will not solve the problem; innovative treatment approaches are also needed. This review analyses the main characteristics of biofilm formation and drug resistance in recurrent uropathogen-induced UTIs. The importance of innovative and alternative therapies for combatting biofilm-caused UTI is emphasised.
Collapse
Affiliation(s)
- Giuseppe Mancuso
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Marilena Trinchera
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Angelina Midiri
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Sebastiana Zummo
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Giulia Vitale
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| | - Carmelo Biondo
- Department of Human Pathology, University of Messina, 98125 Messina, Italy
| |
Collapse
|
14
|
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: 22] [Impact Index Per Article: 22.0] [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
|
15
|
Flores-Percino D, Osorio-Llanes E, Sepulveda Y, Castellar-López J, Madera RB, Rada WR, Meléndez CM, Mendoza-Torres E. Mechanisms of the Quorum Sensing Systems of Pseudomonas aeruginosa: Host and Bacteria. Curr Med Chem 2024; 31:5755-5767. [PMID: 37605403 DOI: 10.2174/0929867331666230821110440] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/25/2023] [Accepted: 07/25/2023] [Indexed: 08/23/2023]
Abstract
Quorum-sensing is a communication mechanism between bacteria with the ability to activate signaling pathways in the bacterium and in the host cells. Pseudomonas aeruginosa is a pathogen with high clinical relevance due to its vast virulence factors repertory and wide antibiotic resistance mechanisms. Due to this, it has become a pathogen of interest for developing new antimicrobial agents in recent years. P. aeruginosa has three major QS systems that regulate a wide gene range linked with virulence factors, metabolic regulation, and environment adaption. Consequently, inhibiting this communication mechanism would be a strategy to prevent the pathologic progression of the infections caused by this bacterium. In this review, we aim to overview the current studies about the signaling mechanisms of the QS system of P. aeruginosa and its effects on this bacterium and the host.
Collapse
Affiliation(s)
- Diana Flores-Percino
- Department of Medicine, Benemérita Universidad Autónoma de Puebla, Puebla, México
| | - Estefanie Osorio-Llanes
- Department of Microbiology, Faculty of Exact and Natural Sciences, Grupo de Investigación Avanzada en Biomedicina, Universidad Libre Barranquilla, Barranquilla, Colombia
- Department of Medicine, Faculty of Health Sciences, Grupo de Investigación Avanzada en Biomedicina, Universidad Libre Seccional Barranquilla, Barranquilla, Colombia
| | - Yanireth Sepulveda
- Department of Microbiology, Faculty of Exact and Natural Sciences, Grupo de Investigación Avanzada en Biomedicina, Universidad Libre Barranquilla, Barranquilla, Colombia
| | - Jairo Castellar-López
- Department of Microbiology, Faculty of Exact and Natural Sciences, Grupo de Investigación Avanzada en Biomedicina, Universidad Libre Barranquilla, Barranquilla, Colombia
| | - Ricardo Belón Madera
- Department of Medicine, Faculty of Health Sciences, Grupo de Investigación Avanzada en Biomedicina, Universidad Libre Seccional Barranquilla, Barranquilla, Colombia
| | - Wendy Rosales Rada
- Department of Microbiology, Faculty of Exact and Natural Sciences, Grupo de Investigación Avanzada en Biomedicina, Universidad Libre Barranquilla, Barranquilla, Colombia
| | - Carlos Mario Meléndez
- Department of Chemistry, Faculty of Basic Sciencies, Grupo de Investigación en Química Orgánica y Biomédica, Universidad del Atlántico, Barranquilla, Colombia
| | - Evelyn Mendoza-Torres
- Department of Medicine, Faculty of Health Sciences, Grupo de Investigación Avanzada en Biomedicina, Universidad Libre Seccional Barranquilla, Barranquilla, Colombia
| |
Collapse
|
16
|
Giovagnorio F, De Vito A, Madeddu G, Parisi SG, Geremia N. Resistance in Pseudomonas aeruginosa: A Narrative Review of Antibiogram Interpretation and Emerging Treatments. Antibiotics (Basel) 2023; 12:1621. [PMID: 37998823 PMCID: PMC10669487 DOI: 10.3390/antibiotics12111621] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/02/2023] [Accepted: 11/10/2023] [Indexed: 11/25/2023] Open
Abstract
Pseudomonas aeruginosa is a ubiquitous Gram-negative bacterium renowned for its resilience and adaptability across diverse environments, including clinical settings, where it emerges as a formidable pathogen. Notorious for causing nosocomial infections, P. aeruginosa presents a significant challenge due to its intrinsic and acquired resistance mechanisms. This comprehensive review aims to delve into the intricate resistance mechanisms employed by P. aeruginosa and to discern how these mechanisms can be inferred by analyzing sensitivity patterns displayed in antibiograms, emphasizing the complexities encountered in clinical management. Traditional monotherapies are increasingly overshadowed by the emergence of multidrug-resistant strains, necessitating a paradigm shift towards innovative combination therapies and the exploration of novel antibiotics. The review accentuates the critical role of accurate antibiogram interpretation in guiding judicious antibiotic use, optimizing therapeutic outcomes, and mitigating the propagation of antibiotic resistance. Misinterpretations, it cautions, can inadvertently foster resistance, jeopardizing patient health and amplifying global antibiotic resistance challenges. This paper advocates for enhanced clinician proficiency in interpreting antibiograms, facilitating informed and strategic antibiotic deployment, thereby improving patient prognosis and contributing to global antibiotic stewardship efforts.
Collapse
Affiliation(s)
- Federico Giovagnorio
- Department of Molecular Medicine, University of Padua, 35121 Padua, Italy; (F.G.); (S.G.P.)
| | - Andrea De Vito
- Unit of Infectious Diseases, Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy;
| | - Giordano Madeddu
- Unit of Infectious Diseases, Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy;
| | | | - Nicholas Geremia
- Unit of Infectious Diseases, Department of Clinical Medicine, Ospedale “dell’Angelo”, 30174 Venice, Italy
- Unit of Infectious Diseases, Department of Clinical Medicine, Ospedale Civile “S.S. Giovanni e Paolo”, 30122 Venice, Italy
| |
Collapse
|
17
|
Guo Y, Mao Z, Ran F, Sun J, Zhang J, Chai G, Wang J. Nanotechnology-Based Drug Delivery Systems to Control Bacterial-Biofilm-Associated Lung Infections. Pharmaceutics 2023; 15:2582. [PMID: 38004561 PMCID: PMC10674810 DOI: 10.3390/pharmaceutics15112582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/09/2023] [Accepted: 10/17/2023] [Indexed: 11/26/2023] Open
Abstract
Airway mucus dysfunction and impaired immunological defenses are hallmarks of several lung diseases, including asthma, cystic fibrosis, and chronic obstructive pulmonary diseases, and are mostly causative factors in bacterial-biofilm-associated respiratory tract infections. Bacteria residing within the biofilm architecture pose a complex challenge in clinical settings due to their increased tolerance to currently available antibiotics and host immune responses, resulting in chronic infections with high recalcitrance and high rates of morbidity and mortality. To address these unmet clinical needs, potential anti-biofilm therapeutic strategies are being developed to effectively control bacterial biofilm. This review focuses on recent advances in the development and application of nanoparticulate drug delivery systems for the treatment of biofilm-associated respiratory tract infections, especially addressing the respiratory barriers of concern for biofilm accessibility and the various types of nanoparticles used to combat biofilms. Understanding the obstacles facing pulmonary drug delivery to bacterial biofilms and nanoparticle-based approaches to combatting biofilm may encourage researchers to explore promising treatment modalities for bacterial-biofilm-associated chronic lung infections.
Collapse
Affiliation(s)
- Yutong Guo
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zeyuan Mao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Fang Ran
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jihong Sun
- Department of Radiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Jingfeng Zhang
- The Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo 315000, China
| | - Guihong Chai
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510180, China
| |
Collapse
|
18
|
Bhowmik P, Modi B, Roy P, Chowdhury A. Strategies to combat Gram-negative bacterial resistance to conventional antibacterial drugs: a review. Osong Public Health Res Perspect 2023; 14:333-346. [PMID: 37920891 PMCID: PMC10626324 DOI: 10.24171/j.phrp.2022.0323] [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: 12/27/2022] [Revised: 07/11/2023] [Accepted: 08/15/2023] [Indexed: 11/04/2023] Open
Abstract
The emergence of antimicrobial resistance raises the fear of untreatable diseases. Antimicrobial resistance is a multifaceted and dynamic phenomenon that is the cumulative result of different factors. While Gram-positive pathogens, such as methicillin-resistant Staphylococcus aureus and Clostridium difficile, were previously the most concerning issues in the field of public health, Gram-negative pathogens are now of prime importance. The World Health Organization's priority list of pathogens mostly includes multidrug-resistant Gram-negative organisms particularly carbapenem-resistant Enterobacterales, carbapenem-resistant Pseudomonas aeruginosa, and extensively drug-resistant Acinetobacter baumannii. The spread of Gram-negative bacterial resistance is a global issue, involving a variety of mechanisms. Several strategies have been proposed to control resistant Gram-negative bacteria, such as the development of antimicrobial auxiliary agents and research into chemical compounds with new modes of action. Another emerging trend is the development of naturally derived antibacterial compounds that aim for targets novel areas, including engineered bacteriophages, probiotics, metal-based antibacterial agents, odilorhabdins, quorum sensing inhibitors, and microbiome-modifying agents. This review focuses on the current status of alternative treatment regimens against multidrug-resistant Gram-negative bacteria, aiming to provide a snapshot of the situation and some information on the broader context.
Collapse
Affiliation(s)
- Priyanka Bhowmik
- Department of Biological Sciences, School of Life Science & Biotechnology, Adamas University, Kolkata, India
| | - Barkha Modi
- Department of Microbiology, Techno India University, Kolkata, India
| | - Parijat Roy
- Department of Biological Sciences, School of Life Science & Biotechnology, Adamas University, Kolkata, India
| | - Antarika Chowdhury
- Department of Biological Sciences, School of Life Science & Biotechnology, Adamas University, Kolkata, India
| |
Collapse
|
19
|
Thakur M, Khushboo, Kumar Y, Yadav V, Pramanik A, Dubey KK. Understanding resistance acquisition by Pseudomonas aeruginosa and possible pharmacological approaches in palliating its pathogenesis. Biochem Pharmacol 2023; 215:115689. [PMID: 37481132 DOI: 10.1016/j.bcp.2023.115689] [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: 05/06/2023] [Revised: 07/03/2023] [Accepted: 07/12/2023] [Indexed: 07/24/2023]
Abstract
Pseudomonas aeruginosa can utilize various virulence factors necessary for host infection and persistence. These virulence factors include pyocyanin, proteases, exotoxins, 2-heptyl-4-hydroxyquinoline N-oxide (HQNO), phospholipases, and siderophores that enable the bacteria to cause severe infections in immunocompromised individuals. P. aeruginosa falls into the category of nosocomial pathogens that are typically resistant to available antibiotics and therapeutic approaches. P. aeruginosa bio-film formation is a major concern in hospitals because it can cause chronic infection and increase the risk of mortality. Therefore, the development of new strategies to disrupt biofilm formation and improve antibiotic efficacy for the treatment of P. aeruginosa infections is crucial. Anti-biofilm and anti-quorum sensing (QS) activity can be viewed as an anti-virulence approach to control the infectious nature of P. aeruginosa. Inhibition of QS and biofilm formation can be achieved through pharmacological approaches such as phytochemicals and essential oils, which have shown promising results in laboratory studies. A regulatory protein called LasR plays a key role in QS signaling to coordinate gene expression. Designing an antagonist molecule that mimics the natural autoinducer might be the best approach for LasR inhibition. Here we reviewed the mechanism behind antibiotic resistance and alternative approaches to combat the pathogenicity of P. aeruginosa.
Collapse
Affiliation(s)
- Mony Thakur
- Department of Microbiology, Central University of Haryana, Mahendergarh, Haryana 123031, India
| | - Khushboo
- Department of Biotechnology, Central University of Haryana, Mahendergarh, Haryana 123031, India
| | - Yatin Kumar
- Department of Microbiology, Central University of Haryana, Mahendergarh, Haryana 123031, India
| | - Vinod Yadav
- Department of Microbiology, Central University of Haryana, Mahendergarh, Haryana 123031, India
| | - Avijit Pramanik
- Department of Microbiology, Central University of Haryana, Mahendergarh, Haryana 123031, India
| | - Kashyap Kumar Dubey
- Biomanufacturing and Process Development Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi-67, India.
| |
Collapse
|
20
|
Shandil S, Yu TT, Sabir S, Black DS, Kumar N. Synthesis of Novel Quinazolinone Analogues for Quorum Sensing Inhibition. Antibiotics (Basel) 2023; 12:1227. [PMID: 37508323 PMCID: PMC10376653 DOI: 10.3390/antibiotics12071227] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/17/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
As bacteria continue to develop resistance mechanisms against antimicrobials, an alternative method to tackle this global concern must be developed. As the pqs system is the most well-known and responsible for biofilm and pyocyanin production, quinazolinone inhibitors of the pqs system in P. aeruginosa were developed. Molecular docking following a rationalised medicinal chemistry approach was adopted to design these analogues. An analysis of docking data suggested that compound 6b could bind with the key residues in the ligand binding domain of PqsR in a similar fashion to the known antagonist M64. The modification of cyclic groups at the 3-position of the quinazolinone core, the introduction of a halogen at the aromatic core and the modification of the terminal group with aromatic and aliphatic chains were investigated to guide the synthesis of a library of 16 quinazolinone analogues. All quinazolinone analogues were tested in vitro for pqs inhibition, with the most active compounds 6b and 6e being tested for biofilm and growth inhibition in P. aeruginosa (PAO1). Compound 6b displayed the highest pqs inhibitory activity (73.4%, 72.1% and 53.7% at 100, 50 and 25 µM, respectively) with no bacterial growth inhibition. However, compounds 6b and 6e only inhibited biofilm formation by 10% and 5%, respectively.
Collapse
Affiliation(s)
- Sahil Shandil
- School of Chemistry, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Tsz Tin Yu
- School of Chemistry, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Shekh Sabir
- School of Chemistry, The University of New South Wales, Sydney, NSW 2052, Australia
| | - David StC Black
- School of Chemistry, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Naresh Kumar
- School of Chemistry, The University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
21
|
Li L, Xu Z, Cao R, Li J, Wu CJ, Wang Y, Zhu H. Effects of hydroxyl group in cyclo(Pro-Tyr)-like cyclic dipeptides on their anti-QS activity and self-assembly. iScience 2023; 26:107048. [PMID: 37360689 PMCID: PMC10285644 DOI: 10.1016/j.isci.2023.107048] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/08/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023] Open
Abstract
We investigated the influence of hydroxyl groups on the anti-quorum-sensing (anti-QS) and anti-biofilm activity of structurally similar cyclic dipeptides, namely cyclo(L-Pro-L-Tyr), cyclo(L-Hyp-L-Tyr), and cyclo(L-Pro-L-Phe), against Pseudomonas aeruginosa PAO1. Cyclo(L-Pro-L-Phe), lacking hydroxyl groups, displayed higher virulence factor inhibition and cytotoxicity, but showed less inhibitory ability in biofilm formation. Cyclo(L-Pro-L-Tyr) and cyclo(L-Hyp-L-Tyr) suppressed genes in both the las and rhl systems, whereas cyclo(L-Pro-L-Phe) mainly downregulated rhlI and pqsR expression. These cyclic dipeptides interacted with the QS-related protein LasR, with similar binding efficiency to the autoinducer 3OC12-HSL, except for cyclo(L-Pro-L-Phe) which had lower affinity. In addition, the introduction of hydroxyl groups significantly improved the self-assembly ability of these peptides. Both cyclo(L-Pro-L-Tyr) and cyclo(L-Hyp-L-Tyr) formed assembly particles at the highest tested concentration. The findings revealed the structure-function relationship of this kind of cyclic dipeptides and provided basis for our follow-up research in the design and modification of anti-QS compounds.
Collapse
Affiliation(s)
- Li Li
- Fujian-Taiwan Science and Technology Cooperation Base of Biomedical Materials and Tissue Engineering, Engineering Research Center of Industrial Biocatalysis, Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian Provincial Key Laboratory of Polymer Materials, College of Chemistry and Materials Science, Fujian Normal University, Fuzhou 350007, China
| | - Zuxian Xu
- Fujian-Taiwan Science and Technology Cooperation Base of Biomedical Materials and Tissue Engineering, Engineering Research Center of Industrial Biocatalysis, Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian Provincial Key Laboratory of Polymer Materials, College of Chemistry and Materials Science, Fujian Normal University, Fuzhou 350007, China
| | - Ruipin Cao
- Fujian-Taiwan Science and Technology Cooperation Base of Biomedical Materials and Tissue Engineering, Engineering Research Center of Industrial Biocatalysis, Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian Provincial Key Laboratory of Polymer Materials, College of Chemistry and Materials Science, Fujian Normal University, Fuzhou 350007, China
| | - Jiaxin Li
- Fujian-Taiwan Science and Technology Cooperation Base of Biomedical Materials and Tissue Engineering, Engineering Research Center of Industrial Biocatalysis, Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian Provincial Key Laboratory of Polymer Materials, College of Chemistry and Materials Science, Fujian Normal University, Fuzhou 350007, China
| | - Chang-Jer Wu
- Department of Food Science, National Taiwan Ocean University, Keelung 20224, Taiwan
| | - Yinglu Wang
- Fujian-Taiwan Science and Technology Cooperation Base of Biomedical Materials and Tissue Engineering, Engineering Research Center of Industrial Biocatalysis, Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian Provincial Key Laboratory of Polymer Materials, College of Chemistry and Materials Science, Fujian Normal University, Fuzhou 350007, China
| | - Hu Zhu
- Fujian-Taiwan Science and Technology Cooperation Base of Biomedical Materials and Tissue Engineering, Engineering Research Center of Industrial Biocatalysis, Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian Provincial Key Laboratory of Polymer Materials, College of Chemistry and Materials Science, Fujian Normal University, Fuzhou 350007, China
- College of Chemical Engineering and Materials Science, Quanzhou Normal University, Quanzhou 362000, China
| |
Collapse
|
22
|
Zhang Y, Liu X, Wen H, Cheng Z, Zhang Y, Zhang H, Mi Z, Fan X. Anti-Biofilm Enzymes-Assisted Antibiotic Therapy against Burn Wound Infection by Pseudomonas aeruginosa. Antimicrob Agents Chemother 2023; 67:e0030723. [PMID: 37272814 PMCID: PMC10353415 DOI: 10.1128/aac.00307-23] [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: 03/07/2023] [Accepted: 05/10/2023] [Indexed: 06/06/2023] Open
Abstract
Pseudomonas aeruginosa can form biofilms at the site of burn wound, leading to infection and the failure of treatment regimens. The previous in vitro study demonstrated that a combination of the quorum-quenching enzyme AidHA147G and the extracellular matrix hydrolase PslG was effective in inhibiting biofilm and promoting antibiotic synergy. The aim of the present study was to evaluate the efficacy of this combination of enzymes in conjunction with tobramycin in treating burn wound infected with P. aeruginosa. The results showed that this treatment was effective in quorum-quenching and biofilm inhibition on infected wounds. Compared with the tobramycin treatment only, simultaneous treatment with the enzymes and antibiotics significantly reduced the severity of tissue damage, decreased the bacterial load, and reduced the expression of the inflammatory indicators myeloperoxidase (MPO) and malondialdehyde (MDA). Topical application of the enzymes also reduced the bacterial load and inflammation to some extent. These results indicate that the combined-enzyme approach is a potentially effective treatment for P. aeruginosa biofilm infections of burn wounds.
Collapse
Affiliation(s)
- Yixin Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Xiaolong Liu
- University of Science and Technology of China, Hefei, Anhui, China
| | - Huamei Wen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Zhongle Cheng
- The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yanyu Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Haichuan Zhang
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, China
| | - Zhongwen Mi
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Xinjiong Fan
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
23
|
Goel N, Ghosh M, Jain D, Sinha R, Khare SK. Inhibition and eradication of Pseudomonas aeruginosa biofilms by secondary metabolites of Nocardiopsis lucentensis EMB25. RSC Med Chem 2023; 14:745-756. [PMID: 37122537 PMCID: PMC10131674 DOI: 10.1039/d2md00439a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/02/2023] [Indexed: 03/09/2023] Open
Abstract
Millions of people worldwide have been impacted by biofilm-associated disorders, which are impregnable owing to frequent changes in surface antigens and gene expression. Globally, about 11% of nosocomial infections, including cystic fibrosis, chronic wound infections, and post-surgical infections, are caused by Pseudomonas aeruginosa, the most prevalent Gram-negative bacterial species. Moreover, biofilms are highly resistant to the host's immune system, and exhibit increased tolerance to stress factors such as starvation, dehydration, and antimicrobials. Here, we have isolated a rare halophilic actinobacteria, Nocardiopsis lucentensis EMB25, and utilized the secondary metabolites for inhibition and eradication of P. aeruginosa biofilm. For the first time, N. lucentensis EMB25 bacteria was explored to study the anti-effect of secondary metabolites on pre-established biofilm. The secondary metabolites targeted the quorum sensing pathway and were found to bind to LasR and RhlR, as confirmed via molecular docking. Also, the reduction in virulence factors, rhamnolipids and pyocyanin further supported the study as these two are regulated by LasR and RhlR. In addition, the downregulation of various QS system genes lasA, lasB, rhlA, rhlB, and pqsA confirmed that the secondary metabolites act on two main regulators of the quorum sensing pathway, LasR, and RhlR. The findings of this study support the bioprospecting of previously unknown and extreme-condition actinobacteria as a rich source of novel bioactives against infections caused by bacterial biofilms.
Collapse
Affiliation(s)
- Nikky Goel
- Enzyme and Microbial Biochemistry Laboratory, Department of Chemistry, Indian Institute of Technology Delhi Hauz Khas New Delhi-110016 India
| | - Moumita Ghosh
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster 3 Milestone, Faridabad-Gurgaon Expressway Faridabad 121001 India
| | - Deepti Jain
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster 3 Milestone, Faridabad-Gurgaon Expressway Faridabad 121001 India
| | - Rajeshwari Sinha
- Enzyme and Microbial Biochemistry Laboratory, Department of Chemistry, Indian Institute of Technology Delhi Hauz Khas New Delhi-110016 India
| | - Sunil Kumar Khare
- Enzyme and Microbial Biochemistry Laboratory, Department of Chemistry, Indian Institute of Technology Delhi Hauz Khas New Delhi-110016 India
| |
Collapse
|
24
|
Zhao A, Sun J, Liu Y. Understanding bacterial biofilms: From definition to treatment strategies. Front Cell Infect Microbiol 2023; 13:1137947. [PMID: 37091673 PMCID: PMC10117668 DOI: 10.3389/fcimb.2023.1137947] [Citation(s) in RCA: 117] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/09/2023] [Indexed: 04/08/2023] Open
Abstract
Bacterial biofilms are complex microbial communities encased in extracellular polymeric substances. Their formation is a multi-step process. Biofilms are a significant problem in treating bacterial infections and are one of the main reasons for the persistence of infections. They can exhibit increased resistance to classical antibiotics and cause disease through device-related and non-device (tissue) -associated infections, posing a severe threat to global health issues. Therefore, early detection and search for new and alternative treatments are essential for treating and suppressing biofilm-associated infections. In this paper, we systematically reviewed the formation of bacterial biofilms, associated infections, detection methods, and potential treatment strategies, aiming to provide researchers with the latest progress in the detection and treatment of bacterial biofilms.
Collapse
Affiliation(s)
- Ailing Zhao
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Jiazheng Sun
- Department of Vasculocardiology, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yipin Liu
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
- *Correspondence: Yipin Liu,
| |
Collapse
|
25
|
Soukarieh F, Gurnani P, Romero M, Halliday N, Stocks M, Alexander C, Cámara M. Design of Quorum Sensing Inhibitor-Polymer Conjugates to Penetrate Pseudomonas aeruginosa Biofilms. ACS Macro Lett 2023; 12:314-319. [PMID: 36790191 PMCID: PMC10035027 DOI: 10.1021/acsmacrolett.2c00699] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/01/2023] [Indexed: 02/16/2023]
Abstract
Antimicrobial resistance (AMR) is a global threat to public health with a forecast of a negative financial impact of one trillion dollars per annum, hence novel therapeutics are urgently needed. The resistance of many bacteria against current drugs is further augmented by the ability of these microbes to form biofilms where cells are encased in a slimy extracellular matrix and either adhered to a surface or forming cell aggregates. Biofilms form physiochemical barriers against the penetration of treatments such as small molecule antibacterials, rendering most treatments ineffective. Pseudomonas aeruginosa, a priority pathogen of immediate concern, controls biofilm formation through multiple layers of gene regulation pathways including quorum sensing (QS), a cell-to-cell signaling system. We have recently reported a series of inhibitors of the PqsR QS regulator from this organism that can potentiate the action of antibiotics. However, these QS inhibitors (QSIs) have shown modest effects on biofilms in contrast with planktonic cultures due to poor penetration through the biofilm matrix. To enhance the delivery of the inhibitors, a small library of polymers was designed as carriers of a specific QSI, with variations in the side chains to introduce either positively charged or neutral moieties to aid penetration into and through the P. aeruginosa biofilm. The synthesized polymers were evaluated in a series of assays to establish their effects on the inhibition of the Pqs QS system in P. aeruginosa, the levels of inhibitor release from polymers, and their impact on biofilm formation. A selected cationic polymer-QSI conjugate was found to penetrate effectively through biofilm layers and to release the QSI. When used in combination with ciprofloxacin, it enhanced the biofilm antimicrobial activity of this antibiotic compared to free QSI and ciprofloxacin under the same conditions.
Collapse
Affiliation(s)
- Fadi Soukarieh
- National
Biofilms Innovation Centre, Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2RD, United
Kingdom
- School
of Life Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2RD, United
Kingdom
| | - Pratik Gurnani
- Division
of Molecular Therapeutics and Formulation, Boots Science Building,
School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
| | - Manuel Romero
- National
Biofilms Innovation Centre, Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2RD, United
Kingdom
- Department
of Microbiology and Parasitology, Faculty of Biology-CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Nigel Halliday
- School
of Life Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2RD, United
Kingdom
| | - Michael Stocks
- School
of Pharmacy, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
| | - Cameron Alexander
- Division
of Molecular Therapeutics and Formulation, Boots Science Building,
School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
| | - Miguel Cámara
- National
Biofilms Innovation Centre, Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2RD, United
Kingdom
- School
of Life Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2RD, United
Kingdom
| |
Collapse
|
26
|
Fernandes S, Borges A, Gomes IB, Sousa SF, Simões M. Curcumin and 10-undecenoic acid as natural quorum sensing inhibitors of LuxS/AI-2 of Bacillus subtilis and LasI/LasR of Pseudomonas aeruginosa. Food Res Int 2023; 165:112519. [PMID: 36869520 DOI: 10.1016/j.foodres.2023.112519] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/06/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023]
Abstract
The quorum sensing (QS) system is related to cell-to-cell communication as a function of population density, which regulates several physiological functions including biofilm formation and virulence gene expression. QS inhibitors have emerged as a promising strategy to tackle virulence and biofilm development. Among a wide variety of phytochemicals, many of them have been described as QS inhibitors. Driven by their promising clues, this study aimed to identify active phytochemicals against LuxS/autoinducer-2 (AI-2) (as the universal QS system) from Bacillus subtilis and LasI/LasR (as a specific QS system) of Pseudomonas aeruginosa, through in silico analysis followed by in vitro validation. The optimized virtual screening protocols were applied to screen a phytochemical database containing 3479 drug-like compounds. The most promising phytochemicals were curcumin, pioglitazone hydrochloride, and 10-undecenoic acid. In vitro analysis corroborated the QS inhibitory activity of curcumin and 10-undecenoic acid, however, pioglitazone hydrochloride showed no relevant effect. Inhibitory effects on LuxS/AI-2 QS system triggered reduction of 33-77% by curcumin (at 1.25-5 µg/mL) and 36-64% by 10-undecenoic acid (at 12.5-50 µg/mL). Inhibition of LasI/LasR QS system was 21% by curcumin (at 200 µg/mL) and 10-54% by 10-undecenoic acid (at 15.625-250 µg/mL). In conclusion, in silico analysis allowed the identification of curcumin and, for the first time, 10-undecenoic acid (showing low cost, high availability, and low toxicity) as alternatives to counteract bacterial pathogenicity and virulence, avoiding the imposition of selective pressure usually related to classic industrial disinfection and antibiotics therapy.
Collapse
Affiliation(s)
- Susana Fernandes
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Anabela Borges
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Inês B Gomes
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Sérgio F Sousa
- UCIBIO/REQUIMTE, BioSIM, Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Manuel Simões
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal.
| |
Collapse
|
27
|
Dua T, Mangal S, Akshita G, Harshdeep, Atri AK, Sharma P, Harjai K, Singh V. Novel Vanillin-based hybrids inhibit quorum sensing and silences phenotypical expressions in Pseudomonas aeruginosa. Drug Dev Res 2023; 84:45-61. [PMID: 36419404 DOI: 10.1002/ddr.22011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/23/2022] [Accepted: 10/31/2022] [Indexed: 11/25/2022]
Abstract
In this study, we report the chemical synthesis, computational analysis, and anti-virulent studies of five Vanillin-based hybrids employing phytochemicals. Vanillin (V) is known to have substantial anti-quorum sensing activity against the gram-negative pathogen Pseudomonas aeruginosa. Therefore, with the aim to further enhance the potency of Vanillin, it was chemically conjugated via a triazole (T) linker with five phytochemicals- Zingerone (Z), Eugenol (E), Guaiacol (G), Cinnamaldehyde (C), and Ferulic acid (F) to form the hybrids named as VTZ (1), VTE (2), VTG (3), VTC (4), and VTF (5), respectively. Molecular docking studies revealed the strong binding affinity of the designed hybrids with quorum-sensing receptors (LasR, Rh1R, and PqsR). The synthesized hybrids were also evaluated for anti-quorum sensing activities to examine the efficacy against P. aeruginosa bacterial strains PAO1. The hybrids VTE (2), VTG (3), and VTC (4) displayed improved anti-quorum activity relative to Vanillin. Furthermore, the attenuation of virulence factors of P. aeruginosa (Las-A protease, Las-B elastase, pyocyanin pigmentation, and motility) in the presence of VTE (2), VTG (3), and VTC (4) further authenticated the anti-virulent activity of the hybrids. The new design strategy of the phytochemical-phytochemical scaffolds and their biological evaluation provides a proof of concept for the simultaneous perturbation of well-established anti-virulent targets. This appears to be highly promising and effective strategy to ameliorate the enigma of antimicrobial resistance.
Collapse
Affiliation(s)
- Tamanna Dua
- Department of Applied Sciences, Punjab Engineering College (Deemed to be University), Chandigarh, India
| | - Surabhi Mangal
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Goel Akshita
- Department of Chemistry, Panjab University, Chandigarh, India
| | - Harshdeep
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Ankit K Atri
- Department of Applied Sciences, Punjab Engineering College (Deemed to be University), Chandigarh, India
| | | | - Kusum Harjai
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Vasundhara Singh
- Department of Applied Sciences, Punjab Engineering College (Deemed to be University), Chandigarh, India
| |
Collapse
|
28
|
Identification of 3-nitrophenol ester derivatives as novel quorum-sensing inhibitors of Pseudomonas aeruginosa. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2022.134284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
29
|
Kushwaha A, Kumar V, Agarwal V. Pseudomonas quinolone signal induces organelle stress and dysregulates inflammation in human macrophages. Biochim Biophys Acta Gen Subj 2023; 1867:130269. [PMID: 36379399 DOI: 10.1016/j.bbagen.2022.130269] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/27/2022] [Accepted: 11/02/2022] [Indexed: 11/15/2022]
Abstract
Pseudomonas quinolone signal (PQS) is a quorum-sensing molecule associated with Pseudomonas aeruginosa that regulates quorum sensing, extracellular vesicle biogenesis, iron acquisition, and the secretion of virulence factors. PQS has been shown to have immunomodulatory effects on the host. It induces oxidative stress, modulates cytokine levels, and activates regulated cell death in the host. In this study, we investigated the effects of PQS (10 μM) on host organelle dynamics and dysfunction in human macrophages at the interphase of endoplasmic reticulum (ER), mitochondria, and lysosome. This study showed that PQS increases cytosolic Ca+2 levels and elevates ER stress, as evidenced by increased expression of BiP and activation of the PERK-CHOP axis of unfolded protein response (UPR). Moreover, PQS also negatively affects mitochondria by disrupting mitochondrial membrane potential and increasing mitochondrial ROS generation (mROS). Additionally, PQS stimulation decreased the number of acridine orange-positive lysosomes, indicating lysosomal destabilization. Furthermore, PQS-induced lysosomal destabilization also induces overexpression of the lysosomal stress-responsive gene TFEB. Besides organelle dysfunction, PQS dysregulates inflammation-related genes by upregulating NLRC4, TMS1, and Caspase 1 while downregulating NLRP3 and IL-1β. Also, PQS increases gene expression of pro-inflammatory cytokines (IL-6, TNF-α, and IFN-γ). In conclusion, our findings suggest that PQS negatively affects human macrophages by interfering with organelle function and dysregulating inflammatory response. Consequently, this study provides crucial insight into PQS-driven macrophage dysfunction and may contribute to a better understanding of Pseudomonas aeruginosa-associated infections.
Collapse
Affiliation(s)
- Ankit Kushwaha
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, Uttar Pradesh 211004, India
| | - Vivek Kumar
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, Uttar Pradesh 211004, India
| | - Vishnu Agarwal
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, Uttar Pradesh 211004, India.
| |
Collapse
|
30
|
Targeting Multidrug-Recalcitrant Pseudomonas aeruginosa Biofilms: Combined-Enzyme Treatment Enhances Antibiotic Efficacy. Antimicrob Agents Chemother 2023; 67:e0135822. [PMID: 36602373 PMCID: PMC9872604 DOI: 10.1128/aac.01358-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that forms biofilms during infection, resulting in recalcitrance to antibiotic treatment. Biofilm inhibition is a promising research direction for the treatment of biofilm-associated infections. Here, a combined-enzyme biofilm-targeted strategy was put forward for the first time to simultaneously prevent biofilm formation and break down preformed biofilms. The N-acylhomoserine lactonase AidH was used as a quorum-sensing inhibitor and was modified to enhance the inhibitory effect on biofilms by rational design. Mutant AidHA147G exerted maximum activity at the human body temperature and pH and could reduce the expression of virulence factors as well as biofilm-related genes of P. aeruginosa. Subsequently, the P. aeruginosa self-produced glycosyl hydrolase PslG joined with AidHA147G to disrupt biofilms. Interestingly, under the combined-enzyme intervention for P. aeruginosa wild-type strain PAO1 and clinical strains, no biofilm was observed on the bottom of NEST glass-bottom cell culture dishes. The combination strategy also helped multidrug-resistant clinical strains change from resistant to intermediate or sensitive to many antibiotics commonly used in clinical practice. These results demonstrated that the combined-enzyme approach for inhibiting biofilms is a potential clinical treatment for P. aeruginosa infection.
Collapse
|
31
|
Gupta MK, Singh R, Rangan L. Phytochemical screening, antibacterial, anti-biofilm and quorum sensing inhibiting activity of Alpinia nigra leaf extract against infectious pathogen Pseudomonas aeruginosa PAO1. Food Control 2023. [DOI: 10.1016/j.foodcont.2022.109327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
32
|
Novel quinazolinone disulfide analogues as pqs quorum sensing inhibitors against Pseudomonas aeruginosa. Bioorg Chem 2023; 130:106226. [DOI: 10.1016/j.bioorg.2022.106226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/09/2022] [Accepted: 10/21/2022] [Indexed: 11/18/2022]
|
33
|
GC-MS Analysis and Microbiological Evaluation of Caraway Essential Oil as a Virulence Attenuating Agent against Pseudomonas aeruginosa. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238532. [PMID: 36500623 PMCID: PMC9741284 DOI: 10.3390/molecules27238532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/27/2022] [Accepted: 11/30/2022] [Indexed: 12/09/2022]
Abstract
The emergence of resistant microbes threatens public health on our planet, and the emergence of resistant bacteria against the most commonly used antibiotics necessitates urgent alternative therapeutic options. One way to fight resistant microbes is to design new antimicrobial agents, however, this approach takes decades of research. An alternative or parallel approach is to target the virulence of bacteria with natural or synthetic agents. Active constituents from medicinal plants represent a wide library to screen for natural anti-virulence agents. Caraway is used as a traditional spice and in some medicinal applications such as carminative, antispasmodic, appetizer, and expectorant. Caraway essential oil is rich in terpenes that were previously reported to have antimicrobial activities. In our study, we tested the caraway essential oil in sub-inhibitory concentration as a virulence agent against the Gram-negative bacteria Pseudomonas aeruginosa. Caraway essential oil in sub-inhibitory concentration dramatically blocked protease activity, pyocyanin production, biofilm formation, and quorum sensing activity of P. aeruginosa. The gas chromatography-mass spectroscopy (GC-MS) profile of caraway fruit oil identified 13 compounds representing 85.4% of the total oil components with carvone and sylvestrene as the main constituents. In conclusion, caraway essential oil is a promising virulence-attenuating agent that can be used against topical infections caused by P. aeruginosa.
Collapse
|
34
|
Shi X, Zarkan A. Bacterial survivors: evaluating the mechanisms of antibiotic persistence. MICROBIOLOGY (READING, ENGLAND) 2022; 168. [PMID: 36748698 DOI: 10.1099/mic.0.001266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bacteria withstand antibiotic onslaughts by employing a variety of strategies, one of which is persistence. Persistence occurs in a bacterial population where a subpopulation of cells (persisters) survives antibiotic treatment and can regrow in a drug-free environment. Persisters may cause the recalcitrance of infectious diseases and can be a stepping stone to antibiotic resistance, so understanding persistence mechanisms is critical for therapeutic applications. However, current understanding of persistence is pervaded by paradoxes that stymie research progress, and many aspects of this cellular state remain elusive. In this review, we summarize the putative persister mechanisms, including toxin-antitoxin modules, quorum sensing, indole signalling and epigenetics, as well as the reasons behind the inconsistent body of evidence. We highlight present limitations in the field and underscore a clinical context that is frequently neglected, in the hope of supporting future researchers in examining clinically important persister mechanisms.
Collapse
Affiliation(s)
- Xiaoyi Shi
- Cambridge Centre for International Research, Cambridge CB4 0PZ, UK
| | - Ashraf Zarkan
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| |
Collapse
|
35
|
Chen W, Zhang P, Guo T, Gu X, Bai B, zhang S, Chang X, Wang Y, Ma S. Design, synthesis and evaluation of oxazolopyridinone derivatives as quorum sensing inhibitors. Bioorg Chem 2022; 130:106266. [DOI: 10.1016/j.bioorg.2022.106266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/30/2022] [Accepted: 11/06/2022] [Indexed: 11/13/2022]
|
36
|
Camberlein V, Jézéquel G, Haupenthal J, Hirsch AKH. The Structures and Binding Modes of Small-Molecule Inhibitors of Pseudomonas aeruginosa Elastase LasB. Antibiotics (Basel) 2022; 11:1060. [PMID: 36009930 PMCID: PMC9404851 DOI: 10.3390/antibiotics11081060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/25/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022] Open
Abstract
Elastase B (LasB) is a zinc metalloprotease and a crucial virulence factor of Pseudomonas aeruginosa. As the need for new strategies to fight antimicrobial resistance (AMR) constantly rises, this protein has become a key target in the development of novel antivirulence agents. The extensive knowledge of the structure of its active site, containing two subpockets and a zinc atom, led to various structure-based medicinal chemistry programs and the optimization of several chemical classes of inhibitors. This review provides a brief reminder of the structure of the active site and a summary of the disclosed P. aeruginosa LasB inhibitors. We specifically focused on the analysis of their binding modes with a detailed representation of them, hence giving an overview of the strategies aiming at targeting LasB by small molecules.
Collapse
Affiliation(s)
- Virgyl Camberlein
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)—Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Gwenaëlle Jézéquel
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)—Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Jörg Haupenthal
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)—Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
| | - Anna K. H. Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)—Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
| |
Collapse
|
37
|
Beenker WAG, Hoeksma J, den Hertog J. Gregatins, a Group of Related Fungal Secondary Metabolites, Inhibit Aspects of Quorum Sensing in Gram-Negative Bacteria. Front Microbiol 2022; 13:934235. [PMID: 35865924 PMCID: PMC9296082 DOI: 10.3389/fmicb.2022.934235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/02/2022] [Indexed: 12/11/2022] Open
Abstract
Quorum sensing (QS) is a process that regulates gene expression based on cell density. In bacteria, QS facilitates collaboration and controls a large number of pathways, including biofilm formation and virulence factor production, which lead to lower sensitivity to antibiotics and higher toxicity in the host, respectively. Inhibition of QS is a promising strategy to combat bacterial infections. In this study, we tested the potential of secondary metabolites from fungi to inhibit bacterial QS using a library derived from more than ten thousand different fungal strains. We used the reporter bacterium, Chromobacterium violaceum, and identified 39 fungal strains that produced QS inhibitor activity. These strains expressed two QS inhibitors that had been described before and eight QS inhibitors that had not been described before. Further testing for QS inhibitor activity against the opportunistic pathogen Pseudomonas aeruginosa led to the identification of gregatins as an interesting family of compounds with QS inhibitor activity. Although various gregatins inhibited QS in P. aeruginosa, these gregatins did not inhibit virulence factor production and biofilm formation. We conclude that gregatins inhibit some, but not all aspects of QS.
Collapse
Affiliation(s)
- Wouter A. G. Beenker
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Jelmer Hoeksma
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Jeroen den Hertog
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
- Institute Biology Leiden, Leiden University, Leiden, Netherlands
- *Correspondence: Jeroen den Hertog
| |
Collapse
|
38
|
Recent Advance in Small Molecules Targeting RhlR of Pseudomonas aeruginosa. Antibiotics (Basel) 2022; 11:antibiotics11020274. [PMID: 35203876 PMCID: PMC8868144 DOI: 10.3390/antibiotics11020274] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 12/02/2022] Open
Abstract
Pseudomonas aeruginosa (P. aeruginosa) is an opportunistic gram-negative pathogen that can cause various infections, particularly in patients with compromised host defenses. P. aeruginosa forms biofilms and produces virulence factors through quorum sensing (QS) network, resulting in resistance to antibiotics. RhlI/RhlR, one of key QS systems in P. aeruginosa, is considered an attractive target for inhibiting biofilm formation and attenuating virulence factors. Several recent studies examined small molecules targeting the RhlI/RhlR system and their in vitro and in vivo biological activities. In this review, RhlR-targeted modulators, including agonists and antagonists, are discussed with particular focus on structure-activity relationship studies and outlook for next-generation anti-biofilm agents.
Collapse
|