51
|
Morgaan HA, Omar HMG, Zakaria AS, Mohamed NM. Repurposing carvacrol, cinnamaldehyde, and eugenol as potential anti-quorum sensing agents against uropathogenic Escherichia coli isolates in Alexandria, Egypt. BMC Microbiol 2023; 23:300. [PMID: 37872476 PMCID: PMC10591344 DOI: 10.1186/s12866-023-03055-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 10/11/2023] [Indexed: 10/25/2023] Open
Abstract
BACKGROUND Urinary tract infections represent one of the most frequent hospital and community-acquired infections with uropathogenic Escherichia coli (UPEC) being the main causative agent. The global increase in the emergence of multidrug-resistant (MDR) UPEC necessitates exploring novel approaches. Repurposing natural products as anti-quorum sensing (QS) agents to impede bacterial virulence is gaining momentum nowadays. Hence, this study investigates the anti-QS potentials of carvacrol, cinnamaldehyde, and eugenol against E. coli isolated from urine cultures of Egyptian patients. RESULTS Antibiotic susceptibility testing was performed for 67 E. coli isolates and 94% of the isolates showed MDR phenotype. The usp gene was detected using PCR and accordingly, 45% of the isolates were categorized as UPEC. Phytochemicals, at their sub-inhibitory concentrations, inhibited the swimming and twitching motilities of UPEC isolates, with eugenol showing the highest inhibitory effect. The agents hindered the biofilm-forming ability of the tested isolates, at two temperature sets, 37 and 30 °C, where eugenol succeeded in significantly inhibiting the biofilm formation by > 50% at both investigated temperatures, as compared with untreated controls. The phytochemicals were shown to downregulate the expression of the QS gene (luxS) and critical genes related to motility, asserting their anti-QS potential. Further, the combinatory activity of the phytoproducts with five antibiotics was assessed by checkerboard assay. The addition of the phytoproducts significantly reduced the minimum inhibitory concentrations of the antibiotics and generated several synergistic or partially synergistic combinations, some of which have not been previously explored. CONCLUSIONS Overall, carvacrol, cinnamaldehyde, and eugenol could be repurposed as potential anti-QS agents, which preferentially reduce the QS-based communication and attenuate the cascades of gene expression, thus decreasing the production of virulence factors in UPEC, and eventually, subsiding their pathogenicity. Furthermore, the synergistic combinations of these agents with antibiotics might provide a new perspective to circumvent the side effects brought about by high antibiotic doses, thereby paving the way for overcoming antibiotic resistance.
Collapse
Affiliation(s)
- Hadeer A Morgaan
- Microbiology and Immunology Department, Faculty of Pharmacy, Alexandria University, El-Khartoom Square, Azarita, Alexandria, Egypt
| | - Hoda M G Omar
- Microbiology and Immunology Department, Faculty of Pharmacy, Alexandria University, El-Khartoom Square, Azarita, Alexandria, Egypt
| | - Azza S Zakaria
- Microbiology and Immunology Department, Faculty of Pharmacy, Alexandria University, El-Khartoom Square, Azarita, Alexandria, Egypt
| | - Nelly M Mohamed
- Microbiology and Immunology Department, Faculty of Pharmacy, Alexandria University, El-Khartoom Square, Azarita, Alexandria, Egypt.
| |
Collapse
|
52
|
Schroven K, Putzeys L, Swinnen AL, Hendrix H, Paeshuyse J, Lavigne R. The phage-encoded protein PIT2 impacts Pseudomonas aeruginosa quorum sensing by direct interaction with LasR. iScience 2023; 26:107745. [PMID: 37736037 PMCID: PMC10509696 DOI: 10.1016/j.isci.2023.107745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/07/2023] [Accepted: 08/24/2023] [Indexed: 09/23/2023] Open
Abstract
In recent decades, there has been a notable increase in antibiotic-resistant Pseudomonas aeruginosa isolates, necessitating the development of innovative treatments to combat this pathogen. This manuscript explores the potential of different phage proteins to attenuate virulence factors of P. aeruginosa, particularly the type II secretion system (T2SS). PIT2, a protein derived from the lytic Pseudomonas phage LMA2 inhibits the T2SS effectors PrpL and LasA and attenuates the bacterial virulence toward HeLa cells and Galleria mellonella. Using RNAseq-based differential gene expression analysis, PIT2's impact on the LasR regulatory network is revealed, which plays a key role in bacterial quorum sensing. This discovery expands our knowledge on phage-encoded modulators of the bacterial metabolism and offers a promising anti-virulence target in P. aeruginosa. As such, it lays the foundation for a new phage-inspired anti-virulence strategy to combat multidrug resistant pathogens and opens the door for SynBio applications.
Collapse
Affiliation(s)
- Kaat Schroven
- Laboratory of Gene Technology, KU Leuven, 3000 Heverlee, Belgium
| | - Leena Putzeys
- Laboratory of Gene Technology, KU Leuven, 3000 Heverlee, Belgium
| | | | - Hanne Hendrix
- Laboratory of Gene Technology, KU Leuven, 3000 Heverlee, Belgium
| | - Jan Paeshuyse
- Laboratory for Host Pathogen Interactions in Livestock, KU Leuven, 3000 Heverlee, Belgium
| | - Rob Lavigne
- Laboratory of Gene Technology, KU Leuven, 3000 Heverlee, Belgium
| |
Collapse
|
53
|
Buder C, Meemken D, Fürstenberg R, Langforth S, Kirse A, Langkabel N. Drinking Pipes and Nipple Drinkers in Pig Abattoir Lairage Pens-A Source of Zoonotic Pathogens as a Hazard to Meat Safety. Microorganisms 2023; 11:2554. [PMID: 37894212 PMCID: PMC10609512 DOI: 10.3390/microorganisms11102554] [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: 09/29/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
The water distribution system in the lairage pens of abattoirs could act as a route of contamination for produced meat. In this study, biofilm formation and the occurrence of specific pathogens in drinking equipment was investigated in different lairage pens in a German commercial pig abattoir. Samples of the water and the drinkers in different locations were microbiologically cultivated and examined. After new drinking equipment had been installed for one month, three months and five years, biofilm formation was detectable, and retrograde growth from the nipple drinkers was seen up to the connection with the main water distribution system. In particular, Enterobacteriaceae and Pseudomonas spp. were found in all samplings of the nipple drinkers. Zoonotic pathogens, Salmonella, pathogenic Yersinia enterocolitica and methicillin-resistant Staphylococcus aureus, were also isolated from the nipple drinkers, while Listeria monocytogenes was not detected via microbial cultivation methods in any of the samples. Since the pigs take the contaminated nipple drinkers into their mouths to drink, or drink contaminated water containing the pathogens, transmission and even infection of the pigs in the lairage can be assumed. This could consequently lead to contamination or cross-contamination of the meat during slaughter and processing and to a public health risk.
Collapse
Affiliation(s)
- Celine Buder
- Working Group Meat Hygiene, Institute of Food Safety and Food Hygiene, School of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany; (C.B.); (R.F.); (S.L.); (N.L.)
- Veterinary Centre for Resistance Research, School of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany
| | - Diana Meemken
- Working Group Meat Hygiene, Institute of Food Safety and Food Hygiene, School of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany; (C.B.); (R.F.); (S.L.); (N.L.)
- Veterinary Centre for Resistance Research, School of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany
| | - Roland Fürstenberg
- Working Group Meat Hygiene, Institute of Food Safety and Food Hygiene, School of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany; (C.B.); (R.F.); (S.L.); (N.L.)
- Veterinary Centre for Resistance Research, School of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany
| | - Susann Langforth
- Working Group Meat Hygiene, Institute of Food Safety and Food Hygiene, School of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany; (C.B.); (R.F.); (S.L.); (N.L.)
- Veterinary Centre for Resistance Research, School of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany
| | - Alina Kirse
- Department of Biometry, Epidemiology and Information Processing, WHO Collaborating Centre for Research and Training for Health at the Human-Animal-Environment Interface, University of Veterinary Medicine Hannover, 30559 Hannover, Germany;
| | - Nina Langkabel
- Working Group Meat Hygiene, Institute of Food Safety and Food Hygiene, School of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany; (C.B.); (R.F.); (S.L.); (N.L.)
- Veterinary Centre for Resistance Research, School of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany
| |
Collapse
|
54
|
Chadha J, Khullar L, Gulati P, Chhibber S, Harjai K. Anti-virulence prospects of Metformin against Pseudomonas aeruginosa: A new dimension to a multifaceted drug. Microb Pathog 2023; 183:106281. [PMID: 37541553 DOI: 10.1016/j.micpath.2023.106281] [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: 05/02/2023] [Revised: 07/29/2023] [Accepted: 07/31/2023] [Indexed: 08/06/2023]
Abstract
Metformin (MeT) is an FDA-approved drug with a myriad of health benefits. Besides being used as an anti-diabetic drug, MeT is also effective against various cancers, liver-, cardiovascular-, and renal diseases. This study was undertaken to examine its unique potential as an anti-virulence drug against an opportunistic bacterial pathogen, Pseudomonas aeruginosa. Due to the menace of multidrug resistance in pathogenic microorganisms, many novel or repurposed drugs with anti-virulence prospects are emerging as next-generation therapies with the aim to overshadow the application of existing antimicrobial regimens. The quorum sensing (QS) mechanisms of P. aeruginosa are an attractive drug target for attenuating bacterial virulence. In this context, the anti-QS potential of MeT was scrutinized using biosensor assays. MeT was comprehensively evaluated for its effects on different motility phenotypes, virulence factor production (phenotypic and genotypic expression) along with biofilm development in P. aeruginosa in vitro. At sub-lethal concentrations, MeT displayed prolific quorum quenching (QQ) ability and remarkably inhibited AHL biosynthesis in P. aeruginosa. Moreover, MeT (1/8 MIC) effectively downregulated the expression levels of various QS- and virulence genes in P. aeruginosa, which coincided with a notable reduction in the levels of alginate, hemolysin, pyocyanin, pyochelin, elastase, and protease production. In silico analysis through molecular docking also predicted strong associations between MeT and QS receptors of P. aeruginosa. MeT also compromised the motility phenotypes and successfully abrogated biofilm formation by inhibiting EPS production in P. aeruginosa. Hence, MeT may be repurposed as an anti-virulence drug against P. aeruginosa in clinical settings.
Collapse
Affiliation(s)
- Jatin Chadha
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Lavanya Khullar
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Pallavi Gulati
- Department of Microbiology, University of Delhi South Campus, New Delhi, India
| | - Sanjay Chhibber
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Kusum Harjai
- Department of Microbiology, Panjab University, Chandigarh, India.
| |
Collapse
|
55
|
Hajdú G, Somogyvári M, Csermely P, Sőti C. Lysosome-related organelles promote stress and immune responses in C. elegans. Commun Biol 2023; 6:936. [PMID: 37704756 PMCID: PMC10499889 DOI: 10.1038/s42003-023-05246-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 08/15/2023] [Indexed: 09/15/2023] Open
Abstract
Lysosome-related organelles (LROs) play diverse roles and their dysfunction causes immunodeficiency. However, their primordial functions remain unclear. Here, we report that C. elegans LROs (gut granules) promote organismal defenses against various stresses. We find that toxic benzaldehyde exposure induces LRO autofluorescence, stimulates the expression of LRO-specific genes and enhances LRO transport capacity as well as increases tolerance to benzaldehyde, heat and oxidative stresses, while these responses are impaired in glo-1/Rab32 and pgp-2 ABC transporter LRO biogenesis mutants. Benzaldehyde upregulates glo-1- and pgp-2-dependent expression of heat shock, detoxification and antimicrobial effector genes, which requires daf-16/FOXO and/or pmk-1/p38MAPK. Finally, benzaldehyde preconditioning increases resistance against Pseudomonas aeruginosa PA14 in a glo-1- and pgp-2-dependent manner, and PA14 infection leads to the deposition of fluorescent metabolites in LROs and induction of LRO genes. Our study suggests that LROs may play a role in systemic responses to stresses and in pathogen resistance.
Collapse
Affiliation(s)
- Gábor Hajdú
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Milán Somogyvári
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Péter Csermely
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Csaba Sőti
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
56
|
Suresh S, Saldanha J, Bhaskar Shetty A, Premanath R, Akhila DS, Mohan Raj JR. Comparison of Antibiofilm Activity of Pseudomonas aeruginosa Phages on Isolates from Wounds of Diabetic and Non-Diabetic Patients. Microorganisms 2023; 11:2230. [PMID: 37764074 PMCID: PMC10536433 DOI: 10.3390/microorganisms11092230] [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: 06/19/2023] [Revised: 08/03/2023] [Accepted: 08/15/2023] [Indexed: 09/29/2023] Open
Abstract
The persistence of organisms as biofilms and the increase in antimicrobial resistance has raised the need for alternative strategies. The study objective was to compare the ability of isolated bacteriophages to remove in vitro biofilms formed by Pseudomonas aeruginosa isolated from the environment with those isolated from diabetic and non-diabetic wounds. P. aeruginosa were isolated from clinical and environmental sites, and antimicrobial susceptibility was tested. Bacteriophages were isolated and characterized based on plaque morphology and host range. A reduction in the viable count assayed the lytic ability of candidate phages. The crystal violet method was used to determine the residual biofilm after 24 h of phage treatment on 72-h-old biofilms. The statistical significance of phage treatment was tested by one-way ANOVA. Of 35 clinical isolates, 17 showed resistance to 1 antibiotic at least, and 7 were multidrug resistant. Nineteen environmental isolates and 11 clinical isolates were drug-sensitive. Nine phages showed 91.2% host coverage, including multidrug-resistant isolates. Phages eradicated 85% of biofilms formed by environmental isolates compared to 58% of biofilms of diabetic isolates and 56% of biofilms of non-diabetic isolates. Clinical isolates are susceptible to phage infection in planktonic form. Biofilms of P. aeruginosa isolated from diabetic wounds and non-diabetic wounds resist removal by phages compared to biofilms formed by environmental isolates. All phages were efficient in dispersing PAO1 biofilms. However, there was a significant difference in their ability to disperse PAO1 biofilms across the different surfaces tested. Partial eradication of biofilm by phages can aid in complementing antibiotics that are unable to penetrate biofilms in a clinical set-up.
Collapse
Affiliation(s)
| | | | | | | | | | - Juliet Roshini Mohan Raj
- Division of Infectious Diseases, Nitte University Center for Science Education and Research, Paneer Campus, Nitte (Deemed to be University), Derelakatte, Mangaluru 575018, India (R.P.); (D.S.A.)
| |
Collapse
|
57
|
Hernández-Moreno LV, Pabón-Baquero LC, Prieto-Rodriguez JA, Patiño-Ladino OJ. Bioactive Compounds from P. pertomentellum That Regulate QS, Biofilm Formation and Virulence Factor Production of P. aeruginosa. Molecules 2023; 28:6181. [PMID: 37687010 PMCID: PMC10488431 DOI: 10.3390/molecules28176181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/27/2023] [Accepted: 07/27/2023] [Indexed: 09/10/2023] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen responsible for many nosocomial infections. This bacterium uses Quorum Sensing (QS) to generate antimicrobial resistance (AMR) so its disruption is considered a novel approach. The current study describes the antibiofilm and QS inhibitory potential of extract and chemical components from Piper pertomentellum. The methodo- logy included the phytochemical study on the aerial part of the species, the determination of QS inhibition efficacy on Chromobacterium violaceum and the evaluation of the effect on biofilm formation and virulence factors on P. aeruginosa. The phytochemical study led to the isolation and identification of a new piperamide (ethyltembamide 1), together with four known amides (tembamide acetate 2, cepharadione B 3, benzamide 4 and tembamide 5). The results indicated that the ethanolic extract and some fractions reduced violacein production in C. violaceum, however, only the ethanolic extract caused inhibition of biofilm formation of P. aeruginosa on polystyrene microtiter plates. Finally, the investigation determined that molecules (1-5) inhibited the formation of biofilms (50% approximately), while compounds 2-4 can inhibit pyocyanin and elastase production (30-50% approximately). In this way, the study contributes to the determination of the potential of extract and chemical constituents from P pertomentellum to regulate the QS system in P. aeruginosa.
Collapse
Affiliation(s)
- Lida V. Hernández-Moreno
- Departamento de Química, Facultad de Ciencias, Universidad Nacional de Colombia, Sede Bogotá, Bogotá 111321, Colombia; (L.V.H.-M.); (O.J.P.-L.)
| | - Ludy C. Pabón-Baquero
- Escuela de Ciencias Básicas y Aplicadas, Universidad de La Salle, Bogotá 111711, Colombia;
| | - Juliet A. Prieto-Rodriguez
- Departamento de Química, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Oscar J. Patiño-Ladino
- Departamento de Química, Facultad de Ciencias, Universidad Nacional de Colombia, Sede Bogotá, Bogotá 111321, Colombia; (L.V.H.-M.); (O.J.P.-L.)
| |
Collapse
|
58
|
Gerner E, Giraldo-Osorno PM, Johansson Loo A, Firdaus R, Ben Amara H, Werthén M, Palmquist A, Thomsen P, Omar O, Almqvist S, Trobos M. Targeting Pseudomonas aeruginosa quorum sensing with sodium salicylate modulates immune responses in vitro and in vivo. Front Cell Infect Microbiol 2023; 13:1183959. [PMID: 37614559 PMCID: PMC10442818 DOI: 10.3389/fcimb.2023.1183959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 07/18/2023] [Indexed: 08/25/2023] Open
Abstract
Introduction Chronic infections are a major clinical challenge in hard-to-heal wounds and implanted devices. Pseudomonas aeruginosa is a common causative pathogen that produces numerous virulence factors. Due to the increasing problem of antibiotic resistance, new alternative treatment strategies are needed. Quorum sensing (QS) is a bacterial communication system that regulates virulence and dampens inflammation, promoting bacterial survival. QS inhibition is a potent strategy to reduce bacterial virulence and alleviate the negative impact on host immune response. Aim This study investigates how secreted factors from P. aeruginosa PAO1, cultured in the presence or absence of the QS inhibitor sodium salicylate (NaSa), influence host immune response. Material and methods In vitro, THP-1 macrophages and neutrophil-like HL-60 cells were used. In vivo, discs of titanium were implanted in a subcutaneous rat model with local administration of P. aeruginosa culture supernatants. The host immune response to virulence factors contained in culture supernatants (+/-NaSa) was characterized through cell viability, migration, phagocytosis, gene expression, cytokine secretion, and histology. Results In vitro, P. aeruginosa supernatants from NaSa-containing cultures significantly increased THP-1 phagocytosis and HL-60 cell migration compared with untreated supernatants (-NaSa). Stimulation with NaSa-treated supernatants in vivo resulted in: (i) significantly increased immune cell infiltration and cell attachment to titanium discs; (ii) increased gene expression of IL-8, IL-10, ARG1, and iNOS, and (iii) increased GRO-α protein secretion and decreased IL-1β, IL-6, and IL-1α secretion, as compared with untreated supernatants. Conclusion In conclusion, treating P. aeruginosa with NaSa reduces the production of virulence factors and modulates major immune events, such as promoting phagocytosis and cell migration, and decreasing the secretion of several pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Erik Gerner
- Department of Biomaterials, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Centre for Antibiotic Resistance Research in Gothenburg (CARe), Gothenburg, Sweden
- Mölnlycke Health Care AB, Gothenburg, Sweden
| | - Paula Milena Giraldo-Osorno
- Department of Biomaterials, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Centre for Antibiotic Resistance Research in Gothenburg (CARe), Gothenburg, Sweden
| | - Anna Johansson Loo
- Department of Biomaterials, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Rininta Firdaus
- Department of Biomaterials, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Centre for Antibiotic Resistance Research in Gothenburg (CARe), Gothenburg, Sweden
| | - Heithem Ben Amara
- Department of Biomaterials, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maria Werthén
- Department of Biomaterials, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Centre for Antibiotic Resistance Research in Gothenburg (CARe), Gothenburg, Sweden
| | - Anders Palmquist
- Department of Biomaterials, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Peter Thomsen
- Department of Biomaterials, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Omar Omar
- Department of Biomedical Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | | | - Margarita Trobos
- Department of Biomaterials, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Centre for Antibiotic Resistance Research in Gothenburg (CARe), Gothenburg, Sweden
| |
Collapse
|
59
|
Rahimi M, Piroozmand A, Shayestehpour M, Salamat S, Peik Falak F, Shakerimoghaddam A, Moosavi GA, Khaledi A. Effect of curcumin nanoparticles and alcoholic extract of Falcaria vulgaris on the growth rate, biofilm, and gene expression in Pseudomonas aeruginosa isolated from burn wound infection. Mol Biol Rep 2023; 50:6681-6690. [PMID: 37378742 DOI: 10.1007/s11033-023-08559-2] [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: 04/05/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023]
Abstract
PURPOSE This study aimed to investigate the effect of Curcumin nanoparticles and alcoholic extract of Falcaria vulgaris on the growth rate, biofilm, and gene expression in Pseudomonas aeruginosa isolated from burn wound infection. METHODS The alcoholic extract of Falcaria vulgaris was purchased from Pasargad Company. Curcumin nanoparticles were synthesized. Antibacterial activity of Curcumin nanoparticles and alcoholic extract of Falcaria vulgaris was investigated by microdilution method alone and in combination. Biofilm inhibitory was investigated by microtitrplate method. Effect of Curcumin nanoparticles and alcoholic extract of Falcaria vulgaris were evaluated on algD gene expression via Real-Time PCR. Cytotoxicity was evaluated by MTT assay on HDF cell line. Then, the data were analyzed using SPSS software. RESULTS Synthesized Curcumin nanoparticles were approved by Fourier Transform Infrared (FTIR), and Scanning Electron Microscope. The alcoholic extract of Falcaria Vulgaris showed significant antibacterial activity against multidrug resistance (MDR) P. aeruginosa isolates at a concentration of 156.25 µg/mL. Moreover, MIC of the curcumin nanoparticle for isolates was 625 µg/mL. Based on fraction inhibition concentration, synergy, and the additive effect were shown against %7.7, and %93.3 of MDRs, respectively. The sub-MIC concentration of the binary compound reduced biofilms and algD gene expression in P. aeruginosa isolates. The Biological function of HDF cell lines was desirable after the effect of the binary compound. CONCLUSIONS Regarding our results, this combination can be suggested as a promising agent in terms of biofilm inhibitory and antimicrobial properties.
Collapse
Affiliation(s)
- Mohammadreza Rahimi
- Department of Microbiology and Immunology, Faculty of Medicine, Kashan University of Medical Science, Kashan, Iran
- Infectious Diseases Research Center, Kashan University of Medical Science, Kashan, Iran
| | - Ahmad Piroozmand
- Department of Microbiology and Immunology, Faculty of Medicine, Kashan University of Medical Science, Kashan, Iran
- Infectious Diseases Research Center, Kashan University of Medical Science, Kashan, Iran
- Autoimmune Diseases Research Center, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Shayestehpour
- Department of Microbiology and Immunology, Faculty of Medicine, Kashan University of Medical Science, Kashan, Iran
- Infectious Diseases Research Center, Kashan University of Medical Science, Kashan, Iran
- Autoimmune Diseases Research Center, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Sarvin Salamat
- Pharm D, Pharmacy School, Islamic Azad University, Tehran Branch, Tehran, Iran
| | - Farzaneh Peik Falak
- Department of Internal Medicine, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Ali Shakerimoghaddam
- Medical Biotechnology Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Gholam Abbas Moosavi
- Department of Vital Statistics and Epidemiology, School of Health, Kashan University of Medical Sciences, Kashan, Iran
| | - Azad Khaledi
- Department of Microbiology and Immunology, Faculty of Medicine, Kashan University of Medical Science, Kashan, Iran.
- Infectious Diseases Research Center, Kashan University of Medical Science, Kashan, Iran.
| |
Collapse
|
60
|
Wiehlmann L, Klockgether J, Hammerbacher AS, Salunkhe P, Horatzek S, Munder A, Peilert JF, Gulbins E, Eberl L, Tümmler B. A VirB4 ATPase of the mobile accessory genome orchestrates core genome-encoded features of physiology, metabolism, and virulence of Pseudomonas aeruginosa TBCF10839. Front Cell Infect Microbiol 2023; 13:1234420. [PMID: 37577372 PMCID: PMC10413270 DOI: 10.3389/fcimb.2023.1234420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 07/10/2023] [Indexed: 08/15/2023] Open
Abstract
Pseudomonas aeruginosa TBCF10839 is a highly virulent strain that can persist and replicate in human neutrophils. Screening of a signature-tagged mutagenesis (STM) TBCF10839 transposon library in phagocytosis tests identified a mutant that carried the transposon in the VirB4 homolog 5PG21 of an integrative and conjugative element (ICE)-associated type IV secretion system of the pKLC102 subtype. 5P21 TBCF10839 insertion mutants were deficient in metabolic versatility, secretion, quorum sensing, and virulence. The mutants were efficiently killed in phagocytosis tests in vitro and were avirulent in an acute murine airway infection model in vivo. The inactivation of 5PG21 silenced the rhl, las, and pqs operons and the gene expression for the synthesis of hydrogen cyanide, the antimetabolite l-2-amino-4-methoxy-trans-3-butenoic acid, and the H2- and H3-type VI secretion systems and their associated effectors. The mutants were impaired in the utilization of carbon sources and stored compounds that are not funneled into intermediary metabolism. This showcase demonstrates that a single gene of the mobile accessory genome can become an essential element to operate the core genome-encoded features of metabolism and virulence.
Collapse
Affiliation(s)
- Lutz Wiehlmann
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Research Core Unit Genomics, Hannover Medical School, Hannover, Germany
| | - Jens Klockgether
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Anna-Silke Hammerbacher
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Prabhakar Salunkhe
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Sonja Horatzek
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Antje Munder
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease, German Center for Lung Research, Hannover, Germany
| | | | - Erich Gulbins
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Institute of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Leo Eberl
- Department of Plant and Microbial Biology, University of Zurich, Zurich, Switzerland
| | - Burkhard Tümmler
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease, German Center for Lung Research, Hannover, Germany
| |
Collapse
|
61
|
Sánchez-Jiménez A, Llamas MA, Marcos-Torres FJ. Transcriptional Regulators Controlling Virulence in Pseudomonas aeruginosa. Int J Mol Sci 2023; 24:11895. [PMID: 37569271 PMCID: PMC10418997 DOI: 10.3390/ijms241511895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/21/2023] [Accepted: 07/22/2023] [Indexed: 08/13/2023] Open
Abstract
Pseudomonas aeruginosa is a pathogen capable of colonizing virtually every human tissue. The host colonization competence and versatility of this pathogen are powered by a wide array of virulence factors necessary in different steps of the infection process. This includes factors involved in bacterial motility and attachment, biofilm formation, the production and secretion of extracellular invasive enzymes and exotoxins, the production of toxic secondary metabolites, and the acquisition of iron. Expression of these virulence factors during infection is tightly regulated, which allows their production only when they are needed. This process optimizes host colonization and virulence. In this work, we review the intricate network of transcriptional regulators that control the expression of virulence factors in P. aeruginosa, including one- and two-component systems and σ factors. Because inhibition of virulence holds promise as a target for new antimicrobials, blocking the regulators that trigger the production of virulence determinants in P. aeruginosa is a promising strategy to fight this clinically relevant pathogen.
Collapse
Affiliation(s)
| | - María A. Llamas
- Department of Biotechnology and Environmental Protection, Estación Experimental del Zaidín-Consejo Superior de Investigaciones Científicas, 18008 Granada, Spain;
| | - Francisco Javier Marcos-Torres
- Department of Biotechnology and Environmental Protection, Estación Experimental del Zaidín-Consejo Superior de Investigaciones Científicas, 18008 Granada, Spain;
| |
Collapse
|
62
|
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
|
63
|
Das T, Das B, Young BC, Aldilla V, Sabir S, Almohaywi B, Willcox M, Manefield M, Kumar N. Ascorbic acid modulates the structure of the Pseudomonas aeruginosa virulence factor pyocyanin and ascorbic acid-furanone-30 combination facilitate biofilm disruption. Front Microbiol 2023; 14:1166607. [PMID: 37520362 PMCID: PMC10381918 DOI: 10.3389/fmicb.2023.1166607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 06/26/2023] [Indexed: 08/01/2023] Open
Abstract
The production of pyocyanin by Pseudomonas aeruginosa increases its virulence, fitness and biofilm formation. Pyocyanin is also a redox molecule and we hypothesize that ascorbic acid being an antioxidant will interact with pyocyanin. The main objective of this study was to investigate the potential interaction of ascorbic acid with pyocyanin, and also to investigate the impact of ascorbic acid in combination with Furanone-30 on quorum sensing and biofilm formation of P. aeruginosa. When incubated with ascorbic acid, hyperchromic and hypsochromic shifts in pyocyanin absorbance peaks at 385 nm and 695 nm were observed. In the presence of dehydroascorbic acid and citric acid, these shifts were absent, indicating that the intrinsic antioxidant property of ascorbic acid was probably essential in binding to pyocyanin. NMR spectroscopy showed shifts in 1H NMR pyocyanin peaks between 8.2 to 5.8 ppm when incubated in the presence of ascorbic acid. Density Functional Theory (DFT) supported potential interactions between the -CH2OH or -OH moieties of ascorbic acid with the -C=O moiety of pyocyanin. The pyocyanin-ascorbic acid complex impaired pyocyanin binding to DNA. Ascorbic acid combined with furanone-30 elevated quorum-sensing inhibition in P. aeruginosa, which was directly associated with significantly reduced P. aeruginosa virulence, adhesion, aggregation and biofilm formation and enhanced antibiotic-mediated bacterial killing. This study demonstrated that the antioxidant ascorbic acid directly binds to pyocyanin, modulates its structure and results in disruption of biofilm formation and associated tolerance to antibiotics.
Collapse
Affiliation(s)
- Theerthankar Das
- Infection Immunity and Inflammation, Charles Perkins Centre, School of Medical Science, The University of Sydney, Sydney, NSW, Australia
- Sydney Institute for Infectious Diseases, School of Medical Science, The University of Sydney, Sydney, NSW, Australia
| | - Biswanath Das
- Department of Organic Chemistry, Arrhenius Laboratory Stockholm University, Stockholm, Sweden
- School of Chemistry, University of New South Wales (UNSW), Sydney, NSW, Australia
| | - Brandon Clark Young
- Infection Immunity and Inflammation, Charles Perkins Centre, School of Medical Science, The University of Sydney, Sydney, NSW, Australia
| | - Vina Aldilla
- School of Chemistry, University of New South Wales (UNSW), Sydney, NSW, Australia
| | - Shekh Sabir
- School of Chemistry, University of New South Wales (UNSW), Sydney, NSW, Australia
| | - Basmah Almohaywi
- College of Pharmacy, King Khalid University (KKU), Abha, Saudi Arabia
| | - Mark Willcox
- School of Optometry and Vision Science, The University of New South Wales, Sydney, NSW, Australia
| | - Mike Manefield
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, NSW, Australia
| | - Naresh Kumar
- School of Chemistry, University of New South Wales (UNSW), Sydney, NSW, Australia
| |
Collapse
|
64
|
Zhao X, Xu C, Qu J, Jin Y, Bai F, Cheng Z, Wu W, Pan X. PitA Controls the H2- and H3-T6SSs through PhoB in Pseudomonas aeruginosa. Appl Environ Microbiol 2023; 89:e0209422. [PMID: 37184394 PMCID: PMC10304775 DOI: 10.1128/aem.02094-22] [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: 12/10/2022] [Accepted: 04/21/2023] [Indexed: 05/16/2023] Open
Abstract
Pseudomonas aeruginosa possesses three type VI secretion systems (T6SSs) that are involved in interspecies competition, internalization into epithelial cells, and virulence. Host-derived mucin glycans regulate the T6SSs through RetS, and attacks from other species activate the H1-T6SS. However, other environmental signals that control the T6SSs remain to be explored. Previously, we determined PitA to be a constitutive phosphate transporter, whose mutation reduces the intracellular phosphate concentration. Here, we demonstrate that mutation in the pitA gene increases the expression of the H2- and H3-T6SS genes and enhances bacterial uptake by A549 cells. We further found that mutation of pitA results in activation of the quorum sensing (QS) systems, which contributes to the upregulation of the H2- and H3-T6SS genes. Overexpression of the phosphate transporter complex genes pstSCAB or knockdown of the phosphate starvation response regulator gene phoB in the ΔpitA mutant reduces the expression of the QS genes and subsequently the H2- and H3-T6SS genes and bacterial internalization. Furthermore, growth of wild-type PA14 in a low-phosphate medium results in upregulation of the QS and H2- and H3-T6SS genes and bacterial internalization compared to those in cells grown in a high-phosphate medium. Deletion of the phoB gene abolished the differences in the expression of the QS and T6SS genes as well as bacterial internalization in the low- and high- phosphate media. Overall, our results elucidate the mechanism of PitA-mediated regulation on the QS system and H2- and H3-T6SSs and reveal a novel pathway that regulates the T6SSs in response to phosphate starvation. IMPORTANCE Pseudomonas aeruginosa is an opportunistic pathogenic bacterium that causes acute and chronic infections in humans. The type VI secretion systems (T6SSs) have been shown to associate with chronic infections. Understanding the mechanism used by the bacteria to sense environmental signals and regulate virulence factors will provide clues for developing novel effective treatment strategies. Here, we demonstrate a relationship between a phosphate transporter and the T6SSs and reveal a novel regulatory pathway that senses phosphate limitation and controls bacterial virulence factors in P. aeruginosa.
Collapse
Affiliation(s)
- Xinrui Zhao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Congjuan Xu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Junze Qu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Yongxin Jin
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Fang Bai
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Zhihui Cheng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Weihui Wu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaolei Pan
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
65
|
Naga NG, El-Badan DE, Ghanem KM, Shaaban MI. It is the time for quorum sensing inhibition as alternative strategy of antimicrobial therapy. Cell Commun Signal 2023; 21:133. [PMID: 37316831 DOI: 10.1186/s12964-023-01154-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/29/2023] [Indexed: 06/16/2023] Open
Abstract
Multiple drug resistance poses a significant threat to public health worldwide, with a substantial increase in morbidity and mortality rates. Consequently, searching for novel strategies to control microbial pathogenicity is necessary. With the aid of auto-inducers (AIs), quorum sensing (QS) regulates bacterial virulence factors through cell-to-cell signaling networks. AIs are small signaling molecules produced during the stationary phase. When bacterial cultures reach a certain level of growth, these molecules regulate the expression of the bound genes by acting as mirrors that reflect the inoculum density.Gram-positive bacteria use the peptide derivatives of these signaling molecules, whereas Gram-negative bacteria use the fatty acid derivatives, and the majority of bacteria can use both types to modulate the expression of the target gene. Numerous natural and synthetic QS inhibitors (QSIs) have been developed to reduce microbial pathogenesis. Applications of QSI are vital to human health, as well as fisheries and aquaculture, agriculture, and water treatment. Video Abstract.
Collapse
Affiliation(s)
- Nourhan G Naga
- Botany and Microbiology Department, Faculty of Science, Alexandria University, Alexandria, Egypt.
| | - Dalia E El-Badan
- Botany and Microbiology Department, Faculty of Science, Alexandria University, Alexandria, Egypt
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Beirut, Lebanon
| | - Khaled M Ghanem
- Botany and Microbiology Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Mona I Shaaban
- Microbiology and Immunology Department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
66
|
Ahamad Khan M, Lone SA, Shahid M, Zeyad MT, Syed A, Ehtram A, Elgorban AM, Verma M, Danish M. Phytogenically Synthesized Zinc Oxide Nanoparticles (ZnO-NPs) Potentially Inhibit the Bacterial Pathogens: In Vitro Studies. TOXICS 2023; 11:toxics11050452. [PMID: 37235266 DOI: 10.3390/toxics11050452] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/02/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023]
Abstract
The usefulness of nanoparticles (NPs) in biological applications, such as nanomedicine, is becoming more widely acknowledged. Zinc oxide nanoparticles (ZnO-NPs) are a type of metal oxide nanoparticle with an extensive use in biomedicine. Here, ZnO-NPs were synthesized using Cassia siamea (L.) leaf extract and characterized using state-of-the-art techniques; UV-vis spectroscopy, XRD, FTIR, and SEM. At sub-minimum inhibitory concentration (MIC) levels, the ability of ZnO@Cs-NPs to suppress quorum-mediated virulence factors and biofilm formation against clinical MDR isolates (Pseudomonas aeruginosa PAO1 and Chromobacterium violaceum MCC-2290) was tested. The ½MIC of ZnO@Cs-NPs reduced violacein production by C. violaceum. Furthermore, ZnO@Cs-NPs sub-MIC significantly inhibited virulence factors such aspyoverdin, pyocyanin, elastase, exoprotease, rhamnolipid, and the swimming motility of P. aeruginosa PAO1 by 76.9, 49.0, 71.1, 53.3, 89.5, and 60%, respectively. Moreover, ZnO@Cs-NPs also showed wide anti-biofilm efficacy, inhibiting a maximum of 67 and 56% biofilms in P. aeruginosa and C. violaceum, respectively. In addition, ZnO@Cs-NPs suppressed extra polymeric substances (EPS) produced by isolates. Additionally, under confocal microscopy, propidium iodide-stained cells of P. aeruginosa and C. violaceum show ZnO@Cs-NP-induced impairment in membrane permeability, revealing strong anti-bacterial efficacy. This research demonstrates that newly synthesized ZnO@Cs-NPs demonstrate a strong efficacy against clinical isolates. In a nutshell, ZnO@Cs-NPs can be used as an alternative therapeutic agent for managing pathogenic infections.
Collapse
Affiliation(s)
- Mo Ahamad Khan
- Department of Microbiology, Jawahar Lal Nehru Medical College (JNMC), Aligarh Muslim University, Aligarh 202002, India
| | - Showkat Ahmad Lone
- Department of Microbiology, Government Medical College, Baramulla 19310, India
| | - Mohammad Shahid
- Department of Agricultural Microbiology, Faculty of Agriculture Science, Aligarh Muslim University, Aligarh 202002, India
| | - Mohammad Tarique Zeyad
- Department of Agricultural Microbiology, Faculty of Agriculture Science, Aligarh Muslim University, Aligarh 202002, India
| | - Asad Syed
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Aquib Ehtram
- La Jolla Institute for Immunology, San Diego, CA 92037, USA
| | - Abdallah M Elgorban
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Meenakshi Verma
- Centre of Research & Development, Department of Chemistry, Chandigarh University, Mohali 160055, India
| | - Mohammad Danish
- Department of Botany, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, India
| |
Collapse
|
67
|
Chakraborty S, Bashir Y, Sirotiya V, Ahirwar A, Das S, Vinayak V. Role of bacterial quorum sensing and quenching mechanism in the efficient operation of microbial electrochemical technologies: A state-of-the-art review. Heliyon 2023; 9:e16205. [PMID: 37215776 PMCID: PMC10199210 DOI: 10.1016/j.heliyon.2023.e16205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 05/06/2023] [Accepted: 05/09/2023] [Indexed: 05/24/2023] Open
Abstract
Microbial electrochemical technologies (METs) are a group of innovative technologies that produce valuables like bioelectricity and biofuels with the simultaneous treatment of wastewater from microorganisms known as electroactive microorganisms. The electroactive microorganisms are capable of transferring electrons to the anode of a MET through various metabolic pathways such as direct (via cytochrome or pili) or indirect (through transporters) transfer. Though this technology is promising, the inferior yield of valuables and the high cost of reactor fabrication are presently impeding the large-scale application of this technology. Therefore, to overcome these major bottlenecks, a lot of research has been dedicated to the application of bacterial signalling, for instance, quorum sensing (QS) and quorum quenching (QQ) mechanisms in METs to improve its efficacy in order to achieve a higher power density and to make it more cost-effective. The QS circuit in bacteria produces auto-inducer signal molecules, which enhances the biofilm-forming ability and regulates the bacterial attachment on the electrode of METs. On the other hand, the QQ circuit can effectively function as an antifouling agent for the membranes used in METs and microbial membrane bioreactors, which is imperative for their stable long-term operation. This state-of-the-art review thus distinctly describes in detail the interaction between the QQ and QS systems in bacteria employed in METs to generate value-added by-products, antifouling strategies, and the recent applications of the signalling mechanisms in METs to improve their yield. Further, the article also throws some light on the recent advancements and the challenges faced while incorporating QS and QQ mechanisms in various types of METs. Thus, this review article will help budding researchers in upscaling METs with the integration of the QS signalling mechanism in METs.
Collapse
Affiliation(s)
- Sukanya Chakraborty
- Diatom Nanoengineering and Metabolism Laboratory (DNM), School of Applied Science, Dr. Harisingh Gour Central University, Sagar, MP, 470003, India
| | - Yasser Bashir
- Department of Civil Engineering, Indian Institute of Technology Delhi, New Delhi, 110016, India
| | - Vandana Sirotiya
- Diatom Nanoengineering and Metabolism Laboratory (DNM), School of Applied Science, Dr. Harisingh Gour Central University, Sagar, MP, 470003, India
| | - Ankesh Ahirwar
- Diatom Nanoengineering and Metabolism Laboratory (DNM), School of Applied Science, Dr. Harisingh Gour Central University, Sagar, MP, 470003, India
- Metabolism, Bioengineering of Microalgal Metabolism and Applications (MIMMA), Mer Molecules Santé, Le Mans University, IUML - FR 3473 CNRS, Le Mans, France
| | - Sovik Das
- Department of Civil Engineering, Indian Institute of Technology Delhi, New Delhi, 110016, India
| | - Vandana Vinayak
- Diatom Nanoengineering and Metabolism Laboratory (DNM), School of Applied Science, Dr. Harisingh Gour Central University, Sagar, MP, 470003, India
| |
Collapse
|
68
|
Transcriptome Dynamics of Pseudomonas aeruginosa during Transition from Overlapping To Non-Overlapping Cell Cycles. mSystems 2023; 8:e0113022. [PMID: 36786632 PMCID: PMC10134858 DOI: 10.1128/msystems.01130-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
Abstract
Bacteria either duplicate their chromosome once per cell division or a new round of replication is initiated before the cells divide, thus cell cycles overlap. Here, we show that the opportunistic pathogen Pseudomonas aeruginosa switches from fast growth with overlapping cell cycles to sustained slow growth with only one replication round per cell division when cultivated under standard laboratory conditions. The transition was characterized by fast-paced, sequential changes in transcriptional activity along the ori-ter axis of the chromosome reflecting adaptation to the metabolic needs during both growth phases. Quorum sensing (QS) activity was highest at the onset of the slow growth phase with non-overlapping cell cycles. RNA sequencing of subpopulations of these cultures sorted based on their DNA content, revealed a strong gene dosage effect as well as specific expression patterns for replicating and nonreplicating cells. Expression of flagella and mexE, involved in multidrug efflux was restricted to cells that did not replicate, while those that did showed a high activity of the cell division locus and recombination genes. A possible role of QS in the formation of these subpopulations upon switching to non-overlapping cell cycles could be a subject of further research. IMPORTANCE The coordination of gene expression with the cell cycle has so far been studied only in a few bacteria, the bottleneck being the need for synchronized cultures. Here, we determined replication-associated effects on transcription by comparing Pseudomonas aeruginosa cultures that differ in their growth mode and number of replicating chromosomes. We further show that cell cycle-specific gene regulation can be principally identified by RNA sequencing of subpopulations from cultures that replicate only once per cell division and that are sorted according to their DNA content. Our approach opens the possibility to study asynchronously growing bacteria from a wide phylogenetic range and thereby enhance our understanding of the evolution of cell cycle control on the transcriptional level.
Collapse
|
69
|
Jeong GJ, Khan F, Khan S, Tabassum N, Mehta S, Kim YM. Pseudomonas aeruginosa virulence attenuation by inhibiting siderophore functions. Appl Microbiol Biotechnol 2023; 107:1019-1038. [PMID: 36633626 DOI: 10.1007/s00253-022-12347-6] [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: 12/16/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 01/13/2023]
Abstract
Pseudmonas aeruginosa is a Gram-negative bacterium known to be ubiquitous and recognized as one of the leading causes of infections such as respiratory, urinary tract, burns, cystic fibrosis, and in immunocompromised individuals. Failure of antimicrobial therapy has been documented to be attributable due to the development of various resistance mechanisms, with a proclivity to develop additional resistance mechanisms rapidly. P. aeruginosa virulence attenuation is an alternate technique for disrupting pathogenesis without impacting growth. The iron-scavenging siderophores (pyoverdine and pyochelin) generated by P. aeruginosa have various properties like scavenging iron, biofilm formation, quorum sensing, increasing virulence, and toxicity to the host. As a result, developing an antivirulence strategy, specifically inhibiting the P. aeruginosa siderophore, has been a promising therapeutic option to limit their infection. Several natural, synthetic compounds and nanoparticles have been identified as potent inhibitors of siderophore production/biosynthesis, function, and transport system. The current review discussed pyoverdine and pyochelin's synthesis and transport system in P. aeruginosa. Furthermore, it is also focused on the role of several natural and synthetic compounds in reducing P. aeruginosa virulence by inhibiting siderophore synthesis, function, and transport. The underlying mechanism involved in inhibiting the siderophore by natural and synthetic compounds has also been explained. KEY POINTS: • Pseudomonas aeruginosa is an opportunistic pathogen linked to chronic respiratory, urinary tract, and burns infections, as well as cystic fibrosis and immunocompromised patients. • P. aeruginosa produces two virulent siderophores forms: pyoverdine and pyochelin, which help it to survive in iron-deficient environments. • The inhibition of siderophore production, transport, and activity using natural and synthesized drugs has been described as a potential strategy for controlling P. aeruginosa infection.
Collapse
Affiliation(s)
- Geum-Jae Jeong
- Department of Food Science and 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.
| | - Sohail Khan
- Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector-62, Noida, Uttar Pradesh, 201309, India
| | - 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
| | - Sonu Mehta
- Anthem Biosciences Private Limited, Bommasandra, Bangalore, Karnataka, 56009, India
| | - 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
|
70
|
Chadha J, Ravi, Singh J, Harjai K. α-Terpineol synergizes with gentamicin to rescue Caenorhabditis elegans from Pseudomonas aeruginosa infection by attenuating quorum sensing-regulated virulence. Life Sci 2023; 313:121267. [PMID: 36481167 DOI: 10.1016/j.lfs.2022.121267] [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: 09/14/2022] [Revised: 11/24/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
AIMS This study scrutinized α-Terpineol (α-T) for its anti-virulence and anti-fouling potential against P. aeruginosa PAO1 in conjunction with gentamicin (GeN) using in-vitro, in-silico, and in-vivo approaches. MAIN METHODS The quorum quenching (QQ) potential of the drug combination was studied using a quorum sensing (QS) biosensor strain and tested for synergy using chequerboard and time-kill kinetics assays. The effect of α-T and GeN on bacterial motility, QS-regulated virulence factor production, and biofilm formation was assessed in P. aeruginosa PAO1 along with molecular docking analysis. The protective effects of α-T-GeN combination were also examined in a Caenorhabditis elegans infection model through slow-killing (SK) assays. KEY FINDINGS The drug combination displayed synergy, enhanced QQ activity, and suppressed AHL production in PAO1. At sub-inhibitory concentrations, the drug combination suppressed the expression of genes regulating QS and pseudomonal virulence, thereby inhibiting the production of virulence factors in PAO1. The drug combination compromised all forms of pseudomonal motility, strongly inhibited biofilm formation, and successfully eradicated preformed biofilms. Based on these findings, it is concluded that GeN (alone) does not harbor any QQ properties, but enhances the QQ potential of α-T. Moreover, combinational treatment protected C. elegans from pseudomonal infection and improved survival rates by 73 % at 96 h. SIGNIFICANCE For the first time, the molecular mechanism responsible for the anti-QS activity of α-T was unraveled through a comprehensive investigation, thereby asserting its potential as an anti-virulent drug against P. aeruginosa.
Collapse
Affiliation(s)
- Jatin Chadha
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Ravi
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Mohali, Punjab, India
| | - Jogender Singh
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Mohali, Punjab, India
| | - Kusum Harjai
- Department of Microbiology, Panjab University, Chandigarh, India.
| |
Collapse
|
71
|
Mohamad F, Alzahrani RR, Alsaadi A, Alrfaei BM, Yassin AEB, Alkhulaifi MM, Halwani M. An Explorative Review on Advanced Approaches to Overcome Bacterial Resistance by Curbing Bacterial Biofilm Formation. Infect Drug Resist 2023; 16:19-49. [PMID: 36636380 PMCID: PMC9830422 DOI: 10.2147/idr.s380883] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 12/06/2022] [Indexed: 01/05/2023] Open
Abstract
The continuous emergence of multidrug-resistant pathogens evoked the development of innovative approaches targeting virulence factors unique to their pathogenic cascade. These approaches aimed to explore anti-virulence or anti-infective therapies. There are evident concerns regarding the bacterial ability to create a superstructure, the biofilm. Biofilm formation is a crucial virulence factor causing difficult-to-treat, localized, and systemic infections. The microenvironments of bacterial biofilm reduce the efficacy of antibiotics and evade the host's immunity. Producing a biofilm is not limited to a specific group of bacteria; however, Pseudomonas aeruginosa, Acinetobacter baumannii, and Staphylococcus aureus biofilms are exemplary models. This review discusses biofilm formation as a virulence factor and the link to antimicrobial resistance. In addition, it explores insights into innovative multi-targeted approaches and their physiological mechanisms to combat biofilms, including natural compounds, phages, antimicrobial photodynamic therapy (aPDT), CRISPR-Cas gene editing, and nano-mediated techniques.
Collapse
Affiliation(s)
- F Mohamad
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Raghad R Alzahrani
- Nanomedicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia,Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Ahlam Alsaadi
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Bahauddeen M Alrfaei
- Stem Cells and Regenerative Medicine, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Alaa Eldeen B Yassin
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Manal M Alkhulaifi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia,Manal M Alkhulaifi, P.O. Box 55670, Riyadh, 11544, Tel +966 (11) 805-1685, Email
| | - Majed Halwani
- Nanomedicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia,Correspondence: Majed Halwani, P.O. Box 3660, Mail Code 1515 (KAIMRC), Riyadh, 11481, Tel +966 (11) 429-4433, Fax +966 (11) 429-4440, Email ;
| |
Collapse
|
72
|
Juhas M. Communication Between Microorganisms. BRIEF LESSONS IN MICROBIOLOGY 2023:27-41. [DOI: 10.1007/978-3-031-29544-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
73
|
Ghanem SM, Abd El-Baky RM, Abourehab MAS, Fadl GFM, Gamil NGFM. Prevalence of Quorum Sensing and Virulence Factor Genes Among Pseudomonas aeruginosa Isolated from Patients Suffering from Different Infections and Their Association with Antimicrobial Resistance. Infect Drug Resist 2023; 16:2371-2385. [PMID: 37113530 PMCID: PMC10128085 DOI: 10.2147/idr.s403441] [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: 01/18/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Purpose Antimicrobial resistance and virulence genes play important roles in increasing the severity of Pseudomonas aeruginosa infections, especially in hospitalized patients with high antibiotic pressure. Most genes that encode Pseudomonas aeruginosa virulence factors are controlled and regulated by the quorum sensing (QS) system. The aim of this study was to investigate the frequency of some virulence genes (rhlR, rhlI, lasR, lasI, lasB, toxA, aprA, algD, ExoS, and plcH genes) and their association with antibiotic resistance. Methods Antimicrobial susceptibility was determined by Kirby-Bauer agar disk diffusion method. A total of 125 clinical isolates of P. aeruginosa were tested for some virulence genes using polymerase chain reaction (PCR). Results The highest resistance was observed against cefepime (92.8%). Multi-drug resistant (MDR) P. aeruginosa represented 63.2% of total isolates with high distribution among wound isolates (21/79, 26.3% of MDR isolates). LasB was the most prevalent virulence gene among the tested isolates (89.6%) followed by aprA (85.6%), exoS (84%), algD (80%), toxA (76.8%), and plcH (75.2). Furthermore, a significant association (P < 0.05) among most of the tested virulence genes and MDR isolates was found. The presence of more than 5 virulence genes was highly observed among wound infections, otitis media, and respiratory tract infection isolates. Conclusion The complex association of virulence genes including QS system regulating genes with antibiotic resistance indicates the importance of the tested factors in the progression of infections, which is considered a great challenge for the health-care team with the need for specific studies for each area having different antibiotic resistance profiles and the development of effective treatment strategies such as anti-virulent and quorum sensing inhibiting drugs against P. aeruginosa infections.
Collapse
Affiliation(s)
- Shimaa M Ghanem
- Department of Microbiology and Immunology, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Rehab Mahmoud Abd El-Baky
- Department of Microbiology and Immunology, Faculty of Pharmacy, Minia University, Minia, Egypt
- Department of Microbiology and Immunology, Faculty of Pharmacy, Deraya University, Minia, Egypt
| | - Mohamed A S Abourehab
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
- Correspondence: Mohamed AS Abourehab, Email
| | - Gamal F M Fadl
- Department of Microbiology and Immunology, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Nancy G F M Gamil
- Department of Microbiology and Immunology, Faculty of Pharmacy, Minia University, Minia, Egypt
| |
Collapse
|
74
|
Virulence Induction in Pseudomonas aeruginosa under Inorganic Phosphate Limitation: a Proteomics Perspective. Microbiol Spectr 2022; 10:e0259022. [PMID: 36354317 PMCID: PMC9769906 DOI: 10.1128/spectrum.02590-22] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Inorganic phosphate (Pi) is a central nutrient and signal molecule for bacteria. Pi limitation was shown to increase the virulence of several phylogenetically diverse pathogenic bacteria with different lifestyles. Hypophosphatemia enhances the risk of death in patients due to general bacteremia and was observed after surgical injury in humans. Phosphate therapy, or the reduction of bacterial virulence by the administration of Pi or phosphate-containing compounds, is a promising anti-infective therapy approach that will not cause cytotoxicity or the emergence of antibiotic-resistant strains. The proof of concept of phosphate therapy has been obtained using primarily Pseudomonas aeruginosa (PA). However, a detailed understanding of Pi-induced changes at protein levels is missing. Using pyocyanin production as proxy, we show that the Pi-mediated induction of virulence is a highly cooperative process that occurs between 0.2 to 0.6 mM Pi. We present a proteomics study of PA grown in minimal medium supplemented with either 0.2 mM or 1 mM Pi and rich medium. About half of the predicted PA proteins could be quantified. Among the 1,471 dysregulated proteins comparing growth in 0.2 mM to 1 mM Pi, 1,100 were depleted under Pi-deficient conditions. Most of these proteins are involved in general and energy metabolism, different biosynthetic and catabolic routes, or transport. Pi depletion caused accumulation of proteins that belong to all major families of virulence factors, including pyocyanin synthesis, secretion systems, quorum sensing, chemosensory signaling, and the secretion of proteases, phospholipases, and phosphatases, which correlated with an increase in exoenzyme production and antibacterial activity. IMPORTANCE Antibiotics are our main weapons to fight pathogenic bacteria, but the increase in antibiotic-resistant strains and their consequences represents a major global health challenge, revealing the necessity to develop alternative antimicrobial strategies that do not involve the bacterial killing or growth inhibition. P. aeruginosa has been placed second on the global priority list to guide research on the development of new antibiotics. One of the most promising alternative strategies is the phosphate therapy for which the proof of concept has been obtained for P. aeruginosa. This article reports the detailed changes at the protein levels comparing P. aeruginosa grown under Pi-abundant and Pi-depleted conditions. These data describe in detail the molecular mechanisms underlying phosphate therapy. Apart from Pi, several other phosphate-containing compounds have been used for phosphate therapy and this study will serve as a reference for comparative studies aimed at evaluating the effect of alternative compounds.
Collapse
|
75
|
Taylor IR, Murray-Nerger LA, Greco TM, Liu D, Cristea IM, Bassler BL. Protein Interaction Networks of Catalytically Active and Catalytically Inactive PqsE in Pseudomonas aeruginosa. mBio 2022; 13:e0155922. [PMID: 36073810 PMCID: PMC9600345 DOI: 10.1128/mbio.01559-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 08/11/2022] [Indexed: 11/24/2022] Open
Abstract
Pseudomonas aeruginosa is a human pathogen that relies on quorum sensing to establish infections. The PqsE quorum-sensing protein is required for P. aeruginosa virulence factor production and infection. PqsE has a reported enzymatic function in the biosynthesis of the quorum-sensing autoinducer called PQS. However, this activity is redundant because, in the absence of PqsE, this role is fulfilled by alternative thioesterases. Rather, PqsE drives P. aeruginosa pathogenic traits via a protein-protein interaction with the quorum-sensing receptor/transcription factor RhlR, an interaction that enhances the affinity of RhlR for target DNA sequences. PqsE catalytic activity is dispensable for interaction with RhlR. Thus, the virulence function of PqsE can be decoupled from its catalytic function. Here, we present an immunoprecipitation-mass spectrometry method employing enhanced green fluorescent protein-PqsE fusions to define the protein interactomes of wild-type PqsE and the catalytically inactive PqsE(D73A) variant in P. aeruginosa and their dependence on RhlR. Several proteins were identified to have specific interactions with wild-type PqsE while not forming associations with PqsE(D73A). In the ΔrhlR strain, an increased number of specific PqsE interactors were identified, including the partner autoinducer synthase for RhlR, called RhlI. Collectively, these results suggest that specific protein-protein interactions depend on PqsE catalytic activity and that RhlR may prevent proteins from interacting with PqsE, possibly due to competition between RhlR and other proteins for PqsE binding. Our results provide a foundation for the identification of the in vivo PqsE catalytic function and, potentially, new proteins involved in P. aeruginosa quorum sensing. IMPORTANCE Pseudomonas aeruginosa causes hospital-borne infections in vulnerable patients, including immunocompromised individuals, burn victims, and cancer patients undergoing chemotherapy. There are no effective treatments for P. aeruginosa infections, which are usually broadly resistant to antibiotics. Animal models show that, to establish infection and to cause illness, P. aeruginosa relies on an interaction between two proteins, namely, PqsE and RhlR. There could be additional protein-protein interactions involving PqsE, which, if defined, could be exploited for the design of new therapeutic strategies to combat P. aeruginosa. Here, we reveal previously unknown protein interactions in which PqsE participates, which will be investigated for potential roles in pathogenesis.
Collapse
Affiliation(s)
- Isabelle R. Taylor
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | | | - Todd M. Greco
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Dawei Liu
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Ileana M. Cristea
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Bonnie L. Bassler
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| |
Collapse
|
76
|
Grace A, Sahu R, Owen DR, Dennis VA. Pseudomonas aeruginosa reference strains PAO1 and PA14: A genomic, phenotypic, and therapeutic review. Front Microbiol 2022; 13:1023523. [PMID: 36312971 PMCID: PMC9607943 DOI: 10.3389/fmicb.2022.1023523] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/28/2022] [Indexed: 11/25/2022] Open
Abstract
Pseudomonas aeruginosa is a ubiquitous, motile, gram-negative bacterium that has been recently identified as a multi-drug resistant pathogen in critical need of novel therapeutics. Of the approximately 5,000 strains, PAO1 and PA14 are common laboratory reference strains, modeling moderately and hyper-virulent phenotypes, respectively. PAO1 and PA14 have been instrumental in facilitating the discovery of novel drug targets, testing novel therapeutics, and supplying critical genomic information on the bacterium. While the two strains have contributed to a wide breadth of knowledge on the natural behaviors and therapeutic susceptibilities of P. aeruginosa, they have demonstrated significant deviations from observations in human infections. Many of these deviations are related to experimental inconsistencies in laboratory strain environment that complicate and, at times, terminate translation from laboratory results to clinical applications. This review aims to provide a comparative analysis of the two strains and potential methods to improve their clinical relevance.
Collapse
Affiliation(s)
- Amber Grace
- Department of Biological Sciences, Alabama State University, Montgomery, AL, United States
| | - Rajnish Sahu
- Department of Biological Sciences, Alabama State University, Montgomery, AL, United States
| | | | - Vida A. Dennis
- Department of Biological Sciences, Alabama State University, Montgomery, AL, United States
- *Correspondence: Vida A. Dennis,
| |
Collapse
|
77
|
Potential of Impatiens balsamina Leaf Extract against Quorum Sensing in Pseudomonas aeruginosa PA01. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2022. [DOI: 10.22207/jpam.16.3.40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bacteria control gene expression by quorum sensing (QS) mechanism owing to producing small signal molecules associated with population density. Both gram-positive and gram-negative bacteria use QS to manage various physiological characteristics, including bioluminescence, virulence gene expression, biofilm formation, and antibiotic resistance. Impatience balsamina is a flowering, perennial and annual herb indigenous to southern Asia in India. All parts of Impatience balsamina have a therapeutic effect on different diseases. This study evaluated the anti-quorum sensing activity of leaf extract of Impatience balsamina by examining its action on Violacein production by Chromobacterium violaceum, a biosensor strain, and Biofilm, Pyocyanin, Protease, and Chitinase production by the reference strain Pseudomonas aeruginosa PA 01. Minimum inhibitory concentration (MIC) for Pseudomonas aeruginosa PA 01was 3.125mg/ml. A concentration of 1.563mg/ml (sub-MIC) showed inhibition of 100% on Las A protease, 78.42% on chitinase, 30.75% on biofilm, and 93.33% on pyocyanin production by Pseudomonas aeruginosa PA 01. This article displayed the quorum quenching activity of Impatience balsamina by hindering the quorum-sensing controlled characteristics of bacteria without killing it, which reduces the proneness of drug resistance in bacteria, a globally accepted emerging problem in the medical field.
Collapse
|
78
|
Effect of Biogenic Silver Nanoparticles on the Quorum-Sensing System of Pseudomonas aeruginosa PAO1 and PA14. Microorganisms 2022; 10:microorganisms10091755. [PMID: 36144357 PMCID: PMC9504124 DOI: 10.3390/microorganisms10091755] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/24/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
The increase in multidrug-resistant microorganisms represents a global threat requiring the development novel strategies to fight bacterial infection. This study aimed to assess the effect of silver nanoparticles (bio-AgNPs) on bacterial growth, biofilm formation, production of virulence factors, and expression of genes related to the quorum-sensing (QS) system of P. aeruginosa PAO1 and PA14. Biofilm formation and virulence assays were performed with bio-AgNPs. RT-qPCR was carried out to determine the effect of bio-AgNPs on the QS regulatory genes lasI, lasR, rhlI, rhlR, pqsA, and mvfR. Bio-AgNPs had an MIC value of 62.50 μM, for both strains. Phenotypic and genotypic assays were carried out using sub-MIC values. Experimental results showed that treatment with sub-MICs of bio-AgNPs reduced (p < 0.05) the motility and rhamnolipids and elastase production in P. aeruginosa PAO1. In PA14, bio-AgNPs stimulated swarming and twitching motilities as well as biofilm formation and elastase and pyocyanin production. Bio-AgNP treatment increased (p < 0.05) the expression of QS genes in PAO1 and PA14. Despite the different phenotypic behaviors in both strains, both showed an increase in the expression of QS genes. Demonstrating that the bio-AgNPs acted in the induction of regulation. The possible mechanism underlying the action of bio-AgNPs involves the induction of the rhl and/or pqs system of PAO1 and of the las and/or pqs system of PA14. These results suggest that exposure to low concentrations of bio-AgNPs may promote the expression of QS regulatory genes in P. aeruginosa, consequently inducing the production of virulence factors such as elastase, pyocyanin, and biofilms.
Collapse
|
79
|
Wang G, Brunel JM, Preusse M, Mozaheb N, Willger SD, Larrouy-Maumus G, Baatsen P, Häussler S, Bolla JM, Van Bambeke F. The membrane-active polyaminoisoprenyl compound NV716 re-sensitizes Pseudomonas aeruginosa to antibiotics and reduces bacterial virulence. Commun Biol 2022; 5:871. [PMID: 36008485 PMCID: PMC9411590 DOI: 10.1038/s42003-022-03836-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 08/11/2022] [Indexed: 11/09/2022] Open
Abstract
Pseudomonas aeruginosa is intrinsically resistant to many antibiotics due to the impermeability of its outer membrane and to the constitutive expression of efflux pumps. Here, we show that the polyaminoisoprenyl compound NV716 at sub-MIC concentrations re-sensitizes P. aeruginosa to abandoned antibiotics by binding to the lipopolysaccharides (LPS) of the outer membrane, permeabilizing this membrane and increasing antibiotic accumulation inside the bacteria. It also prevents selection of resistance to antibiotics and increases their activity against biofilms. No stable resistance could be selected to NV716-itself after serial passages with subinhibitory concentrations, but the transcriptome of the resulting daughter cells shows an upregulation of genes involved in the synthesis of lipid A and LPS, and a downregulation of quorum sensing-related genes. Accordingly, NV716 also reduces motility, virulence factors production, and biofilm formation. NV716 shows a unique and highly promising profile of activity when used alone or in combination with antibiotics against P. aeruginosa, combining in a single molecule anti-virulence and potentiator effects. Additional work is required to more thoroughly understand the various functions of NV716. The polyaminoisoprenyl compound NV716 re-sensitizes Pseudomonas aeruginosa to antibiotics through permeabilizing the outer membrane and increases the activity of antibiotics on biofilms.
Collapse
Affiliation(s)
- Gang Wang
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Jean-Michel Brunel
- Aix Marseille Université, INSERM, SSA, Membranes et Cibles thérapeutiques (MCT), Marseille, France
| | - Matthias Preusse
- Department of Molecular Bacteriology, Helmoltz Centre for Infection Research, Braunschweig, Germany
| | - Negar Mozaheb
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Sven D Willger
- Department of Molecular Bacteriology, Helmoltz Centre for Infection Research, Braunschweig, Germany.,Department of Molecular Bacteriology, Twincore, Hannover, Germany.,Institute for Medical Biometry and Bioinformatics, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Gerald Larrouy-Maumus
- Department of Life Sciences, Faculty of Natural Sciences, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Pieter Baatsen
- Electron Microscopy Platform & Bio Imaging Core, VIB & KULeuven Center for Brain & Disease Research, KULeuven, Leuven, Belgium
| | - Susanne Häussler
- Department of Molecular Bacteriology, Helmoltz Centre for Infection Research, Braunschweig, Germany.,Department of Molecular Bacteriology, Twincore, Hannover, Germany.,Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark.,Cluster of Excellence RESIST, Hannover Medical School, Hannover, Germany
| | - Jean-Michel Bolla
- Aix Marseille Université, INSERM, SSA, Membranes et Cibles thérapeutiques (MCT), Marseille, France
| | - Françoise Van Bambeke
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
80
|
Taylor IR, Jeffrey PD, Moustafa DA, Goldberg JB, Bassler BL. The PqsE Active Site as a Target for Small Molecule Antimicrobial Agents against Pseudomonas aeruginosa. Biochemistry 2022; 61:1894-1903. [PMID: 35985643 PMCID: PMC9454246 DOI: 10.1021/acs.biochem.2c00334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
![]()
The opportunistic pathogen Pseudomonas
aeruginosa causes antibiotic-resistant, nosocomial
infections in immuno-compromised
individuals and is a high priority for antimicrobial development.
Key to pathogenicity in P. aeruginosa are biofilm formation and virulence factor production. Both traits
are controlled by the cell-to-cell communication process called quorum
sensing (QS). QS involves the synthesis, release, and population-wide
detection of signal molecules called autoinducers. We previously reported
that the activity of the RhlR QS transcription factor depends on a
protein–protein interaction with the hydrolase, PqsE, and PqsE
catalytic activity is dispensable for this interaction. Nonetheless,
the PqsE–RhlR interaction could be disrupted by the substitution
of an active site glutamate residue with tryptophan [PqsE(E182W)].
Here, we show that disruption of the PqsE–RhlR interaction
via either the E182W change or alteration of PqsE surface residues
that are essential for the interaction with RhlR attenuates P. aeruginosa infection in a murine host. We use
crystallography to characterize the conformational changes induced
by the PqsE(E182W) substitution to define the mechanism underlying
disruption of the PqsE–RhlR interaction. A loop rearrangement
that repositions the E280 residue in PqsE(E182W) is responsible for
the loss of interaction. We verify the implications garnered from
the PqsE(E182W) structure using mutagenic, biochemical, and additional
structural analyses. We present the next generation of molecules targeting
the PqsE active site, including a structure of the tightest binding
of these compounds, BB584, in complex with PqsE. The findings presented
here provide insights into drug discovery against P.
aeruginosa with PqsE as the target.
Collapse
Affiliation(s)
- Isabelle R Taylor
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, United States
| | - Philip D Jeffrey
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, United States
| | - Dina A Moustafa
- School of Medicine, Children's Healthcare of Atlanta, Inc., Department of Pediatrics, and Center for Cystic Fibrosis and Airway Diseases Research, Emory University, Atlanta, Georgia 30322, United States
| | - Joanna B Goldberg
- School of Medicine, Children's Healthcare of Atlanta, Inc., Department of Pediatrics, and Center for Cystic Fibrosis and Airway Diseases Research, Emory University, Atlanta, Georgia 30322, United States
| | - Bonnie L Bassler
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, United States.,Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, United States
| |
Collapse
|
81
|
Qin S, Xiao W, Zhou C, Pu Q, Deng X, Lan L, Liang H, Song X, Wu M. Pseudomonas aeruginosa: pathogenesis, virulence factors, antibiotic resistance, interaction with host, technology advances and emerging therapeutics. Signal Transduct Target Ther 2022; 7:199. [PMID: 35752612 PMCID: PMC9233671 DOI: 10.1038/s41392-022-01056-1] [Citation(s) in RCA: 467] [Impact Index Per Article: 155.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 06/04/2022] [Accepted: 06/08/2022] [Indexed: 02/05/2023] Open
Abstract
Pseudomonas aeruginosa (P. aeruginosa) is a Gram-negative opportunistic pathogen that infects patients with cystic fibrosis, burn wounds, immunodeficiency, chronic obstructive pulmonary disorder (COPD), cancer, and severe infection requiring ventilation, such as COVID-19. P. aeruginosa is also a widely-used model bacterium for all biological areas. In addition to continued, intense efforts in understanding bacterial pathogenesis of P. aeruginosa including virulence factors (LPS, quorum sensing, two-component systems, 6 type secretion systems, outer membrane vesicles (OMVs), CRISPR-Cas and their regulation), rapid progress has been made in further studying host-pathogen interaction, particularly host immune networks involving autophagy, inflammasome, non-coding RNAs, cGAS, etc. Furthermore, numerous technologic advances, such as bioinformatics, metabolomics, scRNA-seq, nanoparticles, drug screening, and phage therapy, have been used to improve our understanding of P. aeruginosa pathogenesis and host defense. Nevertheless, much remains to be uncovered about interactions between P. aeruginosa and host immune responses, including mechanisms of drug resistance by known or unannotated bacterial virulence factors as well as mammalian cell signaling pathways. The widespread use of antibiotics and the slow development of effective antimicrobials present daunting challenges and necessitate new theoretical and practical platforms to screen and develop mechanism-tested novel drugs to treat intractable infections, especially those caused by multi-drug resistance strains. Benefited from has advancing in research tools and technology, dissecting this pathogen's feature has entered into molecular and mechanistic details as well as dynamic and holistic views. Herein, we comprehensively review the progress and discuss the current status of P. aeruginosa biophysical traits, behaviors, virulence factors, invasive regulators, and host defense patterns against its infection, which point out new directions for future investigation and add to the design of novel and/or alternative therapeutics to combat this clinically significant pathogen.
Collapse
Affiliation(s)
- Shugang Qin
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wen Xiao
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Chuanmin Zhou
- State Key Laboratory of Virology, School of Public Health, Wuhan University, Wuhan, 430071, P.R. China
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | - Qinqin Pu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | - Xin Deng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, People's Republic of China
| | - Lefu Lan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Haihua Liang
- College of Life Sciences, Northwest University, Xi'an, ShaanXi, 710069, China
| | - Xiangrong Song
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Min Wu
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58203, USA.
| |
Collapse
|
82
|
Sionov RV, Steinberg D. Targeting the Holy Triangle of Quorum Sensing, Biofilm Formation, and Antibiotic Resistance in Pathogenic Bacteria. Microorganisms 2022; 10:1239. [PMID: 35744757 PMCID: PMC9228545 DOI: 10.3390/microorganisms10061239] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/12/2022] [Accepted: 06/14/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic and recurrent bacterial infections are frequently associated with the formation of biofilms on biotic or abiotic materials that are composed of mono- or multi-species cultures of bacteria/fungi embedded in an extracellular matrix produced by the microorganisms. Biofilm formation is, among others, regulated by quorum sensing (QS) which is an interbacterial communication system usually composed of two-component systems (TCSs) of secreted autoinducer compounds that activate signal transduction pathways through interaction with their respective receptors. Embedded in the biofilms, the bacteria are protected from environmental stress stimuli, and they often show reduced responses to antibiotics, making it difficult to eradicate the bacterial infection. Besides reduced penetration of antibiotics through the intricate structure of the biofilms, the sessile biofilm-embedded bacteria show reduced metabolic activity making them intrinsically less sensitive to antibiotics. Moreover, they frequently express elevated levels of efflux pumps that extrude antibiotics, thereby reducing their intracellular levels. Some efflux pumps are involved in the secretion of QS compounds and biofilm-related materials, besides being important for removing toxic substances from the bacteria. Some efflux pump inhibitors (EPIs) have been shown to both prevent biofilm formation and sensitize the bacteria to antibiotics, suggesting a relationship between these processes. Additionally, QS inhibitors or quenchers may affect antibiotic susceptibility. Thus, targeting elements that regulate QS and biofilm formation might be a promising approach to combat antibiotic-resistant biofilm-related bacterial infections.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- The Biofilm Research Laboratory, The Institute of Biomedical and Oral Research, The Faculty of Dental Medicine, Hadassah Medical School, The Hebrew University, Jerusalem 9112102, Israel;
| | | |
Collapse
|
83
|
Chadha J, Ravi, Singh J, Chhibber S, Harjai K. Gentamicin Augments the Quorum Quenching Potential of Cinnamaldehyde In Vitro and Protects Caenorhabditis elegans From Pseudomonas aeruginosa Infection. Front Cell Infect Microbiol 2022; 12:899566. [PMID: 35782125 PMCID: PMC9240785 DOI: 10.3389/fcimb.2022.899566] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/29/2022] [Indexed: 11/22/2022] Open
Abstract
The quorum sensing (QS) circuitry of Pseudomonas aeruginosa represents an attractive target to attenuate bacterial virulence and antibiotic resistance. In this context, phytochemicals harboring anti-virulent properties have emerged as an alternative medicine to combat pseudomonal infections. Hence, this study was undertaken to investigate the synergistic effects and quorum quenching (QQ) potential of cinnamaldehyde (CiNN) in combination with gentamicin (GeN) against P. aeruginosa. The QQ activity of this novel combination was evaluated using a QS reporter strain and synergism was studied using chequerboard assays. Further, the genotypic and phenotypic expression of pseudomonal virulence factors was examined alongside biofilm formation. The combination of CiNN and GeN exhibited synergy and promising anti-QS activity. This drug combination was shown to suppress AHL production and downregulate the expression of critical QS genes in P. aeruginosa PAO1. Molecular docking revealed strong interactions between the QS receptors and CiNN, asserting its QQ potential. Bacterial motility was compromised along with a significant reduction in pyocyanin (72.3%), alginate (58.7%), rhamnolipid (33.6%), hemolysin (82.6%), protease (70.9%), and elastase (63.9%) production. The drug combination successfully eradicated preformed biofilms and inhibited biofilm formation by abrogating EPS production. Our findings suggest that although GeN alone could not attenuate QS, but was able to augment the anti-QS potential of CiNN. To validate our results using an infection model, we quantified the survival rates of Caenorhabditis elegans following PAO1 challenge. The combination significantly rescued C. elegans from PAO1 infection and improved its survival rate by 54% at 96 h. In summary, this study is the first to elucidate the mechanism behind the QQ prospects of CiNN (augmented in presence of GeN) by abrogating AHL production and increasing the survival rate of C. elegans, thereby highlighting its anti-virulent properties.
Collapse
|
84
|
Lal R, Singh BK, Sar P, Phale P. The assessment of microbial ecology: a special emphasis on the Indian scenario. ENVIRONMENTAL MICROBIOLOGY REPORTS 2022; 14:325-329. [PMID: 35362197 DOI: 10.1111/1758-2229.13067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 03/26/2022] [Indexed: 06/14/2023]
Affiliation(s)
- Rup Lal
- The Energy & Resources Institute, New Delhi, 110003, India
| | - Brajesh K Singh
- Global Centre for Land-Based Innovation, Western Sydney University, Penrith, NSW, 2751, Australia
- Hawkesbury Institute for the Environment, Western Sydney University, Penrith, NSW, 2751, Australia
| | - Pinaki Sar
- Environmental Microbiology and Genomics Laboratory, Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Prashant Phale
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| |
Collapse
|
85
|
Xu N, Zhao Q, Zhang Z, Zhang Q, Wang Y, Qin G, Ke M, Qiu D, Peijnenburg WJGM, Lu T, Qian H. Phyllosphere Microorganisms: Sources, Drivers, and Their Interactions with Plant Hosts. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:4860-4870. [PMID: 35435673 DOI: 10.1021/acs.jafc.2c01113] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The leaves of plants are colonized by various microorganisms. In comparison to the rhizosphere, less is known about the characteristics and ecological functions of phyllosphere microorganisms. Phyllosphere microorganisms mainly originate from soil, air, and seeds. The composition of phyllosphere microorganisms is mainly affected by ecological and abiotic factors. Phyllosphere microorganisms execute multiple ecological functions by influencing leaf functions and longevity, seed mass, fruit development, and homeostasis of host growth. A plant can respond to phyllosphere microorganisms by secondary metabolite secretion and its immune system. Meanwhile, phyllosphere microorganisms play an important role in ecological stability and environmental safety assessment. However, as a result of the instability of the phyllosphere environment and the poor cultivability of phyllosphere microorganisms in the current research, there are still many limitations, such as the lack of insight into the mechanisms of plant-microorganism interactions, the roles of phyllosphere microorganisms in plant growth processes, the responses of phyllosphere microorganisms to plant metabolites, etc. This review summarizes the latest progress made in the research of the phyllosphere in recent years. This is beneficial for deepening our understanding of phyllosphere microorganisms and promoting the research of plant-atmosphere interactions, plant pathogens, and plant biological control.
Collapse
Affiliation(s)
- Nuohan Xu
- College of Environment, Zhejiang University of Technology, Hangzhou, Zhejiang 310032, People's Republic of China
| | - Qianqiu Zhao
- Xinjiang Institute of Ecology and Geography, Chinese Academy of Science, Urumqi, Xinjiang 830011, People's Republic of China
| | - Zhenyan Zhang
- College of Environment, Zhejiang University of Technology, Hangzhou, Zhejiang 310032, People's Republic of China
| | - Qi Zhang
- College of Environment, Zhejiang University of Technology, Hangzhou, Zhejiang 310032, People's Republic of China
| | - Yan Wang
- College of Environment, Zhejiang University of Technology, Hangzhou, Zhejiang 310032, People's Republic of China
| | - Guoyan Qin
- College of Environment, Zhejiang University of Technology, Hangzhou, Zhejiang 310032, People's Republic of China
| | - Mingjing Ke
- College of Environment, Zhejiang University of Technology, Hangzhou, Zhejiang 310032, People's Republic of China
| | - Danyan Qiu
- College of Environment, Zhejiang University of Technology, Hangzhou, Zhejiang 310032, People's Republic of China
| | - W J G M Peijnenburg
- Institute of Environmental Sciences (CML), Leiden University, 2300 RA Leiden, Netherlands
- National Institute of Public Health and the Environment (RIVM), Center for Safety of Substances and Products, Post Office Box 1, 3720 BA Bilthoven, Netherlands
| | - Tao Lu
- College of Environment, Zhejiang University of Technology, Hangzhou, Zhejiang 310032, People's Republic of China
| | - Haifeng Qian
- College of Environment, Zhejiang University of Technology, Hangzhou, Zhejiang 310032, People's Republic of China
| |
Collapse
|
86
|
Khullar L, Harjai K, Chhibber S. Exploring the therapeutic potential of staphylococcal phage formulations: Current challenges and applications in phage therapy. J Appl Microbiol 2022; 132:3515-3532. [DOI: 10.1111/jam.15462] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/21/2021] [Accepted: 01/17/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Lavanya Khullar
- Department of Microbiology Panjab University Chandigarh India
| | - Kusum Harjai
- Department of Microbiology Panjab University Chandigarh India
| | - Sanjay Chhibber
- Department of Microbiology Panjab University Chandigarh India
| |
Collapse
|
87
|
Park Y, Koo SH. Epidemiology, Molecular Characteristics, and Virulence Factors of Carbapenem-Resistant Pseudomonas aeruginosa Isolated from Patients with Urinary Tract Infections. Infect Drug Resist 2022; 15:141-151. [PMID: 35058697 PMCID: PMC8765443 DOI: 10.2147/idr.s346313] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/30/2021] [Indexed: 12/20/2022] Open
Abstract
Purpose Pseudomonas aeruginosa is an important pathogen that causes urinary tract infections. Carbapenems are the drugs of choice for the treatment of P. aeruginosa infections. However, the emergence and spread of carbapenem-resistant P. aeruginosa (CRPA) is a serious global health threat. In this study, we investigated the epidemiology, molecular characteristics, drug resistance, and virulence factors of CRPA isolated from urine samples. Methods A total of 124 P. aeruginosa isolates were obtained from urine samples collected between March 2020 and February 2021. Clonal relatedness was evaluated using multilocus sequence typing (MLST) and pulsed-field gel electrophoresis (PFGE). We performed antimicrobial susceptibility tests and investigated the presence of carbapenemase genes and virulence factors in CRPA isolates. Results The carbapenem resistance rate of the isolated P. aeruginosa was 46.7% (59/124). A total of 54 (91.5%) out of the 59 CRPA isolates were identified as multidrug-resistant. The majority of the CRPA isolates (81.4%, 48/59) harbored carbapenemase genes, such as blaIMP-6 or blaNDM-1. In an epidemiological analysis using MLST, 88.1% of CRPA isolates were confirmed to be ST773 (50.8%, 30/59) or ST235 (37.3%, 22/59). The CRPA isolates harboring blaIMP-6 and blaNDM-1 belonged to ST235 (PFGE pulsotypes E1-E18, F) and ST773 (PFGE pulsotypes H1-H2, I1-I16) subtypes, respectively. The studied CRPA isolates simultaneously harbored 10 to 14 virulence factors of the 16 virulence factors examined. Nine virulence factor genes (toxA, exoT, plcH, plcN, phzM, phzS, lasB, aprA, and algD) were identified in all CRPA isolates. Conclusion Our study shows that P. aeruginosa ST235 harboring blaIMP-6 and ST773 harboring blaNDM-1—known internationally as high-risk clones with multiple virulence factors—are widely spread in the study area. These findings suggest that continuous monitoring is necessary to prevent the further spread of carbapenemase-producing CRPA.
Collapse
Affiliation(s)
- Yumi Park
- Department of Laboratory Medicine, Konyang University College of Medicine and Myunggok Medical Research Center, Daejeon, Republic of Korea
- Department of Laboratory Medicine, Konyang University Hospital, Daejeon, Republic of Korea
| | - Sun Hoe Koo
- Department of Laboratory Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
- Correspondence: Sun Hoe Koo Department of Laboratory Medicine, Chungnam National University Hospital, 282 Munhwa-ro, Jung-gu, Daejeon, 35015, Republic of KoreaTel +82-42-280-7798Fax +82-42-280-5365 Email
| |
Collapse
|
88
|
Li X, Gu N, Huang TY, Zhong F, Peng G. Pseudomonas aeruginosa: A typical biofilm forming pathogen and an emerging but underestimated pathogen in food processing. Front Microbiol 2022; 13:1114199. [PMID: 36762094 PMCID: PMC9905436 DOI: 10.3389/fmicb.2022.1114199] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 12/30/2022] [Indexed: 01/26/2023] Open
Abstract
Pseudomonas aeruginosa (P. aeruginosa) is a notorious gram-negative pathogenic microorganism, because of several virulence factors, biofilm forming capability, as well as antimicrobial resistance. In addition, the appearance of antibiotic-resistant strains resulting from the misuse and overuse of antibiotics increases morbidity and mortality in immunocompromised patients. However, it has been underestimated as a foodborne pathogen in various food groups for instance water, milk, meat, fruits, and vegetables. Chemical preservatives that are commonly used to suppress the growth of food source microorganisms can cause problems with food safety. For these reasons, finding effective, healthy safer, and natural alternative antimicrobial agents used in food processing is extremely important. In this review, our ultimate goal is to cover recent advances in food safety related to P. aeruginosa including antimicrobial resistance, major virulence factors, and prevention measures. It is worth noting that food spoilage caused by P. aeruginosa should arouse wide concerns of consumers and food supervision department.
Collapse
Affiliation(s)
- Xuejie Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou, China
- Research Institute for Food Nutrition and Human Health, Guangzhou, China
| | - Nixuan Gu
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou, China
| | - Teng Yi Huang
- Department of Diagnostics, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Feifeng Zhong
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou, China
| | - Gongyong Peng
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- *Correspondence: Gongyong Peng, ✉
| |
Collapse
|
89
|
Iron Homeostasis in Pseudomonas aeruginosa: Targeting Iron Acquisition and Storage as an Antimicrobial Strategy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1386:29-68. [DOI: 10.1007/978-3-031-08491-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
90
|
Chadha J, Harjai K, Chhibber S. Repurposing phytochemicals as anti-virulent agents to attenuate quorum sensing-regulated virulence factors and biofilm formation in Pseudomonas aeruginosa. Microb Biotechnol 2021; 15:1695-1718. [PMID: 34843159 PMCID: PMC9151347 DOI: 10.1111/1751-7915.13981] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 11/14/2021] [Indexed: 12/18/2022] Open
Abstract
Unregulated consumption and overexploitation of antibiotics have paved the way for emergence of antibiotic‐resistant strains and ‘superbugs’. Pseudomonas aeruginosa is among the opportunistic nosocomial pathogens causing devastating infections in clinical set‐ups globally. Its artillery equipped with diversified virulence elements, extensive antibiotic resistance and biofilms has made it a ‘hard‐to‐treat’ pathogen. The pathogenicity of P. aeruginosa is modulated by an intricate cell density‐dependent mechanism called quorum sensing (QS). The virulence artillery of P. aeruginosa is firmly controlled by QS genes, and their expression drives the aggressiveness of the infection. Attempts to identify and develop novel antimicrobials have seen a sharp rise in the past decade. Among different proposed mechanisms, a novel anti‐virulence approach to target pseudomonal infections by virtue of anti‐QS and anti‐biofilm drugs appears to occupy the centre stage. In this respect, bioactive phytochemicals have gained prominence among the scientific community owing to their significant quorum quenching (QQ) properties. Recent studies have shed light on the QQ activities of various phytochemicals and other drugs in perturbing the QS‐dependent virulence in P. aeruginosa. This review highlights the recent evidences that reinforce the application of plant bioactives for combating pseudomonal infections, their advantages and shortcomings in anti‐virulence therapy.
Collapse
Affiliation(s)
- Jatin Chadha
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Kusum Harjai
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Sanjay Chhibber
- Department of Microbiology, Panjab University, Chandigarh, India
| |
Collapse
|
91
|
Wagener BM, Hu R, Wu S, Pittet JF, Ding Q, Che P. The Role of Pseudomonas aeruginosa Virulence Factors in Cytoskeletal Dysregulation and Lung Barrier Dysfunction. Toxins (Basel) 2021; 13:776. [PMID: 34822560 PMCID: PMC8625199 DOI: 10.3390/toxins13110776] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/24/2021] [Accepted: 10/28/2021] [Indexed: 12/19/2022] Open
Abstract
Pseudomonas (P.) aeruginosa is an opportunistic pathogen that causes serious infections and hospital-acquired pneumonia in immunocompromised patients. P. aeruginosa accounts for up to 20% of all cases of hospital-acquired pneumonia, with an attributable mortality rate of ~30-40%. The poor clinical outcome of P. aeruginosa-induced pneumonia is ascribed to its ability to disrupt lung barrier integrity, leading to the development of lung edema and bacteremia. Airway epithelial and endothelial cells are important architecture blocks that protect the lung from invading pathogens. P. aeruginosa produces a number of virulence factors that can modulate barrier function, directly or indirectly, through exploiting cytoskeleton networks and intercellular junctional complexes in eukaryotic cells. This review summarizes the current knowledge on P. aeruginosa virulence factors, their effects on the regulation of the cytoskeletal network and associated components, and molecular mechanisms regulating barrier function in airway epithelial and endothelial cells. A better understanding of these processes will help to lay the foundation for new therapeutic approaches against P. aeruginosa-induced pneumonia.
Collapse
Affiliation(s)
- Brant M. Wagener
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (B.M.W.); (R.H.); (S.W.); (J.-F.P.); (Q.D.)
- Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Division of Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ruihan Hu
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (B.M.W.); (R.H.); (S.W.); (J.-F.P.); (Q.D.)
- Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Internal Medicine, Guiqian International General Hospital, Guiyang 550024, China
| | - Songwei Wu
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (B.M.W.); (R.H.); (S.W.); (J.-F.P.); (Q.D.)
- Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jean-Francois Pittet
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (B.M.W.); (R.H.); (S.W.); (J.-F.P.); (Q.D.)
- Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Division of Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Qiang Ding
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (B.M.W.); (R.H.); (S.W.); (J.-F.P.); (Q.D.)
- Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Pulin Che
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (B.M.W.); (R.H.); (S.W.); (J.-F.P.); (Q.D.)
- Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|