1
|
Boughroud H, Chgari O, Wahnou H, Amrani AE, Faqer OE, Amarir F, Mtairag EM, Rais S, Bourjilat F. Assessing Syzygium Aromaticum: From Chemical Profiling to Antioxidant, Antistaphylococcal, and Biocompatibility Attributes. Chem Biodivers 2025; 22:e202402605. [PMID: 39602658 DOI: 10.1002/cbdv.202402605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 11/29/2024]
Abstract
Staphylococcal infections challenge current treatments due to adverse effects and drug resistance, prompting the exploration of plant-derived compounds for their promising properties. This study aimed to analyze Syzygium aromaticum essential oil (SA-EO), aqueous (AE-SA) and ethyl acetate (EAE-SA) extracts for antioxidant and antistaphylococcal effects against sensitive and resistant clinical isolates. Erythrocytes were used as model cells for biocompatibility. The SA-EO was extracted via hydrodistillation and compounds were identified using GC-MS. The antioxidant activity was assessed using various tests, while antimicrobial activity was evaluated through disk diffusion and microdilution assays. Results showed Eugenol as the major compound in SA-EO. The AE exhibited high phenolic and flavonoid content. SA-EO demonstrated antioxidant activity with IC50 values of 84.63 ± 1.94 µg/mL (TAC) and 52.88 ± 1.44 µg/mL (FRAP). The AE-SA showed scavenging effects with EC50 values of 266.52 ± 50.62 µg/mL (DPPH) and 140.67 ± 44.36 µg/mL (NO). The antistaphylococcal activity revealed significant sensitivity of SA-EO with MIC and MBC values ranging from 0.316 to 1.266 mg/mL. SA-EO showed low hemolytic activity even at high concentrations, indicating its safe therapeutic application. Overall, these findings highlight the potential of Syzygium aromaticum in combating multidrug-resistant bacteria and underscore its role as a source of bioactive compounds with antioxidant properties.
Collapse
Affiliation(s)
- Hajar Boughroud
- Immunology and Biodiversity Laboratory, Faculty of Sciences Ain Chock, Hassan II University, Casablanca, Morocco
- Medical Microbiology Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Oumaima Chgari
- Immunology and Biodiversity Laboratory, Faculty of Sciences Ain Chock, Hassan II University, Casablanca, Morocco
| | - Hicham Wahnou
- Immunology and Biodiversity Laboratory, Faculty of Sciences Ain Chock, Hassan II University, Casablanca, Morocco
| | - Abdelaziz El Amrani
- Laboratory of Organic Synthesis, Extraction, and Valorization, Faculty of Sciences Ain Chock, Hassan II University, Casablanca, Morocco
| | - Othman El Faqer
- Immunology and Biodiversity Laboratory, Faculty of Sciences Ain Chock, Hassan II University, Casablanca, Morocco
| | - Fatima Amarir
- Immunology and Biodiversity Laboratory, Faculty of Sciences Ain Chock, Hassan II University, Casablanca, Morocco
| | - El Mostafa Mtairag
- Immunology and Biodiversity Laboratory, Faculty of Sciences Ain Chock, Hassan II University, Casablanca, Morocco
| | - Samira Rais
- Immunology and Biodiversity Laboratory, Faculty of Sciences Ain Chock, Hassan II University, Casablanca, Morocco
- Department of Biology, Faculty of Science Ben M'Sick, Hassan II University, Casablanca, Morocco
| | - Fatna Bourjilat
- Medical Microbiology Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| |
Collapse
|
2
|
Cao J, Li H, Han Q, Li Z, Zhuang J, Dong C, Li A. The accessory secretion system in Streptococcus agalactiae regulates protein secretion, stress resistance, adhesion, immune evasion, and virulence. FISH & SHELLFISH IMMUNOLOGY 2025; 158:110172. [PMID: 39909122 DOI: 10.1016/j.fsi.2025.110172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/21/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
Streptococcus agalactiae is a significant co-pathogenic bacterium in humans and animals, including fish. Bacteria secrete a variety of proteins through an accessory secretion system to modulate their interactions with the host. To investigate the role of the accessory secretion system in S. agalactiae, a deletion mutant strain (ΔaccSec) was constructed via homologous recombination. The accessory secretion system was found to be essential for the viability of S. agalactiae, and its absence led to increased cell death and lysis. In the extracellular fraction of the ΔaccSec mutant, a reduction in the secretion of 33 proteins was observed. Analyses of biological properties indicated that ΔaccSec exhibited significantly reduced stress tolerance and envelope stability. Pathogenicity experiments demonstrated that the ΔaccSec mutant had significantly lower adhesion to cells and fish tissues, as well as decreased resistance to whole blood killing and phagocytosis by macrophages. The cumulative mortality of ΔaccSec in tilapia after intraperitoneal injection was reduced by 55.3-74.2 %. The ΔaccSec mutant exhibited a markedly diminished capacity for colonization in tilapia. Furthermore, we found that ΔaccSec mutant induced higher macrophage reactive oxygen species (ROS) levels and significantly upregulated MHC-II, TLR-2 transcript levels in tilapia spleens compared to the wild-type. Overall, these findings underscore the importance of the accessory secretion system in S. agalactiae pathogenicity, particularly in stabilizing the bacterial envelope, facilitating adhesion, and evading host immunity. The results of this study provide new insights into the mechanisms of virulence regulation in S. agalactiae and lay a foundation for developing live attenuated vaccines.
Collapse
Affiliation(s)
- Jizhen Cao
- State Key Laboratory of Biocontrol/Guangdong Provincial Key Laboratory of Improved Variety Reproduction in Aquatic Economic Animals and Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, PR China
| | - Han Li
- State Key Laboratory of Biocontrol/Guangdong Provincial Key Laboratory of Improved Variety Reproduction in Aquatic Economic Animals and Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, PR China
| | - Qing Han
- State Key Laboratory of Biocontrol/Guangdong Provincial Key Laboratory of Improved Variety Reproduction in Aquatic Economic Animals and Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, PR China
| | - Zhicheng Li
- State Key Laboratory of Biocontrol/Guangdong Provincial Key Laboratory of Improved Variety Reproduction in Aquatic Economic Animals and Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, PR China
| | - Jingyu Zhuang
- State Key Laboratory of Biocontrol/Guangdong Provincial Key Laboratory of Improved Variety Reproduction in Aquatic Economic Animals and Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, PR China
| | - Chuanfu Dong
- State Key Laboratory of Biocontrol/Guangdong Provincial Key Laboratory of Improved Variety Reproduction in Aquatic Economic Animals and Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, PR China
| | - Anxing Li
- State Key Laboratory of Biocontrol/Guangdong Provincial Key Laboratory of Improved Variety Reproduction in Aquatic Economic Animals and Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, PR China.
| |
Collapse
|
3
|
Mapar M, Rydzak T, Hommes JW, Surewaard BGJ, Lewis IA. Diverse molecular mechanisms underpinning Staphylococcus aureus small colony variants. Trends Microbiol 2025; 33:223-232. [PMID: 39393939 DOI: 10.1016/j.tim.2024.09.007] [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: 05/01/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/13/2024]
Abstract
Small colony variants (SCVs) of Staphylococcus aureus are a relatively rare but clinically significant growth morphotype. Infections with SCVs are frequently difficult to treat, inherently antibiotic-resistant, and can lead to persistent infections. Despite a long history of research, the molecular underpinnings of this morphotype and their impact on the clinical trajectory of infections remain unclear. However, a growing body of literature indicates that SCVs are caused by a diverse range of molecular factors. These recent findings suggest that SCVs should be thought of as an ensemble collection of loosely related phenotypes, and not as a single phenomenon. This review describes the diverse mechanisms currently known to contribute to SCVs and proposes an ensemble model for conceptualizing this morphotype.
Collapse
Affiliation(s)
- Maryam Mapar
- Alberta Centre for Advanced Diagnostics, Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Thomas Rydzak
- Alberta Centre for Advanced Diagnostics, Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Josefien W Hommes
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Bas G J Surewaard
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ian A Lewis
- Alberta Centre for Advanced Diagnostics, Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
4
|
Zelmer AR, Yang D, Gunn NJ, Solomon LB, Nelson R, Kidd SP, Richter K, Atkins GJ. Osteomyelitis-relevant antibiotics at clinical concentrations show limited effectivity against acute and chronic intracellular S. aureus infections in osteocytes. Antimicrob Agents Chemother 2024; 68:e0080824. [PMID: 39194210 PMCID: PMC11459924 DOI: 10.1128/aac.00808-24] [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: 06/02/2024] [Accepted: 08/09/2024] [Indexed: 08/29/2024] Open
Abstract
Osteomyelitis caused by Staphylococcus aureus can involve the persistent infection of osteocytes. We sought to determine if current clinically utilized antibiotics were capable of clearing an intracellular osteocyte S. aureus infection. Rifampicin, vancomycin, levofloxacin, ofloxacin, amoxicillin, oxacillin, doxycycline, linezolid, gentamicin, and tigecycline were assessed for their minimum inhibitory concentration (MIC) and minimum bactericidal concentrations against 12 S. aureus strains, at pH 5.0 and 7.2 to mimic lysosomal and cytoplasmic environments, respectively. Those antibiotics whose bone estimated achievable concentration was commonly above their respective MIC for the strains tested were further assayed in a human osteocyte infection model under acute and chronic conditions. Osteocyte-like cells were treated at 1×, 4×, and 10× the MIC for 1 and 7 days following infection (acute model), or at 15 and 21 days of infection (chronic model). The intracellular effectivity of each antibiotic was measured in terms of CFU reduction, small colony variant formation, and bacterial mRNA expression change. Only rifampicin, levofloxacin, and linezolid reduced intracellular CFU numbers significantly in the acute model. Consistent with the transition to a non-culturable state, few if any CFU could be recovered from the chronic model. However, no treatment in either model reduced the quantity of bacterial mRNA or prevented non-culturable bacteria from returning to a culturable state. These findings indicate that S. aureus adapts phenotypically during intracellular infection of osteocytes, adopting a reversible quiescent state that is protected against antibiotics, even at 10× their MIC. Thus, new therapeutic approaches are necessary to cure S. aureus intracellular infections in osteomyelitis.
Collapse
Affiliation(s)
- Anja R. Zelmer
- Center for Orthopedic and Trauma Research, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| | - Dongqing Yang
- Center for Orthopedic and Trauma Research, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| | - Nicholas J. Gunn
- Center for Orthopedic and Trauma Research, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| | - L. Bogdan Solomon
- Center for Orthopedic and Trauma Research, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
- Department of Orthopedics and Trauma, Royal Adelaide Hospital, Adelaide, Australia
| | - Renjy Nelson
- Department of Infectious Diseases, Central Adelaide Local Health Network, Adelaide, Australia
| | - Stephen P. Kidd
- Australian Center for Antimicrobial Resistance Ecology, University of Adelaide, Adelaide, Australia
- Research Center for Infectious Disease, School of Biological Science, University of Adelaide, Adelaide, Australia
| | - Katharina Richter
- Department of Surgery, Richter Lab, Basil Hetzel Institute for Translational Health Research, University of Adelaide, Adelaide, Australia
- Institute for Photonics and Advanced Sensing, University of Adelaide, Adelaide, Australia
| | - Gerald J. Atkins
- Center for Orthopedic and Trauma Research, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| |
Collapse
|
5
|
Gaudreau A, Watson DW, Flannagan RS, Roy P, Shen C, Abdelmoneim A, Beavers WN, Gillies ER, El-Halfawy OM, Heinrichs DE. Mechanistic insights and in vivo efficacy of thiosemicarbazones against methicillin-resistant Staphylococcus aureus. J Biol Chem 2024; 300:107689. [PMID: 39159815 PMCID: PMC11492055 DOI: 10.1016/j.jbc.2024.107689] [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: 06/05/2024] [Revised: 07/27/2024] [Accepted: 08/07/2024] [Indexed: 08/21/2024] Open
Abstract
Staphylococcus aureus poses a significant threat in both community and hospital settings due to its infective and pathogenic nature combined with its ability to resist the action of chemotherapeutic agents. Methicillin-resistant S. aureus (MRSA) represents a critical challenge. Metal-chelating thiosemicarbazones (TSCs) have shown promise in combating MRSA and while previous studies hinted at the antimicrobial potential of TSCs, their mechanisms of action against MRSA are still under investigation. We screened a chemical library for anti-staphylococcal compounds and identified a potent molecule named R91 that contained the NNSN structural motif found within TSCs. We identified that R91 and several structural analogs exhibited antimicrobial activity against numerous S. aureus isolates as well as other Gram-positive bacteria. RNAseq analysis revealed that R91 induces copper and oxidative stress responses. Checkerboard assays demonstrated synergy of R91 with copper, nickel, and zinc. Mutation of the SrrAB two-component regulatory system sensitizes S. aureus to R91 killing, further linking the oxidative stress response to R91 resistance. Moreover, R91 was found to induce hydrogen peroxide production, which contributed to its antimicrobial activity. Remarkably, no mutants with elevated R91 resistance were identified, despite extensive attempts. We further demonstrate that R91 can be used to effectively treat an intracellular reservoir of S. aureus in cell culture and can reduce bacterial burdens in a murine skin infection model. Combined, these data position R91 as a potent TSC effective against MRSA and other Gram-positive bacteria, with implications for future therapeutic development.
Collapse
Affiliation(s)
- Avery Gaudreau
- Department of Microbiology and Immunology, The University of Western Ontario, London, Ontario, Canada
| | - David W Watson
- Department of Microbiology and Immunology, The University of Western Ontario, London, Ontario, Canada
| | - Ronald S Flannagan
- Department of Microbiology and Immunology, The University of Western Ontario, London, Ontario, Canada
| | - Paroma Roy
- Department of Microbiology and Immunology, The University of Western Ontario, London, Ontario, Canada
| | - Chenfangfei Shen
- Department of Chemistry, The University of Western Ontario, London, Ontario, Canada
| | - Ahmed Abdelmoneim
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA; Louisiana Animal Disease Diagnostic Laboratory, Louisiana State University, Baton Rouge, Louisiana, USA
| | - William N Beavers
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Elizabeth R Gillies
- Department of Chemistry, The University of Western Ontario, London, Ontario, Canada; Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario, Canada
| | - Omar M El-Halfawy
- Department of Chemistry and Biochemistry, University of Regina, Regina, Saskatchewan, Canada; Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - David E Heinrichs
- Department of Microbiology and Immunology, The University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
6
|
Burford-Gorst CM, Kidd SP. Phenotypic Variation in Staphylococcus aureus during Colonisation Involves Antibiotic-Tolerant Cell Types. Antibiotics (Basel) 2024; 13:845. [PMID: 39335018 PMCID: PMC11428495 DOI: 10.3390/antibiotics13090845] [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: 07/31/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
Staphylococcus aureus is a bacterial species that is commonly found colonising healthy individuals but that presents a paradoxical nature: simultaneously, it can migrate within the body and cause a range of diseases. Many of these become chronic by resisting immune responses, antimicrobial treatment, and medical intervention. In part, this ability to persist can be attributed to the adoption of multiple cell types within a single cellular population. These dynamics in the S. aureus cell population could be the result of its interplay with host cells or other co-colonising bacteria-often coagulase-negative Staphylococcal (CoNS) species. Further understanding of the unique traits of S. aureus alternative cell types, the drivers for their selection or formation during disease, as well as their presence even during non-pathological colonisation could advance the development of diagnostic tools and drugs tailored to target specific cells that are eventually responsible for chronic infections.
Collapse
Affiliation(s)
- Chloe M Burford-Gorst
- Department of Molecular and Biomedical Sciences, School of Biological Sciences, The University of Adelaide, Adelaide, SA 5005, Australia
- Research Centre for Infectious Diseases (RCID), The University of Adelaide, Adelaide, SA 5005, Australia
| | - Stephen P Kidd
- Department of Molecular and Biomedical Sciences, School of Biological Sciences, The University of Adelaide, Adelaide, SA 5005, Australia
- Research Centre for Infectious Diseases (RCID), The University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
7
|
Yang R, Xu Y, Xu J, Li Y, Wan X, Kong R, Ding C, Tao H, Wang HL. The transcriptional changes of LrgA discriminates the responsiveness of Staphylococcus aureus towards blue light from that of photodynamic inactivation. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 258:112967. [PMID: 38996773 DOI: 10.1016/j.jphotobiol.2024.112967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 05/28/2024] [Accepted: 06/25/2024] [Indexed: 07/14/2024]
Abstract
Antimicrobial blue light (aBL) is utilized as a new approach to inhibit the growth of Staphylococcus aureus (S. aureus). Mediated by the endogenous chromophore, aBL possesses the similar photokilling property with aPDI (antimicrobial photodynamic inactivation), however, their mechanistic discrepancies in triggering the death of staphylococcal cells are not yet understood. Here, we describe the use of a 460-nm-LED to curb the viability of S. aureus. According to the results, the bacterial survival was sharply decreased when blue light was applied, reaching a maximum of 4.11 ± 0.04 log10 units. Moreover, the membrane integrity was damaged by aBL, causing the leakage of intracellular DNA. Transcriptomic analysis indicates the divergent gene expression upon either aBL or aPDI, with pathways such as transport, DNA repair, expression regulation and porphyrin massively affected by aBL. Among the commonly regulated genes, LrgA was underpinned on account of its involvement with biofilm formation and protein transport. By comparing the wildtype with the LrgA-overexpressing (LrgA+) strain, the survival rate, membrane penetration, surface structure and biofilm formation were, to a varying degree, improved for LrgA+, which may suggest that LrgA plays essential roles in modulating the responsiveness of S. aureus. Besides, LrgA may function through regulating the expression of autolysis-related systems. Finally, LrgA overexpression did not attenuate but aggravate the impairment induced by aPDI, showcasing a distinct responsive strategy from aBL. Taken together, this study unveils a unique molecular alteration for the aBL-mediated inactivation, providing the basis of utilizing blue light to reduce the harm brought by S. aureus.
Collapse
Affiliation(s)
- Ruili Yang
- School of Food Science and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Yi Xu
- School of Food Science and Biological Engineering, Hefei University of Technology, Hefei 230009, China.
| | - Jinchun Xu
- School of Food Science and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Yali Li
- School of Food Science and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Xiaoxiao Wan
- School of Food Science and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Rui Kong
- School of Food Science and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Chao Ding
- School of Food Science and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Han Tao
- School of Food Science and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Hui-Li Wang
- School of Food Science and Biological Engineering, Hefei University of Technology, Hefei 230009, China.
| |
Collapse
|
8
|
Cheng K, Sun Y, Yu H, Hu Y, He Y, Shen Y. Staphylococcus aureus SOS response: Activation, impact, and drug targets. MLIFE 2024; 3:343-366. [PMID: 39359682 PMCID: PMC11442139 DOI: 10.1002/mlf2.12137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/17/2024] [Accepted: 04/10/2024] [Indexed: 10/04/2024]
Abstract
Staphylococcus aureus is a common cause of diverse infections, ranging from superficial to invasive, affecting both humans and animals. The widespread use of antibiotics in clinical treatments has led to the emergence of antibiotic-resistant strains and small colony variants. This surge presents a significant challenge in eliminating infections and undermines the efficacy of available treatments. The bacterial Save Our Souls (SOS) response, triggered by genotoxic stressors, encompasses host immune defenses and antibiotics, playing a crucial role in bacterial survival, invasiveness, virulence, and drug resistance. Accumulating evidence underscores the pivotal role of the SOS response system in the pathogenicity of S. aureus. Inhibiting this system offers a promising approach for effective bactericidal treatments and curbing the evolution of antimicrobial resistance. Here, we provide a comprehensive review of the activation, impact, and key proteins associated with the SOS response in S. aureus. Additionally, perspectives on therapeutic strategies targeting the SOS response for S. aureus, both individually and in combination with traditional antibiotics are proposed.
Collapse
Affiliation(s)
- Kaiying Cheng
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of MedicineZhejiang UniversityHangzhouChina
| | - Yukang Sun
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
| | - Huan Yu
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
| | - Yingxuan Hu
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
| | - Yini He
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
| | - Yuanyuan Shen
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouChina
| |
Collapse
|
9
|
Niu H, Gu J, Zhang Y. Bacterial persisters: molecular mechanisms and therapeutic development. Signal Transduct Target Ther 2024; 9:174. [PMID: 39013893 PMCID: PMC11252167 DOI: 10.1038/s41392-024-01866-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 07/18/2024] Open
Abstract
Persisters refer to genetically drug susceptible quiescent (non-growing or slow growing) bacteria that survive in stress environments such as antibiotic exposure, acidic and starvation conditions. These cells can regrow after stress removal and remain susceptible to the same stress. Persisters are underlying the problems of treating chronic and persistent infections and relapse infections after treatment, drug resistance development, and biofilm infections, and pose significant challenges for effective treatments. Understanding the characteristics and the exact mechanisms of persister formation, especially the key molecules that affect the formation and survival of the persisters is critical to more effective treatment of chronic and persistent infections. Currently, genes related to persister formation and survival are being discovered and confirmed, but the mechanisms by which bacteria form persisters are very complex, and there are still many unanswered questions. This article comprehensively summarizes the historical background of bacterial persisters, details their complex characteristics and their relationship with antibiotic tolerant and resistant bacteria, systematically elucidates the interplay between various bacterial biological processes and the formation of persister cells, as well as consolidates the diverse anti-persister compounds and treatments. We hope to provide theoretical background for in-depth research on mechanisms of persisters and suggest new ideas for choosing strategies for more effective treatment of persistent infections.
Collapse
Affiliation(s)
- Hongxia Niu
- School of Basic Medical Science and Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Jiaying Gu
- School of Basic Medical Science and Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Ying Zhang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250022, Shandong, China.
| |
Collapse
|
10
|
Sivasubramaniam BP, Washer BM, Watanabe Y, Ragheb KE, Robinson JP, Wei A. Photodynamic treatment of Staphylococcus aureus with non-iron hemin analogs in the presence of hydrogen peroxide. RSC Med Chem 2024; 15:2138-2145. [PMID: 38911164 PMCID: PMC11187572 DOI: 10.1039/d4md00148f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/02/2024] [Indexed: 06/25/2024] Open
Abstract
Bacteria subjected to antiseptic or antibiotic stress often develop tolerance, a trait that can lead to permanent resistance. To determine whether photodynamic agents could be used to counter tolerance, we evaluated three non-iron hemin analogs (M-PpIX; M = Al, Ga, In) as targeted photosensitizers for antimicrobial photodynamic inactivation (aPDI) following exposure to sublethal H2O2. Al-PpIX is an active producer of ROS whereas Ga- and In-PpIX are more efficient at generating singlet oxygen. Al- and Ga-PpIX are highly potent aPDI agents against S. aureus and methicillin-resistant strains (MRSA) with antimicrobial activity (3 log reduction in colony-forming units) at nanomolar concentrations. The aPDI activities of Al- and Ga-PpIX against S. aureus were tested in the presence of 1 mM H2O2 added at different stages of growth. Bacteria exposed to H2O2 during log-phase growth were less susceptible to aPDI but bacteria treated with H2O2 in their postgrowth phase exhibited aPDI hypersensitivity, with no detectable colony growth after treatment with 15 nM Ga-PpIX.
Collapse
Affiliation(s)
| | - Benjamin M Washer
- Department of Chemistry, Purdue University 560 Oval Drive West Lafayette IN 47907 USA
| | - Yuichiro Watanabe
- Department of Chemistry, Purdue University 560 Oval Drive West Lafayette IN 47907 USA
| | - Kathryn E Ragheb
- College of Veterinary Medicine, Purdue University 625 Harrison Street West Lafayette IN 47907 USA
| | - J Paul Robinson
- College of Veterinary Medicine, Purdue University 625 Harrison Street West Lafayette IN 47907 USA
| | - Alexander Wei
- Department of Chemistry, Purdue University 560 Oval Drive West Lafayette IN 47907 USA
| |
Collapse
|
11
|
Aizamddin MF, Zainal Ariffin Z, Nor Amdan NA, Nawawi MA, Jani NA, Safian MF, Shaffee SNA, Nik Mohamed Daud NMR, Myo Thant MM, Mahat MM. Highly Durable Antibacterial Textiles: Cross-Linked Protonated Polyaniline-Polyacrylic Acid with Prolonged Electrical Stability. ACS OMEGA 2024; 9:23303-23315. [PMID: 38854582 PMCID: PMC11154899 DOI: 10.1021/acsomega.3c09871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 02/24/2024] [Accepted: 02/29/2024] [Indexed: 06/11/2024]
Abstract
This study addressed the limited antibacterial durability of textile materials, which has suppressed their applications in preventing infectious disease transmission. A class of highly durable antibacterial textiles was developed by incorporating protonated polyaniline (PANI) textile with poly(acrylic acid) (PAA) as the functional binder via cross-linking polymerization. The resulting PAA-PANI textile exhibits exceptional electrical conductivity, reaching 8.33 ± 0.04 × 10-3 S/cm when cross-linked with 30% PAA. Remarkably, this textile maintains its electrical stability at 10-3 S/cm even after 50 washing cycles, demonstrating unparalleled durability. Furthermore, the PANI-PAA textile showcases remarkable antibacterial efficacy, with 95.48% efficiency against Pseudomonas aeruginosa and 92.35% efficiency against Staphylococcus aureus bacteria, even after 50 washing cycles. Comparatively, the PAA-PANI textile outperforms its PANI counterpart by achieving an astounding 80% scavenging activity rate, whereas the latter only displayed a rate of 3.22%. This result suggests a solid integration of PAA-PANI into the textile, leading to sustainable antioxidant release. The successful cross-linking of PAA-PANI in textiles holds significant implications for various industries, offering a foundation for the development of wearable textiles with unprecedented antibacterial durability and electrical stability. This breakthrough opens new avenues for combating infectious diseases and enhancing the performance of wearable technologies.
Collapse
Affiliation(s)
- Muhammad Faiz Aizamddin
- Group
Research and Technology, PETRONAS Research
Sdn. Bhd., Bandar Baru Bangi, 43000 Selangor, Malaysia
- School
of Physics and Material Studies, Faculty of Applied Sciences, Universiti Teknologi MARA, Shah Alam 40450, Malaysia
| | - Zaidah Zainal Ariffin
- School
of Biology, Faculty of Applied Sciences, Universiti Teknologi MARA, Shah
Alam 40450, Malaysia
| | - Nur Asyura Nor Amdan
- Bacteriology
Unit, Infectious Disease Research Centre, Institute for Medical Research, National Institutes of Health, Setia Alam, Shah Alam 40170, Malaysia
| | - Mohd Azizi Nawawi
- School
of Chemistry and Environment, Faculty of Applied Sciences, Universiti Teknologi MARA, Shah Alam, Selangor 40450, Malaysia
| | - Nur Aimi Jani
- School
of Physics and Material Studies, Faculty of Applied Sciences, Universiti Teknologi MARA, Shah Alam 40450, Malaysia
| | - Muhd Fauzi Safian
- School
of Chemistry and Environment, Faculty of Applied Sciences, Universiti Teknologi MARA, Shah Alam, Selangor 40450, Malaysia
| | - Siti Nur Amira Shaffee
- Group
Research and Technology, PETRONAS Research
Sdn. Bhd., Bandar Baru Bangi, 43000 Selangor, Malaysia
| | | | - Maung Maung Myo Thant
- Group
Research and Technology, PETRONAS Research
Sdn. Bhd., Bandar Baru Bangi, 43000 Selangor, Malaysia
| | - Mohd Muzamir Mahat
- School
of Physics and Material Studies, Faculty of Applied Sciences, Universiti Teknologi MARA, Shah Alam 40450, Malaysia
- Textile Research
Group, Faculty of Applied Sciences, Universiti
Teknologi MARA, Shah Alam 40450, Malaysia
| |
Collapse
|
12
|
Wongdontree P, Millan-Oropeza A, Upfold J, Lavergne JP, Halpern D, Lambert C, Page A, Kénanian G, Grangeasse C, Henry C, Fouet A, Gloux K, Anba-Mondoloni J, Gruss A. Oxidative stress is intrinsic to staphylococcal adaptation to fatty acid synthesis antibiotics. iScience 2024; 27:109505. [PMID: 38577105 PMCID: PMC10993138 DOI: 10.1016/j.isci.2024.109505] [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: 07/26/2023] [Revised: 12/08/2023] [Accepted: 03/13/2024] [Indexed: 04/06/2024] Open
Abstract
Antibiotics inhibiting the fatty acid synthesis pathway (FASII) of the major pathogen Staphylococcus aureus reach their enzyme targets, but bacteria continue growth by using environmental fatty acids (eFAs) to produce phospholipids. We assessed the consequences and effectors of FASII-antibiotic (anti-FASII) adaptation. Anti-FASII induced lasting expression changes without genomic rearrangements. Several identified regulators affected the timing of adaptation outgrowth. Adaptation resulted in decreased expression of major virulence factors. Conversely, stress responses were globally increased and adapted bacteria were more resistant to peroxide killing. Importantly, pre-exposure to peroxide led to faster anti-FASII-adaptation by stimulating eFA incorporation. This adaptation differs from reports of peroxide-stimulated antibiotic efflux, which leads to tolerance. In vivo, anti-FASII-adapted S. aureus killed the insect host more slowly but continued multiplying. We conclude that staphylococcal adaptation to FASII antibiotics involves reprogramming, which decreases virulence and increases stress resistance. Peroxide, produced by the host to combat infection, favors anti-FASII adaptation.
Collapse
Affiliation(s)
- Paprapach Wongdontree
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
| | - Aaron Millan-Oropeza
- PAPPSO Platform, Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Jennifer Upfold
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
| | - Jean-Pierre Lavergne
- Bacterial Pathogens and Protein Phosphorylation, Molecular Microbiology and Structural Biology, UMR 5086 - CNRS / Université de Lyon, Building IBCP, 7 Passage du Vercors, Lyon, France
| | - David Halpern
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
| | - Clara Lambert
- Université Paris Cité, Institut Cochin, INSERM, U1016, CNRS, UMR8104, Paris, France
| | - Adeline Page
- Protein Science Facility, SFR BioSciences, CNRS, UMS3444, INSERM US8, Université de Lyon, Lyon, France
| | - Gérald Kénanian
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
| | - Christophe Grangeasse
- Bacterial Pathogens and Protein Phosphorylation, Molecular Microbiology and Structural Biology, UMR 5086 - CNRS / Université de Lyon, Building IBCP, 7 Passage du Vercors, Lyon, France
| | - Céline Henry
- PAPPSO Platform, Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Agnès Fouet
- Université Paris Cité, Institut Cochin, INSERM, U1016, CNRS, UMR8104, Paris, France
| | - Karine Gloux
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
| | - Jamila Anba-Mondoloni
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
| | - Alexandra Gruss
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
| |
Collapse
|
13
|
La Manna P, De Carluccio M, Oliva G, Vigliotta G, Rizzo L. Urban wastewater disinfection by iron chelates mediated solar photo-Fenton: Effects on seven pathogens and antibiotic resistance transfer potential. WATER RESEARCH 2024; 249:120966. [PMID: 38070340 DOI: 10.1016/j.watres.2023.120966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/25/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024]
Abstract
The effects of solar photo-Fenton (SPF) process mediated by the iron chelate Fe3+ imminodisuccinic acid (Fe:IDS) on both the inactivation of seven relevant pathogens and the potential for antibiotic resistance transfer (degradation of antibiotic resistance genes (ARGs) and after treatment regrowth), in real secondary treated urban wastewater, were investigated for the first time. A comparison with results obtained by sunlight/H2O2 process and Fe3+ ethylenediaminedisuccinic acid (Fe:EDDS) SPF was also carried out. ARGs were quantified by polymerase chain reaction (PCR) in samples before and after (3 h) the treatment. The persistence of the selected pathogens and ARGs was also evaluated in regrowth tests (72 h) under environmentally mimicking conditions. Fe:IDS SPF resulted to be more effective (from 1.4 log removal for Staphylococcus spp. to 4.3 log removal for Escherichia coli) than Fe:EDDS SPF (from 0.8 log removal for Pseudomonas aeruginosa to 2.0 log removal for Total coliphages) and sunlight/H2O2 (from 1.2 log removal for Clostridium perfringens to 3.3 log removal for E. coli) processes for the seven pathogens investigated. Potential pathogens regrowth was also severely affected, as no substantial regrowth was observed, both in presence and absence of catalase. A similar trend was observed for ARGs removal too (until 0.001 fold change expression for qnrS after 3 h). However, a poor effect and a slight increase in fold change was observed after treatment especially for gyrA, mefA and intl1. Overall, the effect of the investigated processes on ARGs was found to be ARG dependent. Noteworthy, coliphages can regrow after sunlight/H2O2 treatment unlike SPF processes, increasing the risk of antibiotic resistance transfer by transduction mechanism. In conclusion, Fe:IDS SPF is an attractive solution for tertiary treatment of urban wastewater in small wastewater treatment plants as it can provide effective disinfection and a higher protection against antibiotic resistance transfer than the other investigated processes.
Collapse
Affiliation(s)
- Pellegrino La Manna
- Water Science and Technology group (WaSTe), Department of Civil Engineering, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, SA, Italy
| | - Marco De Carluccio
- Water Science and Technology group (WaSTe), Department of Civil Engineering, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, SA, Italy
| | - Gianmaria Oliva
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, SA, Italy
| | - Giovanni Vigliotta
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, SA, Italy
| | - Luigi Rizzo
- Water Science and Technology group (WaSTe), Department of Civil Engineering, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, SA, Italy.
| |
Collapse
|
14
|
Goormaghtigh F, Van Bambeke F. Understanding Staphylococcus aureus internalisation and induction of antimicrobial tolerance. Expert Rev Anti Infect Ther 2024; 22:87-101. [PMID: 38180805 DOI: 10.1080/14787210.2024.2303018] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/04/2024] [Indexed: 01/07/2024]
Abstract
INTRODUCTION Staphylococcus aureus, a human commensal, is also one of the most common and serious pathogens for humans. In recent years, its capacity to survive and replicate in phagocytic and non-phagocytic cells has been largely demonstrated. In these intracellular niches, bacteria are shielded from the immune response and antibiotics, turning host cells into long-term infectious reservoirs. Moreover, neutrophils carry intracellular bacteria in the bloodstream, leading to systemic spreading of the disease. Despite the serious threat posed by intracellular S. aureus to human health, the molecular mechanisms behind its intracellular survival and subsequent antibiotic treatment failure remain elusive. AREA COVERED We give an overview of the killing mechanisms of phagocytes and of the impressive arsenal of virulence factors, toxins and stress responses deployed by S. aureus as a response. We then discuss the different barriers to antibiotic activity in this intracellular niche and finally describe innovative strategies to target intracellular persisting reservoirs. EXPERT OPINION Intracellular niches represent a challenge in terms of diagnostic and treatment. Further research using ad-hoc in-vivo models and single cell approaches are needed to better understand the molecular mechanisms underlying intracellular survival and tolerance to antibiotics in order to identify strategies to eliminate these persistent bacteria.
Collapse
Affiliation(s)
- Frédéric Goormaghtigh
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Françoise Van Bambeke
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
15
|
Baz A, Bakri A, Butcher M, Short B, Ghimire B, Gaur N, Jenkins T, Short RD, Riggio M, Williams C, Ramage G, Brown JL. Staphylococcus aureus strains exhibit heterogenous tolerance to direct cold atmospheric plasma therapy. Biofilm 2023; 5:100123. [PMID: 37138646 PMCID: PMC10149328 DOI: 10.1016/j.bioflm.2023.100123] [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: 12/23/2022] [Revised: 04/04/2023] [Accepted: 04/12/2023] [Indexed: 05/05/2023] Open
Abstract
The global clinical and socioeconomic impact of chronic wounds is substantial. The main difficulty that clinicians face during the treatment of chronic wounds is the risk of infection at the wound site. Infected wounds arise from an accumulation of microbial aggregates in the wound bed, leading to the formation of polymicrobial biofilms that can be largely resistant to antibiotic therapy. Therefore, it is essential for studies to identify novel therapeutics to alleviate biofilm infections. One innovative technique is the use of cold atmospheric plasma (CAP) which has been shown to possess promising antimicrobial and immunomodulatory properties. Here, different clinically relevant biofilm models will be treated with cold atmospheric plasma to assess its efficacy and killing effects. Biofilm viability was assessed using live dead qPCR, and morphological changes associated with CAP evaluated using scanning electron microscopy (SEM). Results indicated that CAP was effective against Candida albicans and Pseudomonas aeruginosa, both as mono-species biofilms and when grown in a triadic model system. CAP also significantly reduced viability in the nosocomial pathogen, Candida auris. Staphylococcus aureus Newman exhibited a level of tolerance to CAP therapy, both when grown alone or in the triadic model when grown alongside C. albicans and P. aeruginosa. However, this degree of tolerance exhibited by S. aureus was strain dependent. At a microscopic level, biofilm treatment led to subtle changes in morphology in the susceptible biofilms, with evidence of cellular deflation and shrinkage. Taken together, these results indicate a promising application of direct CAP therapy in combatting wound and skin-related biofilm infections, although biofilm composition may affect the treatment efficacy.
Collapse
Affiliation(s)
- Abdullah Baz
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, Dentistry & Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, United Kingdom
- Glasgow Biofilm Research Network, 378 Sauchiehall Street, Glasgow, G2 3JZ, United Kingdom
| | - Ahmed Bakri
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, Dentistry & Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, United Kingdom
- Glasgow Biofilm Research Network, 378 Sauchiehall Street, Glasgow, G2 3JZ, United Kingdom
| | - Mark Butcher
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, Dentistry & Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, United Kingdom
- Glasgow Biofilm Research Network, 378 Sauchiehall Street, Glasgow, G2 3JZ, United Kingdom
| | - Bryn Short
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, Dentistry & Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, United Kingdom
- Glasgow Biofilm Research Network, 378 Sauchiehall Street, Glasgow, G2 3JZ, United Kingdom
| | - Bhagirath Ghimire
- Department of Chemistry and Material Science Institute, University of Lancaster, Lancaster, LA1 4YB, United Kingdom
| | - Nishtha Gaur
- Department of Chemistry and Material Science Institute, University of Lancaster, Lancaster, LA1 4YB, United Kingdom
| | - Toby Jenkins
- Department of Chemistry, University of Bath, Bath, BA2 7AY, United Kingdom
| | - Robert D. Short
- Department of Chemistry and Material Science Institute, University of Lancaster, Lancaster, LA1 4YB, United Kingdom
| | - Marcello Riggio
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, Dentistry & Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, United Kingdom
- Glasgow Biofilm Research Network, 378 Sauchiehall Street, Glasgow, G2 3JZ, United Kingdom
| | - Craig Williams
- Glasgow Biofilm Research Network, 378 Sauchiehall Street, Glasgow, G2 3JZ, United Kingdom
- Microbiology Department, Lancaster Royal Infirmary, University of Lancaster, Lancaster, LA1 4YW, United Kingdom
| | - Gordon Ramage
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, Dentistry & Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, United Kingdom
- Glasgow Biofilm Research Network, 378 Sauchiehall Street, Glasgow, G2 3JZ, United Kingdom
| | - Jason L. Brown
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, Dentistry & Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, United Kingdom
- Glasgow Biofilm Research Network, 378 Sauchiehall Street, Glasgow, G2 3JZ, United Kingdom
- Corresponding author. Oral Sciences Research Group, Glasgow Dental School, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, 378 Sauchiehall Street, Glasgow, G2 3JZ, UK.
| |
Collapse
|
16
|
Roese KHC, Torlone C, Cooper LA, Esposito L, Deveau AM, Röse USR, Burkholder KM. Pyrogallol impairs staphylococcal biofilm formation via induction of bacterial oxidative stress. J Appl Microbiol 2023; 134:lxad270. [PMID: 37974055 DOI: 10.1093/jambio/lxad270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/13/2023] [Accepted: 11/15/2023] [Indexed: 11/19/2023]
Abstract
AIMS To examine the effect of the phenolic compound pyrogallol on staphylococcal biofilm formation. METHODS AND RESULTS In crystal violet biofilm assays, pyrogallol-reduced biofilm formation in Staphylococcus epidermidis ATCC 35984, Staph. epidermidis NRRL-B41021, Staphylococcus aureus USA300, and Staph. aureus Newman, without significantly impairing bacterial viability. Pyrogallol-mediated impairment of biofilm formation was likely due to induction of bacterial oxidative stress, as its effect was greater in catalase-deficient versus WT Staph. aureus, and biofilm production was rescued by exogenous catalase. The effect of pyrogallol on staphylococcal biofilm formation mirrored that of the known oxidant hydrogen peroxide, which also reduced biofilm formation in a dose-dependent manner. CONCLUSIONS Pyrogallol reduces biofilm formation in S. aureus and Staph. epidermidis in a mechanism involving induction of bacterial oxidative stress.
Collapse
Affiliation(s)
- Katharina H C Roese
- School of Biological Sciences, University of New England, Biddeford, ME 04005, USA
| | - Christina Torlone
- School of Biological Sciences, University of New England, Biddeford, ME 04005, USA
| | - Lauren A Cooper
- School of Biological Sciences, University of New England, Biddeford, ME 04005, USA
| | - Lee Esposito
- School of Biological Sciences, University of New England, Biddeford, ME 04005, USA
| | - Amy M Deveau
- School of Mathematical and Physical Sciences, University of New England, Biddeford, ME 04005, USA
| | - Ursula S R Röse
- School of Biological Sciences, University of New England, Biddeford, ME 04005, USA
| | - Kristin M Burkholder
- School of Biological Sciences, University of New England, Biddeford, ME 04005, USA
| |
Collapse
|
17
|
Silva E, Teixeira JA, Pereira MO, Rocha CMR, Sousa AM. Evolving biofilm inhibition and eradication in clinical settings through plant-based antibiofilm agents. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 119:154973. [PMID: 37499434 DOI: 10.1016/j.phymed.2023.154973] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 07/05/2023] [Accepted: 07/15/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND After almost 100 years since evidence of biofilm mode of growth and decades of intensive investigation about their formation, regulatory pathways and mechanisms of antimicrobial tolerance, nowadays there are still no therapeutic solutions to eradicate bacterial biofilms and their biomedical related issues. PURPOSE This review intends to provide a comprehensive summary of the recent and most relevant published studies on plant-based products, or their isolated compounds with antibiofilm activity mechanisms of action or identified molecular targets against bacterial biofilms. The objective is to offer a new perspective of most recent data for clinical researchers aiming to prevent or eliminate biofilm-associated infections caused by bacterial pathogens. METHODS The search was performed considering original research articles published on PubMed, Web of Science and Scopus from 2015 to April 2023, using keywords such as "antibiofilm", "antivirulence", "phytochemicals" and "plant extracts". RESULTS Over 180 articles were considered for this review with a focus on the priority human pathogens listed by World Health Organization, including Pseudomonas aeruginosa, Staphylococcus aureus, Klebsiella pneumoniae and Escherichia coli. Inhibition and detachment or dismantling of biofilms formed by these pathogens were found using plant-based extract/products or derivative compounds. Although combination of plant-based products and antibiotics were recorded and discussed, this topic is currently poorly explored and only for a reduced number of bacterial species. CONCLUSIONS This review clearly demonstrates that plant-based products or derivative compounds may be a promising therapeutic strategy to eliminate bacterial biofilms and their associated infections. After thoroughly reviewing the vast amount of research carried out over years, it was concluded that plant-based products are mostly able to prevent biofilm formation through inhibition of quorum sensing signals, but also to disrupt mature biofilms developed by multidrug resistant bacteria targeting the biofilm extracellular polymeric substance. Flavonoids and phenolic compounds seemed the most effective against bacterial biofilms.
Collapse
Affiliation(s)
- Eduarda Silva
- Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal
| | - José A Teixeira
- Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal; LABBELS - Associate Laboratory, Guimarães, Braga, Portugal
| | - Maria Olivia Pereira
- Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal; LABBELS - Associate Laboratory, Guimarães, Braga, Portugal
| | - Cristina M R Rocha
- Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal; LABBELS - Associate Laboratory, Guimarães, Braga, Portugal
| | - Ana Margarida Sousa
- Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal; LABBELS - Associate Laboratory, Guimarães, Braga, Portugal.
| |
Collapse
|
18
|
Stachurski P, Świątkowski W, Ciszewski A, Sarna-Boś K, Michalak A. A Short Review of the Toxicity of Dentifrices-Zebrafish Model as a Useful Tool in Ecotoxicological Studies. Int J Mol Sci 2023; 24:14339. [PMID: 37762640 PMCID: PMC10531698 DOI: 10.3390/ijms241814339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/12/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
This review aims to summarize the literature data regarding the effects of different toothpaste compounds in the zebrafish model. Danio rerio provides an insight into the mechanisms of the ecotoxicity of chemicals as well as an assessment of their fate in the environment to determine long-term environmental impact. The regular use of adequate toothpaste with safe active ingredients possessing anti-bacterial, anti-inflammatory, anti-oxidant, and regenerative properties is one of the most effective strategies for oral healthcare. In addition to water, a typical toothpaste consists of a variety of components, among which three are of predominant importance, i.e., abrasive substances, fluoride, and detergents. These ingredients provide healthy teeth, but their environmental impact on living organisms are often not well-known. Each of them can influence a higher level of organization: subcellular, cellular, tissue, organ, individual, and population. Therefore, it is very important that the properties of a chemical are detected before it is released into the environment to minimize damage. An important part of a chemical risk assessment is the estimation of the ecotoxicity of a compound. The zebrafish model has unique advantages in environmental ecotoxicity research and has been used to study vertebrate developmental biology. Among others, the advantages of this model include its external, visually accessible development, which allows for providing many experimental manipulations. The zebrafish has a significant genetic similarity with other vertebrates. Nevertheless, translating findings from zebrafish studies to human risk assessment requires careful consideration of these differences.
Collapse
Affiliation(s)
- Piotr Stachurski
- Department of Paediatric Dentistry, Medical University of Lublin, 20-059 Lublin, Poland
| | - Wojciech Świątkowski
- Department of Oral Surgery, Medical University of Lublin, 20-059 Lublin, Poland;
| | - Andrzej Ciszewski
- Department of Paediatric Orthopaedics and Rehabilitation, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Katarzyna Sarna-Boś
- Department of Dental Prosthetics, Medical University of Lublin, 20-059 Lublin, Poland;
| | - Agnieszka Michalak
- Independent Laboratory of Behavioral Studies, Medical University of Lublin, 20-059 Lublin, Poland;
| |
Collapse
|
19
|
Becker K. Detection, Identification and Diagnostic Characterization of the Staphylococcal Small Colony-Variant (SCV) Phenotype. Antibiotics (Basel) 2023; 12:1446. [PMID: 37760742 PMCID: PMC10525764 DOI: 10.3390/antibiotics12091446] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
While modern molecular methods have decisively accelerated and improved microbiological diagnostics, phenotypic variants still pose a challenge for their detection, identification and characterization. This particularly applies if they are unstable and hard to detect, which is the case for the small-colony-variant (SCV) phenotype formed by staphylococci. On solid agar media, staphylococcal SCVs are characterized by tiny colonies with deviant colony morphology. Their reduced growth rate and fundamental metabolic changes are the result of their adaptation to an intracellular lifestyle, regularly leading to specific auxotrophies, such as for menadione, hemin or thymidine. These alterations make SCVs difficult to recognize and render physiological, biochemical and other growth-based methods such as antimicrobial susceptibility testing unreliable or unusable. Therefore, diagnostic procedures require prolonged incubation times and, if possible, confirmation by molecular methods. A special approach is needed for auxotrophy testing. However, standardized protocols for SCV diagnostics are missing. If available, SCVs and their putative parental isolates should be genotyped to determine clonality. Since their detection has significant implications for the treatment of the infection, which is usually chronic and relapsing, SCV findings should be specifically reported, commented on, and managed in close collaboration with the microbiological laboratory and the involved clinicians.
Collapse
Affiliation(s)
- Karsten Becker
- Friedrich Loeffler-Institute of Medical Microbiology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße 1, 17489 Greifswald, Germany
| |
Collapse
|
20
|
Pei H, Zhu C, Shu F, Lu Z, Wang H, Ma K, Wang J, Lan R, Shang F, Xue T. CodY: An Essential Transcriptional Regulator Involved in Environmental Stress Tolerance in Foodborne Staphylococcus aureus RMSA24. Foods 2023; 12:3166. [PMID: 37685098 PMCID: PMC10486358 DOI: 10.3390/foods12173166] [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: 06/30/2023] [Revised: 08/04/2023] [Accepted: 08/15/2023] [Indexed: 09/10/2023] Open
Abstract
Staphylococcus aureus (S. aureus), as the main pathogen in milk and dairy products, usually causes intoxication with vomiting and various kinds of inflammation after entering the human body. CodY, an important transcriptional regulator in S. aureus, plays an important role in regulating metabolism, growth, and virulence. However, little is known about the role of CodY on environmental stress tolerance. In this research, we revealed the role of CodY in environmental stress tolerance in foodborne S. aureus RMSA24. codY mutation significantly reduced the tolerance of S. aureus to desiccation and oxidative, salt, and high-temperature stresses. However, S. aureus was more tolerant to low temperature stress due to mutation of codY. We found that the expressions of two important heat shock proteins-GroEL and DanJ-were significantly down-regulated in the mutant codY. This suggests that CodY may indirectly regulate the high- and low-temperature tolerance of S. aureus by regulating the expressions of groEL and danJ. This study reveals a new mechanism of environmental stress tolerance in S. aureus and provides new insights into controlling the contamination and harm caused by S. aureus in the food industry.
Collapse
Affiliation(s)
- Hao Pei
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China; (H.P.); (C.Z.); (F.S.); (Z.L.); (H.W.); (K.M.); (J.W.); (R.L.); (F.S.)
| | - Chengfeng Zhu
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China; (H.P.); (C.Z.); (F.S.); (Z.L.); (H.W.); (K.M.); (J.W.); (R.L.); (F.S.)
| | - Fang Shu
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China; (H.P.); (C.Z.); (F.S.); (Z.L.); (H.W.); (K.M.); (J.W.); (R.L.); (F.S.)
| | - Zhengfei Lu
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China; (H.P.); (C.Z.); (F.S.); (Z.L.); (H.W.); (K.M.); (J.W.); (R.L.); (F.S.)
| | - Hui Wang
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China; (H.P.); (C.Z.); (F.S.); (Z.L.); (H.W.); (K.M.); (J.W.); (R.L.); (F.S.)
| | - Kai Ma
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China; (H.P.); (C.Z.); (F.S.); (Z.L.); (H.W.); (K.M.); (J.W.); (R.L.); (F.S.)
| | - Jun Wang
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China; (H.P.); (C.Z.); (F.S.); (Z.L.); (H.W.); (K.M.); (J.W.); (R.L.); (F.S.)
| | - Ranxiang Lan
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China; (H.P.); (C.Z.); (F.S.); (Z.L.); (H.W.); (K.M.); (J.W.); (R.L.); (F.S.)
| | - Fei Shang
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China; (H.P.); (C.Z.); (F.S.); (Z.L.); (H.W.); (K.M.); (J.W.); (R.L.); (F.S.)
- Food Procession Research Institute, Anhui Agricultural University, Hefei 230036, China
| | - Ting Xue
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China; (H.P.); (C.Z.); (F.S.); (Z.L.); (H.W.); (K.M.); (J.W.); (R.L.); (F.S.)
- Food Procession Research Institute, Anhui Agricultural University, Hefei 230036, China
| |
Collapse
|
21
|
Oliveira M, Cunha E, Tavares L, Serrano I. P. aeruginosa interactions with other microbes in biofilms during co-infection. AIMS Microbiol 2023; 9:612-646. [PMID: 38173971 PMCID: PMC10758579 DOI: 10.3934/microbiol.2023032] [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: 06/15/2023] [Revised: 07/10/2023] [Accepted: 07/26/2023] [Indexed: 01/05/2024] Open
Abstract
This review addresses the topic of biofilms, including their development and the interaction between different counterparts. There is evidence that various diseases, such as cystic fibrosis, otitis media, diabetic foot wound infections, and certain cancers, are promoted and aggravated by the presence of polymicrobial biofilms. Biofilms are composed by heterogeneous communities of microorganisms protected by a matrix of polysaccharides. The different types of interactions between microorganisms gives rise to an increased resistance to antimicrobials and to the host's defense mechanisms, with the consequent worsening of disease symptoms. Therefore, infections caused by polymicrobial biofilms affecting different human organs and systems will be discussed, as well as the role of the interactions between the gram-negative bacteria Pseudomonas aeruginosa, which is at the base of major polymicrobial infections, and other bacteria, fungi, and viruses in the establishment of human infections and diseases. Considering that polymicrobial biofilms are key to bacterial pathogenicity, it is fundamental to evaluate which microbes are involved in a certain disease to convey an appropriate and efficacious antimicrobial therapy.
Collapse
Affiliation(s)
- Manuela Oliveira
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Eva Cunha
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Luís Tavares
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Isa Serrano
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisboa, Portugal
| |
Collapse
|
22
|
Millette G, Séguin DL, Isabelle C, Chamberland S, Lucier JF, Rodrigue S, Cantin AM, Malouin F. Staphylococcus aureus Small-Colony Variants from Airways of Adult Cystic Fibrosis Patients as Precursors of Adaptive Antibiotic-Resistant Mutations. Antibiotics (Basel) 2023; 12:1069. [PMID: 37370388 PMCID: PMC10294822 DOI: 10.3390/antibiotics12061069] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Prototypic Staphylococcus aureus and their small-colony variants (SCVs) are predominant in cystic fibrosis (CF), but the interdependence of these phenotypes is poorly understood. We characterized S. aureus isolates from adult CF patients over several years. Of 18 S. aureus-positive patients (58%), 13 (72%) were positive for SCVs. Characterization included genotyping, SCCmec types, auxotrophy, biofilm production, antibiotic susceptibilities and tolerance, and resistance acquisition rates. Whole-genome sequencing revealed that several patients were colonized with prototypical and SCV-related clones. Some clonal pairs showed acquisition of aminoglycoside resistance that was not explained by aminoglycoside-modifying enzymes, suggesting a mutation-based process. The characteristics of SCVs that could play a role in resistance acquisition were thus investigated further. For instance, SCV isolates produced more biofilm (p < 0.05) and showed a higher survival rate upon exposure to ciprofloxacin and vancomycin compared to their prototypic associated clones. SCVs also developed spontaneous rifampicin resistance mutations at a higher frequency. Accordingly, a laboratory-derived SCV (ΔhemB) acquired resistance to ciprofloxacin and gentamicin faster than its parent counterpart after serial passages in the presence of sub-inhibitory concentrations of antibiotics. These results suggest a role for SCVs in the establishment of persistent antibiotic-resistant clones in adult CF patients.
Collapse
Affiliation(s)
- Guillaume Millette
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (G.M.); (D.L.S.); (C.I.); (S.C.); (J.-F.L.); (S.R.)
| | - David Lalonde Séguin
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (G.M.); (D.L.S.); (C.I.); (S.C.); (J.-F.L.); (S.R.)
| | - Charles Isabelle
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (G.M.); (D.L.S.); (C.I.); (S.C.); (J.-F.L.); (S.R.)
| | - Suzanne Chamberland
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (G.M.); (D.L.S.); (C.I.); (S.C.); (J.-F.L.); (S.R.)
| | - Jean-François Lucier
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (G.M.); (D.L.S.); (C.I.); (S.C.); (J.-F.L.); (S.R.)
| | - Sébastien Rodrigue
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (G.M.); (D.L.S.); (C.I.); (S.C.); (J.-F.L.); (S.R.)
| | - André M. Cantin
- Service de Pneumologie, Département de Médecine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - François Malouin
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada; (G.M.); (D.L.S.); (C.I.); (S.C.); (J.-F.L.); (S.R.)
| |
Collapse
|
23
|
Knobling B, Franke G, Carlsen L, Belmar Campos C, Büttner H, Klupp EM, Maurer PM, Knobloch JK. Phenotypic Variation in Clinical S. aureus Isolates Did Not Affect Disinfection Efficacy Using Short-Term UV-C Radiation. Microorganisms 2023; 11:1332. [PMID: 37317306 PMCID: PMC10223295 DOI: 10.3390/microorganisms11051332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/10/2023] [Accepted: 05/16/2023] [Indexed: 06/16/2023] Open
Abstract
Pigmentation, catalase activity and biofilm formation are virulence factors that cause resistance of Staphylococcus aureus to environmental stress factors including disinfectants. In recent years, automatic UV-C room disinfection gained greater importance in enhanced disinfection procedures to improve disinfection success in hospitals. In this study, we evaluated the effect of naturally occurring variations in the expression of virulence factors in clinical S. aureus isolates on tolerance against UV-C radiation. Quantification of staphyloxanthin expression, catalase activity and biofilm formation for nine genetically different clinical S. aureus isolates as well as reference strain S. aureus ATCC 6538 were performed using methanol extraction, a visual approach assay and a biofilm assay, respectively. Log10 reduction values (LRV) were determined after irradiation of artificially contaminated ceramic tiles with 50 and 22 mJ/cm2 UV-C using a commercial UV-C disinfection robot. A wide variety of virulence factor expression was observed, indicating differential regulation of global regulatory networks. However, no direct correlation with the strength of expression with UV-C tolerance was observed for either staphyloxanthin expression, catalase activity or biofilm formation. All isolates were effectively reduced with LRVs of 4.75 to 5.94. UV-C disinfection seems therefore effective against a wide spectrum of S. aureus strains independent of occurring variations in the expression of the investigated virulence factors. Due to only minor differences, the results of frequently used reference strains seem to be representative also for clinical isolates in S. aureus.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Johannes K. Knobloch
- Department Infection Prevention and Control, Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany; (B.K.); (L.C.); (P.M.M.)
| |
Collapse
|
24
|
Carrilero L, Urwin L, Ward E, Choudhury NR, Monk IR, Turner CE, Stinear TP, Corrigan RM. Stringent Response-Mediated Control of GTP Homeostasis Is Required for Long-Term Viability of Staphylococcus aureus. Microbiol Spectr 2023; 11:e0044723. [PMID: 36877013 PMCID: PMC10101089 DOI: 10.1128/spectrum.00447-23] [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: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 03/07/2023] Open
Abstract
Staphylococcus aureus is an opportunistic bacterial pathogen that often results in difficult-to-treat infections. One mechanism used by S. aureus to enhance survival during infection is the stringent response. This is a stress survival pathway that utilizes the nucleotides (p)ppGpp to reallocate bacterial resources, shutting down growth until conditions improve. Small colony variants (SCVs) of S. aureus are frequently associated with chronic infections, and this phenotype has previously been linked to a hyperactive stringent response. Here, we examine the role of (p)ppGpp in the long-term survival of S. aureus under nutrient-restricted conditions. When starved, a (p)ppGpp-null S. aureus mutant strain ((p)ppGpp0) initially had decreased viability. However, after 3 days we observed the presence and dominance of a population of small colonies. Similar to SCVs, these small colony isolates (p0-SCIs) had reduced growth but remained hemolytic and sensitive to gentamicin, phenotypes that have been tied to SCVs previously. Genomic analysis of the p0-SCIs revealed mutations arising within gmk, encoding an enzyme in the GTP synthesis pathway. We show that a (p)ppGpp0 strain has elevated levels of GTP, and that the mutations in the p0-SCIs all lower Gmk enzyme activity and consequently cellular GTP levels. We further show that in the absence of (p)ppGpp, cell viability can be rescued using the GuaA inhibitor decoyinine, which artificially lowers the intracellular GTP concentration. Our study highlights the role of (p)ppGpp in GTP homeostasis and underscores the importance of nucleotide signaling for long-term survival of S. aureus in nutrient-limiting conditions, such as those encountered during infections. IMPORTANCE Staphylococcus aureus is a human pathogen that upon invasion of a host encounters stresses, such as nutritional restriction. The bacteria respond by switching on a signaling cascade controlled by the nucleotides (p)ppGpp. These nucleotides function to shut down bacterial growth until conditions improve. Therefore, (p)ppGpp are important for bacterial survival and have been implicated in promoting chronic infections. Here, we investigate the importance of (p)ppGpp for long-term survival of bacteria in nutrient-limiting conditions similar to those in a human host. We discovered that in the absence of (p)ppGpp, bacterial viability decreases due to dysregulation of GTP homeostasis. However, the (p)ppGpp-null bacteria were able to compensate by introducing mutations in the GTP synthesis pathway that led to a reduction in GTP build-up and a rescue of viability. This study therefore highlights the importance of (p)ppGpp for the regulation of GTP levels and for long-term survival of S. aureus in restricted environments.
Collapse
Affiliation(s)
- Laura Carrilero
- The Florey Institute, School of Biosciences, University of Sheffield, Sheffield, United Kingdom
| | - Lucy Urwin
- The Florey Institute, School of Biosciences, University of Sheffield, Sheffield, United Kingdom
| | - Ezra Ward
- The Florey Institute, School of Biosciences, University of Sheffield, Sheffield, United Kingdom
| | - Naznin R. Choudhury
- The Florey Institute, School of Biosciences, University of Sheffield, Sheffield, United Kingdom
| | - Ian R. Monk
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Claire E. Turner
- The Florey Institute, School of Biosciences, University of Sheffield, Sheffield, United Kingdom
| | - Timothy P. Stinear
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Rebecca M. Corrigan
- The Florey Institute, School of Biosciences, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
25
|
Wilkinson M, Wilkinson OJ, Feyerherm C, Fletcher EE, Wigley DB, Dillingham MS. Structures of RecBCD in complex with phage-encoded inhibitor proteins reveal distinctive strategies for evasion of a bacterial immunity hub. eLife 2022; 11:e83409. [PMID: 36533901 PMCID: PMC9836394 DOI: 10.7554/elife.83409] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 12/18/2022] [Indexed: 12/23/2022] Open
Abstract
Following infection of bacterial cells, bacteriophage modulate double-stranded DNA break repair pathways to protect themselves from host immunity systems and prioritise their own recombinases. Here, we present biochemical and structural analysis of two phage proteins, gp5.9 and Abc2, which target the DNA break resection complex RecBCD. These exemplify two contrasting mechanisms for control of DNA break repair in which the RecBCD complex is either inhibited or co-opted for the benefit of the invading phage. Gp5.9 completely inhibits RecBCD by preventing it from binding to DNA. The RecBCD-gp5.9 structure shows that gp5.9 acts by substrate mimicry, binding predominantly to the RecB arm domain and competing sterically for the DNA binding site. Gp5.9 adopts a parallel coiled-coil architecture that is unprecedented for a natural DNA mimic protein. In contrast, binding of Abc2 does not substantially affect the biochemical activities of isolated RecBCD. The RecBCD-Abc2 structure shows that Abc2 binds to the Chi-recognition domains of the RecC subunit in a position that might enable it to mediate the loading of phage recombinases onto its single-stranded DNA products.
Collapse
Affiliation(s)
- Martin Wilkinson
- Section of Structural Biology, Department of Infectious Disease, Faculty of Medicine, Imperial College LondonLondonUnited Kingdom
| | - Oliver J Wilkinson
- DNA:protein Interactions Unit, School of Biochemistry, University of BristolBristolUnited Kingdom
| | - Connie Feyerherm
- DNA:protein Interactions Unit, School of Biochemistry, University of BristolBristolUnited Kingdom
| | - Emma E Fletcher
- DNA:protein Interactions Unit, School of Biochemistry, University of BristolBristolUnited Kingdom
| | - Dale B Wigley
- Section of Structural Biology, Department of Infectious Disease, Faculty of Medicine, Imperial College LondonLondonUnited Kingdom
| | - Mark S Dillingham
- DNA:protein Interactions Unit, School of Biochemistry, University of BristolBristolUnited Kingdom
| |
Collapse
|
26
|
Islam S, Callender AC, Ho QN, Wakeman CA. Iron restriction induces the small-colony variant phenotype in Staphylococcus aureus. Front Microbiol 2022; 13:978859. [PMID: 36569073 PMCID: PMC9772265 DOI: 10.3389/fmicb.2022.978859] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
Pathogens such as Staphylococcus aureus must overcome host-induced selective pressures, including limited iron availability. To cope with the harsh conditions of the host environment, S. aureus can adapt its physiology in multiple ways. One of these adaptations is the fermenting small-colony variant (SCV) phenotype, which is known to be inherently tolerant to certain classes of antibiotics and heme toxicity. We hypothesized that SCVs might also behave uniquely in response to iron starvation since one of the major cellular uses of iron is the respiration machinery. In this study, a respiring strain of S. aureus and fermenting SCV strains were treated with different concentrations of the iron chelator, 2,2' dipyridyl (DIP). Our data demonstrate that a major impact of iron starvation in S. aureus is the repression of respiration and the induction of the SCV phenotype. We demonstrate that the SCV phenotype transiently induced by iron starvation mimics the aminoglycoside recalcitrance exhibited by genetic SCVs. Furthermore, prolonged growth in iron starvation promotes increased emergence of stable aminoglycoside-resistant SCVs relative to the naturally occurring subpopulation of SCVs within an S. aureus community. These findings may have relevance to physiological and evolutionary processes occurring within bacterial populations infecting iron-limited host environments.
Collapse
Affiliation(s)
| | | | | | - Catherine A. Wakeman
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, United States
| |
Collapse
|
27
|
Shahriarinour M, Divsar F. Release Kinetics and Antibacterial Property of Curcumin-Loaded Date Palm (Phoenix dactylifera L.) Pollen. ARABIAN JOURNAL FOR SCIENCE AND ENGINEERING 2022. [DOI: 10.1007/s13369-022-07301-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
28
|
Functional mgrA Influences Genetic Changes within a Staphylococcus aureus Cell Population over Time. J Bacteriol 2022; 204:e0013822. [PMID: 36154359 DOI: 10.1128/jb.00138-22] [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: 11/20/2022] Open
Abstract
Prolonged survival in the host-bacteria microenvironment drives the selection of alternative cell types in Staphylococcus aureus, permitting quasi-dormant sub-populations to develop. These facilitate antibiotic tolerance, long-term growth, and relapse of infection. Small Colony Variants (SCV) are an important cell type associated with persistent infection but are difficult to study in vitro due to the instability of the phenotype and reversion to the normal cell type. We have previously reported that under conditions of growth in continuous culture over a prolonged culture time, SCVs dominated a heterogenous population of cell types and these SCVs harbored a mutation in the DNA binding domain of the gene for the transcription factor, mgrA. To investigate this specific cell type further, S. aureus WCH-SK2-ΔmgrA itself was assessed with continuous culture. Compared to the wild type, the mgrA mutant strain required fewer generations to select for SCVs. There was an increased rate of mutagenesis within the ΔmgrA strain compared to the wild type, which we postulate is the mechanism explaining the increased emergence of SCV selection. The mgrA derived SCVs had impeded metabolism, altered MIC to specific antibiotics and an increased biofilm formation compared to non-SCV strain. Whole genomic sequencing detected single nucleotide polymorphisms (SNP) in phosphoglucosamine mutase glmM and tyrosine recombinase xerC. In addition, several genomic rearrangements were detected which affected genes involved in important functions such as antibiotic and toxic metal resistance and pathogenicity. Thus, we propose a direct link between mgrA and the SCV phenotype. IMPORTANCE Within a bacterial population, a stochastically generated heterogeneity of phenotypes allows continual survival against current and future stressors. The generation of a sub-population of quasi-dormant Small Colony Variants (SCV) in Staphylococcus aureus is such a mechanism, allowing for persistent or relapse of infection despite initial intervention seemingly clearing the infection. The use of continuous culture under clinically relevant conditions has allowed us to introduce time to the growth system and selects SCV within the population. This study provides valuable insights into the generation of SCV which are not addressed in standard laboratory generated models and reveals new pathways for understanding persistent S. aureus infection which can potentially be targeted in future treatments of persistent S. aureus infection.
Collapse
|
29
|
Turgimbayeva A, Zein U, Zharkov DO, Ramankulov Y, Saparbaev M, Abeldenov S. Cloning and characterization of the major AP endonuclease from Staphylococcus aureus. DNA Repair (Amst) 2022; 119:103390. [DOI: 10.1016/j.dnarep.2022.103390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/19/2022] [Accepted: 08/20/2022] [Indexed: 11/03/2022]
|
30
|
Durand BARN, Pouget C, Magnan C, Molle V, Lavigne JP, Dunyach-Remy C. Bacterial Interactions in the Context of Chronic Wound Biofilm: A Review. Microorganisms 2022; 10:microorganisms10081500. [PMID: 35893558 PMCID: PMC9332326 DOI: 10.3390/microorganisms10081500] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/15/2022] [Accepted: 07/21/2022] [Indexed: 02/04/2023] Open
Abstract
Chronic wounds, defined by their resistance to care after four weeks, are a major concern, affecting millions of patients every year. They can be divided into three types of lesions: diabetic foot ulcers (DFU), pressure ulcers (PU), and venous/arterial ulcers. Once established, the classical treatment for chronic wounds includes tissue debridement at regular intervals to decrease biofilm mass constituted by microorganisms physiologically colonizing the wound. This particular niche hosts a dynamic bacterial population constituting the bed of interaction between the various microorganisms. The temporal reshuffle of biofilm relies on an organized architecture. Microbial community turnover is mainly associated with debridement (allowing transitioning from one major representant to another), but also with microbial competition and/or collaboration within wounds. This complex network of species and interactions has the potential, through diversity in antagonist and/or synergistic crosstalk, to accelerate, delay, or worsen wound healing. Understanding these interactions between microorganisms encountered in this clinical situation is essential to improve the management of chronic wounds.
Collapse
Affiliation(s)
- Benjamin A. R. N. Durand
- Bacterial Virulence and Chronic Infections, UMR 1047, Université Montpellier, INSERM, Service de Microbiologie et Hygiène Hospitalière, CHU Nîmes, 30908 Nîmes, France; (B.A.R.N.D.); (C.P.); (C.M.); (J.-P.L.)
| | - Cassandra Pouget
- Bacterial Virulence and Chronic Infections, UMR 1047, Université Montpellier, INSERM, Service de Microbiologie et Hygiène Hospitalière, CHU Nîmes, 30908 Nîmes, France; (B.A.R.N.D.); (C.P.); (C.M.); (J.-P.L.)
| | - Chloé Magnan
- Bacterial Virulence and Chronic Infections, UMR 1047, Université Montpellier, INSERM, Service de Microbiologie et Hygiène Hospitalière, CHU Nîmes, 30908 Nîmes, France; (B.A.R.N.D.); (C.P.); (C.M.); (J.-P.L.)
| | - Virginie Molle
- Laboratory of Pathogen Host Interactions, Université de Montpellier, CNRS, UMR 5235, 34000 Montpellier, France;
| | - Jean-Philippe Lavigne
- Bacterial Virulence and Chronic Infections, UMR 1047, Université Montpellier, INSERM, Service de Microbiologie et Hygiène Hospitalière, CHU Nîmes, 30908 Nîmes, France; (B.A.R.N.D.); (C.P.); (C.M.); (J.-P.L.)
| | - Catherine Dunyach-Remy
- Bacterial Virulence and Chronic Infections, UMR 1047, Université Montpellier, INSERM, Service de Microbiologie et Hygiène Hospitalière, CHU Nîmes, 30908 Nîmes, France; (B.A.R.N.D.); (C.P.); (C.M.); (J.-P.L.)
- Correspondence: ; Tel.: +33-466-683-202
| |
Collapse
|
31
|
Biswas L, Götz F. Molecular Mechanisms of Staphylococcus and Pseudomonas Interactions in Cystic Fibrosis. Front Cell Infect Microbiol 2022; 11:824042. [PMID: 35071057 PMCID: PMC8770549 DOI: 10.3389/fcimb.2021.824042] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 12/16/2021] [Indexed: 11/15/2022] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive genetic disorder that is characterized by recurrent and chronic infections of the lung predominantly by the opportunistic pathogens, Gram-positive Staphylococcus aureus and Gram-negative Pseudomonas aeruginosa. While S. aureus is the main colonizing bacteria of the CF lungs during infancy and early childhood, its incidence declines thereafter and infections by P. aeruginosa become more prominent with increasing age. The competitive and cooperative interactions exhibited by these two pathogens influence their survival, antibiotic susceptibility, persistence and, consequently the disease progression. For instance, P. aeruginosa secretes small respiratory inhibitors like hydrogen cyanide, pyocyanin and quinoline N-oxides that block the electron transport pathway and suppress the growth of S. aureus. However, S. aureus survives this respiratory attack by adapting to respiration-defective small colony variant (SCV) phenotype. SCVs cause persistent and recurrent infections and are also resistant to antibiotics, especially aminoglycosides, antifolate antibiotics, and to host antimicrobial peptides such as LL-37, human β-defensin (HBD) 2 and HBD3; and lactoferricin B. The interaction between P. aeruginosa and S. aureus is multifaceted. In mucoid P. aeruginosa strains, siderophores and rhamnolipids are downregulated thus enhancing the survival of S. aureus. Conversely, protein A from S. aureus inhibits P. aeruginosa biofilm formation while protecting both P. aeruginosa and S. aureus from phagocytosis by neutrophils. This review attempts to summarize the current understanding of the molecular mechanisms that drive the competitive and cooperative interactions between S. aureus and P. aeruginosa in the CF lungs that could influence the disease outcome.
Collapse
Affiliation(s)
- Lalitha Biswas
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Friedrich Götz
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, Tübingen, Germany
| |
Collapse
|
32
|
Lakshmi SA, Prasath KG, Tamilmuhilan K, Srivathsan A, Shafreen RMB, Kasthuri T, Pandian SK. Suppression of Thiol-Dependent Antioxidant System and Stress Response in Methicillin-Resistant Staphylococcus aureus by Docosanol: Explication Through Proteome Investigation. Mol Biotechnol 2022; 64:575-589. [PMID: 35018617 DOI: 10.1007/s12033-021-00434-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 12/01/2021] [Indexed: 11/28/2022]
Abstract
The present study was aimed to investigate the effect of docosanol on the protein expression profile of methicillin-resistant Staphylococcus aureus (MRSA). Thus, two-dimensional gel electrophoresis coupled with MALDI-TOF MS technique was utilized to identify the differentially regulated proteins in the presence of docosanol. A total of 947 protein spots were identified from the intracellular proteome of both control and docosanol treated samples among which 40 spots were differentially regulated with a fold change greater than 1.0. Prominently, the thiol-dependent antioxidant system and stress response proteins are downregulated in MRSA, which are critical for survival during oxidative stress. In particular, docosanol downregulated the expression of Tpx, AhpC, BshC, BrxA, and YceI with a fold change of 1.4 (p = 0.02), 1.4 (p = 0.01), 1.6 (p = 0.002), 4.9 (p = 0.02), and 1.4 (p = 0.02), respectively. In addition, docosanol reduced the expression of proteins involved in purine metabolic pathways, biofilm growth cycle, and virulence factor production. Altogether, these findings suggest that docosanol could efficiently target the antioxidant pathway by reducing the expression of bacillithiol and stress-associated proteins.
Collapse
Affiliation(s)
- Selvaraj Alagu Lakshmi
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630003, India
| | - Krishnan Ganesh Prasath
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630003, India
- Department of Biotechnology, Sri Venkateswara College of Engineering, Sriperumbudur, Tamil Nadu, 602117, India
| | - Kannapiran Tamilmuhilan
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630003, India
| | - Adimoolam Srivathsan
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630003, India
| | - Raja Mohamed Beema Shafreen
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630003, India
- Department of Biotechnology, Dr. Umayal Ramanathan College for Women, Alagappapuram, Karaikudi, Tamil Nadu, 630003, India
| | - Thirupathi Kasthuri
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630003, India
| | | |
Collapse
|
33
|
Shimamura Y, Yui T, Horiike H, Masuda S. Toxicity of combined exposure to acrylamide and Staphylococcus aureus. Toxicol Rep 2022; 9:876-882. [DOI: 10.1016/j.toxrep.2022.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 11/26/2022] Open
|
34
|
Genetic Factors Affect the Survival and Behaviors of Selected Bacteria during Antimicrobial Blue Light Treatment. Int J Mol Sci 2021; 22:ijms221910452. [PMID: 34638788 PMCID: PMC8508746 DOI: 10.3390/ijms221910452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 12/26/2022] Open
Abstract
Antimicrobial resistance is a global, mounting and dynamic issue that poses an immediate threat to human, animal, and environmental health. Among the alternative antimicrobial treatments proposed to reduce the external use of antibiotics is electromagnetic radiation, such as blue light. The prevailing mechanistic model is that blue light can be absorbed by endogenous porphyrins within the bacterial cell, inducing the production of reactive oxygen species, which subsequently inflict oxidative damages upon different cellular components. Nevertheless, it is unclear whether other mechanisms are involved, particularly those that can affect the efficacy of antimicrobial blue light treatments. In this review, we summarize evidence of inherent factors that may confer protection to a selected group of bacteria against blue light-induced oxidative damages or modulate the physiological characteristics of the treated bacteria, such as virulence and motility. These include descriptions of three major photoreceptors in bacteria, chemoreceptors, SOS-dependent DNA repair and non-SOS protective mechanisms. Future directions are also provided to assist with research efforts to increase the efficacy of antimicrobial blue light and to minimize the development of blue light-tolerant phenotypes.
Collapse
|
35
|
Abdul Hamid AI, Cara A, Diot A, Laurent F, Josse J, Gueirard P. Differential Early in vivo Dynamics and Functionality of Recruited Polymorphonuclear Neutrophils After Infection by Planktonic or Biofilm Staphylococcus aureus. Front Microbiol 2021; 12:728429. [PMID: 34526981 PMCID: PMC8435793 DOI: 10.3389/fmicb.2021.728429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/05/2021] [Indexed: 12/28/2022] Open
Abstract
Staphylococcus aureus is a human pathogen known for its capacity to shift between the planktonic and biofilm lifestyles. In vivo, the antimicrobial immune response is characterized by the recruitment of inflammatory phagocytes, namely polymorphonuclear neutrophils (PMNs) and monocytes/macrophages. Immune responses to planktonic bacteria have been extensively studied, but many questions remain about how biofilms can modulate inflammatory responses and cause recurrent infections in live vertebrates. Thus, the use of biologically sound experimental models is essential to study the specific immune signatures elicited by biofilms. Here, a mouse ear pinna model of infection was used to compare early innate immune responses toward S. aureus planktonic or biofilm bacteria. Flow cytometry and cytokine assays were carried out to study the inflammatory responses in infected tissues. These data were complemented with intravital confocal imaging analyses, allowing the real-time observation of the dynamic interactions between EGFP + phagocytes and bacteria in the ear pinna tissue of LysM-EGFP transgenic mice. Both bacterial forms induced an early and considerable recruitment of phagocytes in the ear tissue, associated with a predominantly pro-inflammatory cytokine profile. The inflammatory response was mostly composed of PMNs in the skin and the auricular lymph node. However, the kinetics of PMN recruitment were different between the 2 forms in the first 2 days post-infection (pi). Two hours pi, biofilm inocula recruited more PMNs than planktonic bacteria, but with decreased motility parameters and capacity to emit pseudopods. Inversely, biofilm inocula recruited less PMNs 2 days pi, but with an “over-activated” status, illustrated by an increased phagocytic activity, CD11b level of expression and ROS production. Thus, the mouse ear pinna model allowed us to reveal specific differences in the dynamics of recruitment and functional properties of phagocytes against biofilms. These differences would influence the specific adaptive immune responses to biofilms elicited in the lymphoid tissues.
Collapse
Affiliation(s)
- Aizat Iman Abdul Hamid
- Laboratoire Microorganismes: Génome et Environnement, CNRS UMR 6023, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Andréa Cara
- Centre International de Recherche et Infectiologie, Inserm U1111, CNRS UMR 5308, École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Alan Diot
- Centre International de Recherche et Infectiologie, Inserm U1111, CNRS UMR 5308, École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Frédéric Laurent
- Centre International de Recherche et Infectiologie, Inserm U1111, CNRS UMR 5308, École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Jérôme Josse
- Centre International de Recherche et Infectiologie, Inserm U1111, CNRS UMR 5308, École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Pascale Gueirard
- Laboratoire Microorganismes: Génome et Environnement, CNRS UMR 6023, Université Clermont Auvergne, Clermont-Ferrand, France
| |
Collapse
|
36
|
Abstract
Staphylococcus aureus is a common cause of both superficial and invasive infections of humans and animals. Despite a potent host response and apparently appropriate antibiotic therapy, staphylococcal infections frequently become chronic or recurrent, demonstrating a remarkable ability of S. aureus to withstand the hostile host environment. There is growing evidence that staphylococcal DNA repair makes important contributions to the survival of the pathogen in host tissues, as well as promoting the emergence of mutants that resist host defenses and antibiotics. While much of what we know about DNA repair in S. aureus is inferred from studies with model organisms, the roles of specific repair mechanisms in infection are becoming clear and differences with Bacillus subtilis and Escherichia coli have been identified. Furthermore, there is growing interest in staphylococcal DNA repair as a target for novel therapeutics that sensitize the pathogen to host defenses and antibiotics. In this review, we discuss what is known about staphylococcal DNA repair and its role in infection, examine how repair in S. aureus is similar to, or differs from, repair in well-characterized model organisms, and assess the potential of staphylococcal DNA repair as a novel therapeutic target.
Collapse
|
37
|
Budimir M, Marković Z, Vajdak J, Jovanović S, Kubat P, Humpoliček P, Mičušik M, Danko M, Barras A, Milivojević D, Špitalsky Z, Boukherroub R, Marković BT. Enhanced visible light-triggered antibacterial activity of carbon quantum dots/polyurethane nanocomposites by gamma rays induced pre-treatment. Radiat Phys Chem Oxf Engl 1993 2021. [DOI: 10.1016/j.radphyschem.2021.109499] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
38
|
RexAB promotes the survival of Staphylococcus aureus exposed to multiple classes of antibiotics. Antimicrob Agents Chemother 2021; 65:e0059421. [PMID: 34310219 PMCID: PMC8448105 DOI: 10.1128/aac.00594-21] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Antibiotics inhibit essential bacterial processes, resulting in arrest of growth and, in some cases, cell death. Many antibiotics are also reported to trigger endogenous production of reactive oxygen species (ROS), which damage DNA, leading to induction of the mutagenic SOS response associated with the emergence of drug resistance. However, the type of DNA damage that arises and how this triggers the SOS response are largely unclear. We found that several different classes of antibiotic triggered dose-dependent induction of the SOS response in Staphylococcus aureus, indicative of DNA damage, including some bacteriostatic drugs. The SOS response was heterogenous and varied in magnitude between strains and antibiotics. However, in many cases, full induction of the SOS response was dependent upon the RexAB helicase/nuclease complex, which processes DNA double-strand breaks to produce single-stranded DNA and facilitate RecA nucleoprotein filament formation. The importance of RexAB in repair of DNA was confirmed by measuring bacterial survival during antibiotic exposure, with most drugs having significantly greater bactericidal activity against rexB mutants than against wild-type strains. For some, but not all, antibiotics there was no difference in bactericidal activity between wild type and rexB mutant under anaerobic conditions, indicative of a role for reactive oxygen species in mediating DNA damage. Taken together, this work confirms previous observations that several classes of antibiotics cause DNA damage in S. aureus and extends them by showing that processing of DNA double-strand breaks by RexAB is a major trigger of the mutagenic SOS response and promotes bacterial survival.
Collapse
|
39
|
Catalase Protects Biofilm of Staphylococcus aureus against Daptomycin Activity. Antibiotics (Basel) 2021; 10:antibiotics10050511. [PMID: 33946290 PMCID: PMC8146090 DOI: 10.3390/antibiotics10050511] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/22/2021] [Accepted: 04/27/2021] [Indexed: 12/21/2022] Open
Abstract
Daptomycin is recommended for the treatment of Staphylococcus aureus infections due to its bactericidal activity. However, its mechanism of action is poorly understood. The involvement of reactive oxygen species (ROS) in the bactericidal activity of daptomycin has been proved against planktonic S. aureus, but not against the biofilm of S. aureus. Therefore, we evaluated if ROS contributes to the effect of daptomycin against biofilm of S. aureus. Biofilms of wild type, catalase deficient and daptomycin-resistant S. aureus strains were grown in microtiter-plates. After three days, the biofilms were exposed to daptomycin with or without thiourea in the presence of a ROS indicator. After overnight incubation, the amount of ROS and the percentage of surviving bacteria were determined. The bacterial survival was higher and the amount of ROS was lower in the wild type than in the catalase deficient biofilm, demonstrating a protective effect of catalase against daptomycin. The induction of cytotoxic ROS formation by daptomycin was verified by the addition of thiourea, which reduced the amount of ROS and protected the wild type biofilm against high concentrations of daptomycin. Accordingly, only the highest concentration of daptomycin reduced the bacterial survival and increased the ROS formation in the resistant biofilm. In conclusion, daptomycin induced the production of cytotoxic levels of endogenous ROS in S. aureus biofilm and the presence of catalase protected the biofilm against the lethality of the induced ROS.
Collapse
|
40
|
Greninger AL, Addetia A, Tao Y, Adler A, Qin X. Inactivation of genes in oxidative respiration and iron acquisition pathways in pediatric clinical isolates of Small colony variant Enterobacteriaceae. Sci Rep 2021; 11:7457. [PMID: 33811225 PMCID: PMC8018945 DOI: 10.1038/s41598-021-86764-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 03/11/2021] [Indexed: 02/01/2023] Open
Abstract
Isolation of bacterial small colony variants (SCVs) from clinical specimens is not uncommon and can fundamentally change the outcome of the associated infections. Bacterial SCVs often emerge with their normal colony phenotype (NCV) co-isolates in the same sample. The basis of SCV emergence in vivo is not well understood in Gram-negative bacteria. In this study, we interrogated the causal genetic lesions of SCV growth in three pairs of NCV and SCV co-isolates of Escherichia coli, Citrobacter freundii, and Enterobacter hormaechei. We confirmed SCV emergence was attributed to limited genomic mutations: 4 single nucleotide variants in the E. coli SCV, 5 in C. freundii, and 8 in E. hormaechei. In addition, a 10.2 kb chromosomal segment containing 11 genes was deleted in the E. hormaechei SCV isolate. Each SCV had at least one coding change in a gene associated with bacterial oxidative respiration and another involved in iron capture. Chemical and genetic rescue confirmed defects in heme biosynthesis for E. coli and C. freundii and lipoic acid biosynthesis in E. hormaachei were responsible for the SCV phenotype. Prototrophic growth in all 3 SCV Enterobacteriaceae species was unaffected under anaerobic culture conditions in vitro, illustrating how SCVs may persist in vivo.
Collapse
Affiliation(s)
- Alexander L. Greninger
- grid.34477.330000000122986657Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA USA ,grid.240741.40000 0000 9026 4165Seattle Children’s Hospital, Seattle, WA USA
| | - Amin Addetia
- grid.34477.330000000122986657Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA USA ,grid.240741.40000 0000 9026 4165Seattle Children’s Hospital, Seattle, WA USA
| | - Yue Tao
- grid.16821.3c0000 0004 0368 8293Shanghai Children’s Medical Center, Translational Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Amanda Adler
- grid.240741.40000 0000 9026 4165Seattle Children’s Hospital, Seattle, WA USA
| | - Xuan Qin
- grid.34477.330000000122986657Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA USA ,grid.240741.40000 0000 9026 4165Seattle Children’s Hospital, Seattle, WA USA
| |
Collapse
|
41
|
Mora-Hernández Y, Vera Murguía E, Stinenbosch J, Hernández Jauregui P, van Dijl JM, Buist G. Molecular typing and antimicrobial resistance profiling of 33 mastitis-related Staphylococcus aureus isolates from cows in the Comarca Lagunera region of Mexico. Sci Rep 2021; 11:6912. [PMID: 33767356 PMCID: PMC7994548 DOI: 10.1038/s41598-021-86453-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 03/10/2021] [Indexed: 11/26/2022] Open
Abstract
Mastitis in cows is a major cause of economic losses and it is commonly associated with Staphylococcus aureus. Little is known about the S. aureus lineages causing mastitis in Mexican cattle. The aim of this study was to type S. aureus isolates causing mastitis in cows from the Comarca Lagunera region in Mexico in 2015-2016. Multi-locus variable number tandem repeat fingerprinting (MLVF) of 33 S. aureus isolates obtained from 210 milk samples revealed the MLVF clusters A (n = 1), B (n = 26), C (n = 5) and D (n = 1). Spa-typing showed that clusters A and B represent the spa-type t224, cluster C includes spa-types t3196 and t416, and cluster D represents spa-type t114. The different spa-types were mirrored by the masses of protein A bands as detected by Western blotting. Antimicrobial susceptibility testing showed that one isolate was susceptible to all antimicrobials tested, whereas all other strains were resistant only to benzylpenicillin. These findings show that only four S. aureus lineages, susceptible to most antimicrobials, were responsible for causing mastitis at the time of sampling. Lastly, many isolates carried the same small plasmid, designated pSAM1. The high prevalence of pSAM1 amongst the antimicrobial-susceptible isolates suggests an association with bovine colonization or mastitis rather than antimicrobial resistance.
Collapse
Affiliation(s)
- Y Mora-Hernández
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, 9700 RB, Groningen, The Netherlands
| | - E Vera Murguía
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, 9700 RB, Groningen, The Netherlands
| | - J Stinenbosch
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, 9700 RB, Groningen, The Netherlands
| | | | - Jan Maarten van Dijl
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, 9700 RB, Groningen, The Netherlands.
| | - G Buist
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, 9700 RB, Groningen, The Netherlands
| |
Collapse
|
42
|
Maliszewska I, Goldeman W. Pentamidine enhances photosensitization of Acinetobacter baumannii using diode lasers with emission of light at wavelength of ʎ = 405 nm and ʎ = 635 nm. Photodiagnosis Photodyn Ther 2021; 34:102242. [PMID: 33662618 DOI: 10.1016/j.pdpdt.2021.102242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/22/2021] [Accepted: 02/26/2021] [Indexed: 12/01/2022]
Abstract
Antimicrobial photodynamic inactivation is currently one of the most promising trends in the modern bactericidal protocols. Under the conditions defined in our studies, we found that in vitro photosensitization of A. baumannii with 5-ALA as a precursor of protoporphyrin IX (photosensitizer) reduces the concentration of viable cells in planktonic cultures, and this process can be strongly enhanced by pentamidine. Diode lasers with the peak-power wavelength of ʎ = 405 nm (radiation intensity of 26 mW cm-2) and ʎ = 635 nm (radiation intensity of 55 mW cm-2) were used in this study. It was found that a blue laser light (energy fluence of 64 J cm-2; no external photosensitizer) in the presence of pentamidine resulted in a reduction of CFU of 99.992 % compared to 99.97 % killing without pentamidine. When a red laser light was used in the experiments (energy fluence of 136 J cm-2; no external photosensitizer), the mortality rate was 99.98 % in the presence of pentamidine compared to 99.93 % of those killed without the addition of this drug. The lethal effect with 5-ALA was achieved under blue light fluence of 16 J cm-2 (in the presence of pentamidine) and 32 J cm-2 (without pentamidine). In the case of laser light of 635 nm, the lethal effect with 5-ALA was attained with energy fluence of 51 J cm-2 (with pentamidine) and 102 J cm-2 (without pentamidine). The possible roles of pentamidine in enhancing photodynamic inactivation of A. baumannii have been discussed.
Collapse
Affiliation(s)
- Irena Maliszewska
- Department of Organic and Medicinal Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370, Wrocław, Poland.
| | - Waldemar Goldeman
- Department of Organic and Medicinal Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370, Wrocław, Poland
| |
Collapse
|
43
|
Rapacka-Zdonczyk A, Wozniak A, Nakonieczna J, Grinholc M. Development of Antimicrobial Phototreatment Tolerance: Why the Methodology Matters. Int J Mol Sci 2021; 22:2224. [PMID: 33672375 PMCID: PMC7926562 DOI: 10.3390/ijms22042224] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 12/12/2022] Open
Abstract
Due to rapidly growing antimicrobial resistance, there is an urgent need to develop alternative, non-antibiotic strategies. Recently, numerous light-based approaches, demonstrating killing efficacy regardless of microbial drug resistance, have gained wide attention and are considered some of the most promising antimicrobial modalities. These light-based therapies include five treatments for which high bactericidal activity was demonstrated using numerous in vitro and in vivo studies: antimicrobial blue light (aBL), antimicrobial photodynamic inactivation (aPDI), pulsed light (PL), cold atmospheric plasma (CAP), and ultraviolet (UV) light. Based on their multitarget activity leading to deleterious effects to numerous cell structures-i.e., cell envelopes, proteins, lipids, and genetic material-light-based treatments are considered to have a low risk for the development of tolerance and/or resistance. Nevertheless, the most recent studies indicate that repetitive sublethal phototreatment may provoke tolerance development, but there is no standard methodology for the proper evaluation of this phenomenon. The statement concerning the lack of development of resistance to these modalities seem to be justified; however, the most significant motivation for this review paper was to critically discuss existing dogma concerning the lack of tolerance development, indicating that its assessment is more complex and requires better terminology and methodology.
Collapse
Affiliation(s)
- Aleksandra Rapacka-Zdonczyk
- Laboratory of Molecular Diagnostics, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland; (A.R.-Z.); (A.W.); (J.N.)
- Department of Pharmaceutical Microbiology, The Faculty of Pharmacy, Medical University of Gdansk, Hallera 107, 80-416 Gdansk, Poland
| | - Agata Wozniak
- Laboratory of Molecular Diagnostics, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland; (A.R.-Z.); (A.W.); (J.N.)
| | - Joanna Nakonieczna
- Laboratory of Molecular Diagnostics, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland; (A.R.-Z.); (A.W.); (J.N.)
| | - Mariusz Grinholc
- Laboratory of Molecular Diagnostics, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland; (A.R.-Z.); (A.W.); (J.N.)
| |
Collapse
|
44
|
Targeting the bacterial SOS response for new antimicrobial agents: drug targets, molecular mechanisms and inhibitors. Future Med Chem 2021; 13:143-155. [PMID: 33410707 DOI: 10.4155/fmc-2020-0310] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Antimicrobial resistance is a pressing threat to global health, with multidrug-resistant pathogens becoming increasingly prevalent. The bacterial SOS pathway functions in response to DNA damage that occurs during infection, initiating several pro-survival and resistance mechanisms, such as DNA repair and hypermutation. This makes SOS pathway components potential targets that may combat drug-resistant pathogens and decrease resistance emergence. This review discusses the mechanism of the SOS pathway; the structure and function of potential targets AddAB, RecBCD, RecA and LexA; and efforts to develop selective small-molecule inhibitors of these proteins. These inhibitors may serve as valuable tools for target validation and provide the foundations for desperately needed novel antibacterial therapeutics.
Collapse
|
45
|
Pieranski M, Sitkiewicz I, Grinholc M. Increased photoinactivation stress tolerance of Streptococcus agalactiae upon consecutive sublethal phototreatments. Free Radic Biol Med 2020; 160:657-669. [PMID: 32916279 DOI: 10.1016/j.freeradbiomed.2020.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/24/2020] [Accepted: 09/02/2020] [Indexed: 02/08/2023]
Abstract
Streptococcus agalactiae (Group B Streptococcus, GBS) is a common commensal bacterium in adults but remains a leading source of invasive infections in newborns, pregnant women, and the elderly, and more recently, causes an increased incidence of invasive disease in nonpregnant adults. Reduced penicillin susceptibility and emerging resistance to non-β-lactams pose challenges for the development and implementation of novel, nonantimicrobial strategies to reduce the burden of GBS infections. Antimicrobial photodynamic inactivation (aPDI) via the production of singlet oxygen or other reactive oxygen species leads to the successful eradication of pathogenic bacteria, affecting numerous cellular targets of microbial pathogens and indicating a low risk of resistance development. Nevertheless, we have previously reported possible aPDI tolerance development upon repeated sublethal aPDI applications; thus, the current work was aimed at investigating whether aPDI tolerance could be observed for GBS and what mechanisms could cause it. To address this problem, 10 cycles of sublethal aPDI treatments employing rose bengal as a photosensitizer, were applied to the S. agalactiae ATCC 27956 reference strain and two clinical isolates (2306/02 and 2974/07, serotypes III and V, respectively). We demonstrated aPDI tolerance development and stability after 5 cycles of subculturing with no aPDI exposure. Though the treatment resulted in a stable phenotype, no increases in mutation rate or accumulated genetic alterations were observed (employing a RIF-, CIP-, STR-resistant mutant selection assay and cyl sequencing, respectively). qRT-PCR analysis demonstrated that 10 sublethal aPDI exposures led to increased expression of all tested major oxidative stress response elements; changes in sodA, ahpC, npx, cylE, tpx and recA expression indicate possible mechanisms of developed tolerance. Increased expression upon sublethal aPDI treatment was reported for all but two genes, namely, ahpC and cylE. aPDI targeting cylE was further supported by colony morphology changes induced with 10 cycles of aPDI (increased SCV population, increased hemolysis, increased numbers of dark- and unpigmented colonies). In oxidant killing assays, aPDI-tolerant strains demonstrated no increased tolerance to hypochlorite, superoxide (paraquat), singlet oxygen (new methylene blue) or oxidative stress induced by aPDI employing a structurally different photosensitizer, i.e., zinc phthalocyanine, indicating a lack of cross resistance. The results indicate that S. agalactiae may develop stable aPDI tolerance but not resistance when subjected to multiple sublethal phototreatments, and this risk should be considered significant when defining efficient anti-S. agalactiae aPDI protocols.
Collapse
Affiliation(s)
- Michal Pieranski
- Intercollegiate Faculty of Biotechnology, Laboratory of Molecular Diagnostics, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307, Gdansk, Poland
| | - Izabela Sitkiewicz
- Department of Drug Biotechnology and Bioinformatics, National Medicines Institute, Chelmska 30/34, 00-725, Warszawa, Poland
| | - Mariusz Grinholc
- Intercollegiate Faculty of Biotechnology, Laboratory of Molecular Diagnostics, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307, Gdansk, Poland.
| |
Collapse
|
46
|
Ha KP, Clarke RS, Kim GL, Brittan JL, Rowley JE, Mavridou DAI, Parker D, Clarke TB, Nobbs AH, Edwards AM. Staphylococcal DNA Repair Is Required for Infection. mBio 2020; 11:e02288-20. [PMID: 33203752 PMCID: PMC7683395 DOI: 10.1128/mbio.02288-20] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/14/2020] [Indexed: 01/07/2023] Open
Abstract
To cause infection, Staphylococcus aureus must withstand damage caused by host immune defenses. However, the mechanisms by which staphylococcal DNA is damaged and repaired during infection are poorly understood. Using a panel of transposon mutants, we identified the rexBA operon as being important for the survival of Staphylococcus aureus in whole human blood. Mutants lacking rexB were also attenuated for virulence in murine models of both systemic and skin infections. We then demonstrated that RexAB is a member of the AddAB family of helicase/nuclease complexes responsible for initiating the repair of DNA double-strand breaks. Using a fluorescent reporter system, we were able to show that neutrophils cause staphylococcal DNA double-strand breaks through reactive oxygen species (ROS) generated by the respiratory burst, which are repaired by RexAB, leading to the induction of the mutagenic SOS response. We found that RexAB homologues in Enterococcus faecalis and Streptococcus gordonii also promoted the survival of these pathogens in human blood, suggesting that DNA double-strand break repair is required for Gram-positive bacteria to survive in host tissues. Together, these data demonstrate that DNA is a target of host immune cells, leading to double-strand breaks, and that the repair of this damage by an AddAB-family enzyme enables the survival of Gram-positive pathogens during infection.IMPORTANCE To cause infection, bacteria must survive attack by the host immune system. For many bacteria, including the major human pathogen Staphylococcus aureus, the greatest threat is posed by neutrophils. These immune cells ingest the invading organisms and try to kill them with a cocktail of chemicals that includes reactive oxygen species (ROS). The ability of S. aureus to survive this attack is crucial for the progression of infection. However, it was not clear how the ROS damaged S. aureus and how the bacterium repaired this damage. In this work, we show that ROS cause breaks in the staphylococcal DNA, which must be repaired by a two-protein complex known as RexAB; otherwise, the bacterium is killed, and it cannot sustain infection. This provides information on the type of damage that neutrophils cause S. aureus and the mechanism by which this damage is repaired, enabling infection.
Collapse
Affiliation(s)
- Kam Pou Ha
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Rebecca S Clarke
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Gyu-Lee Kim
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Jane L Brittan
- Bristol Dental School, University of Bristol, Bristol, United Kingdom
| | - Jessica E Rowley
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Despoina A I Mavridou
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - Dane Parker
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Thomas B Clarke
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Angela H Nobbs
- Bristol Dental School, University of Bristol, Bristol, United Kingdom
| | - Andrew M Edwards
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| |
Collapse
|
47
|
Ranganathan N, Johnson R, Edwards AM. The general stress response of Staphylococcus aureus promotes tolerance of antibiotics and survival in whole human blood. MICROBIOLOGY (READING, ENGLAND) 2020; 166:1088-1094. [PMID: 33095698 PMCID: PMC7723259 DOI: 10.1099/mic.0.000983] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/06/2020] [Indexed: 12/29/2022]
Abstract
Staphylococcus aureus is a frequent cause of invasive human infections such as bacteraemia and infective endocarditis. These infections frequently relapse or become chronic, suggesting that the pathogen has mechanisms to tolerate the twin threats of therapeutic antibiotics and host immunity. The general stress response of S. aureus is regulated by the alternative sigma factor B (σB) and provides protection from multiple stresses including oxidative, acidic and heat. σB also contributes to virulence, intracellular persistence and chronic infection. However, the protective effect of σB on bacterial survival during exposure to antibiotics or host immune defences is poorly characterized. We found that σB promotes the survival of S. aureus exposed to the antibiotics gentamicin, ciprofloxacin, vancomycin and daptomycin, but not oxacillin or clindamycin. We also found that σB promoted staphylococcal survival in whole human blood, most likely via its contribution to oxidative stress resistance. Therefore, we conclude that the general stress response of S. aureus may contribute to the development of chronic infection by conferring tolerance to both antibiotics and host immune defences.
Collapse
Affiliation(s)
- Nisha Ranganathan
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Rd, London, SW7 2AZ, UK
- Present address: Charing Cross Hospital, Fulham, Palace Road, W6 8RF, UK
| | - Rebecca Johnson
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Rd, London, SW7 2AZ, UK
- Present address: Horizon Discovery, Waterbeach, Cambridge, CB25 9TL, UK
| | - Andrew M. Edwards
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Rd, London, SW7 2AZ, UK
| |
Collapse
|
48
|
Memar MY, Yekani M, Celenza G, Poortahmasebi V, Naghili B, Bellio P, Baghi HB. The central role of the SOS DNA repair system in antibiotics resistance: A new target for a new infectious treatment strategy. Life Sci 2020; 262:118562. [PMID: 33038378 DOI: 10.1016/j.lfs.2020.118562] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/15/2020] [Accepted: 10/01/2020] [Indexed: 01/19/2023]
Abstract
Bacteria have a considerable ability and potential to acquire resistance against antimicrobial agents by acting diverse mechanisms such as target modification or overexpression, multidrug transporter systems, and acquisition of drug hydrolyzing enzymes. Studying the mechanisms of bacterial cell physiology is mandatory for the development of novel strategies to control the antimicrobial resistance phenomenon, as well as for the control of infections in clinics. The SOS response is a cellular DNA repair mechanism that has an essential role in the bacterial biologic process involved in resistance to antibiotics. The activation of the SOS network increases the resistance and tolerance of bacteria to stress and, as a consequence, to antimicrobial agents. Therefore, SOS can be an applicable target for the discovery of new antimicrobial drugs. In the present review, we focus on the central role of SOS response in bacterial resistance mechanisms and its potential as a new target for control of resistant pathogens.
Collapse
Affiliation(s)
- Mohammad Yousef Memar
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Students' Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mina Yekani
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Giuseppe Celenza
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Vahdat Poortahmasebi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Behrooz Naghili
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pierangelo Bellio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Hossein Bannazadeh Baghi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
49
|
Cobb LH, McCabe EM, Priddy LB. Therapeutics and delivery vehicles for local treatment of osteomyelitis. J Orthop Res 2020; 38:2091-2103. [PMID: 32285973 PMCID: PMC8117475 DOI: 10.1002/jor.24689] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/07/2020] [Accepted: 04/11/2020] [Indexed: 02/04/2023]
Abstract
Osteomyelitis, or the infection of the bone, presents a major complication in orthopedics and may lead to prolonged hospital visits, implant failure, and in more extreme cases, amputation of affected limbs. Typical treatment for this disease involves surgical debridement followed by long-term, systemic antibiotic administration, which contributes to the development of antibiotic-resistant bacteria and has limited ability to eradicate challenging biofilm-forming pathogens including Staphylococcus aureus-the most common cause of osteomyelitis. Local delivery of high doses of antibiotics via traditional bone cement can reduce systemic side effects of an antibiotic. Nonetheless, growing concerns over burst release (then subtherapeutic dose) of antibiotics, along with microbial colonization of the nondegradable cement biomaterial, further exacerbate antibiotic resistance and highlight the need to engineer alternative antimicrobial therapeutics and local delivery vehicles with increased efficacy against, in particular, biofilm-forming, antibiotic-resistant bacteria. Furthermore, limited guidance exists regarding both standardized formulation protocols and validated assays to predict efficacy of a therapeutic against multiple strains of bacteria. Ideally, antimicrobial strategies would be highly specific while exhibiting a broad spectrum of bactericidal activity. With a focus on S. aureus infection, this review addresses the efficacy of novel therapeutics and local delivery vehicles, as alternatives to the traditional antibiotic regimens. The aim of this review is to discuss these components with regards to long bone osteomyelitis and to encourage positive directions for future research efforts.
Collapse
Affiliation(s)
- Leah H. Cobb
- Department of Agricultural and Biological Engineering, Mississippi State University, Mississippi State, MS, USA
| | - Emily M. McCabe
- Department of Agricultural and Biological Engineering, Mississippi State University, Mississippi State, MS, USA,Department of Mechanical Engineering, Mississippi State University, Mississippi State, MS, USA
| | - Lauren B. Priddy
- Department of Agricultural and Biological Engineering, Mississippi State University, Mississippi State, MS, USA,corresponding author: Contact: , (662) 325-5988, Department of Agricultural and Biological Engineering, Mississippi State University, 130 Creelman Street, Mississippi State, MS, USA 39762
| |
Collapse
|
50
|
A comprehensive review of bacterial osteomyelitis with emphasis on Staphylococcus aureus. Microb Pathog 2020; 148:104431. [PMID: 32801004 DOI: 10.1016/j.micpath.2020.104431] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 02/04/2023]
Abstract
Osteomyelitis, a significant infection of bone tissue, gives rise to two main groups of infection: acute and chronic. These groups are further categorized in terms of the duration of infection. Usually, children and adults are more susceptible to acute and chronic infections, respectively. The aforementioned groups of osteomyelitis share almost 80% of the corresponding bacterial pathogens. Among all bacteria, Staphylococcus aureus (S. aureus) is a significant pathogen and is associated with a high range of osteomyelitis symptoms. S. aureus has many strategies for interacting with host cells including Small Colony Variant (SCV), biofilm formation, and toxin secretion. In addition, it induces an inflammatory response and causes host cell death by apoptosis and necrosis. However, any possible step to take in this respect is dependent on the conditions and host responses. In the absence of any immune responses and antibiotics, bacteria actively duplicate themselves; however, in the presence of phagocytic cell and harassing conditions, they turn into a SCV, remaining sustainable for a long time. SCV is characterized by notable advantages such as (a) intracellular life that mediates a dam against immune cells and (b) low ATP production that mediates resistance against antibiotics.
Collapse
|