1
|
Silva-Santana G, Baêta Júnior ES, Silva Conceição GM, Aguiar-Alves F, Lima Brandão ML, Lopes-Torres EJ, Mattos-Guaraldi AL. Intervention of Corynebacterium striatum in the sessile lifestyle of Staphylococcus aureus wild-type and mutants for ica genes in polymicrobial biofilms. Microb Pathog 2025; 204:107577. [PMID: 40222568 DOI: 10.1016/j.micpath.2025.107577] [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: 06/29/2024] [Revised: 04/05/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
This study investigated the interactions between Corynebacterium striatum and Staphylococcus aureus, two bacterial species commonly found in the human microbiota, particularly colonizing the skin and mucous membranes. Both organisms, however, are also capable of causing acute and chronic infections. While S. aureus is widely recognized as a clinically significant pathogen, C. striatum is frequently underestimated and often regarded as a contaminant-even when isolated in pure culture from nosocomial infections. The ability of these microorganisms to develop multidrug resistance and form biofilms complicates the management of the infections they cause. This study focused on the interaction between C. striatum and S. aureus, particularly the influence of the former on the pathogenic potential of the latter, emphasizing biofilm formation in S. aureus mutants deficient in the icaR and icaC genes. Antimicrobial susceptibility testing revealed that 85.7 % of the S. aureus strains were multidrug-resistant, with all strains resistant to β-lactam antibiotics. Additionally, 55.6 % of the strains produced strong slime on Congo Red agar, indicating a high potential for biofilm formation. In monomicrobial assays, both C. striatum and S. aureus exhibited enhanced adhesion to hydrophilic surfaces. In polymicrobial settings, C. striatum predominated in most cases: on glass surfaces, 70 % of biofilms were dominated by C. striatum, 20 % by S. aureus, and 10 % showed an even distribution. On polystyrene, 80 % of the biofilms were dominated by C. striatum, while 20 % were dominated by S. aureus. Analysis of extracellular polymeric substances (EPS) revealed distinct compositional profiles: C. striatum primarily produced proteinaceous matrices, whereas S. aureus biofilms were rich in polysaccharides. Ultrastructural examination showed that S. aureus formed dense clusters embedded in large amounts of EPS, while C. striatum biofilms exhibited lower EPS production overall. Furthermore, the effect of C. striatum-derived compounds on S. aureus biofilms was assessed. In 90 % of co-cultured strains, a progressive decrease in sessile cell populations was observed, accompanied by an increase in planktonic cells. This finding suggests that C. striatum can disrupt the biofilm integrity of S. aureus, potentially modulating its pathogenic phenotype. In conclusion, the results demonstrate that C. striatum competes effectively with S. aureus for surface colonization and, under certain conditions, may induce a transition of S. aureus from a sessile to a planktonic state. These findings highlight the complexity of interspecies interactions in polymicrobial communities and suggest that C. striatum may play a modulatory role in S. aureus virulence. Such insights have important implications for the understanding and treatment of polymicrobial infections.
Collapse
Affiliation(s)
- Giorgio Silva-Santana
- Health Science Center, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Laboratory of Diphtheria and Corynebacteria of Clinical Relevance, Faculty of Medical Sciences, University of the State of Rio de Janeiro. The Collaborating Centre for Reference and Research on Diphtheria/National Health Foundation/Ministry of Health, Rio de Janeiro, Brazil.
| | - Eustáquio Souza Baêta Júnior
- Department of Mechanical Engineering, Postgraduate Program in Mechanical Engineering, FEN, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Greice Maria Silva Conceição
- Systems, Analytical Indicators and Data Section, Department of Quality Control, Bio- Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Fabio Aguiar-Alves
- Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, FL, 33401, USA
| | - Marcelo Luiz Lima Brandão
- Laboratory of Microbiology Control, Department of Quality Control, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Eduardo José Lopes-Torres
- Laboratório de Helmintologia Romero Lascasas Porto, Departamento de Microbiologia, Imunologia e Parasitologia, Universidade do Estado do Rio de Janeiro, Brazil; Laboratório Multiusuário de Parasitologia, Departamento de Microbiologia, Imunologia e Parasitologia, Universidade do Estado do Rio de Janeiro, Brazil
| | - Ana Luiza Mattos-Guaraldi
- Health Science Center, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Laboratory of Diphtheria and Corynebacteria of Clinical Relevance, Faculty of Medical Sciences, University of the State of Rio de Janeiro. The Collaborating Centre for Reference and Research on Diphtheria/National Health Foundation/Ministry of Health, Rio de Janeiro, Brazil
| |
Collapse
|
2
|
He X, Zhang W, Liu J, Liu J, Chen Y, Luan C, Zhang J, Bao G, Lin X, Muh F, Lin T, Lu F. The global regulatory role of RsbUVW in virulence and biofilm formation in MRSA. Microb Pathog 2025; 203:107508. [PMID: 40158706 DOI: 10.1016/j.micpath.2025.107508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 03/22/2025] [Accepted: 03/23/2025] [Indexed: 04/02/2025]
Abstract
The widespread prevalence of methicillin-resistant Staphylococcus aureus (MRSA) has caused serious challenges to clinical treatment. This study was designed to explore effective targets for MRSA prevention and control. The key virulence regulator was screened through the correlation analysis between virulence and various regulatory factors in the main clinical epidemic MRSA. The potential key factors were inactivated to further evaluate the inhibitory effect on the virulence of MRSA standard strain S. aureus ATCC43300 and its influence on drug resistance and biofilm formation. Enterobacterial repetitive intergenic consensus-PCR was used to analyze the clinical epidemic genotypes of MRSA. The virulence of MRSA was evaluated mainly by measuring its adhesion and invasion ability to A549 cells, the lethality to Galleria mellonella larvae, and the transcription level of related genes. The biofilm formation was assessed by crystal violet staining on polystyrene microplates. The results showed that most virulence factors of clinical representative MRSA strain were significantly positively correlated with RsbUVW system. After knocking out the rsbV gene, a key component of the rsbUVW system, the virulence of S. aureus ATCC43300 was significantly reduced (P < 0.05), as indicated by a significant decrease in lethality against G. mellonella larvae and invasion against A549 cells, and a significant decrease in the expression of immune escape related virulence factors polysaccharide intercellular adhesin (PIA) and staphyloxanthin. The biomass and stability of protein-dependent biofilm by S. aureus ATCC43300 were significantly increased. This study will provide useful information for the effective prevention and control of MRSA.
Collapse
Affiliation(s)
- Xinlong He
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, 225001, China; The Key Laboratory of the Jiangsu Higher Education Institutions for Nucleic Acid & Cell Fate Regulation (Yangzhou University), Yangzhou, 225001, China; Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225001, China
| | - Wenwen Zhang
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; Department of Clinical Laboratory, Changning Maternity and Infant Health Hospital, Affiliated Hospital of East China Normal University, Shanghai, 200050, China
| | - Jie Liu
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Jiali Liu
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Yinsong Chen
- Department of Lung, Third People's Hospital of Yangzhou, Yangzhou, China
| | - Changjiao Luan
- Department of Lung, Third People's Hospital of Yangzhou, Yangzhou, China
| | - Jun Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China
| | - Guangyu Bao
- Department of Clinical Laboratory, First Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China
| | - Xiangfang Lin
- Department of Clinical Laboratory, First Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China
| | - Fauzi Muh
- Department of Epidemiology & Tropical Diseases, Faculty of Public Health, Universitas Diponegoro, Tembalang, Semarang, 50275, Indonesia
| | - Tao Lin
- Department of Clinical Laboratory, First Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China.
| | - Feng Lu
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China.
| |
Collapse
|
3
|
Elbaiomy RG, Luo X, Guo R, Deng S, Du M, El-Sappah AH, Bakeer M, Azzam MM, Elolimy AA, Madkour M, Li Z, Zhang Z. Antibiotic resistance in Helicobacter pylori: a genetic and physiological perspective. Gut Pathog 2025; 17:35. [PMID: 40410811 PMCID: PMC12102891 DOI: 10.1186/s13099-025-00704-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 04/25/2025] [Indexed: 05/25/2025] Open
Abstract
The identification of Helicobacter pylori (H. pylori) infection as the primary etiology of gastroduodenal diseases represents a significant advancement in the field of gastroenterology. The management of these diseases has undergone a substantial transformation, and antibiotic treatment is now universally applicable. H. pylori has been the subject of numerous investigations to determine the prevalence of antibiotic resistance. However, many of these studies are limited, particularly regarding the number and representativeness of the strains assessed. Genetic and physiological modifications, such as gene mutations, efflux pump alterations, biofilm formation, and coccoid formation, contribute to the observed resistance. Our review focuses on the emergence of antibiotic-resistant strains, particularly emphasizing the various modifications of H. pylori that confer this resistance. In conclusion, we elucidate the challenges, potential solutions, and prospects in this field, providing researchers with the knowledge necessary to overcome the resistance exhibited by H. pylori.
Collapse
Affiliation(s)
- Rania G Elbaiomy
- Department of Biological Engineering, Sichuan University of Science & Engineering, Zigong, 643000, China
| | - Xiaoling Luo
- Department of Gastroenterology, FuShun People's Hospital, Zigong, 643000, China
| | - Rong Guo
- Department of Gastroenterology, FuShun People's Hospital, Zigong, 643000, China
| | - Shiyuan Deng
- Department of Biological Engineering, Sichuan University of Science & Engineering, Zigong, 643000, China
| | - Meifang Du
- Department of Biological Engineering, Sichuan University of Science & Engineering, Zigong, 643000, China
| | - Ahmed H El-Sappah
- School of Agriculture, Forestry and Food Engineering, Yibin University, Yibin, 644000, Sichuan, China
- Department of Genetics, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | - Mohammed Bakeer
- Division of Hematology and Medical Oncology, Mayo Clinic, Jacksonville, FL, USA
- Division of Internal Medicine-Clinical Hematology, Al-Azhar University, Cairo, 11765, Egypt
| | - Mahmoud M Azzam
- Department of Animal Production, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed A Elolimy
- Department of Integrative Agriculture, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain, Abu Dhabi, 15551, United Arab Emirates.
| | - Mahmoud Madkour
- Animal Production Department, National Research Centre, Dokki, 12622, Giza, Egypt
| | - Zaixin Li
- Department of Biological Engineering, Sichuan University of Science & Engineering, Zigong, 643000, China.
| | - Zhi Zhang
- Department of Biological Engineering, Sichuan University of Science & Engineering, Zigong, 643000, China.
| |
Collapse
|
4
|
Silva-Santana G. Staphylococcus aureus: Dynamics of pathogenicity and antimicrobial-resistance in hospital and community environments - Comprehensive overview. Res Microbiol 2025; 176:104267. [PMID: 39805330 DOI: 10.1016/j.resmic.2025.104267] [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: 10/24/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
This study reviews Staphylococcus aureus, a significant pathogen in both hospital and community-acquired infections, addressing its epidemiology, pathogenesis, and antimicrobial resistance. It highlights virulence mechanisms, such as adhesion factors, toxins, enzymes, and biofilms, which contribute to survival and immune evasion. The spread of resistance occurs through the transfer of mobile genetic elements like SCCmec and genetic mutations. The analysis also compares hospital and community strains, including multidrug-resistant lineages like MRSA, VISA, and VRSA. The study concludes that S. aureus presents a major public health challenge, requiring new therapeutic approaches and preventive strategies.
Collapse
Affiliation(s)
- Giorgio Silva-Santana
- Health Science Center, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro (RJ), Brazil.
| |
Collapse
|
5
|
Tian LL, Li Y, Yang R, Jiang Y, He JJ, Wang H, Chen LQ, Zhu WY, Xue T, Li BB. Low concentrations of tetrabromobisphenol A promote the biofilm formation of methicillin-resistant Staphylococcus aureus. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 283:116853. [PMID: 39137468 DOI: 10.1016/j.ecoenv.2024.116853] [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: 03/31/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024]
Abstract
The effect and underlying mechanism of tetrabromobisphenol A (TBBPA), a plastic additive, on biofilm formation of methicillin-resistant Staphylococcus aureus (MRSA USA300) remain unknown. This study first investigated the impact of different concentrations of TBBPA on the growth and biofilm formation of USA300. The results indicated that a low concentration (0.5 mg/L) of TBBPA promoted the growth and biofilm formation of USA300, whereas high concentrations (5 mg/L and 10 mg/L) of TBBPA had inhibitory effects. Further exploration revealed that the low concentration of TBBPA enhance biofilm formation by promoting the synthesis of extracellular proteins, release of extracellular DNA (eDNA), and production of staphyloxanthin. RTqPCR analysis demonstrated that the low concentration of TBBPA upregulated genes associated with extracellular protein synthesis (sarA, fnbA, fnbB, aur) and eDNA formation (atlA) and increased the expression of genes involved in staphyloxanthin biosynthesis (crtM), suggesting a potential mechanism for enhanced resistance of USA300 to adverse conditions. These findings shed light on how low concentrations of TBBPA facilitate biofilm formation in USA300 and highlight the indirect impact of plastic additives on pathogenic bacteria in terms of human health. In the future, in-depth studies about effects of plastic additives on pathogenicity of pathogenic bacteria should be conducted. CAPSULE: The protein and eDNA contents in biofilms of methicillin-resistant Staphylococcus aureus are increased by low concentrations of TBBPA.
Collapse
Affiliation(s)
- Lin-Lin Tian
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yun Li
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Rui Yang
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Ying Jiang
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Jiao-Jiao He
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Hui Wang
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Li-Qi Chen
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Wen-Ya Zhu
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Ting Xue
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China; Food Procession Research Institute, Anhui Agricultural University, Hefei, Anhui 230036, China.
| | - Bing-Bing Li
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China.
| |
Collapse
|
6
|
Huang Q, Zhu L, Huang F, Zhao Y, Wang H, Luan S, Xiao C. Novel quinazolin-6-yl Isoindolinone: Altering polysaccharide chemstructure for antibacterial efficacy against Staphylococcus aureus. Int J Biol Macromol 2024; 280:135650. [PMID: 39278453 DOI: 10.1016/j.ijbiomac.2024.135650] [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: 08/02/2024] [Revised: 09/03/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
The ongoing development of novel strategies to combat Staphylococcus aureus and eliminate its biofilm formation has gained significant attention for human health. Antibiotic-resistant S. aureus necessitates the development of novel antibacterial agents with new mechanism of action. This study introduced a promising recently synthesized quinazolin-6-yl isoindolinone (IQE-X1), which exhibited potent antibacterial and antibiofilm efficacy with average median inhibitory concentration (IC50) of 3.37 μg mL-1 and minimal inhibitory concentration (MIC) of 12.5 μg mL-1, coupled with its ability to reduce cell surface hydrophobicity. IQE-X1 dose-dependently decreased extracellular polysaccharides (EPS) and its component monosaccharides, including rhamnose, arabinose, glucosamine, galactose, glucose, xylose, mannose, and ribose, accompanied by an increase in capsular polysaccharides (CP) and its individual monosaccharides, especially glucosamine. IQE-X1 demonstrated specificity in modulating the structural profiles of EPS and CP by altering the compositional ratios of their component monosaccharides. The potential mechanism of polysaccharide modulation was preliminarily elucidated through the response of β-N-acetylaminoglucosidase to IQE-X1 and their direct binding interaction. These findings provide new insights into the potential manipulation of the chemstructure of these biologically important macromolecules, EPS and CP, and highlight the antibacterial potential of IQE-X1 as a polysaccharide modulator for the development of more effective polysaccharide-targeted strategies against S. aureus.
Collapse
Affiliation(s)
- Qingchun Huang
- Shanghai Key Lab of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, PR China.
| | - Lisong Zhu
- Shanghai Key Lab of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, PR China
| | - Fengcheng Huang
- Shanghai Key Lab of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, PR China
| | - Yanjun Zhao
- Shanghai Key Lab of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, PR China
| | - Hongye Wang
- Shanghai Key Lab of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, PR China
| | - Shaorong Luan
- School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Ciying Xiao
- School of Biotechnology, East China University of Science and Technology, Shanghai 200237, PR China
| |
Collapse
|
7
|
Ali A, Riaz S. Emerging threats of high biofilm formation and antibiotic resistance in clinical methicillin-resistant Staphylococcus aureus (MRSA) isolates from Pakistan. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2024; 121:105592. [PMID: 38614413 DOI: 10.1016/j.meegid.2024.105592] [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: 02/21/2024] [Revised: 04/05/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
OBJECTIVES This multicenter study, conducted from a One Health perspective, aimed to comprehensively examine the prevalence of methicillin-resistant Staphylococcus aureus (MRSA) infections and their biofilm-forming capabilities in Pakistan. Phylogenetic analysis of the study isolates was also performed. METHODS A total of 150 MRSA isolates were screened from various clinical samples using Cefoxitin antibiotic discs. Genotypic confirmation was conducted through mecA, S. aureus-specific nuc, and 16S rRNA genes. Biofilm formation was assessed using Congo red agar (CRA) and slime layer quantification methods. The intercellular adhesion (ica) operon genes, specifically icaA and icaD, were detected. Phylogenetic analysis utilized the 16S rRNA sequences. Statistical associations between various parameters were determined using chi-square analysis. RESULTS The presence of the mecA gene was observed in 131 out of 150 isolates (87.3%). CRA identified 28% and 40% of isolates as strong and moderate biofilm producers, respectively, while 9.3% were classified as non-biofilm producers. The slime layer assay exhibited higher sensitivity, classifying only 4.7% of isolates as non-biofilm producers. Biofilm-forming genes icaA and icaD were detected in 85.3% and 86.7% of the isolates, respectively. Antibiotic resistance was more prevalent among biofilm-forming isolates, particularly against ciprofloxacin, levofloxacin, erythromycin, trimethoprim-sulfamethoxazole, and fosfomycin. Ceftaroline demonstrated efficacy irrespective of biofilm-forming abilities. Conversely, non-biofilm producers exhibited complete susceptibility to clarithromycin and tigecycline. CONCLUSIONS Clinical MRSA strains exhibit a substantial potential for biofilm formation, contributing to a resistant phenotype. Routine antibiotic testing in clinical settings that overlook the biofilm aspect may lead to the failure of empiric antibiotic therapy.
Collapse
Affiliation(s)
- Asad Ali
- Institute of Microbiology and Molecular Genetics, University of the Punjab, Quaid-i-Azam Campus, Lahore 54590, Pakistan.
| | - Saba Riaz
- Institute of Microbiology and Molecular Genetics, University of the Punjab, Quaid-i-Azam Campus, Lahore 54590, Pakistan; CitiLab and Research Centre, 525-A Faisal Town, Lahore, Pakistan.
| |
Collapse
|
8
|
Watanabe S, Nsofor CA, Thitiananpakorn K, Tan XE, Aiba Y, Takenouchi R, Kiga K, Sasahara T, Miyanaga K, Veeranarayanan S, Shimamori Y, Lian AYS, Nguyen TM, Nguyen HM, Alessa O, Kumwenda GP, Jayathilake S, Revilleza JEC, Baranwal P, Nishikawa Y, Li FY, Kawaguchi T, Sankaranarayanan S, Arbaah M, Zhang Y, Maniruzzaman, Liu Y, Sarah H, Li J, Sugano T, Ho TMD, Batbold A, Nayanjin T, Cui L. Metabolic remodeling by RNA polymerase gene mutations is associated with reduced β-lactam susceptibility in oxacillin-susceptible MRSA. mBio 2024; 15:e0033924. [PMID: 38988221 PMCID: PMC11237739 DOI: 10.1128/mbio.00339-24] [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: 02/01/2024] [Accepted: 03/27/2024] [Indexed: 07/12/2024] Open
Abstract
The emergence of oxacillin-susceptible methicillin-resistant Staphylococcus aureus (OS-MRSA) has imposed further challenges to the clinical management of MRSA infections. When exposed to β-lactam antibiotics, these strains can easily acquire reduced β-lactam susceptibility through chromosomal mutations, including those in RNA polymerase (RNAP) genes such as rpoBC, which may then lead to treatment failure. Despite the increasing prevalence of such strains and the apparent challenges they pose for diagnosis and treatment, there is limited information available on the actual mechanisms underlying such chromosomal mutation-related transitions to reduced β-lactam susceptibility, as it does not directly associate with the expression of mecA. This study investigated the cellular physiology and metabolism of six missense mutants with reduced oxacillin susceptibility, each carrying respective mutations on RpoBH929P, RpoBQ645H, RpoCG950R, RpoCG498D, RpiAA64E, and FruBA211E, using capillary electrophoresis-mass spectrometry-based metabolomics analysis. Our results showed that rpoBC mutations caused RNAP transcription dysfunction, leading to an intracellular accumulation of ribonucleotides. These mutations also led to the accumulation of UDP-Glc/Gal and UDP-GlcNAc, which are precursors of UTP-associated peptidoglycan and wall teichoic acid. Excessive amounts of building blocks then contributed to the cell wall thickening of mutant strains, as observed in transmission electron microscopy, and ultimately resulted in decreased susceptibility to β-lactam in OS-MRSA. IMPORTANCE The emergence of oxacillin-susceptible methicillin-resistant Staphylococcus aureus (OS-MRSA) strains has created new challenges for treating MRSA infections. These strains can become resistant to β-lactam antibiotics through chromosomal mutations, including those in the RNA polymerase (RNAP) genes such as rpoBC, leading to treatment failure. This study investigated the mechanisms underlying reduced β-lactam susceptibility in four rpoBC mutants of OS-MRSA. The results showed that rpoBC mutations caused RNAP transcription dysfunction, leading to an intracellular accumulation of ribonucleotides and precursors of peptidoglycan as well as wall teichoic acid. This, in turn, caused thickening of the cell wall and ultimately resulted in decreased susceptibility to β-lactam in OS-MRSA. These findings provide insights into the mechanisms of antibiotic resistance in OS-MRSA and highlight the importance of continued research in developing effective treatments to combat antibiotic resistance.
Collapse
Affiliation(s)
- Shinya Watanabe
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Chijioke A Nsofor
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
- Department of Biotechnology, School of Biological Sciences, Federal University of Technology Owerri Nigeria, Owerri, Nigeria
| | - Kanate Thitiananpakorn
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Xin-Ee Tan
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Yoshifumi Aiba
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Remi Takenouchi
- School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Kotaro Kiga
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Teppei Sasahara
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Kazuhiko Miyanaga
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Srivani Veeranarayanan
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Yuzuki Shimamori
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Adeline Yeo Syin Lian
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Thuy Minh Nguyen
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Huong Minh Nguyen
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Ola Alessa
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | | | - Sarangi Jayathilake
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | | | - Priyanka Baranwal
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Yutaro Nishikawa
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Feng-Yu Li
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Tomofumi Kawaguchi
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Sowmiya Sankaranarayanan
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Mahmoud Arbaah
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Yuancheng Zhang
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Maniruzzaman
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Yi Liu
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Hossain Sarah
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Junjie Li
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Takashi Sugano
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Thi My Duyen Ho
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Anujin Batbold
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Tergel Nayanjin
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Longzhu Cui
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
9
|
Lei MG, Jorgenson MA, Robbs EJ, Black IM, Archer-Hartmann S, Shalygin S, Azadi P, Lee CY. Characterization of Ssc, an N-acetylgalactosamine-containing Staphylococcus aureus surface polysaccharide. J Bacteriol 2024; 206:e0004824. [PMID: 38712944 PMCID: PMC11112989 DOI: 10.1128/jb.00048-24] [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: 02/11/2024] [Accepted: 04/08/2024] [Indexed: 05/08/2024] Open
Abstract
Whole genome sequencing has revealed that the genome of Staphylococcus aureus possesses an uncharacterized 5-gene operon (SAOUHSC_00088-00092 in strain 8325 genome) that encodes factors with functions related to polysaccharide biosynthesis and export, indicating the existence of a new extracellular polysaccharide species. We designate this locus as ssc for staphylococcal surface carbohydrate. We found that the ssc genes were weakly expressed and highly repressed by the global regulator MgrA. To characterize Ssc, Ssc was heterologously expressed in Escherichia coli and extracted by heat treatment. Ssc was also conjugated to AcrA from Campylobacter jejuni in E. coli using protein glycan coupling technology (PGCT). Analysis of the heat-extracted Ssc and the purified Ssc-AcrA glycoconjugate by tandem mass spectrometry revealed that Ssc is likely a polymer consisting of N-acetylgalactosamine. We further demonstrated that the expression of the ssc genes in S. aureus affected phage adsorption and susceptibility, suggesting that Ssc is surface-exposed. IMPORTANCE Surface polysaccharides play crucial roles in the biology and virulence of bacterial pathogens. Staphylococcus aureus produces four major types of polysaccharides that have been well-characterized. In this study, we identified a new surface polysaccharide containing N-acetylgalactosamine (GalNAc). This marks the first report of GalNAc-containing polysaccharide in S. aureus. Our discovery lays the groundwork for further investigations into the chemical structure, surface location, and role in pathogenesis of this new polysaccharide.
Collapse
Affiliation(s)
- Mei G. Lei
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Matthew A. Jorgenson
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Emily J. Robbs
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Ian M. Black
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | | | - Sergei Shalygin
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Chia Y. Lee
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
10
|
Zeden MS. mSphere of Influence: Targeting bacterial signaling and metabolism to overcome antimicrobial resistance. mSphere 2024; 9:e0063223. [PMID: 38305167 PMCID: PMC10900877 DOI: 10.1128/msphere.00632-23] [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] [Indexed: 02/03/2024] Open
Abstract
Dr Merve Suzan Zeden works in the field of molecular bacteriology and antibiotic resistance. In this mSphere of Influence article, she reflects on how three papers, entitled "c-di-AMP modulates Listeria monocytogenes central metabolism to regulate growth, antibiotic resistance and osmoregulation," "Amino acid catabolism in Staphylococcus aureus and the function of carbon catabolite repression," and "Evolving MRSA: high-level β-lactam resistance in Staphylococcus aureus is associated with RNA polymerase alterations and fine tuning of gene expression," made an impact on her work on bacterial metabolism and antimicrobial resistance and how it shaped her research in understanding the link in between.
Collapse
Affiliation(s)
- Merve S Zeden
- School of Biological and Chemical Sciences, College of Science and Engineering, University of Galway, Galway, Ireland
| |
Collapse
|
11
|
Fernández-Barat L, López-Aladid R, Vázquez N, Cabrera R, Vila J, Ferrer M, Torres A. Bacterial Adaptive Memory in Methicillin-Resistant Staphylococcus aureus from Endotracheal Tubes. Pathogens 2024; 13:144. [PMID: 38392882 PMCID: PMC10892081 DOI: 10.3390/pathogens13020144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
OBJECTIVES To evaluate the expression dynamics of biofilm genes in methicillin-resistant Staphylococcus aureus (MRSA) retrieved from endotracheal tubes (ETT) and to determine how gene regulation is attenuated in vitro where host-environmental factors are no longer present. METHODS Biofilm was grown (24 h) in tryptic broth soy plus 0.25% glucose for a clinical MRSA isolate in planktonic state and after sessile growth named ETT-MRSA (S2, S3, S4, S5, S6, S7). Gene expression of five biofilm-related genes (icaC, clfB, ebps, fnbB, and RNA III) was assessed consecutively from day 1 to day 4 after ETT growth through real-time PCR. 16S rRNA was used as a control. RESULTS The MRSA isolates retrieved from ETT were capable of producing biofilms dependent on ica. The gene expression dynamics of ETT-MRSA changed progressively compared to planktonic MRSA gene expression under both ambient air (p < 0.001) and ambient air with 5% CO2 (p < 0.001). Dynamic assessment of icaC expression in both atmospheric conditions showed progressive downregulation in vitro compared to in vivo ETT biofilms. The expression patterns of clfB and ebps genes were similar to icaC. In contrast, the expression of the RNA III gene showed progressive upregulation from day 1 to day 4 (p < 0.001). CONCLUSIONS MRSA loses its biofilm gene expression in vitro, by adaptive features across multiple generations, as evidenced by the progressive downregulation of icaC and upregulation of RNA III. These findings underscore the significance of host-environment dependence in regulating bacterial biofilm genes, highlighting its importance in diagnostics. Bacterial strains lose their host-specific characteristics as they are cultured in vitro.
Collapse
Affiliation(s)
- Laia Fernández-Barat
- Centro de Investigación Biomedica En Red-Enfermedades Respiratorias (CibeRes, CB06/06/0028) and Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.L.-A.); (N.V.); (R.C.); (M.F.)
- University of Barcelona, 08193 Barcelona, Spain;
| | - Ruben López-Aladid
- Centro de Investigación Biomedica En Red-Enfermedades Respiratorias (CibeRes, CB06/06/0028) and Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.L.-A.); (N.V.); (R.C.); (M.F.)
- University of Barcelona, 08193 Barcelona, Spain;
| | - Nil Vázquez
- Centro de Investigación Biomedica En Red-Enfermedades Respiratorias (CibeRes, CB06/06/0028) and Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.L.-A.); (N.V.); (R.C.); (M.F.)
- University of Barcelona, 08193 Barcelona, Spain;
| | - Roberto Cabrera
- Centro de Investigación Biomedica En Red-Enfermedades Respiratorias (CibeRes, CB06/06/0028) and Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.L.-A.); (N.V.); (R.C.); (M.F.)
- University of Barcelona, 08193 Barcelona, Spain;
| | - Jordi Vila
- University of Barcelona, 08193 Barcelona, Spain;
- Microbiology Service at Hospital Clinic and Institute of Global Health (ISGlobal), 08036 Barcelona, Spain
| | - Miquel Ferrer
- Centro de Investigación Biomedica En Red-Enfermedades Respiratorias (CibeRes, CB06/06/0028) and Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.L.-A.); (N.V.); (R.C.); (M.F.)
- University of Barcelona, 08193 Barcelona, Spain;
- Pulmonary and Critical Care Unit, Respiratory Institute, Hospital Clinic, 08036 Barcelona, Spain
| | - Antoni Torres
- Centro de Investigación Biomedica En Red-Enfermedades Respiratorias (CibeRes, CB06/06/0028) and Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (R.L.-A.); (N.V.); (R.C.); (M.F.)
- University of Barcelona, 08193 Barcelona, Spain;
- Pulmonary and Critical Care Unit, Respiratory Institute, Hospital Clinic, 08036 Barcelona, Spain
| |
Collapse
|
12
|
Wright MJ, Bai G. Bacterial second messenger cyclic di-AMP in streptococci. Mol Microbiol 2023; 120:791-804. [PMID: 37898560 DOI: 10.1111/mmi.15187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/10/2023] [Accepted: 10/15/2023] [Indexed: 10/30/2023]
Abstract
Cyclic dimeric adenosine monophosphate (c-di-AMP) has been well studied in bacteria, including those of the genus Streptococcus, since the first recognition of this dinucleotide in 2008. Streptococci possess a sole diadenylate cyclase, CdaA, and distinct c-di-AMP phosphodiesterases. Interestingly, cdaA is required for viability of some streptococcal species but not all when streptococci are grown in standard laboratory media. Bacteria of this genus also have distinct c-di-AMP effector proteins, diverse c-di-AMP-signaling pathways, and subsequent biological outcomes. In streptococci, c-di-AMP may influence bacterial growth, morphology, biofilm formation, competence program, drug resistance, and bacterial pathogenesis. c-di-AMP secreted by streptococci has also been shown to interact with the mammalian host and induces immune responses including type I interferon production. In this review, we summarize the reported c-di-AMP networks in seven species of the genus Streptococcus, which cause diverse clinical manifestations, and propose future perspectives to investigate the signaling molecule in these streptococcal pathogens.
Collapse
Affiliation(s)
- Michael J Wright
- Department of Internal Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Guangchun Bai
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| |
Collapse
|
13
|
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
|
14
|
Francis D, Veeramanickathadathil Hari G, Koonthanmala Subash A, Bhairaddy A, Joy A. The biofilm proteome of Staphylococcus aureus and its implications for therapeutic interventions to biofilm-associated infections. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 138:327-400. [PMID: 38220430 DOI: 10.1016/bs.apcsb.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Staphylococcus aureus is a major healthcare concern due to its ability to inflict life-threatening infections and evolve antibiotic resistance at an alarming pace. It is frequently associated with hospital-acquired infections, especially device-associated infections. Systemic infections due to S. aureus are difficult to treat and are associated with significant mortality and morbidity. The situation is worsened by the ability of S. aureus to form social associations called biofilms. Biofilms embed a community of cells with the ability to communicate with each other and share resources within a polysaccharide or protein matrix. S. aureus establish biofilms on tissues and conditioned abiotic surfaces. Biofilms are hyper-tolerant to antibiotics and help evade host immune responses. Biofilms exacerbate the severity and recalcitrance of device-associated infections. The development of a biofilm involves various biomolecules, such as polysaccharides, proteins and nucleic acids, contributing to different structural and functional roles. Interconnected signaling pathways and regulatory molecules modulate the expression of these molecules. A comprehensive understanding of the molecular biology of biofilm development would help to devise effective anti-biofilm therapeutics. Although bactericidal agents, antimicrobial peptides, bacteriophages and nano-conjugated anti-biofilm agents have been employed with varying levels of success, there is still a requirement for effective and clinically viable anti-biofilm therapeutics. Proteins that are expressed and utilized during biofilm formation, constituting the biofilm proteome, are a particularly attractive target for anti-biofilm strategies. The proteome can be explored to identify potential anti-biofilm drug targets and utilized for rational drug discovery. With the aim of uncovering the biofilm proteome, this chapter explores the mechanism of biofilm formation and its regulation. Furthermore, it explores the antibiofilm therapeutics targeted against the biofilm proteome.
Collapse
Affiliation(s)
- Dileep Francis
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India.
| | | | | | - Anusha Bhairaddy
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India
| | - Atheene Joy
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India
| |
Collapse
|
15
|
Patel H, Rawat S. A genetic regulatory see-saw of biofilm and virulence in MRSA pathogenesis. Front Microbiol 2023; 14:1204428. [PMID: 37434702 PMCID: PMC10332168 DOI: 10.3389/fmicb.2023.1204428] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/30/2023] [Indexed: 07/13/2023] Open
Abstract
Staphylococcus aureus is one of the most common opportunistic human pathogens causing several infectious diseases. Ever since the emergence of the first methicillin-resistant Staphylococcus aureus (MRSA) strain decades back, the organism has been a major cause of hospital-acquired infections (HA-MRSA). The spread of this pathogen across the community led to the emergence of a more virulent subtype of the strain, i.e., Community acquired Methicillin resistant Staphylococcus aureus (CA-MRSA). Hence, WHO has declared Staphylococcus aureus as a high-priority pathogen. MRSA pathogenesis is remarkable because of the ability of this "superbug" to form robust biofilm both in vivo and in vitro by the formation of polysaccharide intercellular adhesin (PIA), extracellular DNA (eDNA), wall teichoic acids (WTAs), and capsule (CP), which are major components that impart stability to a biofilm. On the other hand, secretion of a diverse array of virulence factors such as hemolysins, leukotoxins, enterotoxins, and Protein A regulated by agr and sae two-component systems (TCS) aids in combating host immune response. The up- and downregulation of adhesion genes involved in biofilm formation and genes responsible for synthesizing virulence factors during different stages of infection act as a genetic regulatory see-saw in the pathogenesis of MRSA. This review provides insight into the evolution and pathogenesis of MRSA infections with a focus on genetic regulation of biofilm formation and virulence factors secretion.
Collapse
Affiliation(s)
| | - Seema Rawat
- Microbiology Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat, India
| |
Collapse
|
16
|
Papadopoulou V, Sidders AE, Lu KY, Velez AZ, Durham PG, Bui DT, Angeles-Solano M, Dayton PA, Rowe SE. Overcoming biological barriers to improve treatment of a Staphylococcus aureus wound infection. Cell Chem Biol 2023; 30:513-526.e5. [PMID: 37148883 PMCID: PMC10198964 DOI: 10.1016/j.chembiol.2023.04.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/19/2023] [Accepted: 04/17/2023] [Indexed: 05/08/2023]
Abstract
Chronic wounds frequently become infected with bacterial biofilms which respond poorly to antibiotic therapy. Aminoglycoside antibiotics are ineffective at treating deep-seated wound infections due to poor drug penetration, poor drug uptake into persister cells, and widespread antibiotic resistance. In this study, we combat the two major barriers to successful aminoglycoside treatment against a biofilm-infected wound: limited antibiotic uptake and limited biofilm penetration. To combat the limited antibiotic uptake, we employ palmitoleic acid, a host-produced monounsaturated fatty acid that perturbs the membrane of gram-positive pathogens and induces gentamicin uptake. This novel drug combination overcomes gentamicin tolerance and resistance in multiple gram-positive wound pathogens. To combat biofilm penetration, we examined the ability of sonobactericide, a non-invasive ultrasound-mediated-drug delivery technology to improve antibiotic efficacy using an in vivo biofilm model. This dual approach dramatically improved antibiotic efficacy against a methicillin-resistant Staphylococcus aureus (MRSA) wound infection in diabetic mice.
Collapse
Affiliation(s)
- Virginie Papadopoulou
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA.
| | - Ashelyn E Sidders
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kuan-Yi Lu
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Amanda Z Velez
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Phillip G Durham
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA; Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Duyen T Bui
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Michelle Angeles-Solano
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Paul A Dayton
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA; Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Sarah E Rowe
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
17
|
Zhao H, Wu X, Wang B, Shen L, Rao L, Wang X, Zhang J, Xiao Y, Xu Y, Yu J, Guo Y, Zhou Y, Wan B, Wu C, Chen L, Yu F. Phenotypic and genomic analysis of the hypervirulent ST22 methicillin-resistant Staphylococcus aureus in China. mSystems 2023:e0124222. [PMID: 37184650 DOI: 10.1128/msystems.01242-22] [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] [Indexed: 05/16/2023] Open
Abstract
ST22 MRSA (methicillin-resistant Staphylococcus aureus) strains are only sporadically reported in China. Through the phylogenetic reconstruction of 30 ST22 strains from China and 480 ST22 strains from global sources, we found that the global ST22 strains can be divided into three clades (I, II, and III). The China ST22 strains were found primarily in clade II (IIb and IIc) and also in clade III, indicating that the China ST22-MRSA clones have different origins. The China subclade IIb strains (SCCmec Vb-t309) may evolve from the native ST22 MSSA clone, while the China IIc strains may have spread from other countries. Subclade IIc (SCCmecIVa-t309) strains exhibited particularly strong lethality and invasiveness in Galleria mellonella infection and mouse skin abscess models in comparison to USA300 and other dominant China HA-MRSA (ST5 and ST239) or CA-MRSA (ST59) strains. This study described the emergence of a highly virulent ST22 MRSA subclade and improved our insight into the molecular epidemiology of ST22 strains in China.
Collapse
Affiliation(s)
- Huilin Zhao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaocui Wu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bingjie Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Li Shen
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lulin Rao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xinyi Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiao Zhang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yanghua Xiao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yanlei Xu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jingyi Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yinjuan Guo
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ying Zhou
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Baoshan Wan
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chunyang Wu
- First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liang Chen
- Hackensack Meridian Health Center for Discovery and Innovation, Nutley, New Jersey, USA
- Hackensack Meridian School of Medicine, Nutley, New Jersey, USA
| | - Fangyou Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
18
|
Sarquis A, Bajrami D, Mizaikoff B, Ladero V, Alvarez MA, Fernandez M. Characterization of the Biofilms Formed by Histamine-Producing Lentilactobacillus parabuchneri Strains in the Dairy Environment. Foods 2023; 12:foods12071503. [PMID: 37048324 PMCID: PMC10093819 DOI: 10.3390/foods12071503] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/29/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
Lentilactobacillus parabuchneri, a lactic acid bacterium, is largely responsible for the production and accumulation of histamine, a toxic biogenic amine, in cheese. L. parabuchneri strains can form biofilms on the surface of industry equipment. Since they are resistant to cleaning and disinfection, they may act as reservoirs of histamine-producing contaminants in cheese. The aim of this study was to investigate the biofilm-producing capacity of L. parabuchneri strains. Using the crystal violet technique, the strains were first categorized as weak, moderate or strong biofilm producers. Analysis of their biofilm matrices revealed them to be mainly composed of proteins. Two strains of each category were then selected to analyze the influence on the biofilm-forming capacity of temperature, pH, carbon source, NaCl concentration and surface material (i.e., focusing on those used in the dairy industry). In general, low temperature (8 °C), high NaCl concentrations (2–3% w/v) and neutral pH (pH 6) prevented biofilm formation. All strains were found to adhere easily to beech wood. These findings increase knowledge of the biofilm-forming capacity of histamine-producing L. parabuchneri strains and how their formation may be prevented for improving food safety.
Collapse
Affiliation(s)
- Agustina Sarquis
- Departmento de Tecnología y Biotecnología de Productos Lácteos, Instituto de Productos Lácteos de Asturias, IPLA, CSIC, 33300 Villaviciosa, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Diellza Bajrami
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Albert Einstein-Allee 11, 89081 Ulm, Germany
| | - Boris Mizaikoff
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Albert Einstein-Allee 11, 89081 Ulm, Germany
- Hahn-Schickard, Institute for Microanalysis Systems, Sedanstrasse 14, 89077 Ulm, Germany
| | - Victor Ladero
- Departmento de Tecnología y Biotecnología de Productos Lácteos, Instituto de Productos Lácteos de Asturias, IPLA, CSIC, 33300 Villaviciosa, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Miguel A. Alvarez
- Departmento de Tecnología y Biotecnología de Productos Lácteos, Instituto de Productos Lácteos de Asturias, IPLA, CSIC, 33300 Villaviciosa, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Maria Fernandez
- Departmento de Tecnología y Biotecnología de Productos Lácteos, Instituto de Productos Lácteos de Asturias, IPLA, CSIC, 33300 Villaviciosa, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
19
|
Ali Alghamdi B, Al-Johani I, Al-Shamrani JM, Musamed Alshamrani H, Al-Otaibi BG, Almazmomi K, Yusnoraini Yusof N. Antimicrobial resistance in methicillin-resistant staphylococcus aureus. Saudi J Biol Sci 2023; 30:103604. [PMID: 36936699 PMCID: PMC10018568 DOI: 10.1016/j.sjbs.2023.103604] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/02/2023] [Accepted: 02/19/2023] [Indexed: 03/02/2023] Open
Abstract
In the medical community, antibiotics are revered as a miracle because they stop diseases brought on by pathogenic bacteria. Antibiotics have become the cornerstone of contemporary medical advancements ever since penicillin was discovered. Antibiotic resistance developed among germs quickly, placing a strain in the medical field. Methicillin-resistant Staphylococcus aureus (MRSA), Since 1961, has emerged as the major general antimicrobial resistant bacteria (AMR) worldwide. MRSA can easily transmit across the hospital system and has mostly gained resistance to medications called beta-lactamases. This enzyme destroys the cell wall of beta-lactam antibiotics resulting in resistance against that respective antibiotic. Daptomycin, linezolid and vancomycin were previously used to treat MRSA infections. However, due to mutations and Single nucleotide polymorphisms (SNPs) in Open reading frames (ORFs) and SCCmec machinery of respective antibody, MRSA developed resistance against those antibiotics. The MRSA strains (USA300, CC398, CC130 etc.), when their pan-genomes were analyzed were found the genes involved in invoking resistance against the antibiotics as well as the epidemiology of that respective strain. PENC (penicillin plus potassium clavulanate) is the new antibiotic showing potential in treatment of MRSA though it is itself resistant against penicillin alone. In this review, our main focus is on mechanism of development of AMR in MRSA, how different ORFs are involved in evoking resistance in MRSA and what is the core-genome of different antimicrobial resistant MRSA.
Collapse
Affiliation(s)
- Bandar Ali Alghamdi
- Department of Cardiac Surgery, King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia
| | - Intisar Al-Johani
- Department of Biotechnology, Taif University, Taif City, Saudi Arabia
| | | | - Hussein Musamed Alshamrani
- Directorate of Health Affairs in Qunfudah Center (Namerah Primary Health care) Pharmacy Department, Saudi Arabia
| | | | - Kholod Almazmomi
- Department of Biotechnology, Taif University, Taif City, Saudi Arabia
| | - Nik Yusnoraini Yusof
- Institute for Research in Molecular Medicine (INFORMM), Health Campus, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
- Corresponding author at.: Institute for Research in Molecular Medicine (INFORMM) Universiti Sains Malaysia Kubang Kerian, Kelantan 16150, Malaysia.
| |
Collapse
|
20
|
Nolan AC, Zeden MS, Kviatkovski I, Campbell C, Urwin L, Corrigan RM, Gründling A, O’Gara JP. Purine Nucleosides Interfere with c-di-AMP Levels and Act as Adjuvants To Re-Sensitize MRSA To β-Lactam Antibiotics. mBio 2023; 14:e0247822. [PMID: 36507833 PMCID: PMC9973305 DOI: 10.1128/mbio.02478-22] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/17/2022] [Indexed: 12/14/2022] Open
Abstract
The purine-derived signaling molecules c-di-AMP and (p)ppGpp control mecA/PBP2a-mediated β-lactam resistance in methicillin-resistant Staphylococcus aureus (MRSA) raise the possibility that purine availability can control antibiotic susceptibility. Consistent with this, exogenous guanosine and xanthosine, which are fluxed through the GTP branch of purine biosynthesis, were shown to significantly reduce MRSA β-lactam resistance. In contrast, adenosine (fluxed to ATP) significantly increased oxacillin resistance, whereas inosine (which can be fluxed to ATP and GTP via hypoxanthine) only marginally increased oxacillin susceptibility. Furthermore, mutations that interfere with de novo purine synthesis (pur operon), transport (NupG, PbuG, PbuX) and the salvage pathway (DeoD2, Hpt) increased β-lactam resistance in MRSA strain JE2. Increased resistance of a nupG mutant was not significantly reversed by guanosine, indicating that NupG is required for guanosine transport, which is required to reduce β-lactam resistance. Suppressor mutants resistant to oxacillin/guanosine combinations contained several purine salvage pathway mutations, including nupG and hpt. Guanosine significantly increased cell size and reduced levels of c-di-AMP, while inactivation of GdpP, the c-di-AMP phosphodiesterase negated the impact of guanosine on β-lactam susceptibility. PBP2a expression was unaffected in nupG or deoD2 mutants, suggesting that guanosine-induced β-lactam susceptibility may result from dysfunctional c-di-AMP-dependent osmoregulation. These data reveal the therapeutic potential of purine nucleosides, as β-lactam adjuvants that interfere with the normal activation of c-di-AMP are required for high-level β-lactam resistance in MRSA. IMPORTANCE The clinical burden of infections caused by antimicrobial resistant (AMR) pathogens is a leading threat to public health. Maintaining the effectiveness of existing antimicrobial drugs or finding ways to reintroduce drugs to which resistance is widespread is an important part of efforts to address the AMR crisis. Predominantly, the safest and most effective class of antibiotics are the β-lactams, which are no longer effective against methicillin-resistant Staphylococcus aureus (MRSA). Here, we report that the purine nucleosides guanosine and xanthosine have potent activity as adjuvants that can resensitize MRSA to oxacillin and other β-lactam antibiotics. Mechanistically, exposure of MRSA to these nucleosides significantly reduced the levels of the cyclic dinucleotide c-di-AMP, which is required for β-lactam resistance. Drugs derived from nucleotides are widely used in the treatment of cancer and viral infections highlighting the clinical potential of using purine nucleosides to restore or enhance the therapeutic effectiveness of β-lactams against MRSA and potentially other AMR pathogens.
Collapse
Affiliation(s)
- Aaron C. Nolan
- Microbiology, School of Biological and Chemical Sciences, University of Galway, Ireland
| | - Merve S. Zeden
- Microbiology, School of Biological and Chemical Sciences, University of Galway, Ireland
| | - Igor Kviatkovski
- Section of Molecular Microbiology and Medical Research Council Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Christopher Campbell
- Microbiology, School of Biological and Chemical Sciences, University of Galway, Ireland
| | - Lucy Urwin
- The Florey Institute, School of Bioscience, University of Sheffield, Sheffield, United Kingdom
| | - Rebecca M. Corrigan
- The Florey Institute, School of Bioscience, University of Sheffield, Sheffield, United Kingdom
| | - Angelika Gründling
- Section of Molecular Microbiology and Medical Research Council Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - James P. O’Gara
- Microbiology, School of Biological and Chemical Sciences, University of Galway, Ireland
| |
Collapse
|
21
|
Sorieul C, Dolce M, Romano MR, Codée J, Adamo R. Glycoconjugate vaccines against antimicrobial resistant pathogens. Expert Rev Vaccines 2023; 22:1055-1078. [PMID: 37902243 DOI: 10.1080/14760584.2023.2274955] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/20/2023] [Indexed: 10/31/2023]
Abstract
INTRODUCTION Antimicrobial resistance (AMR) is responsible for the death of millions worldwide and stands as a major threat to our healthcare systems, which are heavily reliant on antibiotics to fight bacterial infections. The development of vaccines against the main pathogens involved is urgently required as prevention remains essential against the rise of AMR. AREAS COVERED A systematic research review was conducted on MEDLINE database focusing on the six AMR pathogens defined as ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Escherichia coli), which are considered critical or high priority pathogens by the World Health Organization (WHO) and the Centers for Disease Control and Prevention (CDC). The analysis was intersecated with the terms carbohydrate, glycoconjugate, bioconjugate, glyconanoparticle, and multiple presenting antigen system vaccines. EXPERT OPINION Glycoconjugate vaccines have been successful in preventing meningitis and pneumoniae, and there are high expectations that they will play a key role in fighting AMR. We herein discuss the recent technological, preclinical, and clinical advances, as well as the challenges associated with the development of carbohydrate-based vaccines against leading AMR bacteria, with focus on the ESKAPE pathogens. The need of innovative clinical and regulatory approaches to tackle these targets is also highlighted.
Collapse
Affiliation(s)
- Charlotte Sorieul
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Marta Dolce
- GSK, Via Fiorentina 1, Siena, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | | | - Jeroen Codée
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | | |
Collapse
|
22
|
He X, Zhang W, Cao Q, Li Y, Bao G, Lin T, Bao J, Chang C, Yang C, Yin Y, Xu J, Ren Z, Jin Y, Lu F. Global Downregulation of Penicillin Resistance and Biofilm Formation by MRSA Is Associated with the Interaction between Kaempferol Rhamnosides and Quercetin. Microbiol Spectr 2022; 10:e0278222. [PMID: 36354319 PMCID: PMC9769653 DOI: 10.1128/spectrum.02782-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 10/24/2022] [Indexed: 11/12/2022] Open
Abstract
The rapid development of methicillin-resistant Staphylococcus aureus (MRSA) drug resistance and the formation of biofilms seriously challenge the clinical application of classic antibiotics. Extracts of the traditional herb Chenopodium ambrosioides L. were found to have strong antibiofilm activity against MRSA, but their mechanism of action remains poorly understood. This study was designed to investigate the antibacterial and antibiofilm activities against MRSA of flavonoids identified from C. ambrosioides L. in combination with classic antibiotics, including ceftazidime, erythromycin, levofloxacin, penicillin G, and vancomycin. Liquid chromatography-mass spectrometry (LC-MS) was used to analyze the nonvolatile chemical compositions. Reverse transcription (RT)-PCR was used to investigate potential multitargets of flavonoids based on global transcriptional responses of virulence and antibiotic resistance. A synergistic antibacterial and biofilm-inhibiting activity of the alcoholic extract of the ear of C. ambrosioides L. in combination with penicillin G was observed against MRSA, which proved to be closely related to the interaction of the main components of kaempferol rhamnosides with quercetin. In regard to the mechanism, the increased sensitivity of MRSA to penicillin G was shown to be related to the downregulation of penicillinase with SarA as a potential drug target, while the antibiofilm activity was mainly related to downregulation of various virulence factors involved in the initial and mature stages of biofilm development, with SarA and/or σB as drug targets. This study provides a theoretical basis for further exploration of the medicinal activity of kaempferol rhamnosides and quercetin and their application in combination with penicillin G against MRSA biofilm infection. IMPORTANCE In this study, the synergistic antibacterial and antibiofilm effects of the traditional herb C. ambrosioides L. and the classic antibiotic penicillin G on MRSA provide a potential strategy to deal with the rapid development of MRSA antibiotic resistance. This study also provides a theoretical basis for further optimizing the combined effect of kaempferol rhamnosides, quercetin, and penicillin G and exploring anti-MRSA biofilm infection research with SarA and σB as drug targets.
Collapse
Affiliation(s)
- Xinlong He
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Wenwen Zhang
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Qingchao Cao
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, People’s Republic of China
| | - Yinyue Li
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Guangyu Bao
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| | - Tao Lin
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| | - Jiaojiao Bao
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| | - Caiwang Chang
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| | - Changshui Yang
- Department of Pharmacy, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Yi Yin
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Jiahui Xu
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Zhenyu Ren
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Yingshan Jin
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, People’s Republic of China
| | - Feng Lu
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| |
Collapse
|
23
|
Antimicrobial and Antibiofilm Effects of Combinatorial Treatment Formulations of Anti-Inflammatory Drugs-Common Antibiotics against Pathogenic Bacteria. Pharmaceutics 2022; 15:pharmaceutics15010004. [PMID: 36678634 PMCID: PMC9864814 DOI: 10.3390/pharmaceutics15010004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
With the spread of multi-drug-resistant (MDR) bacteria and the lack of effective antibiotics to treat them, developing new therapeutic methods and strategies is essential. In this study, we evaluated the antibacterial and antibiofilm activity of different formulations composed of ibuprofen (IBP), acetylsalicylic acid (ASA), and dexamethasone sodium phosphate (DXP) in combination with ciprofloxacin (CIP), gentamicin (GEN), cefepime (FEP), imipenem (IPM), and meropenem (MEM) on clinical isolates of Staphylococcus aureus (S. aureus) and Pseudomonas aeruginosa (P. aeruginosa) as well as the transcription levels of biofilm-associated genes in the presence of sub-MICs of IBP, ASA, and DXP. The minimal inhibitory concentrations (MICs), minimal biofilm inhibitory concentrations (MBICs), and minimum biofilm eradication concentrations (MBECs) of CIP, GEN, FEP, IPM, and MEM with/without sub-MICs of IBP (200 µg/mL), ASA (200 µg/mL), and DXP (500 µg/mL) for the clinical isolates were determined by the microbroth dilution method. Quantitative real-time-PCR (qPCR) was used to determine the expression levels of biofilm-related genes, including icaA in S. aureus and algD in P. aeruginosa at sub-MICs of IBP, ASA, and DXP. All S. aureus isolates were methicillin-resistant S. aureus (MRSA), and all P. aeruginosa were resistant to carbapenems. IBP decreased the levels of MIC, MBIC, and MBEC for all antibiotic agents in both clinical isolates, except for FEP among P. aeruginosa isolates. In MRSA isolates, ASA decreased the MICs of GEN, FEP, and IPM and the MBICs of IPM and MEM. In P. aeruginosa, ASA decreased the MICs of FEP, IPM, and MEM, the MBICs of FEP and MEM, and the MBEC of FEP. DXP increased the MICs of CIP, GEN, and FEP, and the MBICs of CIP, GEN, and FEP among both clinical isolates. The MBECs of CIP and FEP for MRSA isolates and the MBECs of CIP, GEN, and MEM among P. aeruginosa isolates increased in the presence of DXP. IBP and ASA at 200 µg/mL significantly decreased the transcription level of algD in P. aeruginosa, and IBP significantly decreased the transcription level of icaA in S. aureus. DXP at 500 µg/mL significantly increased the expression levels of algD and icaA genes in S. aureus and P. aeruginosa isolates, respectively. Our findings showed that the formulations containing ASA and IBP have significant effects on decreasing the MIC, MBIC, and MBEC levels of some antibiotics and can down-regulate the expression of biofilm-related genes such as icaA and algD. Therefore, NSAIDs represent appropriate candidates for the design of new antibacterial and antibiofilm therapeutic formulations.
Collapse
|
24
|
Wang S, Huang X, Yang J, Yang D, Zhang Y, Hou Y, Lin L, Hua L, Liang W, Wu B, Peng Z. Biocontrol of methicillin-resistant Staphylococcus aureus using a virulent bacteriophage derived from a temperate one. Microbiol Res 2022; 267:127258. [PMID: 36434990 DOI: 10.1016/j.micres.2022.127258] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/30/2022] [Accepted: 11/10/2022] [Indexed: 11/18/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) poses a serious threat to global public health due to its resistance to specific antibiotics. Bacteriophages particularly the lytic ones are promoted as a potential powerful-tool to combat infections caused by drug resistant bacteria; while several disadvantages limited the application of the temperate ones. In this study, we isolated 14 phages against MRSA strains, and found three ones showed the capacity of killing most of the target MRSA strains. However, whole genome sequencing and generation of lysogens indicated that these three bacteriophage candidates were temperate ones. Therefore, we mutated one (4PHSA25) of them to a virulent bacteriophage (4PHCISA25). Phenotypical characterization assays revealed that 4PHCISA25 had similar lytic spectrum, temperature, pH, and UV sensitivities to 4PHSA25. However, 4PHCISA25 displayed increased lytic activities and decreased bacteriophage insensitive mutant frequency. Biofilm removing assays showed that 4PHCISA25 exhibited a better capacity than 4PHSA25 on eliminating biofilms formed by MRSA strains. Mouse experiments demonstrated that injection of 4PHCISA25 was safe to the mice and treatment with it (109 PFU per mouse) inhibited the development of abscess induced by MRSA within 24 h and promoted the recovery from the clinical signs. Taken together, this study highlights the use of phages combating MRSA.
Collapse
Affiliation(s)
- Shuang Wang
- State Key Laboratory of Agricultural Microbiology, The Cooperative Innovation Centre for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xi Huang
- State Key Laboratory of Agricultural Microbiology, The Cooperative Innovation Centre for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jie Yang
- State Key Laboratory of Agricultural Microbiology, The Cooperative Innovation Centre for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Dan Yang
- State Key Laboratory of Agricultural Microbiology, The Cooperative Innovation Centre for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yue Zhang
- State Key Laboratory of Agricultural Microbiology, The Cooperative Innovation Centre for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yanyan Hou
- State Key Laboratory of Agricultural Microbiology, The Cooperative Innovation Centre for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Lin Lin
- State Key Laboratory of Agricultural Microbiology, The Cooperative Innovation Centre for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Lin Hua
- State Key Laboratory of Agricultural Microbiology, The Cooperative Innovation Centre for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Wan Liang
- State Key Laboratory of Agricultural Microbiology, The Cooperative Innovation Centre for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Bin Wu
- State Key Laboratory of Agricultural Microbiology, The Cooperative Innovation Centre for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.
| | - Zhong Peng
- State Key Laboratory of Agricultural Microbiology, The Cooperative Innovation Centre for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China; Hubei Hongshan Laboratory, Wuhan, China.
| |
Collapse
|
25
|
Antibiofilm Combinatory Strategy: Moxifloxacin-Loaded Nanosystems and Encapsulated N-Acetyl-L-Cysteine. Pharmaceutics 2022; 14:pharmaceutics14112294. [PMID: 36365113 PMCID: PMC9699636 DOI: 10.3390/pharmaceutics14112294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/20/2022] [Accepted: 10/22/2022] [Indexed: 11/29/2022] Open
Abstract
Bacterial biofilms of Staphylococcus aureus, formed on implants, have a massive impact on the increasing number of antimicrobial resistance cases. The current treatment for biofilm-associated infections is based on the administration of antibiotics, failing to target the biofilm matrix. This work is focused on the development of multiple lipid nanoparticles (MLNs) encapsulating the antibiotic moxifloxacin (MOX). The nanoparticles were functionalized with d-amino acids to target the biofilm matrix. The produced formulations exhibited a mean hydrodynamic diameter below 300 nm, a low polydispersity index, and high encapsulation efficiency. The nanoparticles exhibited low cytotoxicity towards fibroblasts and low hemolytic activity. To target bacterial cells and the biofilm matrix, MOX-loaded MLNs were combined with a nanosystem encapsulating a matrix-disruptive agent: N-acetyl-L-cysteine (NAC). The nanosystems alone showed a significant reduction of both S. aureus biofilm viability and biomass, using the microtiter plate biofilm model. Further, biofilms grown inside polyurethane catheters were used to assess the effect of combining MOX-loaded and NAC-loaded nanosystems on biofilm viability. An increased antibiofilm efficacy was observed when combining the functionalized MOX-loaded MLNs and NAC-loaded nanosystems. Thus, nanosystems as carriers of bactericidal and matrix-disruptive agents are a promising combinatory strategy towards the eradication of S. aureus biofilms.
Collapse
|
26
|
Comparative Study of Antibacterial, Antibiofilm, Antiswarming and Antiquorum Sensing Activities of Origanum vulgare Essential Oil and Terpinene-4-ol against Pathogenic Bacteria. Life (Basel) 2022; 12:life12101616. [PMID: 36295051 PMCID: PMC9605346 DOI: 10.3390/life12101616] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/01/2022] [Accepted: 10/08/2022] [Indexed: 11/17/2022] Open
Abstract
Essential oils from aromatic and medicinal plants have many bioactive compounds known for their important biological activities mainly their antibacterial effects. Here we evaluated qualitatively and quantitatively the biofilm formation capability of pathogenic bacterial strains (n = 8). Then, we investigated the antibacterial, antibiofilm, antiquorum-sensing, and antiswarming efficacy of Origanum vulgare essential oil (EO) and terpinene-4-ol. Our results revealed that EO exhibited a more potent inhibitory effect against the tested strains. While the terpinene-4-ol was found to be more effective against developed Staphylococcus aureus biofilm. Regarding the anti quorum-sensing activity, we noticed that O. vulgare displayed better inhibition percentages in violacein production even at a low concentration (MIC/4). Additionally, this EO showed better inhibition of Pseudomonas aeruginosa PAO1 migration in comparison with the terpinene-4-ol. Our findings revealed that using pure O. vulgare EO demonstrated better competitive effects against pathogenic bacteria with a different mode of action when compared to the terpinene-4-ol. Hence, exploration and development of efficient anti-infection agents from natural resources such as full EOs represent promising tools in anti-infective therapy.
Collapse
|
27
|
Cheng X, Ning J, Xu X, Zhou X. The role of bacterial cyclic di-adenosine monophosphate in the host immune response. Front Microbiol 2022; 13:958133. [PMID: 36106081 PMCID: PMC9465037 DOI: 10.3389/fmicb.2022.958133] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
Cyclic di-adenosine monophosphate (c-di-AMP) is a second messenger which is widely used in signal transduction in bacteria and archaea. c-di-AMP plays an important role in the regulation of bacterial physiological activities, such as the cell cycle, cell wall stability, environmental stress response, and biofilm formation. Moreover, c-di-AMP produced by pathogens can be recognized by host cells for the activation of innate immune responses. It can induce type I interferon (IFN) response in a stimulator of interferon genes (STING)-dependent manner, activate the nuclear factor kappa B (NF-κB) pathway, inflammasome, and host autophagy, and promote the production and secretion of cytokines. In addition, c-di-AMP is capable of triggering a host mucosal immune response as a mucosal adjuvant. Therefore, c-di-AMP is now considered to be a new pathogen-associated molecular pattern in host immunity and has become a promising target in bacterial/viral vaccine and drug research. In this review, we discussed the crosstalk between bacteria and host immunity mediated by c-di-AMP and addressed the role of c-di-AMP as a mucosal adjuvant in boosting evoked immune responses of subunit vaccines. The potential application of c-di-AMP in immunomodulation and immunotherapy was also discussed in this review.
Collapse
Affiliation(s)
- Xingqun Cheng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jia Ning
- The School and Hospital of Stomatology, Tianjin Medical University, Tianjin, China
| | - Xin Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Xuedong Zhou,
| |
Collapse
|
28
|
Loganathan A, Nachimuthu R. Antibiotic resistance, biofilm forming ability, and clonal profiling of clinical isolates of Staphylococcus aureus from southern and northeastern India. ASIAN BIOMED 2022; 16:191-199. [PMID: 37551171 PMCID: PMC10321179 DOI: 10.2478/abm-2022-0023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Staphylococcus aureus is a pathogen endemic in India and sometimes deadly for patients in intensive care units. Objectives To determine the antibiotic-resistance pattern, biofilm forming ability, and clonal type of S. aureus from isolates collected in Tamil Nadu (south) and the Mizoram (northeast) regions of India. Methods We collected S. aureus isolates from diagnostic laboratories in Tamil Nadu and Mizoram. An antibiotic susceptibility test was performed according to Clinical Laboratory and Standards Institute methods. Antibiotic-resistant determinants such as mecA, mecC, blaZ, vanA, vanB, and vanC were confirmed by polymerase chain reaction (PCR). All isolates were further studied for biofilm forming ability. Enterobacterial repetitive intergenic consensus (ERIC)-PCR was used for clonal analysis. Results A study of 206 clinical isolates showed 52.9% prevalence of methicillin-resistant S. aureus in Tamil Nadu and 49.4% in Mizoram. Minimum inhibitory concentration tests showed a high prevalence of 67% oxacillin resistance in isolates from Tamil Nadu and 49% in isolates from Mizoram. PCR showed 53% mecA in Tamil Nadu and 49% mecA in Mizoram. Vancomycin-intermediate resistance S. aureus (VISA) prevalence was lower in isolates from Tamil Nadu (4%) and Mizoram (5%). All methicillin-resistant S. aureus (MRSA) isolates formed biofilms. Clonal analysis revealed a genetic relatedness between the isolates. Conclusions The prevalence of MRSA is high in the regions studied, with most of the clinical isolates being multidrug resistant. Adopting appropriate community-based preventive measures and establishing antimicrobial stewardship is highly recommended to minimize the dissemination in antibiotic resistance.
Collapse
Affiliation(s)
- Archana Loganathan
- Department of Bio-Medical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu632014, India
| | - Ramesh Nachimuthu
- Department of Bio-Medical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu632014, India
| |
Collapse
|
29
|
Vlaeminck J, Lin Q, Xavier BB, De Backer S, Berkell M, De Greve H, Hernalsteens JP, Kumar-Singh S, Goossens H, Malhotra-Kumar S. The dynamic transcriptome during maturation of biofilms formed by methicillin-resistant Staphylococcus aureus. Front Microbiol 2022; 13:882346. [PMID: 35966712 PMCID: PMC9366926 DOI: 10.3389/fmicb.2022.882346] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 07/04/2022] [Indexed: 01/21/2023] Open
Abstract
BackgroundMethicillin-resistant Staphylococcus aureus (MRSA), a leading cause of chronic infections, forms prolific biofilms which afford an escape route from antibiotic treatment and host immunity. However, MRSA clones are genetically diverse, and mechanisms underlying biofilm formation remain under-studied. Such studies form the basis for developing targeted therapeutics. Here, we studied the temporal changes in the biofilm transcriptome of three pandemic MRSA clones: USA300, HEMRSA-15, and ST239.MethodsBiofilm formation was assessed using a static model with one representative strain per clone. Total RNA was extracted from biofilm and planktonic cultures after 24, 48, and 72 h of growth, followed by rRNA depletion and sequencing (Illumina Inc., San Diego, CA, United States, NextSeq500, v2, 1 × 75 bp). Differentially expressed gene (DEG) analysis between phenotypes and among early (24 h), intermediate (48 h), and late (72 h) stages of biofilms was performed together with in silico co-expression network construction and compared between clones. To understand the influence of SCCmec and ACME on biofilm formation, isogenic mutants containing deletions of the entire elements or of single genes therein were constructed in USA300.ResultsGenes involved in primarily core genome-encoded KEGG pathways (transporters and others) were upregulated in 24-h biofilm culture compared to 24-h planktonic culture. However, the number of affected pathways in the ST239 24 h biofilm (n = 11) was remarkably lower than that in USA300/EMRSA-15 biofilms (USA300: n = 27, HEMRSA-15: n = 58). The clfA gene, which encodes clumping factor A, was the single common DEG identified across the three clones in 24-h biofilm culture (2.2- to 2.66-fold). In intermediate (48 h) and late (72 h) stages of biofilms, decreased expression of central metabolic and fermentative pathways (glycolysis/gluconeogenesis, fatty acid biosynthesis), indicating a shift to anaerobic conditions, was already evident in USA300 and HEMRSA-15 in 48-h biofilm cultures; ST239 showed a similar profile at 72 h. Last, SCCmec+ACME deletion and opp3D disruption negatively affected USA300 biofilm formation.ConclusionOur data show striking differences in gene expression during biofilm formation by three of the most important pandemic MRSA clones, USA300, HEMRSA-15, and ST239. The clfA gene was the only significantly upregulated gene across all three strains in 24-h biofilm cultures and exemplifies an important target to disrupt early biofilms. Furthermore, our data indicate a critical role for arginine catabolism pathways in early biofilm formation.
Collapse
Affiliation(s)
- Jelle Vlaeminck
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Qiang Lin
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Basil Britto Xavier
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Sarah De Backer
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Matilda Berkell
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
- Molecular Pathology Group, Laboratory of Cell Biology & Histology, University of Antwerp, Antwerp, Belgium
| | - Henri De Greve
- VIB-VUB Center for Structural Biology, Vrije Universiteit Brussel, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Samir Kumar-Singh
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
- Molecular Pathology Group, Laboratory of Cell Biology & Histology, University of Antwerp, Antwerp, Belgium
| | - Herman Goossens
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Surbhi Malhotra-Kumar
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
- *Correspondence: Surbhi Malhotra-Kumar,
| |
Collapse
|
30
|
Lu Y, Ning H, Kang J, Bai G, Zhou L, Kang Y, Wu Z, Tian M, Zhao J, Ma Y, Bai Y. Cyclic-di-AMP Phosphodiesterase Elicits Protective Immune Responses Against Mycobacterium tuberculosis H37Ra Infection in Mice. Front Cell Infect Microbiol 2022; 12:871135. [PMID: 35811674 PMCID: PMC9256937 DOI: 10.3389/fcimb.2022.871135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Many antigens from Mycobacterium tuberculosis (M. tuberculosis) have been demonstrated as strong immunogens and proved to have application potential as vaccine candidate antigens. Cyclic di-AMP (c-di-AMP) as a bacterial second messenger regulates various bacterial processes as well as the host immune responses. Rv2837c, the c-di-AMP phosphodiesterase (CnpB), was found to be relative to virulence of M. tuberculosis and interference with host innate immune response. In this study, recombinant CnpB was administered subcutaneously to mice. We found that CnpB had strong immunogenicity and induced high levels of humoral response and lung mucosal immunity after M. tuberculosis intranasally infection. CnpB immunization stimulated splenocyte proliferation and the increasing number of activated NK cells but had little effects on Th1/Th2 cellular immune responses in spleens. However, CnpB induced significant Th1/Th2 cellular immune responses with a decreased number of T and B cells in the lungs, and significantly recruits of CD4+ and CD8+ T cells after M. tuberculosis attenuated strain H37Ra infection. Besides, we first reported that CnpB could stimulate IFN-β expression transitorily and inhibit the autophagy of macrophages in vitro. In mice intranasally infection model, CnpB immunization alleviated pathological changes and reduced M. tuberculosis H37Ra loads in the lungs. Thus, our results suggested that CnpB interferes with host innate and adaptive immune responses and confers protection against M. tuberculosis respiratory infection, which should be considered in vaccine development as well as a drug target.
Collapse
Affiliation(s)
- Yanzhi Lu
- Department of Microbiology and Pathogen Biology, Basic Medical School, Air Force Medical University, Xi’an, China
| | - Huanhuan Ning
- Department of Microbiology and Pathogen Biology, Basic Medical School, Air Force Medical University, Xi’an, China
| | - Jian Kang
- Department of Microbiology and Pathogen Biology, Basic Medical School, Air Force Medical University, Xi’an, China
| | - Guangchun Bai
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Lei Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital, Air Force Medical University, Xi’an, China
| | - Yali Kang
- Department of Physiology, Basic Medical School, Ningxia Medical University, Yinchuan, China
| | - Zhengfeng Wu
- Student Brigade, Basic Medical School, Air Force Medical University, Xi’an, China
| | - Maolin Tian
- Student Brigade, Basic Medical School, Air Force Medical University, Xi’an, China
| | - Junhao Zhao
- Student Brigade, Basic Medical School, Air Force Medical University, Xi’an, China
| | - Yueyun Ma
- Department of Clinical Laboratory, The First Affiliated Hospital, Air Force Medical University, Xi’an, China
- Department of Clinical Laboratory, Air Force Medical Center, Air Force Medical University, Beijing, China
- *Correspondence: Yinlan Bai, ; Yueyun Ma,
| | - Yinlan Bai
- Department of Microbiology and Pathogen Biology, Basic Medical School, Air Force Medical University, Xi’an, China
- *Correspondence: Yinlan Bai, ; Yueyun Ma,
| |
Collapse
|
31
|
Leshem T, Schnall BS, Azrad M, Baum M, Rokney A, Peretz A. Incidence of biofilm formation among MRSA and MSSA clinical isolates from hospitalized patients in Israel. J Appl Microbiol 2022; 133:922-929. [PMID: 35503533 PMCID: PMC9540986 DOI: 10.1111/jam.15612] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/25/2022] [Accepted: 04/29/2022] [Indexed: 12/01/2022]
Abstract
Aim To assess the biofilm‐producing capacities of Staphylococcus aureus strains isolated from hospitalized patients in Israel. Methods and Results A total of 16 S. aureus (80 MRSA and 83 MSSA) from screening (nasal swab) and clinical samples (blood and wounds) were characterized. Biofilm‐producing capacities were determined using two different biofilm detection assays: Congo Red agar (CRA) and microtiter plate (MtP). In addition, a real‐time PCR analysis was performed to detect the presence of biofilm‐associated genes (icaA and icaD) and mecA gene. The two assays showed similar biofilm production pattern (28.2% agreement). MRSA strains tended to be greater biofilm‐producers than MSSA strains. The presence of mecA was associated with biofilm production (p = 0.030). Additionally, bacteria isolated from blood samples produced less biofilm compared to those from other sources. Finally, no association was found between icaA and icaD presence and biofilm production. Conclusion This study supports earlier assumptions that biofilm formation depends strongly on environmental conditions. Significance and Impact of Study This study significantly improved our knowledge on the biofilm production capacity of S. aureus strains in Israel. Moreover, it revealed an association between the mecA gene and biofilm production. Finally, this study underscores the importance of further research to evaluate risk factors for biofilm production.
Collapse
Affiliation(s)
- Tamar Leshem
- Baruch Padeh Medical Center, Clinical Microbiology Laboratory, Poriya, Tiberias, 1528001, Israel
| | | | - Maya Azrad
- Baruch Padeh Medical Center, Clinical Microbiology Laboratory, Poriya, Tiberias, 1528001, Israel
| | - Motti Baum
- Staphylococcus aureus National Reference Center, Israel Ministry of Health, Jerusalem, Israel
| | - Assaf Rokney
- Staphylococcus aureus National Reference Center, Israel Ministry of Health, Jerusalem, Israel
| | - Avi Peretz
- Baruch Padeh Medical Center, Clinical Microbiology Laboratory, Poriya, Tiberias, 1528001, Israel.,Azrieli Faculty of Medicine, Bar Ilan University, Safed, 1311502, Israel
| |
Collapse
|
32
|
Donadu MG, Ferrari M, Mazzarello V, Zanetti S, Kushkevych I, Rittmann SKMR, Stájer A, Baráth Z, Szabó D, Urbán E, Gajdács M. No Correlation between Biofilm-Forming Capacity and Antibiotic Resistance in Environmental Staphylococcus spp.: In Vitro Results. Pathogens 2022; 11:pathogens11040471. [PMID: 35456146 PMCID: PMC9031815 DOI: 10.3390/pathogens11040471] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/08/2022] [Accepted: 04/13/2022] [Indexed: 01/18/2023] Open
Abstract
The production of biofilms is a critical factor in facilitating the survival of Staphylococcus spp. in vivo and in protecting against various environmental noxa. The possible relationship between the antibiotic-resistant phenotype and biofilm-forming capacity has raised considerable interest. The purpose of the study was to assess the interdependence between biofilm-forming capacity and the antibiotic-resistant phenotype in 299 Staphylococcus spp. (S. aureus n = 143, non-aureus staphylococci [NAS] n = 156) of environmental origin. Antimicrobial susceptibility testing and detection of methicillin resistance (MR) was performed. The capacity of isolates to produce biofilms was assessed using Congo red agar (CRA) plates and a crystal violet microtiter-plate-based (CV-MTP) method. MR was identified in 46.9% of S. aureus and 53.8% of NAS isolates (p > 0.05), with resistance to most commonly used drugs being significantly higher in MR isolates compared to methicillin-susceptible isolates. Resistance rates were highest for clindamycin (57.9%), erythromycin (52.2%) and trimethoprim-sulfamethoxazole (51.1%), while susceptibility was retained for most last-resort drugs. Based on the CRA plates, biofilm was produced by 30.8% of S. aureus and 44.9% of NAS (p = 0.014), while based on the CV-MTP method, 51.7% of S. aureus and 62.8% of NAS were identified as strong biofilm producers, respectively (mean OD570 values: S. aureus: 0.779±0.471 vs. NAS: 1.053±0.551; p < 0.001). No significant differences in biofilm formation were observed based on MR (susceptible: 0.824 ± 0.325 vs. resistant: 0.896 ± 0.367; p = 0.101). However, pronounced differences in biofilm formation were identified based on rifampicin susceptibility (S: 0.784 ± 0.281 vs. R: 1.239 ± 0.286; p = 0.011). The mechanistic understanding of the mechanisms Staphylococcus spp. use to withstand harsh environmental and in vivo conditions is crucial to appropriately address the therapy and eradication of these pathogens.
Collapse
Affiliation(s)
- Matthew Gavino Donadu
- Hospital Pharmacy, Azienda Ospedaliero Universitaria di Sassari, 07100 Sassari, Italy;
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (V.M.); (S.Z.)
| | - Marco Ferrari
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (V.M.); (S.Z.)
- Correspondence:
| | - Vittorio Mazzarello
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (V.M.); (S.Z.)
| | - Stefania Zanetti
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (V.M.); (S.Z.)
| | - Ivan Kushkevych
- Department of Experimental Biology, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic;
| | - Simon K.-M. R. Rittmann
- Archaea Physiology & Biotechnology Group, Department of Functional and Evolutionary Ecology, Universität Wien, 1090 Wien, Austria;
| | - Anette Stájer
- Department of Periodontology, Faculty of Dentistry, University of Szeged, Tisza Lajos körút 62-64, 6720 Szeged, Hungary;
| | - Zoltán Baráth
- Department of Prosthodontics, Faculty of Dentistry, University of Szeged, Tisza Lajos körút 62–64, 6720 Szeged, Hungary;
| | - Dóra Szabó
- Institute of Medical Microbiology, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary;
| | - Edit Urbán
- Department of Medical Microbiology and Immunology, University of Pécs Medical School, Szigeti út 12, 7624 Pécs, Hungary;
| | - Márió Gajdács
- Department of Oral Biology and Experimental Dental Research, Faculty of Dentistry, University of Szeged, Tisza Lajos krt. 63, 6720 Szeged, Hungary;
| |
Collapse
|
33
|
An Interplay of Multiple Positive and Negative Factors Governs Methicillin Resistance in Staphylococcus aureus. Microbiol Mol Biol Rev 2022; 86:e0015921. [PMID: 35420454 PMCID: PMC9199415 DOI: 10.1128/mmbr.00159-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The development of resistance to β-lactam antibiotics has made Staphylococcus aureus a clinical burden on a global scale. MRSA (methicillin-resistant S. aureus) is commonly known as a superbug. The ability of MRSA to proliferate in the presence of β-lactams is attributed to the acquisition of mecA, which encodes the alternative penicillin binding protein, PBP2A, which is insensitive to the antibiotics. Most MRSA isolates exhibit low-level β-lactam resistance, whereby additional genetic adjustments are required to develop high-level resistance. Although several genetic factors that potentiate or are required for high-level resistance have been identified, how these interact at the mechanistic level has remained elusive. Here, we discuss the development of resistance and assess the role of the associated components in tailoring physiology to accommodate incoming mecA.
Collapse
|
34
|
Krzyżek P, Migdał P, Grande R, Gościniak G. Biofilm Formation of Helicobacter pylori in Both Static and Microfluidic Conditions Is Associated With Resistance to Clarithromycin. Front Cell Infect Microbiol 2022; 12:868905. [PMID: 35402304 PMCID: PMC8990135 DOI: 10.3389/fcimb.2022.868905] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/07/2022] [Indexed: 12/18/2022] Open
Abstract
It is widely accepted that production of biofilm is a protective mechanism against various type of stressors, including exposure to antibiotics. However, the impact of this structure on the spread of antibiotic resistance in Helicobacter pylori is still poorly understood. Therefore, the aim of the current research was to determine the relationship between biofilm formation and antibiotic resistance of H. pylori. The study was carried out on 24 clinical strains with different resistance profiles (antibiotic-sensitive, mono-resistant, double-resistant and multidrug-resistant) against clarithromycin (CLR), metronidazole (MTZ) and levofloxacin (LEV). Using static conditions and a crystal violet staining method, a strong correlation was observed between biofilm formation and resistance to CLR but not MTZ or LEV. Based on the obtained results, three the strongest and three the weakest biofilm producers were selected and directed for a set of microfluidic experiments performed in the Bioflux system combined with fluorescence microscopy. Under continuous flow conditions, it was observed that strong biofilm producers formed twice as much of biofilm and created significantly more eDNA and in particular proteins within the biofilm matrix when compared to weak biofilm producers. Additionally, it was noticed that strong biofilm producers had higher tendency for autoaggregation and presented morphostructural differences (a greater cellular packing, shorter cells and a higher amount of both OMVs and flagella) in relation to weak biofilm counterparts. In conclusion, resistance to CLR in clinical H. pylori strains was associated with a broad array of phenotypical features translating to the ability of strong biofilm formation.
Collapse
Affiliation(s)
- Paweł Krzyżek
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
- *Correspondence: Paweł Krzyżek,
| | - Paweł Migdał
- Department of Environment, Hygiene and Animal Welfare, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| | - Rossella Grande
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Grażyna Gościniak
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
35
|
Biofilm Formation by Pathogenic Bacteria: Applying a Staphylococcus aureus Model to Appraise Potential Targets for Therapeutic Intervention. Pathogens 2022; 11:pathogens11040388. [PMID: 35456063 PMCID: PMC9027693 DOI: 10.3390/pathogens11040388] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 01/02/2023] Open
Abstract
Carried in the nasal passages by up to 30% of humans, Staphylococcus aureus is recognized to be a successful opportunistic pathogen. It is a frequent cause of infections of the upper respiratory tract, including sinusitis, and of the skin, typically abscesses, as well as of food poisoning and medical device contamination. The antimicrobial resistance of such, often chronic, health conditions is underpinned by the unique structure of bacterial biofilm, which is the focus of increasing research to try to overcome this serious public health challenge. Due to the protective barrier of an exopolysaccharide matrix, bacteria that are embedded within biofilm are highly resistant both to an infected individual’s immune response and to any treating antibiotics. An in-depth appraisal of the stepwise progression of biofilm formation by S. aureus, used as a model infection for all cases of bacterial antibiotic resistance, has enhanced understanding of this complicated microscopic structure and served to highlight possible intervention targets for both patient cure and community infection control. While antibiotic therapy offers a practical means of treatment and prevention, the most favorable results are achieved in combination with other methods. This review provides an overview of S. aureus biofilm development, outlines the current range of anti-biofilm agents that are used against each stage and summarizes their relative merits.
Collapse
|
36
|
Aswathanarayan JB, Rao P, HM S, GS S, Rai RV. Biofilm-Associated Infections in Chronic Wounds and Their Management. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022. [DOI: 10.1007/5584_2022_738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
37
|
Xu Z, Huang T, Du M, Soteyome T, Lan H, Hong W, Peng F, Fu X, Peng G, Liu J, Kjellerup BV. Regulatory network controls microbial biofilm development, with Candida albicans as a representative: from adhesion to dispersal. Bioengineered 2022; 13:253-267. [PMID: 34709974 PMCID: PMC8805954 DOI: 10.1080/21655979.2021.1996747] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/19/2021] [Indexed: 02/05/2023] Open
Abstract
Microorganisms mainly exist in the form of biofilm in nature. Biofilm can contaminate food and drinking water system, as well as cause chronic wound infections, thereby posing a potential threat to public health safety. In the last two decades, researchers have made efforts to investigate the genetic contributors control different stages of biofilm development (adherence, initiation, maturation, and dispersal). As an opportunistic pathogen, C. albicans causes severe superficial or systemic infections with high morbidity and mortality under conditions of immune dysfunction. It has been reported that 80% of C. albicans infections are directly or indirectly associated with biofilm formation on host or abiotic surfaces including indwelling medical devices, resulting in high morbidity and mortality. Significantly, the outcome of C. albicans biofilm development includes enhanced invasion, exacerbated inflammatory responses and intrinsic resistance to antimicrobial chemotherapy. Thus, this review aimed at providing a comprehensive overview of the regulatory network controls microbial biofilm development, with C. albicans as a representative, served as reference for therapeutic targets.
Collapse
Affiliation(s)
- Zhenbo Xu
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou 510640, China
- State Key Laboratory of Applied Microbiology China Southern; Insititue of Microbiology, Guangdong Academy of Sciences 510070, China
- Department of Civil and Environmental Engineering, University of Maryland, College Park, MD20742,USA
- Department of Laboratory Medicine, the Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Home Economics Technology, Rajamangala University of Technology Phra Nakhon, Bangkok, Thailand
- National Institute of Fundamental Studies, Hantana road, Kandy, Sri Lanka
| | - Tengyi Huang
- Department of Laboratory Medicine, the Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Min Du
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Thanapop Soteyome
- Home Economics Technology, Rajamangala University of Technology Phra Nakhon, Bangkok, Thailand
| | - Haifeng Lan
- Department of Orthopaedic Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wei Hong
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Fang Peng
- Department of Critical Care Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xin Fu
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Gongyong Peng
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Junyan Liu
- Department of Civil and Environmental Engineering, University of Maryland, College Park, MD20742,USA
| | - Birthe V. Kjellerup
- Department of Civil and Environmental Engineering, University of Maryland, College Park, MD20742,USA
| |
Collapse
|
38
|
Mahto KU, Kumari S, Das S. Unraveling the complex regulatory networks in biofilm formation in bacteria and relevance of biofilms in environmental remediation. Crit Rev Biochem Mol Biol 2021; 57:305-332. [PMID: 34937434 DOI: 10.1080/10409238.2021.2015747] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Biofilms are assemblages of bacteria embedded within a matrix of extracellular polymeric substances (EPS) attached to a substratum. The process of biofilm formation is a complex phenomenon regulated by the intracellular and intercellular signaling systems. Various secondary messenger molecules such as cyclic dimeric guanosine 3',5'-monophosphate (c-di-GMP), cyclic adenosine 3',5'-monophosphate (cAMP), and cyclic dimeric adenosine 3',5'-monophosphate (c-di-AMP) are involved in complex signaling networks to regulate biofilm development in several bacteria. Moreover, the cell to cell communication system known as Quorum Sensing (QS) also regulates biofilm formation via diverse mechanisms in various bacterial species. Bacteria often switch to the biofilm lifestyle in the presence of toxic pollutants to improve their survivability. Bacteria within a biofilm possess several advantages with regard to the degradation of harmful pollutants, such as increased protection within the biofilm to resist the toxic pollutants, synthesis of extracellular polymeric substances (EPS) that helps in the sequestration of pollutants, elevated catabolic gene expression within the biofilm microenvironment, higher cell density possessing a large pool of genetic resources, adhesion ability to a wide range of substrata, and metabolic heterogeneity. Therefore, a comprehensive account of the various factors regulating biofilm development would provide valuable insights to modulate biofilm formation for improved bioremediation practices. This review summarizes the complex regulatory networks that influence biofilm development in bacteria, with a major focus on the applications of bacterial biofilms for environmental restoration.
Collapse
Affiliation(s)
- Kumari Uma Mahto
- Department of Life Science, Laboratory of Environmental Microbiology and Ecology (LEnME), National Institute of Technology, Odisha, India
| | - Swetambari Kumari
- Department of Life Science, Laboratory of Environmental Microbiology and Ecology (LEnME), National Institute of Technology, Odisha, India
| | - Surajit Das
- Department of Life Science, Laboratory of Environmental Microbiology and Ecology (LEnME), National Institute of Technology, Odisha, India
| |
Collapse
|
39
|
Wu S, Zhang J, Peng Q, Liu Y, Lei L, Zhang H. The Role of Staphylococcus aureus YycFG in Gene Regulation, Biofilm Organization and Drug Resistance. Antibiotics (Basel) 2021; 10:antibiotics10121555. [PMID: 34943766 PMCID: PMC8698359 DOI: 10.3390/antibiotics10121555] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 02/05/2023] Open
Abstract
Antibiotic resistance is a serious global health concern that may have significant social and financial consequences. Methicillin-resistant Staphylococcus aureus (MRSA) infection is responsible for substantial morbidity and leads to the death of 21.8% of infected patients annually. A lack of novel antibiotics has prompted the exploration of therapies targeting bacterial virulence mechanisms. The two-component signal transduction system (TCS) enables microbial cells to regulate gene expression and the subsequent metabolic processes that occur due to environmental changes. The YycFG TCS in S. aureus is essential for bacterial viability, the regulation of cell membrane metabolism, cell wall synthesis and biofilm formation. However, the role of YycFG-associated biofilm organization in S. aureus antimicrobial drug resistance and gene regulation has not been discussed in detail. We reviewed the main molecules involved in YycFG-associated cell wall biosynthesis, biofilm development and polysaccharide intercellular adhesin (PIA) accumulation. Two YycFG-associated regulatory mechanisms, accessory gene regulator (agr) and staphylococcal accessory regulator (SarA), were also discussed. We highlighted the importance of biofilm formation in the development of antimicrobial drug resistance in S. aureus infections. Data revealed that inhibition of the YycFG pathway reduced PIA production, biofilm formation and bacterial pathogenicity, which provides a potential target for the management of MRSA-induced infections.
Collapse
Affiliation(s)
- Shizhou Wu
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China; (S.W.); (J.Z.); (Q.P.)
| | - Junqi Zhang
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China; (S.W.); (J.Z.); (Q.P.)
| | - Qi Peng
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China; (S.W.); (J.Z.); (Q.P.)
| | - Yunjie Liu
- West China School of Public Health, Sichuan University, Chengdu 610041, China;
| | - Lei Lei
- West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Correspondence: (L.L.); (H.Z.)
| | - Hui Zhang
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China; (S.W.); (J.Z.); (Q.P.)
- Correspondence: (L.L.); (H.Z.)
| |
Collapse
|
40
|
Dietrich A, Steffens U, Sass P, Bierbaum G. The hypersusceptible antibiotic screening strain Staphylococcus aureus SG511-Berlin harbors multiple mutations in regulatory genes. Int J Med Microbiol 2021; 311:151545. [PMID: 34896903 DOI: 10.1016/j.ijmm.2021.151545] [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: 08/17/2021] [Revised: 12/01/2021] [Accepted: 12/05/2021] [Indexed: 10/19/2022] Open
Abstract
The genetic plasticity of Staphylococcus aureus has facilitated the evolution of many virulent and drug-resistant strains. Here we present the sequence of the 2.74 Mbp genome of S. aureus SG511-Berlin, which is frequently used for antibiotic screening. Although S. aureus SG511 and the related methicillin-resistant S. aureus MRSA252 share a high similarity in their core genomes, indicated by an average nucleotide identity (ANI) of 99.83%, the accessory genomes of these strains differed, as nearly no mobile elements and resistance determinants were identified in the genome of S. aureus SG511. Susceptibility testing showed that S. aureus SG511 was susceptible to most of the tested antibiotics of different classes. Intriguingly, and in contrast to the standard laboratory strain S. aureus HG001, S. aureus SG511 was even hyper-susceptible towards cell wall and membrane targeting agents, with the exception of the MurA-inhibitor fosfomycin. In depth comparative genome analysis revealed that, in addition to the loss of function mutation in the antibiotic sensor histidine kinase gene graS, further mutations had occurred in the lysyltransferase gene mprF, the structural giant protein gene ebh, and the regulator genes codY and saeR, which might contribute to antibiotic susceptibility. In addition, an insertion element in agrC abolishes Agr-activity in S. aureus SG511, and the spa and sarS genes, which encode the surface protein SpA and its transcriptional regulator, were deleted. Thus, the lack of mobile resistance genes together with multiple mutations affecting cell envelope morphology may render S. aureus SG511 hyper-susceptible towards most cell wall targeting agents.
Collapse
Affiliation(s)
- Alina Dietrich
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Medical Faculty, 53127 Bonn, Germany.
| | - Ursula Steffens
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Medical Faculty, 53127 Bonn, Germany.
| | - Peter Sass
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Medical Faculty, 53127 Bonn, Germany.
| | - Gabriele Bierbaum
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Medical Faculty, 53127 Bonn, Germany.
| |
Collapse
|
41
|
Loss of GdpP function in Staphylococcus aureus leads to β-lactam tolerance and enhanced evolution of β-lactam resistance. Antimicrob Agents Chemother 2021; 66:e0143121. [PMID: 34843389 DOI: 10.1128/aac.01431-21] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Infections caused by Staphylococcus aureus are a leading cause of mortality. Treating infections caused by S. aureus is difficult due to resistance against most traditional antibiotics, including β-lactams. We previously reported the presence of mutations in gdpP among S. aureus strains that were obtained by serial passaging in β-lactam drugs. Similar mutations have recently been reported in natural S. aureus isolates that are either non-susceptible or resistant to β-lactam antibiotics. gdpP codes for a phosphodiesterase that cleaves cyclic-di-AMP (CDA), a newly discovered second messenger. In this study, we sought to identify the role of gdpP in β-lactam resistance in S. aureus. Our results showed that gdpP associated mutations caused loss of phosphodiesterase function, leading to increased CDA accumulation in the bacterial cytosol. Deletion of gdpP led to an enhanced ability of the bacteria to withstand a β-lactam challenge (two to three log increase in bacterial colony forming units) by promoting tolerance without enhancing MICs of β-lactam antibiotics. Our results demonstrated that increased drug tolerance due to loss of GdpP function can provide a selective advantage in acquisition of high-level β-lactam resistance. Loss of GdpP function thus increases tolerance to β-lactams that can lead to its therapy failure and can permit β-lactam resistance to occur more readily.
Collapse
|
42
|
Two Novel Semisynthetic Lipoglycopeptides Active against Staphylococcus aureus Biofilms and Cells in Late Stationary Growth Phase. Pharmaceuticals (Basel) 2021; 14:ph14111182. [PMID: 34832964 PMCID: PMC8619453 DOI: 10.3390/ph14111182] [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: 10/08/2021] [Revised: 11/06/2021] [Accepted: 11/17/2021] [Indexed: 11/16/2022] Open
Abstract
The increase in antibiotic resistance among Gram-positive bacteria underscores the urgent need to develop new antibiotics. New antibiotics should target actively growing susceptible bacteria that are resistant to clinically accepted antibiotics including bacteria that are not growing or are protected in a biofilm environment. In this paper, we compare the in vitro activities of two new semisynthetic glycopeptide antibiotics, MA79 and ERJ390, with two clinically used glycopeptide antibiotics-vancomycin and teicoplanin. The new antibiotics effectively killed not only exponentially growing cells of Staphylococcus aureus, but also cells in the stationary growth phase and biofilm.
Collapse
|
43
|
Chen JW, Hsu CC, Su CC, Hsu RB, Chiu YL, Chia JS, Jung CJ. Transient bacteremia promotes catheter-related central venous thrombosis through neutrophil extracellular traps. Thromb Haemost 2021; 122:1198-1208. [PMID: 34768303 DOI: 10.1055/a-1695-8612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Formation of intravenous catheter-related thrombosis leads to central venous stenosis in patients requiring renal replacement therapy or chemotherapy infusion, yet the triggering or mechanisms remain unclear, especially in patients without symptoms of infection. In this study, we found that neutrophil extracellular traps (NETs) could be detected in the fibrin sheaths from dialysis patients without clinical manifestations of infection. Confocal microscopy revealed bacteria imbedded in NETs in the fibrin sheaths. Thirty-nine of 50 (78%) fibrin sheath specimens contained bacteria detectable by 16S ribosomal RNA genome typing with a predominance of Staphylococcus aureus (69%). In rat models, transient bacteremia of S. aureus induced NETs in enlarged fibrin sheaths, and treatment with DNase I alone significantly reduced both NET and fibrin sheath formation surrounding the catheter. Therefore, transient bacteremia could be a silent trigger that induces NET-related immunothrombosis enhancing catheter-related central venous stenosis.
Collapse
Affiliation(s)
- Jeng-Wei Chen
- National Taiwan University Hospital, Department of Surgery, National Taiwan University College of Medicine, Taipei, Taiwan.,Graduate institute of clinical medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chih-Chieh Hsu
- School of Dentistry, Graduate Institute of Oral Biology, National Taiwan University, Taipei, Taiwan
| | - Chien-Chia Su
- National Taiwan University Hospital, Department of ophthalmology, National Taiwan University College of Medicine, Taipei, Taiwan.,Graduate institute of clinical medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ron-Bin Hsu
- National Taiwan University Hospital, Department of Surgery, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yen-Ling Chiu
- Graduate institute of clinical medicine, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Medicine, Division of Nephrology, Far Eastern Memorial Hospital, New Taipei City, Taiwan.,College of Informatics, Department of Computer Science and Engineering, Graduate Program in Biomedical Informatics, Yuan Ze University, Chung-Li, Taiwan
| | - Jean-San Chia
- Graduate institute of clinical medicine, National Taiwan University College of Medicine, Taipei, Taiwan.,Graduate Institute of Microbiology, National Taiwan University College of Medicine, Taipei, Taiwan.,School of Dentistry, Graduate Institute of Clinical Dentistry, National Taiwan University, Taipei, Taiwan
| | - Chiau-Jing Jung
- College of Medicine, School of Medicine, Department of Microbiology and Immunology, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
44
|
Extracellular DNA (eDNA). A Major Ubiquitous Element of the Bacterial Biofilm Architecture. Int J Mol Sci 2021; 22:ijms22169100. [PMID: 34445806 PMCID: PMC8396552 DOI: 10.3390/ijms22169100] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 12/22/2022] Open
Abstract
After the first ancient studies on microbial slime (the name by which the biofilm matrix was initially indicated), multitudes of studies on the morphology, composition and physiology of biofilms have arisen. The emergence of the role that biofilms play in the pathogenesis of recalcitrant and persistent clinical infections, such as periprosthetic orthopedic infections, has reinforced scientific interest. Extracellular DNA (eDNA) is a recently uncovered component that is proving to be almost omnipresent in the extracellular polymeric substance (EPS) of biofilm. This macromolecule is eliciting unprecedented consideration for the critical impact on the pathogenesis of chronic clinical infections. After a systematic review of the literature, an updated description of eDNA in biofilms is presented, with a special focus on the latest findings regarding its fundamental structural role and the contribution it makes to the complex architecture of bacterial biofilms through interactions with a variety of other molecular components of the biofilm matrix.
Collapse
|
45
|
Cardoso Guimarães L, Marques de Souza B, de Oliveira Whitaker C, Abreu F, Barreto Rocha Ferreira R, Dos Santos KRN. Increased biofilm formation by Staphylococcus aureus clinical isolates on surfaces covered with plasma proteins. J Med Microbiol 2021; 70. [PMID: 34338626 DOI: 10.1099/jmm.0.001389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Introduction. Biofilm formation is a major virulence factor associated with Staphylococcus aureus infections. However, the influence of plasma proteins on biofilm formation of clinical isolates in vitro remains unclear.Hypotheses. We hypothesized that coating surfaces with plasma proteins might induce biofilm formation by S. aureus of different clonal lineages.Aim. To evaluate biofilm production by clinical S. aureus isolates of different clonal lineages isolated in Rio de Janeiro hospitals and investigated the presence of biofilm-associated genes.Methodology. This study assessed biofilm production of 60 S. aureus isolates in polystyrene microtitre plates with and without fibrinogen or fibronectin. The biochemical composition of the biofilm matrices was determined and the biofilm formation on fibrinogen-coated surfaces was also evaluated by confocal laser scanning microscopy. The presence of biofilm-related genes was detected by PCR, and the typing and functionality of agr operon was also evaluated.Results. Most of the isolates (45 %) were weak biofilm producers or non-producers. However, most of them presented a significant increase in biofilm production on plates covered with plasma proteins. There was no significant difference in biofilm formation between methicillin-resistant and -susceptible S. aureus isolates, or between different clonal lineages, except for ST30-IV (weak producers) and ST239-III (strong producers). The fnbB gene was associated with higher biofilm production.Conclusion. An increase in biofilm production in the presence of plasma proteins highlights the importance of investigating biofilm formation by S. aureus clinical isolates under different conditions since this virulence factor contributes to persistent infections and increased resistance to antimicrobials.
Collapse
Affiliation(s)
- Lorrayne Cardoso Guimarães
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Bruna Marques de Souza
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Fernanda Abreu
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | |
Collapse
|
46
|
Paes Leme RC, da Silva RB. Antimicrobial Activity of Non-steroidal Anti-inflammatory Drugs on Biofilm: Current Evidence and Potential for Drug Repurposing. Front Microbiol 2021; 12:707629. [PMID: 34385992 PMCID: PMC8353384 DOI: 10.3389/fmicb.2021.707629] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/05/2021] [Indexed: 12/20/2022] Open
Abstract
It has been demonstrated that some non-steroidal anti-inflammatory drugs (NSAIDs), like acetylsalicylic acid, diclofenac, and ibuprofen, have anti-biofilm activity in concentrations found in human pharmacokinetic studies, which could fuel an interest in repurposing these well tolerated drugs as adjunctive therapies for biofilm-related infections. Here we sought to review the currently available data on the anti-biofilm activity of NSAIDs and its relevance in a clinical context. We performed a systematic literature review to identify the most commonly tested NSAIDs drugs in the last 5 years, the bacterial species that have demonstrated to be responsive to their actions, and the emergence of resistance to these molecules. We found that most studies investigating NSAIDs' activity against biofilms were in vitro, and frequently tested non-clinical bacterial isolates, which may not adequately represent the bacterial populations that cause clinically-relevant biofilm-related infections. Furthermore, studies concerning NSAIDs and antibiotic resistance are scarce, with divergent outcomes. Although the potential to use NSAIDs to control biofilm-related infections seems to be an exciting avenue, there is a paucity of studies that tested these drugs using appropriate in vivo models of biofilm infections or in controlled human clinical trials to support their repurposing as anti-biofilm agents.
Collapse
Affiliation(s)
- Rodrigo Cuiabano Paes Leme
- Laboratório Especial de Microbiologia Clínica (LEMC), Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, Brazil.,Department of Infectious Diseases, Centro Universitário de Volta Redonda, Volta Redonda, Brazil
| | | |
Collapse
|
47
|
Jin Y, Yu X, Chen Y, Chen W, Shen P, Luo Q, Zhang S, Kong X, Zheng B, Xiao Y. Characterization of highly virulent community-associated methicillin-resistant Staphylococcus aureus ST9-SCC mec XII causing bloodstream infection in China. Emerg Microbes Infect 2021; 9:2526-2535. [PMID: 33174510 PMCID: PMC7717876 DOI: 10.1080/22221751.2020.1848354] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Previous studies have shown that livestock (LA)-MRSA ST398 evolved from a human-adapted methicillin-susceptible S. aureus (MSSA) clone. However, detailed information regarding ST9 is still unclear. Here, we characterized a community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) ST9-SCCmec XII isolate that has not been previously reported to cause serious disease in China. We obtained whole-genome sequences of one ST9-t899-XII isolate—ZY462471—from a patient with bloodstream infection without livestock contact. The antibiotic susceptibilities of ZY462471 were determined and the clinical information was extracted from medical notes and compared with twenty-seven previously sequenced genomes. Phylogenetic reconstruction was performed to investigate the probable host evolutionary origins of ZY462471, and the difference in resistome and virulence factors were investigated. Virulence assay was performed to evaluate the high virulence potential of ZY462471 and compare the virulence between the closest ST9 MSSA neighbours. Clinical data suggested that ZY462471 is a CA-MRSA. Phylogenetic analysis showed a much closer relationship of ZY462471 with human-associated MSSA ST9 isolates than other LA-MRSA ST9 isolates, suggesting that ZY462471 probably evolved from ST9 MSSA predecessors by acquiring an SCCmec cassette. Importantly, virulence assays indicated that ZY462471 was highly virulent and compared with the MSSA ST9 predecessors, ZY462471 did not show attenuated virulence. Finally, we found that ZY462471 harboured an immune evasion cluster (IEC)-carrying βC-Φ, which is typically found in human clinical S. aureus rather than LA-MRSA isolates, suggesting that ZY4762471 obtained the IEC-carrying βC-Φs from human clinical S. aureus strains. Considering its high virulence potential, this strain should be monitored to prevent more widespread dissemination.
Collapse
Affiliation(s)
- Ye Jin
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Yu
- Department of Respiratory and Critical Care Medicine, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yunbo Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weiwei Chen
- Department of Laboratory Medicine, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Pin Shen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qixia Luo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuntian Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyang Kong
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Beiwen Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yonghong Xiao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
48
|
Accumulation of Succinyl Coenzyme A Perturbs the Methicillin-Resistant Staphylococcus aureus (MRSA) Succinylome and Is Associated with Increased Susceptibility to Beta-Lactam Antibiotics. mBio 2021; 12:e0053021. [PMID: 34182779 PMCID: PMC8437408 DOI: 10.1128/mbio.00530-21] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Penicillin binding protein 2a (PBP2a)-dependent resistance to β-lactam antibiotics in methicillin-resistant Staphylococcus aureus (MRSA) is regulated by the activity of the tricarboxylic acid (TCA) cycle via a poorly understood mechanism. We report that mutations in sucC and sucD, but not other TCA cycle enzymes, negatively impact β-lactam resistance without changing PBP2a expression. Increased intracellular levels of succinyl coenzyme A (succinyl-CoA) in the sucC mutant significantly perturbed lysine succinylation in the MRSA proteome. Suppressor mutations in sucA or sucB, responsible for succinyl-CoA biosynthesis, reversed sucC mutant phenotypes. The major autolysin (Atl) was the most succinylated protein in the proteome, and increased Atl succinylation in the sucC mutant was associated with loss of autolytic activity. Although PBP2a and PBP2 were also among the most succinylated proteins in the MRSA proteome, peptidoglycan architecture and cross-linking were unchanged in the sucC mutant. These data reveal that perturbation of the MRSA succinylome impacts two interconnected cell wall phenotypes, leading to repression of autolytic activity and increased susceptibility to β-lactam antibiotics.
Collapse
|
49
|
Manoharan A, Das T, Whiteley GS, Glasbey T, Kriel FH, Manos J. The effect of N-acetylcysteine in a combined antibiofilm treatment against antibiotic-resistant Staphylococcus aureus. J Antimicrob Chemother 2021; 75:1787-1798. [PMID: 32363384 DOI: 10.1093/jac/dkaa093] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 01/31/2020] [Accepted: 02/17/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The WHO declared Staphylococcus aureus as a 'pathogen of high importance' in 2017. One-fifth of all bloodstream-related infections in Australia and 12 000 cases of bacteraemia in the UK (2017-18) were caused by the MRSA variant. To address the need for novel therapies, we investigated several permutations of an innovative combination therapy containing N-acetylcysteine (NAC), an antibiotic and an enzyme of choice in eradicating MRSA and MSSA biofilms. METHODS Biofilm viability (resazurin assay) and colony count methods were used to investigate the effect of NAC, antibiotics and enzymes on S. aureus biofilm disruption and killing. The effects of NAC and enzymes on the polysaccharide content of biofilm matrices were analysed using the phenol/sulphuric acid method and the effect of NAC on DNA cleavage was determined using the Qubit fluorometer technique. Changes in biofilm architecture when subjected to NAC and enzymes were visualized using confocal laser scanning microscopy (CLSM). RESULTS NAC alone displayed bacteriostatic effects when tested on planktonic bacterial growth. Combination treatments containing 30 mM NAC resulted in ≥90% disruption of biofilms across all MRSA and MSSA strains with a 2-3 log10 decrease in cfu/mL in treated biofilms. CLSM showed that NAC treatment drastically disrupted S. aureus biofilm architecture. There was also reduced polysaccharide production in MRSA biofilms in the presence of NAC. CONCLUSIONS Our results indicate that inclusion of NAC in a combination treatment is a promising strategy for S. aureus biofilm eradication. The intrinsic acidity of NAC was identified as key to maximum biofilm disruption and degradation of matrix components.
Collapse
Affiliation(s)
- Arthika Manoharan
- Department of Infectious Diseases and Immunology, Central Clinical School, The University of Sydney, Sydney, Australia
| | - Theerthankar Das
- Department of Infectious Diseases and Immunology, Central Clinical School, The University of Sydney, Sydney, Australia
| | | | - Trevor Glasbey
- Whiteley Corporation, 19-23 Laverick Avenue, Tomago NSW 2319, Australia
| | - Frederik H Kriel
- Whiteley Corporation, 19-23 Laverick Avenue, Tomago NSW 2319, Australia
| | - Jim Manos
- Whiteley Corporation, 19-23 Laverick Avenue, Tomago NSW 2319, Australia
| |
Collapse
|
50
|
The Diadenylate Cyclase CdaA Is Critical for Borrelia turicatae Virulence and Physiology. Infect Immun 2021; 89:IAI.00787-20. [PMID: 33846120 PMCID: PMC8316131 DOI: 10.1128/iai.00787-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/22/2021] [Indexed: 12/16/2022] Open
Abstract
Relapsing fever (RF), caused by spirochetes of the genus Borrelia, is a globally distributed, vector-borne disease with high prevalence in developing countries. To date, signaling pathways required for infection and virulence of RF Borrelia spirochetes are unknown. Cyclic di-AMP (c-di-AMP), synthesized by diadenylate cyclases (DACs), is a second messenger predominantly found in Gram-positive organisms that is linked to virulence and essential physiological processes. Although Borrelia is Gram-negative, it encodes one DAC (CdaA), and its importance remains undefined. To investigate the contribution of c-di-AMP signaling in the RF bacterium Borrelia turicatae, a cdaA mutant was generated. The mutant was significantly attenuated during murine infection, and genetic complementation reversed this phenotype. Because c-di-AMP is essential for viability in many bacteria, whole-genome sequencing was performed on cdaA mutants, and single-nucleotide polymorphisms identified potential suppressor mutations. Additionally, conditional mutation of cdaA confirmed that CdaA is important for normal growth and physiology. Interestingly, mutation of cdaA did not affect expression of homologs of virulence regulators whose levels are impacted by c-di-AMP signaling in the Lyme disease bacterium Borrelia burgdorferi Finally, the cdaA mutant had a significant growth defect when grown with salts, at decreased osmolarity, and without pyruvate. While the salt treatment phenotype was not reversed by genetic complementation, possibly due to suppressor mutations, growth defects at decreased osmolarity and in media lacking pyruvate could be attributed directly to cdaA inactivation. Overall, these results indicate CdaA is critical for B. turicatae pathogenesis and link c-di-AMP to osmoregulation and central metabolism in RF spirochetes.
Collapse
|