1
|
Teixeira IM, de Moraes Assumpção Y, Paletta ACC, Antunes M, da Silva IT, Jaeger LH, Ferreira RF, de Oliveira Ferreira E, de Araújo Penna B. Investigation on biofilm composition and virulence traits of S. pseudintermedius isolated from infected and colonized dogs. Braz J Microbiol 2024; 55:2923-2936. [PMID: 38955982 PMCID: PMC11405647 DOI: 10.1007/s42770-024-01405-y] [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: 02/21/2024] [Accepted: 05/28/2024] [Indexed: 07/04/2024] Open
Abstract
Staphylococcus pseudintermedius, which is part of the skin microbiome of dogs, causes a variety of opportunistic infections. These infections may become more difficult to treat due to the formation of biofilm. The capacity of S. pseudintermedius to form biofilm, as well as the associated genes, has not been elucidated. This study evaluated the production and composition of S. pseudintermedius biofilm. Samples were collected from both infected dogs and asymptomatic dogs. Isolates were identified using mass spectrometry and Multiplex-PCR. Biofilm production and composition were assessed using a quantitative microtiter plate assay. The presence of ica operon genes and sps genes was investigated using conventional PCR. The investigation of Agr type and virulence genes was conducted in silico on 24 sequenced samples. All strains could produce strong biofilms, with most of the isolates presenting a polysaccharide biofilm. 63.6% of the isolates carried the complete ica operon (ADBC). All samples showed the presence of the genes spsK, spsA, and spsL, while the distribution of other genes varied. Agr type III was the most prevalent (52.2%). All sequenced samples carried the cytotoxins hlb, luk-S, luk-F, as well as the exfoliative toxins siet and se_int. No isolate displayed other exfoliative toxins. Only LB1733 presented a set of different enterotoxins (sea, seb, sec_canine, seh, sek, sel, and seq). Our findings suggest that S. pseudintermedius is a strong producer of biofilm and carries virulence genes.
Collapse
Affiliation(s)
- Izabel Mello Teixeira
- Laboratório de Biologia de Anaeróbios, Depto. Microbiologia Médica, IMPPG, UFRJ, Rio de Janeiro, Brazil
- Laboratório de Cocos Gram Positivos, Depto. de Microbiologia e Parasitologia, UFF, Rio de Janeiro, Brazil
| | - Yasmim de Moraes Assumpção
- Laboratório de Cocos Gram Positivos, Depto. de Microbiologia e Parasitologia, UFF, Rio de Janeiro, Brazil
| | - Ana Clara Cabral Paletta
- Laboratório de Cocos Gram Positivos, Depto. de Microbiologia e Parasitologia, UFF, Rio de Janeiro, Brazil
| | - Milena Antunes
- Laboratório de Biologia de Anaeróbios, Depto. Microbiologia Médica, IMPPG, UFRJ, Rio de Janeiro, Brazil
| | - Isabella Thomaz da Silva
- Laboratório de Cocos Gram Positivos, Depto. de Microbiologia e Parasitologia, UFF, Rio de Janeiro, Brazil
| | - Lauren Hubert Jaeger
- Laboratório de Células-Tronco e Parasitologia Molecular, Depto. de Ciências Farmacêuticas, UFJF, Rio de Janeiro, Brazil
| | - Renata Fernandes Ferreira
- Curso de Medicina Veterinária da Universidade de Vassouras - Diagnóstico em medicina veterinária - Vassouras, Rio de Janeiro, Brazil
| | | | - Bruno de Araújo Penna
- Laboratório de Cocos Gram Positivos, Depto. de Microbiologia e Parasitologia, UFF, Rio de Janeiro, Brazil.
| |
Collapse
|
2
|
Nascimento LD, Lopes ACP, Teixeira MM, da Silva JMA, Silva LO, de Almeida JB, Campos GB, Teodósio R, Marques LM. Clinical and Microbiological Profile of Diabetic Foot Ulcers Infected With Staphylococcus aureus in a Regional General Hospital in Bahia, Brazil. INT J LOW EXTR WOUND 2024; 23:252-263. [PMID: 34747264 DOI: 10.1177/15347346211050771] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
It is necessary to know the resistance profile of Staphylococcus aureus to better control diabetic foot ulcer infections, to establish rational antibiotic therapy, and to avoid the development of resistant strains. This cross-sectional study evaluated the clinical parameters, virulence, and antimicrobial resistance profiles of S aureus in patients with diabetic foot disease admitted to a public hospital. S aureus strains were identified in patients with diabetes with amputation indication. Infected tissue samples were collected, microbes were isolated and identified. The microbial resistance profile was determined. Samples were also analyzed for biofilm formation and other virulence markers. The 34 individuals examined were mostly men, black, aged 60 years on average, and generally had a low income and education level. Most individuals had type 2 diabetes, and the mean time since diagnosis was 13.9 years. On an SF-36 (the Medical Outcomes Study 36-item short-form health survey) quality-of-life questionnaire, 75% of individuals obtained a score equal to 0 for physical impairment. S aureus specimens from 17 patients were isolated, corresponding to 50% of samples. Five isolates were classified as methicillin-resistant S aureus (MRSA). Molecular typing revealed that 20% of MRSA strains were SCCmec type V and 80% were type I. All isolates were sensitive to doxycycline; 61.5% were resistant to erythromycin, 38.5% to cefoxitin, 30.7% to clindamycin and ciprofloxacin, 23% to meropenem, 15.3% to gentamicin, 38.5% to oxacillin, and 7.7% (one strain) to vancomycin. Regarding biofilm production, 53% of samples were able to produce biofilms, and 84.6% had icaA and/or icaD genes. Additionally, the following enterotoxin genes were identified in the isolates: seb, sec, seg, and sei (5.9%, 5.9%, 11.8%, and 23.9%, respectively) and agr types 1 (5.9%) and 2 (11.8%). Genotypic evaluation made it possible to understand the pathogenicity of S aureus strains isolated from the diabetic foot; laboratory tests can assist in the monitoring of patients with systemic involvement.
Collapse
Affiliation(s)
| | | | - Mariana Morais Teixeira
- Federal University of Bahia, Multidisciplinary Institute in Health, Vitória da Conquista, Bahia, Brazil
| | | | - Letícia Oliveira Silva
- Federal University of Bahia, Multidisciplinary Institute in Health, Vitória da Conquista, Bahia, Brazil
| | - Jessica Bomfim de Almeida
- Federal University of Bahia, Multidisciplinary Institute in Health, Vitória da Conquista, Bahia, Brazil
| | - Guilherme Barreto Campos
- Federal University of Bahia, Multidisciplinary Institute in Health, Vitória da Conquista, Bahia, Brazil
| | - Rosa Teodósio
- Instituto de Higiene e Medicina Tropical, IHMT, Universidade NOVA de Lisboa, UNL; Global Health and Tropical Medicine,, Lisbon, Portugal
| | - Lucas Miranda Marques
- Federal University of Bahia, Multidisciplinary Institute in Health, Vitória da Conquista, Bahia, Brazil
| |
Collapse
|
3
|
Pokhrel S, Sharma N, Aryal S, Khadka R, Thapa TB, Pandey P, Joshi G. Detection of Biofilm Production and Antibiotic Susceptibility Pattern among Clinically Isolated Staphylococcus aureus. J Pathog 2024; 2024:2342468. [PMID: 38745848 PMCID: PMC11090675 DOI: 10.1155/2024/2342468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 03/16/2024] [Accepted: 04/13/2024] [Indexed: 05/16/2024] Open
Abstract
Aim The increasing antibiotic resistance and the ability to form biofilms in medical devices have become the leading cause of severe infections associated with Staphylococcus aureus (S. aureus). Since the bacteria living in biofilms can exhibit 10- to 1,000-fold increase in antibiotic resistance and implicate chronic infectious diseases, the detection of S. aureus ability to form biofilms is of great importance for managing, minimizing, and effectively treating infections caused by it. This study aimed to compare the tube and tissue culture methods to detect biofilm production and antibiotic susceptibility in MRSA and MSSA. Materials and Methods The S. aureus isolates were identified by the examination of the colony morphology, Gram staining, and various biochemical tests. Antimicrobial susceptibility testing of all isolates was performed by the modified Kirby-Bauer disc diffusion method as recommended by CLSI guidelines. MRSA screening was performed phenotypically using a cefoxitin disc (30 µg). Isolates were tested for inducible resistance using the D-test, and two phenotypic methods detected biofilm formation. Results Among 982 nonrepeated clinical specimens, S. aureus was isolated from 103 (10.48%). Among 103 clinical isolates of S. aureus, 54 (52.42%) isolates were MRSA, and 49 (47.57%) were MSSA. Among 54 MRSA isolates, the inducible MLSB phenotype was observed in 23/54 (42.59%) with a positive D-test. By TCP method, 26 (48.1%) MRSA isolates were strong biofilm producers, whereas, among all MSSA isolates, only 6 (12.2%) were strong biofilm producers. Conclusion MRSA showed strong biofilm production in comparison with MSSA. The TCP method is a recommended reliable method to detect the biofilm among S. aureus isolates, and the TM method could be useful for the screening of biofilm production in S. aureus in the routine clinical laboratory.
Collapse
Affiliation(s)
- Sushant Pokhrel
- Department of Laboratory Medicine, Manmohan Memorial Institute of Health Sciences, Kathmandu, Nepal
| | - Namrata Sharma
- Department of Microbiology, Nobel College, Kathmandu, Nepal
| | - Suraj Aryal
- Department of Microbiology, Chitwan Medical College, Bharatpur, Nepal
| | - Rachita Khadka
- Department of Microbiology, Nobel College, Kathmandu, Nepal
| | - Tika Bahadur Thapa
- Department of Laboratory Medicine, Manmohan Memorial Institute of Health Sciences, Kathmandu, Nepal
| | - Pawan Pandey
- Department of Pharmacology, Nobel College, Kathmandu, Nepal
| | - Govardhan Joshi
- Department of Laboratory Medicine, Manmohan Memorial Institute of Health Sciences, Kathmandu, Nepal
- Department of Microbiology, Global Hospital, Lalitpur, Nepal
| |
Collapse
|
4
|
Aboelnaga N, Elsayed SW, Abdelsalam NA, Salem S, Saif NA, Elsayed M, Ayman S, Nasr M, Elhadidy M. Deciphering the dynamics of methicillin-resistant Staphylococcus aureus biofilm formation: from molecular signaling to nanotherapeutic advances. Cell Commun Signal 2024; 22:188. [PMID: 38519959 PMCID: PMC10958940 DOI: 10.1186/s12964-024-01511-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/01/2024] [Indexed: 03/25/2024] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) represents a global threat, necessitating the development of effective solutions to combat this emerging superbug. In response to selective pressures within healthcare, community, and livestock settings, MRSA has evolved increased biofilm formation as a multifaceted virulence and defensive mechanism, enabling the bacterium to thrive in harsh conditions. This review discusses the molecular mechanisms contributing to biofilm formation across its developmental stages, hence representing a step forward in developing promising strategies for impeding or eradicating biofilms. During staphylococcal biofilm development, cell wall-anchored proteins attach bacterial cells to biotic or abiotic surfaces; extracellular polymeric substances build scaffolds for biofilm formation; the cidABC operon controls cell lysis within the biofilm, and proteases facilitate dispersal. Beside the three main sequential stages of biofilm formation (attachment, maturation, and dispersal), this review unveils two unique developmental stages in the biofilm formation process for MRSA; multiplication and exodus. We also highlighted the quorum sensing as a cell-to-cell communication process, allowing distant bacterial cells to adapt to the conditions surrounding the bacterial biofilm. In S. aureus, the quorum sensing process is mediated by autoinducing peptides (AIPs) as signaling molecules, with the accessory gene regulator system playing a pivotal role in orchestrating the production of AIPs and various virulence factors. Several quorum inhibitors showed promising anti-virulence and antibiofilm effects that vary in type and function according to the targeted molecule. Disrupting the biofilm architecture and eradicating sessile bacterial cells are crucial steps to prevent colonization on other surfaces or organs. In this context, nanoparticles emerge as efficient carriers for delivering antimicrobial and antibiofilm agents throughout the biofilm architecture. Although metal-based nanoparticles have been previously used in combatting biofilms, its non-degradability and toxicity within the human body presents a real challenge. Therefore, organic nanoparticles in conjunction with quorum inhibitors have been proposed as a promising strategy against biofilms. As nanotherapeutics continue to gain recognition as an antibiofilm strategy, the development of more antibiofilm nanotherapeutics could offer a promising solution to combat biofilm-mediated resistance.
Collapse
Affiliation(s)
- Nirmeen Aboelnaga
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Salma W Elsayed
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
- Department of Microbiology & Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Nehal Adel Abdelsalam
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Salma Salem
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Nehal A Saif
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Manar Elsayed
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Shehab Ayman
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Maha Nasr
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mohamed Elhadidy
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt.
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt.
- Department of Bacteriology, Mycology and Immunology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt.
| |
Collapse
|
5
|
Azzam A, Shawky RM, El-Mahdy TS. Sub-inhibitory concentrations of ceftriaxone induce morphological alterations and PIA-independent biofilm formation in Staphylococcus aureus. Braz J Microbiol 2024; 55:297-308. [PMID: 37979131 PMCID: PMC10920565 DOI: 10.1007/s42770-023-01177-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023] Open
Abstract
The exposure of bacteria to sub-inhibitory concentrations of antibiotics is of biological significance since it can occur in vivo under many circumstances, including low-dose treatment, poor adherence to a regimen, poor drug penetration, drug-drug interactions, and antibiotic resistance of the pathogen. In this study, we investigated the effects of subinhibitory concentrations of four antibiotics: ampicillin, ceftriaxone, gentamicin, and norfloxacin, which are commonly used in clinical settings and on cell morphology and biofilm formation in Staphylococcus aureus as one of the leading causes of nosocomial and biofilm-associated infections. Nine clinical S. aureus biofilm-producing isolates and two known biofilm-producing reference strains, S. aureus ATCC 29213 and S. aureus ATCC 6538, were used in this study. Sub-MICs of beta-lactam antibiotics (ampicillin and ceftriaxone) significantly induced biofilm formation in S. aureus ATCC 29213 and S. aureus ATCC 6538 and in six clinical isolates out of the nine selected isolates when compared with the antibiotic-free control group (P < 0.05), with an approximately 2- to 2.5-fold increase. Gentamicin and norfloxacin induced biofilms in S. aureus ATCC 29213 and S. aureus ATCC 6538, while gentamicin and norfloxacin induced biofilms only in three and two of the nine tested isolates, respectively (P < 0.05). The chemical nature of the biofilm matrix produced by half the MIC of ceftriaxone in the six isolates that showed increased biofilm was all non-polysaccharide in composition (PIA-independent). Gene expression of biofilm-encoding genes atl and sarA in biofilms of the two tested strains (S. aureus ATCC 6538) and clinical strain (S. aureus 16) showed a significant upregulation after exposure to half MIC of ceftriaxone. Additionally, the bacterial cell morphological changes in planktonic cells caused by half MIC of ceftriaxone were evaluated by scanning electron microscopy, which demonstrated a significant cell enlargement when compared with the antibiotic-free control (P < 0.05), and some deformed cells were also noticed. In S. aureus clinical isolates, sub-MICs of ampicillin, ceftriaxone, gentamicin, and norfloxacin may stimulate substantial production of biofilm, which could have important clinical significance and make infection treatment challenges. Further, in vivo research is needed to fully comprehend how sub-MIC of antibiotics can affect biofilm formation in clinical settings. Additionally, more research is required to reveal the clinical implications of the morphological alterations in S. aureus brought on by exposure to ceftriaxone at concentrations below its MIC.
Collapse
Affiliation(s)
- Ahmed Azzam
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University, Ain Helwan, Cairo, Egypt
| | - Riham M Shawky
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University, Ain Helwan, Cairo, Egypt
| | - Taghrid S El-Mahdy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University, Ain Helwan, Cairo, Egypt.
- Department of Microbiology and Immunology, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, Egypt.
| |
Collapse
|
6
|
Bouiller K, Jacko NF, Shumaker MJ, Talbot BM, Read TD, David MZ. Factors associated with foreign body infection in methicillin-resistant Staphylococcus aureus bacteremia. Front Immunol 2024; 15:1335867. [PMID: 38433826 PMCID: PMC10904584 DOI: 10.3389/fimmu.2024.1335867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/05/2024] [Indexed: 03/05/2024] Open
Abstract
Background We aimed to compare patient characteristics, MRSA sequence types, and biofilm production of MRSA strains that did and did not cause a foreign body infection in patients with MRSA bloodstream infections (BSI). Methods All adult patients with MRSA BSI hospitalized in two hospitals were identified by clinical microbiology laboratory surveillance. Only patients who had at least one implanted foreign body during the episode of BSI were included. Results In July 2018 - March 2022, of 423 patients identified with MRSA BSI, 118 (28%) had ≥1 foreign body. Among them, 51 (43%) had one or more foreign body infections. In multivariable analysis, factors associated with foreign body infection were history of MRSA infection in the last year (OR=4.7 [1.4-15.5], p=0.012) community-associated BSI (OR=68.1 [4.2-1114.3], p=0.003); surgical site infection as source of infection (OR=11.8 [2-70.4], p=0.007); presence of more than one foreign body (OR=3.4 [1.1-10.7], p=0.033); interval between foreign body implantation and infection <18 months (OR=3.3 [1.1-10], p=0.031); and positive blood culture ≥48h (OR=16.7 [4.3-65.7], p<0.001). The most prevalent sequence type was ST8 (39%), followed by ST5 (29%), and ST105 (20%) with no significant difference between patients with or without foreign body infection. Only 39% of MRSA isolates formed a moderate/strong biofilm. No significant difference was observed between patients with foreign body infection and those without foreign body infection. In multivariable analysis, subjects infected with a MRSA isolate producing moderate/strong in vitro biofilm were more likely to have a history of MRSA infection in the last year (OR=3.41 [1.23-9.43]), interval between foreign body implantation and MRSA BSI <18 months (OR=3.1 [1.05-9.2]) and ST8 (OR=10.64 [2-57.3]). Conclusion Most factors associated with foreign body infection in MRSA BSI were also characteristic of persistent infections. Biofilm-forming isolates were not associated with a higher risk of foreign-body infection but appeared to be associated with MRSA genetic lineage, especially ST8.
Collapse
Affiliation(s)
- Kevin Bouiller
- Division of Infectious Diseases, Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Université de Franche-Comté, CHU Besançon, UMR-CNRS 6249 Chrono-environnement, Department of Infectious and Tropical Diseases, Besançon, France
| | - Natasia F Jacko
- Division of Infectious Diseases, Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Margot J Shumaker
- Division of Infectious Diseases, Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Brooke M Talbot
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Timothy D Read
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Michael Z David
- Division of Infectious Diseases, Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
7
|
McLaughlin M, Fiebig A, Crosson S. XRE transcription factors conserved in Caulobacter and φCbK modulate adhesin development and phage production. PLoS Genet 2023; 19:e1011048. [PMID: 37972151 PMCID: PMC10688885 DOI: 10.1371/journal.pgen.1011048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/30/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023] Open
Abstract
The xenobiotic response element (XRE) family of transcription factors (TFs), which are commonly encoded by bacteria and bacteriophage, regulate diverse features of bacterial cell physiology and impact phage infection dynamics. Through a pangenome analysis of Caulobacter species isolated from soil and aquatic ecosystems, we uncovered an apparent radiation of a paralogous XRE TF gene cluster, several of which have established functions in the regulation of holdfast adhesin development and biofilm formation in C. crescentus. We further discovered related XRE TFs throughout the class Alphaproteobacteria and its phages, including the φCbK Caulophage, suggesting that members of this cluster impact host-phage interactions. Here we show that a closely related group of XRE transcription factors encoded by both C. crescentus and φCbK can physically interact and function to control the transcription of a common gene set, influencing processes including holdfast development and the production of φCbK virions. The φCbK-encoded XRE paralog, tgrL, is highly expressed at the earliest stages of infection and can directly inhibit transcription of host genes including hfiA, a potent holdfast inhibitor, and gafYZ, an activator of prophage-like gene transfer agents (GTAs). XRE proteins encoded from the C. crescentus chromosome also directly repress gafYZ transcription, revealing a functionally redundant set of host regulators that may protect against spurious production of GTA particles and inadvertent cell lysis. Deleting the C. crescentus XRE transcription factors reduced φCbK burst size, while overexpressing these host genes or φCbK tgrL rescued this burst defect. We conclude that this XRE TF gene cluster, shared by C. crescentus and φCbK, plays an important role in adhesion regulation under phage-free conditions, and influences host-phage dynamics during infection.
Collapse
Affiliation(s)
- Maeve McLaughlin
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, United States of America
| | - Aretha Fiebig
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, United States of America
| | - Sean Crosson
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, United States of America
| |
Collapse
|
8
|
Kawasuji H, Ikezawa Y, Morita M, Sugie K, Somekawa M, Ezaki M, Koshiyama Y, Takegoshi Y, Murai Y, Kaneda M, Kimoto K, Nagaoka K, Niimi H, Morinaga Y, Yamamoto Y. High Incidence of Metastatic Infections in Panton-Valentine Leucocidin-Negative, Community-Acquired Methicillin-Resistant Staphylococcus aureus Bacteremia: An 11-Year Retrospective Study in Japan. Antibiotics (Basel) 2023; 12:1516. [PMID: 37887217 PMCID: PMC10604685 DOI: 10.3390/antibiotics12101516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 10/28/2023] Open
Abstract
Panton-Valentine leucocidin (PVL)-negative community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) was originally disseminated in Japan and has since replaced healthcare-associated MRSA (HA-MRSA). However, the clinical characteristics of CA-MRSA bacteremia (CA-MRSAB) compared with those of HA-MRSA bacteremia (HA-MRSAB) are unknown. We aim to clarify differences and investigate associations between the clinical manifestations and virulence genes associated with plasma-biofilm formation in PVL-negative CA-MRSA. From 2011 to 2021, when CA-MRSA dramatically replaced HA-MRSA, 79 MRSA strains were collected from blood cultures and analyzed via SCCmec typing and targeted virulence gene (lukSF-PV, cna, and fnbB) detection. The incidence of metastatic infection was significantly higher in CA-MRSAB than in HA-MRSAB. PVL genes were all negative, although cna and fnbB were positive in 55.6% (20/36) and 50% (18/36) of CA-MRSA strains and 3.7% (1/27) and 7.4% (2/27) of HA-MRSA strains, respectively. cna and fnbB carriage were not associated with the development of metastatic infections in MRSAB; however, the bacteremia duration was significantly longer in CA-MRSAB harboring cna. CA-MRSAB may be more likely to cause metastatic infections than HA-MRSAB. Since CA-MRSA is dominant in Japan, suspected metastatic infection foci should be identified by computed tomography, magnetic resonance imaging, and echocardiography when treating MRSAB.
Collapse
Affiliation(s)
- Hitoshi Kawasuji
- Department of Clinical Infectious Diseases, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Yoshihiro Ikezawa
- Department of Clinical Infectious Diseases, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Mika Morita
- Department of Clinical Laboratory and Molecular Pathology, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Kazushige Sugie
- Department of Clinical Laboratory and Molecular Pathology, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Mayu Somekawa
- Department of Microbiology, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Masayoshi Ezaki
- Department of Clinical Infectious Diseases, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Yuki Koshiyama
- Department of Clinical Infectious Diseases, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Yusuke Takegoshi
- Department of Clinical Infectious Diseases, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Yushi Murai
- Department of Clinical Infectious Diseases, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Makito Kaneda
- Department of Clinical Infectious Diseases, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Kou Kimoto
- Department of Clinical Infectious Diseases, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Kentaro Nagaoka
- Department of Clinical Infectious Diseases, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Hideki Niimi
- Department of Clinical Laboratory and Molecular Pathology, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Yoshitomo Morinaga
- Department of Microbiology, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| | - Yoshihiro Yamamoto
- Department of Clinical Infectious Diseases, Toyama University Graduate School of Medicine and Pharmaceutical Sciences, Toyama 930-0194, Japan
| |
Collapse
|
9
|
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
|
10
|
McLaughlin M, Fiebig A, Crosson S. XRE Transcription Factors Conserved in Caulobacter and φCbK Modulate Adhesin Development and Phage Production. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.20.554034. [PMID: 37645952 PMCID: PMC10462132 DOI: 10.1101/2023.08.20.554034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Upon infection, transcriptional shifts in both a host bacterium and its invading phage determine host and viral fitness. The xenobiotic response element (XRE) family of transcription factors (TFs), which are commonly encoded by bacteria and phages, regulate diverse features of bacterial cell physiology and impact phage infection dynamics. Through a pangenome analysis of Caulobacter species isolated from soil and aquatic ecosystems, we uncovered an apparent radiation of a paralogous XRE TF gene cluster, several of which have established functions in the regulation of holdfast adhesin development and biofilm formation in C. crescentus. We further discovered related XRE TFs across the class Alphaproteobacteria and its phages, including the φCbK Caulophage, suggesting that members of this gene cluster impact host-phage interactions. Here we show that that a closely related group of XRE proteins, encoded by both C. crescentus and φCbK, can form heteromeric associations and control the transcription of a common gene set, influencing processes including holdfast development and the production of φCbK virions. The φCbK XRE paralog, tgrL, is highly expressed at the earliest stages of infection and can directly repress transcription of hfiA, a potent holdfast inhibitor, and gafYZ, a transcriptional activator of prophage-like gene transfer agents (GTAs) encoded on the C. crescentus chromosome. XRE proteins encoded from the C. crescentus chromosome also directly repress gafYZ transcription, revealing a functionally redundant set of host regulators that may protect against spurious production of GTA particles and inadvertent cell lysis. Deleting host XRE transcription factors reduced φCbK burst size, while overexpressing these genes or φCbK tgrL rescued this burst defect. We conclude that an XRE TF gene cluster, shared by C. crescentus and φCbK, plays an important role in adhesion regulation under phage-free conditions, and influences host-phage dynamics during infection.
Collapse
Affiliation(s)
- Maeve McLaughlin
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Aretha Fiebig
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Sean Crosson
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
11
|
Oliveira AMD, Anjos Szczerepa MMD, Bronharo Tognim MC, Abreu Filho BAD, Cardozo-Filho L, Gomes RG, Bergamasco R. Moringa oleifera seed oil extracted by pressurized n-propane and its effect against Staphylococcus aureus biofilms. ENVIRONMENTAL TECHNOLOGY 2023; 44:1083-1098. [PMID: 34704544 DOI: 10.1080/09593330.2021.1994653] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 10/06/2021] [Indexed: 06/13/2023]
Abstract
Staphylococcus aureus is often associated worldwide with foodborne illnesses, and the elimination of biofilms formed by this bacterium from industrial surfaces is very challenging. To date, there have been few attempts to investigate plant oils obtained by recent green technologies, applied against biofilms on usual surfaces of the food industry and bacteria isolated from such environment. Therefore, this study evaluated the activity of Moringa oleifera seed oil (MOSO), extracted with pressurized n-propane, against standard and environmental S. aureus biofilms. Additionally, a genotypic and phenotypic study of the environmental S. aureus was proposed. It was found that this bacterium was a MSSA (methicillin-sensitive S. aureus), a carrier of icaA and icaD genes that has strong adhesion (OD550=1.86 ± 0.19) during biofilm formation. The use of pressurized n-propane as a solvent was efficient in obtaining MOSO, achieving a yield of 60.9%. Gas chromatography analyses revealed the presence of a rich source of fatty acids in MOSO, mainly oleic acid (62.47%), behenic acid (10.5%) and palmitic acid (7.32%). On polystyrene surface, MOSO at 0.5% and 1% showed inhibitory and bactericidal activity, respectively, against S. aureus biofilms. MOSO at 1% allowed a maximum reduction of 2.38 log UFC/cm² of S. aureus biofilms formed on PVC (polyvinyl chloride) surface. Scanning electron microscopy showed disturbances on the surface of S. aureus after exposure to MOSO. These unprecedented findings suggest that MOSO extracted with pressurized n-propane is potentially capable of inhibiting biofilms of different S. aureus strains, thus, contributing to microbiological safety during food processing.
Collapse
Affiliation(s)
| | | | | | | | - Lúcio Cardozo-Filho
- Department of Chemical Engineering, State University of Maringa, Maringa, Brazil
| | | | - Rosângela Bergamasco
- Department of Chemical Engineering, State University of Maringa, Maringa, Brazil
| |
Collapse
|
12
|
Costa MDOCE, do Nascimento APB, Martins YC, dos Santos MT, Figueiredo AMDS, Perez-Rueda E, Nicolás MF. The gene regulatory network of Staphylococcus aureus ST239-SCC mecIII strain Bmb9393 and assessment of genes associated with the biofilm in diverse backgrounds. Front Microbiol 2023; 13:1049819. [PMID: 36704545 PMCID: PMC9871828 DOI: 10.3389/fmicb.2022.1049819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Introduction Staphylococcus aureus is one of the most prevalent and relevant pathogens responsible for a wide spectrum of hospital-associated or community-acquired infections. In addition, methicillin-resistant Staphylococcus aureus may display multidrug resistance profiles that complicate treatment and increase the mortality rate. The ability to produce biofilm, particularly in device-associated infections, promotes chronic and potentially more severe infections originating from the primary site. Understanding the complex mechanisms involved in planktonic and biofilm growth is critical to identifying regulatory connections and ways to overcome the global health problem of multidrug-resistant bacteria. Methods In this work, we apply literature-based and comparative genomics approaches to reconstruct the gene regulatory network of the high biofilm-producing strain Bmb9393, belonging to one of the highly disseminating successful clones, the Brazilian epidemic clone. To the best of our knowledge, we describe for the first time the topological properties and network motifs for the Staphylococcus aureus pathogen. We performed this analysis using the ST239-SCCmecIII Bmb9393 strain. In addition, we analyzed transcriptomes available in the literature to construct a set of genes differentially expressed in the biofilm, covering different stages of the biofilms and genetic backgrounds of the strains. Results and discussion The Bmb9393 gene regulatory network comprises 1,803 regulatory interactions between 64 transcription factors and the non-redundant set of 1,151 target genes with the inclusion of 19 new regulons compared to the N315 transcriptional regulatory network published in 2011. In the Bmb9393 network, we found 54 feed-forward loop motifs, where the most prevalent were coherent type 2 and incoherent type 2. The non-redundant set of differentially expressed genes in the biofilm consisted of 1,794 genes with functional categories relevant for adaptation to the variable microenvironments established throughout the biofilm formation process. Finally, we mapped the set of genes with altered expression in the biofilm in the Bmb9393 gene regulatory network to depict how different growth modes can alter the regulatory systems. The data revealed 45 transcription factors and 876 shared target genes. Thus, the gene regulatory network model provided represents the most up-to-date model for Staphylococcus aureus, and the set of genes altered in the biofilm provides a global view of their influence on biofilm formation from distinct experimental perspectives and different strain backgrounds.
Collapse
Affiliation(s)
| | - Ana Paula Barbosa do Nascimento
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | | | | | - Agnes Marie de Sá Figueiredo
- Instituto de Investigaciones en Matemáticas Aplicadas y en Sistemas, Universidad Nacional Autónoma de México, Unidad Académica Yucatán, Merida, Mexico
| | - Ernesto Perez-Rueda
- Laboratório de Biologia Molecular de Bactérias, Instituto de Microbiologia Paulo de Goés, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil,*Correspondence: Ernesto Perez-Rueda ✉
| | - Marisa Fabiana Nicolás
- Laboratório Nacional de Computação Científica (LNCC), Petrópolis, Brazil,Marisa Fabiana Nicolás ✉
| |
Collapse
|
13
|
McLaughlin M, Hershey DM, Reyes Ruiz LM, Fiebig A, Crosson S. A cryptic transcription factor regulates Caulobacter adhesin development. PLoS Genet 2022; 18:e1010481. [PMID: 36315598 PMCID: PMC9648850 DOI: 10.1371/journal.pgen.1010481] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 11/10/2022] [Accepted: 10/18/2022] [Indexed: 11/07/2022] Open
Abstract
Alphaproteobacteria commonly produce an adhesin that is anchored to the exterior of the envelope at one cell pole. In Caulobacter crescentus this adhesin, known as the holdfast, facilitates attachment to solid surfaces and cell partitioning to air-liquid interfaces. An ensemble of two-component signal transduction (TCS) proteins controls C. crescentus holdfast biogenesis by indirectly regulating expression of HfiA, a potent inhibitor of holdfast synthesis. We performed a genetic selection to discover direct hfiA regulators that function downstream of the adhesion TCS system and identified rtrC, a hypothetical gene. rtrC transcription is directly activated by the adhesion TCS regulator, SpdR. Though its primary structure bears no resemblance to any defined protein family, RtrC binds and regulates dozens of sites on the C. crescentus chromosome via a pseudo-palindromic sequence. Among these binding sites is the hfiA promoter, where RtrC functions to directly repress transcription and thereby activate holdfast development. Either RtrC or SpdR can directly activate transcription of a second hfiA repressor, rtrB. Thus, environmental regulation of hfiA transcription by the adhesion TCS system is subject to control by an OR-gated type I coherent feedforward loop; these regulatory motifs are known to buffer gene expression against fluctuations in regulating signals. We have further assessed the functional role of rtrC in holdfast-dependent processes, including surface adherence to a cellulosic substrate and formation of pellicle biofilms at air-liquid interfaces. Strains harboring insertional mutations in rtrC have a diminished adhesion profile in a competitive cheesecloth binding assay and a reduced capacity to colonize pellicle biofilms in select media conditions. Our results add to an emerging understanding of the regulatory topology and molecular components of a complex bacterial cell adhesion control system.
Collapse
Affiliation(s)
- Maeve McLaughlin
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, United States of America
| | - David M. Hershey
- Department of Bacteriology, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Leila M. Reyes Ruiz
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Aretha Fiebig
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, United States of America
| | - Sean Crosson
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, United States of America
| |
Collapse
|
14
|
Miao F, Tai Z, Wang Y, Zhu Q, Fang JKH, Hu M. Tachyplesin I Analogue Peptide as an Effective Antimicrobial Agent against Candida albicans- Staphylococcus aureus Poly-Biofilm Formation and Mixed Infection. ACS Infect Dis 2022; 8:1839-1850. [PMID: 35998684 DOI: 10.1021/acsinfecdis.2c00080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Microbial biofilms are difficult to tackle in many infectious diseases. Candida albicans and Staphylococcus aureus are prevalent symbiotic strains in polymicrobial biofilms, which showed enhanced antimicrobial resistance and made identifying effective treatment techniques more difficult. The antibiofilm abilities of tachplesin I analogue peptide (TP11A) and tachplesin I were investigated quantitatively in this study. Both inhibited C. albicans monomicrobial, S. aureus monomicrobial, and C. albicans-S. aureus polymicrobial biofilms quite well. TP11A suppressed the biofilm- and virulence-related genes of C. albicans (hwp 1) and S. aureus (ica A, fnb B, agr A, hla, nor A, and sig B) in the mixed biofilm, according to quantitative reverse transcription polymerase chain reaction analysis. We created an injectable thermosensitive in situ PLEL@TP11A gel system by simply adding TP11A into poly(d,l-lactide)-poly(ethylene glycol)-poly(d,l-lactide) (PLEL). Using C. albicans-S. aureus mixed infected wound models of mice, the in vivo therapeutic effect of TP11A and PLEL@TP11A in polymicrobial infections was investigated. The findings revealed that TP11A and PLEL@TP11A could efficiently reduce bacterial and fungal burden in wound infections, as well as accelerated wound healing. Based on above findings, TP11A might be an effective antimicrobial against C. albicans-S. aureus poly-biofilm formation and mixed infection.
Collapse
Affiliation(s)
- Fengze Miao
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai 201306, China.,Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai 201306, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China.,Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai 200443, China
| | - Youji Wang
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai 201306, China.,Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai 201306, China
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China.,Shanghai Engineering Research Center for Topical Chinese Medicine, Shanghai 200443, China
| | - James Kar-Hei Fang
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon 999077, China
| | - Menghong Hu
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai 201306, China.,Shanghai Collaborative Innovation for Aquatic Animal Genetics and Breeding, Shanghai Ocean University, Shanghai 201306, China
| |
Collapse
|
15
|
Liu L, Liu B, Li L, He MX, Zhou XD, Li Q. Myrtenol Inhibits Biofilm Formation and Virulence in the Drug-Resistant Acinetobacter baumannii: Insights into the Molecular Mechanisms. Infect Drug Resist 2022; 15:5137-5148. [PMID: 36082242 PMCID: PMC9447999 DOI: 10.2147/idr.s379212] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/17/2022] [Indexed: 12/04/2022] Open
Abstract
Background blaNDM-1-producing Acinetobacter baumannii (BP-AB) remains a critical problem in nosocomial infections, because of its resistance mediated by the biofilm formation and virulence factors. No studies have confirmed myrtenol’s efficacy in inhibiting the biofilm formation and virulence associated with biofilm of BP-AB. Methods The tested concentrations of myrtenol were wild type (A), 50 μg/mL (B), 100 μg/mL (C), 200 μg/mL (D), 250 μg/mL (E), and 300 μg/mL (F). Results The BP-AB biofilm inhibition was significantly higher in the D, E, and F groups than in the A, B, and C groups. Myrtenol significantly reduced the air-liquid interface ring formation in glass tubes. It also effectively inhibited the attachment of BP-AB strains on polystyrene surfaces as shown by crystal violet staining. Microscopy showed a significant reduction in biofilm formation with dispersed BP-AB strains. The confocal laser scanning microscopy analysis showed a significant reduction in the biofilm’s biomass, covered surface area, and thickness. The scanning electron microscopy analysis revealed significantly fewer BP-AB aggregates on the coverslip surface. In the CompStat analysis, the biofilm’s biomass, maximum thickness, and surface-to-volume ratio were significantly reduced. The qPCR analysis revealed a significant down-regulation of bfmR, bap, csuA/B, and ompA expression, which positively correlated with the biofilm’s biomass, maximum thickness, and surface-to-volume ratio in BP-AB strains. Myrtenol significantly improved the susceptibility of BP-AB to the antibiotics amikacin, piperacillin/tazobactam, cefoperazone/sulbactam, and ceftazidime. Conclusion Myrtenol attenuates the BP-AB biofilm formation and virulence by suppressing the expression of bfmR, bap, csuA/B, and ompA.
Collapse
Affiliation(s)
- Lei Liu
- Department of Respiratory Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, People’s Republic of China
- NHC Key Laboratory of Tropical Disease Control, Hainan Medical University, Haikou, Hainan, People’s Republic of China
| | - Bin Liu
- Department of Respiratory Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, People’s Republic of China
- NHC Key Laboratory of Tropical Disease Control, Hainan Medical University, Haikou, Hainan, People’s Republic of China
| | - Liang Li
- Department of Respiratory Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, People’s Republic of China
- NHC Key Laboratory of Tropical Disease Control, Hainan Medical University, Haikou, Hainan, People’s Republic of China
| | - Ming-Xin He
- NHC Key Laboratory of Tropical Disease Control, Hainan Medical University, Haikou, Hainan, People’s Republic of China
| | - Xiang-Dong Zhou
- Department of Respiratory Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, People’s Republic of China
- NHC Key Laboratory of Tropical Disease Control, Hainan Medical University, Haikou, Hainan, People’s Republic of China
| | - Qi Li
- Department of Respiratory Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, People’s Republic of China
- NHC Key Laboratory of Tropical Disease Control, Hainan Medical University, Haikou, Hainan, People’s Republic of China
- Correspondence: Qi Li, Email
| |
Collapse
|
16
|
Yi L, Jin M, Gao M, Wang H, Fan Q, Grenier D, Sun L, Wang S, Wang Y. Specific quantitative detection of Streptococcus suis and Actinobacillus pleuropneumoniae in co-infection and mixed biofilms. Front Cell Infect Microbiol 2022; 12:898412. [PMID: 35992166 PMCID: PMC9381733 DOI: 10.3389/fcimb.2022.898412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Respiratory infections seriously affect the swine industry worldwide. Co-infections of two vital pathogenic bacteria Streptococcus suis (S. suis) and Actinobacillus pleuropneumoniae (A. pleuropneumoniae), colonizing the respiratory tract often occurs in veterinary clinical practice. Moreover, our previous research found that S. suis and A. pleuropneumoniae can form biofilm in vitro. The formation of a mixed biofilm not only causes persistent infections, but also increases the multiple drug resistance of bacteria, which brings difficulties to disease prevention and control. However, the methods for detecting S. suis and A. pleuropneumoniae in co-infection and biofilm are immature. Therefore, in this study, primers and probes were designed based on the conservative sequence of S. suis gdh gene and A. pleuropneumoniae apxIVA gene. Then, a TaqMan duplex real-time PCR method for simultaneous detection of S. suis and A. pleuropneumoniae was successfully established via optimizing the reaction system and conditions. The specificity analysis results showed that this TaqMan real-time PCR method had strong specificity and high reliability. The sensitivity test results showed that the minimum detection concentration of S. suis and A. pleuropneumoniae recombinant plasmid was 10 copies/μL, which is 100 times more sensitive than conventional PCR methods. The amplification efficiencies of S. suis and A. pleuropneumoniae were 95.9% and 104.4% with R2 value greater than 0.995, respectively. The slopes of the calibration curves of absolute cell abundance of S. suis and A. pleuropneumoniae were 1.02 and 1.09, respectively. The assays were applied to cultivated mixed biofilms and approximately 108 CFUs per biofilm were quantified when 108 CFUs planktonic bacteria of either S. suis or A. pleuropneumoniae were added to biofilms. In summary, this study developed a TaqMan real-time PCR assay for specific, accurate quantification of S. suis or A. pleuropneumoniae in mixed biofilms, which may help for the detection, prevention and control of diseases caused by a bacterial mixed infection involving S. suis and A. pleuropneumoniae.
Collapse
Affiliation(s)
- Li Yi
- College of Life Science, Luoyang Normal University, Luoyang, China
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Manyu Jin
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Mengxia Gao
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Haikun Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Qingying Fan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Daniel Grenier
- Groupe de Recherche en Écologie Buccale (GREB), Faculté de Médecine Dentaire, Université Laval, Quebec City, QC, Canada
| | - Liyun Sun
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Shaohui Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- *Correspondence: Yang Wang, ; Shaohui Wang,
| | - Yang Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- *Correspondence: Yang Wang, ; Shaohui Wang,
| |
Collapse
|
17
|
Functionalized Self-Assembled Monolayers: Versatile Strategies to Combat Bacterial Biofilm Formation. Pharmaceutics 2022; 14:pharmaceutics14081613. [PMID: 36015238 PMCID: PMC9415113 DOI: 10.3390/pharmaceutics14081613] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 11/16/2022] Open
Abstract
Bacterial infections due to biofilms account for up to 80% of bacterial infections in humans. With the increased use of antibiotic treatments, indwelling medical devices, disinfectants, and longer hospital stays, antibiotic resistant infections are sharply increasing. Annual deaths are predicted to outpace cancer and diabetes combined by 2050. In the past two decades, both chemical and physical strategies have arisen to combat biofilm formation on surfaces. One such promising chemical strategy is the formation of a self-assembled monolayer (SAM), due to its small layer thickness, strong covalent bonds, typically facile synthesis, and versatility. With the goal of combating biofilm formation, the SAM could be used to tether an antibacterial agent such as a small-molecule antibiotic, nanoparticle, peptide, or polymer to the surface, and limit the agent’s release into its environment. This review focuses on the use of SAMs to inhibit biofilm formation, both on their own and by covalent grafting of a biocidal agent, with the potential to be used in indwelling medical devices. We conclude with our perspectives on ongoing challenges and future directions for this field.
Collapse
|
18
|
Clearance of mixed biofilms of Streptococcus pneumoniae and methicillin-susceptible/resistant Staphylococcus aureus by antioxidants N-acetyl-L-cysteine and cysteamine. Sci Rep 2022; 12:6668. [PMID: 35461321 PMCID: PMC9035182 DOI: 10.1038/s41598-022-10609-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 04/07/2022] [Indexed: 12/01/2022] Open
Abstract
Biofilm-associated infections are of great concern because they are associated with antibiotic resistance and immune evasion. Co-colonization by Staphylococcus aureus and Streptococcus pneumoniae is possible and a threat in clinical practice. We investigated the interaction between S. aureus and S. pneumoniae in mixed biofilms and tested new antibiofilm therapies with antioxidants N-acetyl-l-cysteine (NAC) and cysteamine (Cys). We developed two in vitro S. aureus–S. pneumoniae mixed biofilms in 96-well polystyrene microtiter plates and we treated in vitro biofilms with Cys and NAC analyzing their effect by CV staining and viable plate counting. S. pneumoniae needed a higher proportion of cells in the inoculum and planktonic culture to reach a similar population rate in the mixed biofilm. We demonstrated the effect of Cys in preventing S. aureus biofilms and S. aureus–S. pneumoniae mixed biofilms. Moreover, administration of 5 mg/ml of NAC nearly eradicated the S. pneumoniae population and killed nearly 94% of MSSA cells and 99% of MRSA cells in the mixed biofilms. The methicillin resistance background did not change the antioxidants effect in S. aureus. These results identify NAC and Cys as promising repurposed drug candidates for the prevention and treatment of mixed biofilms by S. pneumoniae and S. aureus.
Collapse
|
19
|
Caldara M, Belgiovine C, Secchi E, Rusconi R. Environmental, Microbiological, and Immunological Features of Bacterial Biofilms Associated with Implanted Medical Devices. Clin Microbiol Rev 2022; 35:e0022120. [PMID: 35044203 PMCID: PMC8768833 DOI: 10.1128/cmr.00221-20] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The spread of biofilms on medical implants represents one of the principal triggers of persistent and chronic infections in clinical settings, and it has been the subject of many studies in the past few years, with most of them focused on prosthetic joint infections. We review here recent works on biofilm formation and microbial colonization on a large variety of indwelling devices, ranging from heart valves and pacemakers to urological and breast implants and from biliary stents and endoscopic tubes to contact lenses and neurosurgical implants. We focus on bacterial abundance and distribution across different devices and body sites and on the role of environmental features, such as the presence of fluid flow and properties of the implant surface, as well as on the interplay between bacterial colonization and the response of the human immune system.
Collapse
Affiliation(s)
- Marina Caldara
- Interdepartmental Center on Safety, Technologies, and Agri-food Innovation (SITEIA.PARMA), University of Parma, Parma, Italy
| | - Cristina Belgiovine
- IRCCS Humanitas Research Hospital, Rozzano–Milan, Italy
- Scuola di Specializzazione in Microbiologia e Virologia, Università degli Studi di Pavia, Pavia, Italy
| | - Eleonora Secchi
- Institute of Environmental Engineering, ETH Zürich, Zürich, Switzerland
| | - Roberto Rusconi
- IRCCS Humanitas Research Hospital, Rozzano–Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele–Milan, Italy
| |
Collapse
|
20
|
Gao K, Su B, Dai J, Li P, Wang R, Yang X. Anti-Biofilm and Anti-Hemolysis Activities of 10-Hydroxy-2-decenoic Acid against Staphylococcus aureus. Molecules 2022; 27:1485. [PMID: 35268586 PMCID: PMC8912057 DOI: 10.3390/molecules27051485] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 12/03/2022] Open
Abstract
Persistent infections caused by Staphylococcus aureus biofilms pose a major threat to global public health. 10-Hydroxy-2-decenoic acid (10-HDA), a main fatty acid in royal jelly, has been shown to possess various biological activities. The purpose of this study was to explore the effects of 10-HDA on the biofilms and virulence of S. aureus and its potential molecular mechanism. Quantitative crystal violet staining indicated that 10-HDA significantly reduced the biofilm biomass at sub-minimum inhibitory concentration (MIC) levels (1/32MIC to 1/2MIC). Scanning electron microscope (SEM) observations demonstrated that 10-HDA inhibited the secretion of extracellular polymeric substances, decreased bacterial adhesion and aggregation, and disrupted biofilm architecture. Moreover, 10-HDA could significantly decrease the biofilm viability and effectively eradicated the mature biofilms. It was also found that the hemolytic activity of S. aureus was significantly inhibited by 10-HDA. qRT-PCR analyses revealed that the expressions of global regulators sarA, agrA, and α-hemolysin gene hla were downregulated by 10-HDA. These results indicate that 10-HDA could be used as a potential natural antimicrobial agent to control the biofilm formation and virulence of S. aureus.
Collapse
Affiliation(s)
- Kuankuan Gao
- State Key Laboratory of Biobased Material and Green Papermaking (LBMP), Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China; (K.G.); (B.S.); (P.L.); (R.W.)
- Key Laboratory of Shandong Microbial Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China;
| | - Bei Su
- State Key Laboratory of Biobased Material and Green Papermaking (LBMP), Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China; (K.G.); (B.S.); (P.L.); (R.W.)
- Key Laboratory of Shandong Microbial Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China;
| | - Jing Dai
- Key Laboratory of Shandong Microbial Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China;
| | - Piwu Li
- State Key Laboratory of Biobased Material and Green Papermaking (LBMP), Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China; (K.G.); (B.S.); (P.L.); (R.W.)
- Key Laboratory of Shandong Microbial Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China;
| | - Ruiming Wang
- State Key Laboratory of Biobased Material and Green Papermaking (LBMP), Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China; (K.G.); (B.S.); (P.L.); (R.W.)
- Key Laboratory of Shandong Microbial Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China;
| | - Xiaohui Yang
- State Key Laboratory of Biobased Material and Green Papermaking (LBMP), Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China; (K.G.); (B.S.); (P.L.); (R.W.)
- Key Laboratory of Shandong Microbial Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China;
| |
Collapse
|
21
|
Characterization of the Inclusion Complexes of Isothiocyanates with γ-Cyclodextrin for Improvement of Antibacterial Activities against Staphylococcus aureus. Foods 2021; 11:foods11010060. [PMID: 35010184 PMCID: PMC8750663 DOI: 10.3390/foods11010060] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/15/2021] [Accepted: 12/23/2021] [Indexed: 12/23/2022] Open
Abstract
The aim of this study was to develop inclusions formed by γ-cyclodextrin (γ-CD) and three isothiocyanates (ITCs), including benzyl isothiocyanate (BITC), phenethyl isothiocyanate (PEITC), and 3-methylthiopropyl isothiocyanate (MTPITC) to improve their controlled release for the inhibition of Staphylococcus aureus (S. aureus). These inclusion complexes were characterized using X-ray diffraction, Fourier-transform infrared, thermogravimetry, and scanning electron microscopy (SEM), providing appropriate evidence to confirm the formation of inclusion complexes. Preliminary evaluation of the antimicrobial activity of the different inclusion complexes, carried out in vitro by agar diffusion, showed that such activity lasted 5–7 days longer in γ-CD-BITC, in comparison with γ-CD-PEITC and γ-CD-MTPITC. The biofilm formation was less in S. aureus treated with γ-CD-BITC than that of BITC by using crystal violet quantification assay and SEM. The expression of virulence genes, including sarA, agr, cp5D, cp8F, clf, nuc, and spa, showed sustained downregulation in S. aureus treated with γ-CD-BITC for 24 h by quantitative real-time polymerase chain reaction (qRT-PCR). Moreover, the growth of S. aureus in cooked chicken breast treated with γ-CD-BITC and BITC was predicted by the Gompertz model. The lag time of γ-CD-BITC was 1.3–2.4 times longer than that of BITC, and correlation coefficient (R2) of the secondary models was 0.94–0.99, respectively. These results suggest that BITC has a more durable antibacterial effect against S. aureus after encapsulation by γ-CD.
Collapse
|
22
|
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: 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: 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
|
23
|
Evaluation of immune effect of Streptococcus suis biofilm-associated protein PDH. Vet Microbiol 2021; 263:109270. [PMID: 34749282 DOI: 10.1016/j.vetmic.2021.109270] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 10/29/2021] [Indexed: 11/23/2022]
Abstract
As a zoonotic pathogen, Streptococcus suis(S. suis) takes pigs as the main host and is mainly colonizes in the upper respiratory tract and tonsil of pigs, causing septicemia, endocarditis and meningitis in pigs. Pyruvate dehydrogenase (PDH) is an enzyme that catalyzes the conversion of pyruvate to acetyl-CoA. As an immunogenic membrane-associated protein in S. suis, it has been found to be closely related to the formation of biofilm. In this study, the recombinant PDH (rPDH) of S. suis ZY05719 (serotype 2) was expressed and purified in E. coli by His affinity chromatography. Western blotting analysis showed that there was a strong specific reaction between PDH protein and PDH antiserum. Mice were immunized with recombinant PDH and inactivated bacteria, and the relative survival rates were 70 % and 60 %, respectively. In addition, mice immunized with PDH caused high levels of antibodies and high expression of immune-related genes in the spleen, which significantly protected the liver, brain and spleen from pathological damage. In addition, PDH antiserum could significantly inhibit the growth of S. suis and the formation of S. suis biofilm in vitro. These results further suggest that PDH is a promising candidate for S. suis biofilm-related subunit vaccine.
Collapse
|
24
|
Wang J, Zhu J, Meng J, Qiu T, Wang W, Wang R, Liu J. Baicalin inhibits biofilm formation by influencing primary adhesion and aggregation phases in Staphylococcus saprophyticus. Vet Microbiol 2021; 262:109242. [PMID: 34562786 DOI: 10.1016/j.vetmic.2021.109242] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/19/2021] [Indexed: 12/20/2022]
Abstract
The ability to form biofilms on surfaces makes Staphylococcus saprophyticus (S. saprophyticus) becomes the main pathogenic factor in nosocomial infections. Previously, we demonstrated that baicalin (Bac) inhibited azithromycin-resistant S. saprophyticus (ARSS) biofilm formation. This investigation aims to explore the influence of baicalin on primary adhesion and aggregation phases of biofilm formation, and the treatment effect of baicalin and azithromycin on ARSS biofilm-associated infection. Crystal violet (CV) staining and scanning electron microscope (SEM) observations clearly showed that sub-inhibitory concentration baicalin inhibited ARSS biofilm formation when baicalin was added before the adhesion and aggregation phases. Baicalin significantly increased the relative adhesion inhibition rate and decreased the rate of bacteria aggregation in a dose-dependent manner. Moreover, CLSM and cell lysis assays revealed that baicalin inhibited the production of surface proteins and cell autolysis in bacteria adhesion and aggregation phases of biofilm formation. Meanwhile, the relative expressions of adhesion-related and autolysis-related genes were down-regulated by baicalin. In vivo, the combination of baicalin and azithromycin succeeded in eradicating ARSS from the mouse cutaneous infection model and decreasing the pathological injuries, the expressions of cytokines in infected tissue, and the number of inflammatory cells in the blood. Simultaneously, baicalin decreased the bacterial burdens in tubes, the level of TNF-α, and the number of monocytes and neutrophils compared with that of the SS and azithromycin groups. Based on these results, baicalin inhibited the adhesion and aggregation phases of biofilm formation by influenced the production of surface proteins and cell autolysis. Baicalin and azithromycin synergetically treated ARSS biofilm-associated infection.
Collapse
Affiliation(s)
- Jinli Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Traditional Chinese Veterinary Medicine Research Center, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Jinyue Zhu
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Traditional Chinese Veterinary Medicine Research Center, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Jinwu Meng
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Traditional Chinese Veterinary Medicine Research Center, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Tianxin Qiu
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Traditional Chinese Veterinary Medicine Research Center, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Wenjia Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Traditional Chinese Veterinary Medicine Research Center, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Rui Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Traditional Chinese Veterinary Medicine Research Center, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Jiaguo Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Traditional Chinese Veterinary Medicine Research Center, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
25
|
Sapra R, Rajora AK, Kumar P, Maurya GP, Pant N, Haridas V. Chemical Biology of Sortase A Inhibition: A Gateway to Anti-infective Therapeutic Agents. J Med Chem 2021; 64:13097-13130. [PMID: 34516107 DOI: 10.1021/acs.jmedchem.1c00386] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Staphylococcus aureus is the leading cause of hospital-acquired infections. The enzyme sortase A, present on the cell surface of S. aureus, plays a key role in bacterial virulence without affecting the bacterial viability. Inhibition of sortase A activity offers a powerful but clinically less explored therapeutic strategy, as it offers the possibility of not inducing any selective pressure on the bacteria to evolve drug-resistant strains. In this Perspective, we offer a chemical space narrative for the design of sortase A inhibitors, as delineated into three broad domains: peptidomimetics, natural products, and synthetic small molecules. This provides immense opportunities for medicinal chemists to alleviate the ever-growing crisis of antibiotic resistance.
Collapse
Affiliation(s)
- Rachit Sapra
- Department of Chemistry, Indian Institute of Technology Delhi, New Delhi-110016, India
| | - Amit K Rajora
- Department of Chemistry, Indian Institute of Technology Delhi, New Delhi-110016, India
| | - Pushpendra Kumar
- Department of Chemistry, Indian Institute of Technology Delhi, New Delhi-110016, India
| | - Govind P Maurya
- Department of Chemistry, Indian Institute of Technology Delhi, New Delhi-110016, India
| | - Nalin Pant
- Department of Chemistry, Indian Institute of Technology Delhi, New Delhi-110016, India
| | - V Haridas
- Department of Chemistry, Indian Institute of Technology Delhi, New Delhi-110016, India
| |
Collapse
|
26
|
Karimaei S, Kazem Aghamir SM, Foroushani AR, Pourmand MR. Antibiotic tolerance in biofilm persister cells of Staphylococcus aureus and expression of toxin-antitoxin system genes. Microb Pathog 2021; 159:105126. [PMID: 34384900 DOI: 10.1016/j.micpath.2021.105126] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/04/2021] [Accepted: 08/05/2021] [Indexed: 11/17/2022]
Abstract
The ability of Staphylococcus aureus to form biofilm and persister cells is the main cause of recurrent infections. This study aimed to evaluate the expression of toxin-antitoxin (TA) systems in persister cells within S. aureus biofilms. Time-dependent variation in the persister population present in biofilms of S. aureus was examined after treatment with bactericidal antibiotics. Then, the relative expression level of type II TA system (mazF, relE1, and relE2), type I TA system (sprG), and clpP protease genes in S. aureus strains were assessed by Real _Time PCR. Among the sixteen isolates, two isolates were found to be the strongest biofilm producers. The established biofilm of these isolates showed a comparable biphasic pattern at the lethal dose of the antibiotics. The expression level of TA system genes was increased and strain-specific expression patterns were observed under antibiotics stress conditions. Persisters within a biofilm may establish a reservoir for relapsing infection and could contribute to treatment failures. Hence, the possible role of the TA systems should be considered in biofilm and persister cell formation.
Collapse
Affiliation(s)
- Samira Karimaei
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Abbas Rahimi Foroushani
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Pourmand
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
27
|
Diepoltová A, Konečná K, Janďourek O, Nachtigal P. Study of the impact of cultivation conditions and peg surface modification on the in vitro biofilm formation of Staphylococcus aureus and Staphylococcus epidermidis in a system analogous to the Calgary biofilm device. J Med Microbiol 2021; 70. [PMID: 34048334 DOI: 10.1099/jmm.0.001371] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Introduction. Staphylococcus aureus (SA) and Staphylococcus epidermidis (SE) are the most common pathogens from the genus Staphylococcus causing biofilm-associated infections. Generally, biofilm-associated infections represent a clinical challenge. Bacteria in biofilms are difficult to eradicate due to their resistance and serve as a reservoir for recurring persistent infections.Gap Statement. A variety of protocols for in vitro drug activity testing against staphylococcal biofilms have been introduced. However, there are often fundamental differences. All these differences in methodical approaches can then be reflected in the form of discrepancies between results.Aim. In this study, we aimed to develop optimal conditions for staphylococcal biofilm formation on pegs. The impact of peg surface modification was also studied.Methodology. The impact of tryptic soy broth alone or supplemented with foetal bovine serum (FBS) or human plasma (HP), together with the impact of the inoculum density of bacterial suspensions and the shaking versus the static mode of cultivation, on total biofilm biomass production in SA and SE reference strains was studied. The surface of pegs was modified with FBS, HP, or poly-l-lysine (PLL). The impact on total biofilm biomass was evaluated using the crystal violet staining method and statistical data analysis.Results. Tryptic soy broth supplemented with HP together with the shaking mode led to crucial potentiation of biofilm formation on pegs in SA strains. The SE strain did not produce biofilm biomass under the same conditions on pegs. Preconditioning of peg surfaces with FBS and HP led to a statistically significant increase in biofilm biomass formation in the SE strain.Conclusion. Optimal cultivation conditions for robust staphylococcal biofilm formation in vitro might differ among different bacterial strains and methodical approaches. The shaking mode and supplementation of cultivation medium with HP was beneficial for biofilm formation on pegs for SA (ATCC 29213) and methicillin-resistant SA (ATCC 43300). Peg conditioning with HP and PLL had no impact on biofilm formation in either of these strains. Peg coating with FBS showed an adverse effect on the biofilm formation of these strains. By contrast, there was a statistically significant increase in biofilm biomass production on pegs coated with FBS and HP for SE (ATCC 35983).
Collapse
Affiliation(s)
- Adéla Diepoltová
- Charles University, Faculty of Pharmacy in Hradec Králové, Teaching and Research Centre, Czech Republic
| | - Klára Konečná
- Charles University, Faculty of Pharmacy in Hradec Králové, Teaching and Research Centre, Czech Republic
| | - Ondřej Janďourek
- Charles University, Faculty of Pharmacy in Hradec Králové, Teaching and Research Centre, Czech Republic
| | - Petr Nachtigal
- Charles University, Faculty of Pharmacy in Hradec Králové, Teaching and Research Centre, Czech Republic
| |
Collapse
|
28
|
Wu S, Liu Y, Lei L, Zhang H. An Antisense yycF RNA Modulates Biofilm Organization of Methicillin-Resistant Staphylococcus aureus and Pathogenicity in a Rat Model of Osteomyelitis. Antibiotics (Basel) 2021; 10:antibiotics10050603. [PMID: 34069543 PMCID: PMC8160899 DOI: 10.3390/antibiotics10050603] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/06/2021] [Accepted: 03/10/2021] [Indexed: 02/05/2023] Open
Abstract
Staphylococcus aureus (S. aureus) is one of most common opportunistic pathogens and is attributed to several human infections. The increasing incidence of methicillin-resistant S. aureus (MRSA) is a serious clinical threat for osteomyelitis crisis. The YycFG two-component system of S. aureus regulates genes associated with biofilm formation. To investigate the potential role of an antisense yycF RNA in the regulation of transcription levels of yycF and associated effects on biofilm formation and pathogenicity, antisense yycF (ASyycF) RNA was detected by RT-PCR and 5′ RACE assays. ASyycF overexpression mutants were constructed, and the biofilm biomass was determined by crystal violet microtiter assay and scanning electron microscopy (SEM). Quantitative RT-PCR and Western blotting analyses were used to detect whether ASyycF overexpression inhibited the transcription and translation of biofilm-related genes. Then, a rat tibial infective model was used to evaluate the pathogenicity of ASyycF overexpression in vivo. ASyycF transcription led to reductions in YycF production and biofilm formation. Overexpression of ASyycF inhibited the transcription and translation of biofilm-related genes. The sensitivity to vancomycin was improved in ASyycF-overexpressing MRSA. Furthermore, ASyycF inhibited MRSA invasion in a rat tibial infection model. From this study, the expression of the YycF protein was found to be inversely correlated with different levels of ASyycF transcription. The biofilm biomass and pathogenicity decreased in the ASyycF-overexpressing mutant. Thus, the current evidence may support ASyycF as a supplementary strategy for managing S. aureus and MRSA infections.
Collapse
Affiliation(s)
- Shizhou Wu
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Yunjie Liu
- Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health, Sichuan University, Chengdu 610041, China;
| | - Lei Lei
- Department of Preventive Dentistry, 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;
- Correspondence: (L.L.); (H.Z.)
| |
Collapse
|
29
|
Walsh L, Johnson CN, Hill C, Ross RP. Efficacy of Phage- and Bacteriocin-Based Therapies in Combatting Nosocomial MRSA Infections. Front Mol Biosci 2021; 8:654038. [PMID: 33996906 PMCID: PMC8116899 DOI: 10.3389/fmolb.2021.654038] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/07/2021] [Indexed: 12/21/2022] Open
Abstract
Staphylococcus aureus is a pathogen commonly found in nosocomial environments where infections can easily spread - especially given the reduced immune response of patients and large overlap between personnel in charge of their care. Although antibiotics are available to treat nosocomial infections, the increased occurrence of antibiotic resistance has rendered many treatments ineffective. Such is the case for methicillin resistant S. aureus (MRSA), which has continued to be a threat to public health since its emergence. For this reason, alternative treatment technologies utilizing antimicrobials such as bacteriocins, bacteriophages (phages) and phage endolysins are being developed. These antimicrobials provide an advantage over antibiotics in that many have narrow inhibition spectra, enabling treatments to be selected based on the target (pathogenic) bacterium while allowing for survival of commensal bacteria and thus avoiding collateral damage to the microbiome. Bacterial resistance to these treatments occurs less frequently than with antibiotics, particularly in circumstances where combinatory antimicrobial therapies are used. Phage therapy has been well established in Eastern Europe as an effective treatment against bacterial infections. While there are no Randomized Clinical Trials (RCTs) to our knowledge examining phage treatment of S. aureus infections that have completed all trial phases, numerous clinical trials are underway, and several commercial phage preparations are currently available to treat S. aureus infections. Bacteriocins have primarily been used in the food industry for bio-preservation applications. However, the idea of repurposing bacteriocins for human health is an attractive one considering their efficacy against many bacterial pathogens. There are concerns about the ability of bacteriocins to survive the gastrointestinal tract given their proteinaceous nature, however, this obstacle may be overcome by altering the administration route of the therapy through encapsulation, or by bioengineering protease-resistant variants. Obstacles such as enzymatic digestion are less of an issue for topical/local administration, for example, application to the surface of the skin. Bacteriocins have also shown impressive synergistic effects when used in conjunction with other antimicrobials, including antibiotics, which may allow antibiotic-based therapies to be used more sparingly with less resistance development. This review provides an updated account of known bacteriocins, phages and phage endolysins which have demonstrated an impressive ability to kill S. aureus strains. In particular, examples of antimicrobials with the ability to target MRSA strains and their subsequent use in a clinical setting are outlined.
Collapse
Affiliation(s)
- Lauren Walsh
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Crystal N Johnson
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Cork, Ireland
| | - Colin Hill
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - R Paul Ross
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Cork, Ireland
| |
Collapse
|
30
|
Jasińska E, Bogut A, Magryś A, Olender A. Evaluation of the role of staphylococci in the pathomechanism of conjunctivitis. Int Ophthalmol 2021; 41:2585-2600. [PMID: 33778922 PMCID: PMC8238708 DOI: 10.1007/s10792-021-01818-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/16/2021] [Indexed: 11/27/2022]
Abstract
Purpose Determination of the association between ica genes and phenotypic biofilm formation in staphylococcal isolates involved in conjunctivitis, their antibiotic resistance as well as detection of selected virulence characteristics: adhesion to epithelial cells and in vitro cytotoxicity. Methods The study included 26 Staphylococcus aureus (SA) and 26 Staphylococcus epidermidis (SE) isolates. The presence of icaAD genes and ica operon was determined by the PCR assay. Phenotypic biofilm formation was verified using the microtitre plate assay. Antibiotic resistance was performed using the disc diffusion method. Staphylococcal ability to attach to host cells was assessed by flow cytometry. Cytotoxicity on epithelial cells was evaluated by LDH assay. Results The ica genes were detected in 26.9% of SE and in 42.3% of SA isolates. Only 15.3% of isolates (SE) were positive for both the icaAD and the ica operon. Phenotypically, 19.2% of SE isolates were strong biofilm producers, among which three were both icaAD- and ica operon-positive. About 26.9% of SA isolates were strong biofilm producers. Methicillin resistance (MR) was detected in 34.6% of SE and 26.9% of SA isolates. About 75% of MR isolates were multidrug resistant. SA isolates adhered to host cells more extensively than SE. SA isolates released higher level of LDH than SE. Conclusions Adherence abilities were commonly observed in staphylococci associated with conjunctivitis. However, low prevalence of isolates positive for a complete and functional ica locus and low prevalence of strong biofilm producers was detected. SA adhered to a greater extent to eukaryotic cells than SE and were more cytotoxic.
Collapse
Affiliation(s)
- Ewa Jasińska
- Department of General Ophthalmology, Medical University of Lublin, Lublin, Poland
| | - Agnieszka Bogut
- Chair and Department of Medical Microbiology, Medical University of Lublin, Lublin, Poland.
| | - Agnieszka Magryś
- Chair and Department of Medical Microbiology, Medical University of Lublin, Lublin, Poland
| | - Alina Olender
- Chair and Department of Medical Microbiology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
31
|
Liu J, Madec JY, Bousquet-Mélou A, Haenni M, Ferran AA. Destruction of Staphylococcus aureus biofilms by combining an antibiotic with subtilisin A or calcium gluconate. Sci Rep 2021; 11:6225. [PMID: 33737602 PMCID: PMC7973569 DOI: 10.1038/s41598-021-85722-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 02/28/2021] [Indexed: 12/14/2022] Open
Abstract
In S. aureus biofilms, bacteria are embedded in a matrix of extracellular polymeric substances (EPS) and are highly tolerant to antimicrobial drugs. We thus sought to identify non-antibiotic substances with broad-spectrum activity able to destroy the EPS matrix and enhance the effect of antibiotics on embedded biofilm bacteria. Among eight substances tested, subtilisin A (0.01 U/mL) and calcium gluconate (CaG, Ca2+ 1.25 mmol/L) significantly reduced the biomass of biofilms formed by at least 21/24 S. aureus isolates. Confocal laser scanning microscopy confirmed that they both eliminated nearly all the proteins and PNAG from the matrix. By contrast, antibiotics alone had nearly no effect on biofilm biomass and the selected one (oxytetracycline-OTC) could only slightly reduce biofilm bacteria. The combination of OTC with CaG or subtilisin A led to an additive reduction (average of 2 log10 CFU/mL) of embedded biofilm bacteria on the isolates susceptible to OTC (MBC < 10 μg/mL, 11/24). Moreover, these two combinations led to a reduction of the embedded biofilm bacteria higher than 3 log10 CFU/mL for 20–25% of the isolates. Further studies are now required to better understand the factors that cause the biofilm produced by specific isolates (20–25%) to be susceptible to the combinations.
Collapse
Affiliation(s)
- JingJing Liu
- Unité Antibiorésistance et Virulence Bactériennes, Université de Lyon - ANSES Laboratoire de Lyon, Lyon, France.,INTHERES, INRAE, ENVT, Université de Toulouse, Toulouse, France
| | - Jean-Yves Madec
- Unité Antibiorésistance et Virulence Bactériennes, Université de Lyon - ANSES Laboratoire de Lyon, Lyon, France
| | | | - Marisa Haenni
- Unité Antibiorésistance et Virulence Bactériennes, Université de Lyon - ANSES Laboratoire de Lyon, Lyon, France
| | - Aude A Ferran
- INTHERES, INRAE, ENVT, Université de Toulouse, Toulouse, France.
| |
Collapse
|
32
|
Diclofenac May Induce PIA-Independent Biofilm Formation in Staphylococcus aureus Strains. Int J Microbiol 2021. [DOI: 10.1155/2021/8823775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Staphylococcus aureus is a pathogen commonly resistant to antibiotics. Biofilm formation is one of the important factors related to its virulence. Non-antibiotics drugs, such as nonsteroidal anti-inflammatory agents (NSAIDs), have been studied as an alternative for treating infections by multiresistant pathogens and biofilm-associated infections. In this study, the effects of NSAID sodium diclofenac on growth inhibition and biofilm formation of S. aureus were evaluated. The minimum inhibitory concentration (MIC) of diclofenac for fifty isolates ranged from 200 to 400 μg/mL. Diclofenac sub-MICs induced biofilm in 32.3% of biofilm-negative strains in tryptic soy broth. All biofilms induced by the drug showed a PIA- (polysaccharide intercellular adhesion-) independent composition, and the scanning electron microscopy showed that the induced biofilm presented a very discrete matrix. The combination of diclofenac with rifampicin sub-MICs induced strong production of PIA-dependent biofilm in three of four strains, while combination of NSAID with NaCl induced the formation of partially polysaccharide biofilm in two strains and PIA-independent biofilm in another strain. The combination of NSAID with glucose resulted in PIA-independent biofilms in all four strains tested. The results showed that diclofenac can commonly induce biofilm production by a PIA-independent pathway. However, when this NSAID is combined with other types of inducing agents, the composition of the biofilm produced may vary.
Collapse
|
33
|
Yin W, Xu S, Wang Y, Zhang Y, Chou SH, Galperin MY, He J. Ways to control harmful biofilms: prevention, inhibition, and eradication. Crit Rev Microbiol 2020; 47:57-78. [PMID: 33356690 DOI: 10.1080/1040841x.2020.1842325] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Biofilms are complex microbial architectures that encase microbial cells in a matrix comprising self-produced extracellular polymeric substances. Microorganisms living in biofilms are much more resistant to hostile environments than their planktonic counterparts and exhibit enhanced resistance against the microbicides. From the human perspective, biofilms can be classified into beneficial, neutral, and harmful. Harmful biofilms impact food safety, cause plant and animal diseases, and threaten medical fields, making it urgent to develop effective and robust strategies to control harmful biofilms. In this review, we discuss various strategies to control biofilm formation on infected tissues, implants, and medical devices. We classify the current strategies into three main categories: (i) changing the properties of susceptible surfaces to prevent biofilm formation; (ii) regulating signalling pathways to inhibit biofilm formation; (iii) applying external forces to eradicate the biofilm. We hope this review would motivate the development of innovative and effective strategies for controlling harmful biofilms.
Collapse
Affiliation(s)
- Wen Yin
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Siyang Xu
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Yiting Wang
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Yuling Zhang
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Shan-Ho Chou
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Michael Y Galperin
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Jin He
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| |
Collapse
|
34
|
Yu J, Jiang F, Zhang F, Pan Y, Wang J, Han P, Tang J, Shen H. Virtual Screening for Novel SarA Inhibitors to Prevent Biofilm Formation of Staphylococcus aureus in Prosthetic Joint Infections. Front Microbiol 2020; 11:587175. [PMID: 33224124 PMCID: PMC7674313 DOI: 10.3389/fmicb.2020.587175] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 10/15/2020] [Indexed: 02/02/2023] Open
Abstract
Staphylococcus aureus is one of the predominant causes of periprosthetic joint infections (PJIs). Bacterial adhesion and biofilm formation are important factors in the pathogenesis of PJIs. S. aureus biofilm formation is regulated by several factors, including S. aureus regulator A (SarA). Previous studies have found that SarA mutants have limited ability to develop biofilms. In this study, we identified a SarA-targeting antibiofilm compound, ZINC00990144, and evaluated its efficacy and toxicity. According to static biofilm assay, the antibiofilm ability of the compound was concentration dependent. ZINC00990144 reduced biofilm in multiple strains by 40–86% at a concentration of 11.5 μM. Additionally, ZINC00990144 inhibited biofilm formation on different orthopedic implant materials including Titanium and UHMWPE disc. Furthermore, quantitative polymerase chain reaction results demonstrated that ZINC00990144 upregulated the expression of S. aureus exoproteases to inhibit the formation of biofilms. Moreover, ZINC00990144 prevented biofilm formation when exposed to sub-inhibitory doses of vancomycin, which is known to promote biofilm formation. CCK-8 results demonstrated ZINC00990144 has no significant effect on cell viability at concentration of 11.5 μM or below. Finally, we verified the antibiofilm function of the compound in vivo using implant infection mice model with/without exposure to sub-inhibitory vancomycin. In conclusion, ZINC00990144 acts by modulating between biofilm and planktonic state of S. aureus instead of being bactericidal. Therefore, it has the potential to be used in combination with other antibiotics to prevent PJIs.
Collapse
Affiliation(s)
- Jinlong Yu
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Jiang
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Feiyang Zhang
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yunqi Pan
- Department of Clinical Laboratory, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jianqiang Wang
- Department of Clinical Laboratory, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Pei Han
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jin Tang
- Department of Clinical Laboratory, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Shen
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
35
|
Av Sá LGD, Silva CRD, de A Neto JB, Cândido TM, de Oliveira LC, do Nascimento FB, Barroso FD, da Silva LJ, de Mesquita JR, de Moraes MO, Cavalcanti BC, Júnior HV. Etomidate inhibits the growth of MRSA and exhibits synergism with oxacillin. Future Microbiol 2020; 15:1611-1619. [PMID: 33215536 DOI: 10.2217/fmb-2020-0078] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Aim: The purpose of this study was to evaluate the antimicrobial activity of the anesthetic etomidate against strains of MRSA and biofilms. Materials & methods: The antibacterial effect of etomidate was assessed by the broth microdilution method. To investigate the probable action mechanism of the compound flow cytometry techniques were used. Results: MRSA strains showed MIC equal to 500 and 1000 μg/ml of etomidate. Four-fifths (80%) of the tested MRSA strains demonstrated synergistic effect with oxacillin. Etomidate also showed activity against MRSA biofilm at concentration of 250 μg/ml. Cytometric analysis revealed that the cells treated with etomidate leading to cell death, probably by apoptosis. Conclusion: Etomidate showed antibacterial activity against MRSA.
Collapse
Affiliation(s)
- Lívia G do Av Sá
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Cecília R da Silva
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - João B de A Neto
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil.,University Center Christus, Fortaleza, CE, Brazil
| | - Thiago M Cândido
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Leilson C de Oliveira
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Francisca Bsa do Nascimento
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Fátima Dd Barroso
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Lisandra J da Silva
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - Manoel O de Moraes
- Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Bruno C Cavalcanti
- Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Hélio Vn Júnior
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
36
|
Antibiofilm effects of N,O-acetals derived from 2-amino-1,4-naphthoquinone are associated with downregulation of important global virulence regulators in methicillin-resistant Staphylococcus aureus. Sci Rep 2020; 10:19631. [PMID: 33184312 PMCID: PMC7661526 DOI: 10.1038/s41598-020-76372-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023] Open
Abstract
Despite the existing antibiotics, antimicrobial resistance is a major challenge. Consequently, the development of new drugs remains in great demand. Quinones is part of a broad group of molecules that present antibacterial activity besides other biological properties. The main purpose of this study was to evaluate the antibiofilm activities of synthetic N,O-acetals derived from 2-amino-1,4-naphthoquinone [7a: 2-(methoxymethyl)-amino-1,4-naphthoquinone; 7b: 2-(ethoxymethyl)-amino-1,4-naphthoquinone; and 7c: 2-(propynyloxymethyl)-amino-1,4-naphthoquinone] against methicillin-resistant Staphylococcus aureus (MRSA). The derivatives 7b and 7c, specially 7b, caused strong impact on biofilm accumulation. This inhibition was linked to decreased expression of the genes fnbA, spa, hla and psmα3. More importantly, this downregulation was paralleled by the modulation of global virulence regulators. The substitution of 2-ethoxymethyl (7b) in comparison with 2-propynyloxymethyl (7c) enhanced sarA-agr inhibition, decreased fnbA transcripts (positively regulated by sarA) and strongly impaired biofilm accumulation. Indeed, 7b triggered intensive autolysis and was able to eliminate vancomycin-persistent cells. Consequently, 7b is a promising molecule displaying not only antimicrobial effects, but also antibiofilm and antipersistence activities. Therefore, 7b is a good candidate for further studies involving the development of novel and more rational antimicrobials able to act in chronic and recalcitrant infections, associated with biofilm formation.
Collapse
|
37
|
Nunes SDO, Rosa HDS, Canellas ALB, Romanos MTV, Dos Santos KRN, Muricy G, Oelemann WMR, Laport MS. High reduction of staphylococcal biofilm by aqueous extract from marine sponge-isolated Enterobacter sp. Res Microbiol 2020; 172:103787. [PMID: 33049327 DOI: 10.1016/j.resmic.2020.10.002] [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] [Received: 05/06/2020] [Revised: 09/13/2020] [Accepted: 10/05/2020] [Indexed: 10/23/2022]
Abstract
Staphylococcus aureus and Staphylococcus epidermidis are among the most important bacterial species responsible for biofilm formation on indwelling medical devices, including orthopaedic implants. The increasing resistance to antimicrobials, partly attributed to the ability to form biofilms, is a challenge for the development of new antimicrobial agents. In this study, the cell-free supernatant obtained from sponge-associated Enterobacter strain 84.3 culture inhibited biofilm formation (>65%) and dissociated mature biofilm (>85%) formed by S. aureus and S. epidermidis strains. The culture supernatant was subjected to solvent partitioning and the aqueous extract presented a concentration-dependent antibiofilm activity for each strain with a minimum biofilm eradication concentration (MBEC) ranging from 16 to 256 μg/mL. The effect of the aqueous extract on mature S. aureus biofilm was analyzed by confocal scanning laser microscopy, showing a significant reduction of the biofilm layer as well as diminished interactions among the cells. This extract is not toxic for mammalian cells (L929 cell line). Studies targeting substances with antibiofilm activity gained significant attention in recent years due to difficult-to-treat biofilm infections. Here, sponge-associated Enterobacter 84.3 proved to be a source of substances capable of eradicating staphylococcal biofilm, with potential medical use in the future.
Collapse
Affiliation(s)
- Suzanne de Oliveira Nunes
- Instituto de Microbiologia Paulo de Góes, Universidade Federal Do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Cidade Universitária, 21941-590, Rio de Janeiro, RJ, Brazil
| | - Heloisa da Silva Rosa
- Instituto de Microbiologia Paulo de Góes, Universidade Federal Do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Cidade Universitária, 21941-590, Rio de Janeiro, RJ, Brazil
| | - Anna Luiza Bauer Canellas
- Instituto de Microbiologia Paulo de Góes, Universidade Federal Do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Cidade Universitária, 21941-590, Rio de Janeiro, RJ, Brazil
| | - Maria Teresa Villela Romanos
- Instituto de Microbiologia Paulo de Góes, Universidade Federal Do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Cidade Universitária, 21941-590, Rio de Janeiro, RJ, Brazil
| | - Katia R N Dos Santos
- Instituto de Microbiologia Paulo de Góes, Universidade Federal Do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Cidade Universitária, 21941-590, Rio de Janeiro, RJ, Brazil
| | - Guilherme Muricy
- Museu Nacional, Universidade Federal Do Rio de Janeiro, Quinta da Boa Vista s/no., São Cristóvão, 20940-040, Rio de Janeiro, RJ, Brazil
| | - Walter M R Oelemann
- Instituto de Microbiologia Paulo de Góes, Universidade Federal Do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Cidade Universitária, 21941-590, Rio de Janeiro, RJ, Brazil
| | - Marinella Silva Laport
- Instituto de Microbiologia Paulo de Góes, Universidade Federal Do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Cidade Universitária, 21941-590, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
38
|
Salzer A, Keinhörster D, Kästle C, Kästle B, Wolz C. Small Alarmone Synthetases RelP and RelQ of Staphylococcus aureus Are Involved in Biofilm Formation and Maintenance Under Cell Wall Stress Conditions. Front Microbiol 2020; 11:575882. [PMID: 33072039 PMCID: PMC7533549 DOI: 10.3389/fmicb.2020.575882] [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] [Received: 06/24/2020] [Accepted: 08/25/2020] [Indexed: 12/15/2022] Open
Abstract
The stringent response is characterized by the synthesis of the alarmone (p)ppGpp. The phenotypic consequences resulting from (p)ppGpp accumulation vary among species, and for several pathogenic bacteria, it has been shown that the activation of the stringent response strongly affects biofilm formation and maintenance. In Staphylococcus aureus, (p)ppGpp can be synthesized by the RelA/SpoT homolog Rel upon amino acid deprivation or by the two small alarmone synthetases RelP and RelQ under cell wall stress. We found that relP and relQ increase biofilm formation under cell wall stress conditions induced by a subinhibitory vancomycin concentration. However, the effect of (p)ppGpp on biofilm formation is independent of the regulators CodY and Agr. Biofilms formed by the strain HG001 or its (p)ppGpp-defective mutants are mainly composed of extracellular DNA and proteins. Furthermore, the induction of the RelPQ-mediated stringent response contributes to biofilm-related antibiotic tolerance. The proposed (p)ppGpp-inhibiting peptide DJK-5 shows bactericidal and biofilm-inhibitory activity. However, a non-(p)ppGpp-producing strain is even more vulnerable to DJK-5. This strongly argues against the assumption that DJK-5 acts via (p)ppGpp inhibition. In summary, RelP and RelQ play a major role in biofilm formation and maintenance under cell wall stress conditions.
Collapse
Affiliation(s)
| | | | | | | | - Christiane Wolz
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| |
Collapse
|
39
|
Begić M, Josić D. Biofilm formation and extracellular microvesicles-The way of foodborne pathogens toward resistance. Electrophoresis 2020; 41:1718-1739. [PMID: 32901923 DOI: 10.1002/elps.202000106] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/08/2020] [Accepted: 07/15/2020] [Indexed: 12/21/2022]
Abstract
Almost all known foodborne pathogens are able to form biofilms as one of the strategies for survival under harsh living conditions, to ward off the inhibition and the disinfection during food production, transport and storage, as well as during cleaning and sanitation of corresponding facilities. Biofilms are communities where microbial cells live under constant intracellular interaction and communication. Members of the biofilm community are embedded into extracellular matrix that contains polysaccharides, DNA, lipids, proteins, and small molecules that protect microorganisms and enable their intercellular communication under stress conditions. Membrane vesicles (MVs) are produced by both Gram positive and Gram negative bacteria. These lipid membrane-enveloped nanoparticles play an important role in biofilm genesis and in communication between different biofilm members. Furthermore, MVs are involved in other important steps of bacterial life like cell wall modeling, cellular division, and intercellular communication. They also carry toxins and virulence factors, as well as nucleic acids and different metabolites, and play a key role in host infections. After entering host cells, MVs can start many pathologic processes and cause serious harm and cell death. Prevention and inhibition of both biofilm formation and shedding of MVs by foodborne pathogens has a very important role in food production, storage, and food safety in general. Better knowledge of biofilm formation and maintaining, as well as the role of microbial vesicles in this process and in the process of host cells' infection is essential for food safety and prevention of both food spoilage and host infection.
Collapse
Affiliation(s)
- Marija Begić
- Faculty of Medicine, Juraj Dobrila University, Pula, Croatia
| | - Djuro Josić
- Faculty of Medicine, Juraj Dobrila University, Pula, Croatia.,Warren Alpert Medical School, Brown University, Providence, RI, USA
| |
Collapse
|
40
|
Dutta K, Karmakar A, Jana D, Ballav S, Shityakov S, Panda AK, Ghosh C. Benzyl isocyanate isolated from the leaves of Psidium guajava inhibits Staphylococcus aureus biofilm formation. BIOFOULING 2020; 36:1000-1017. [PMID: 33172298 DOI: 10.1080/08927014.2020.1842877] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 09/03/2020] [Accepted: 10/20/2020] [Indexed: 06/11/2023]
Abstract
Benzyl isocyanate (BIC), from methanol extract of Psidium guajava leaves, exhibited substantial anti-biofilm activities against Staphylococcus aureus, the common bacterial pathogen in nosocomial infections. Major components of the extract included eugenol, BIC, phenyl-2-methoxy-4-(1-propenyl)-acetate and 2,5-pyrrolidinedione,1-penta-3-4-dienyl, analyzed by GC-MS and HPLC studies. BIC exhibited substantial anti-biofilm activitiy against S. aureus, established by assaying biofilm formation, biofilm metabolic activity, bacterial adherence to hydrocarbons, exopolysaccharide formation, and optical and scanning electron microscopic studies. BIC significantly downregulated the important biofilm markers of S. aureus, viz., icaAD, sarA and agr, observed by quantitative real time polymerase chain reaction analysis. Molecular docking studies revealed thermodynamically favorable interaction of BIC with IcaA, SarA and Agr, having Gibbs energy values of -8.45, -9.09 and -10.29 kcal mol-1, respectively. BIC after binding to IcaR, the repressor of IcaA, influences its binding to target DNA site (Eshape, -157.27 kcal mol-1). The results are considered to demonstrate anti-biofilm potential of BIC against bacterial infections.
Collapse
Affiliation(s)
- Kunal Dutta
- Department of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India
| | - Amit Karmakar
- Department of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India
| | - Debarati Jana
- Department of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India
| | - Saroj Ballav
- Department of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India
| | - Sergey Shityakov
- Department of Anaesthesia and Critical Care, University of Würzburg, Würzburg, Germany
| | - Amiya Kumar Panda
- Department of Chemistry, Vidyasagar University, Midnapore, West Bengal, India
| | - Chandradipa Ghosh
- Department of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India
| |
Collapse
|
41
|
Gidari A, Sabbatini S, Schiaroli E, Perito S, Francisci D, Baldelli F, Monari C. Tedizolid-Rifampicin Combination Prevents Rifampicin-Resistance on in vitro Model of Staphylococcus aureus Mature Biofilm. Front Microbiol 2020; 11:2085. [PMID: 32983061 PMCID: PMC7484889 DOI: 10.3389/fmicb.2020.02085] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 08/07/2020] [Indexed: 12/14/2022] Open
Abstract
Staphylococcus aureus infections associated with implanted medical devices are difficult to treat and require long-lasting antibiotic therapies, especially when device removal is not possible or easy such as in the case of joint prostheses. Biofilm formation is a major cause of treatment failure and infection recurrence. This study aimed to test, for the first time, the in vitro combination of tedizolid plus rifampicin on methicillin-sensitive (MSSA ATCC 6538) and methicillin-resistant (MRSA ATCC 43300) S. aureus mature biofilm. Here, we demonstrated that the combination of tedizolid with rifampicin significantly disaggregated pre-formed biofilm of both strains, reduced their metabolic activity and exerted bactericidal activity at clinically meaningful concentrations. Notably, tedizolid was able to completely prevent the emergence of resistance to rifampicin. Moreover these effects were similar to those obtained with daptomycin plus rifampicin, a well-known and widely used combination. Preliminary results on some MRSA clinical isolates confirmed the efficacy of this combination in reducing biofilm biomass and preventing rifampicin resistance onset. Further in vivo studies are needed to confirm the validity of this promising therapeutic option that can be useful against biofilm-associated S. aureus infections.
Collapse
Affiliation(s)
- Anna Gidari
- Department of Medicine, Clinic of Infectious Diseases, University of Perugia, Perugia, Italy
| | - Samuele Sabbatini
- Department of Medicine, Medical Microbiology Section, University of Perugia, Perugia, Italy
| | - Elisabetta Schiaroli
- Department of Medicine, Clinic of Infectious Diseases, University of Perugia, Perugia, Italy
| | - Stefano Perito
- Department of Medicine, Medical Microbiology Section, University of Perugia, Perugia, Italy
| | - Daniela Francisci
- Department of Medicine, Clinic of Infectious Diseases, University of Perugia, Perugia, Italy
| | - Franco Baldelli
- Department of Medicine, Clinic of Infectious Diseases, University of Perugia, Perugia, Italy
| | - Claudia Monari
- Department of Medicine, Medical Microbiology Section, University of Perugia, Perugia, Italy
| |
Collapse
|
42
|
Sabbatini S, Monari C, Ballet N, Decherf AC, Bozza S, Camilloni B, Perito S, Vecchiarelli A. Anti-Biofilm Properties of Saccharomyces cerevisiae CNCM I-3856 and Lacticaseibacillus rhamnosus ATCC 53103 Probiotics against G. vaginalis. Microorganisms 2020; 8:microorganisms8091294. [PMID: 32847138 PMCID: PMC7564297 DOI: 10.3390/microorganisms8091294] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 12/24/2022] Open
Abstract
Bacterial vaginosis (BV) is characterized by the presence of a polymicrobial biofilm where Gardnerella vaginalis plays a key role. Previously, we demonstrated that Saccharomyces cerevisiae CNCM (French National Collection of Cultures of Microorganisms) I-3856 is helpful in resolving experimental simulated BV in mice. In this study, we analyzed its capacity to affect G. vaginalis biofilms and to potentiate the activity of standard antimicrobial agents. We also investigated the anti-biofilm activity of Lacticaseibacillus rhamnosus GG (ATCC 53103), a well-known strain for its intestinal healthy benefits. Biofilm biomass was assessed by crystal violet staining, and G. vaginalis viability was assessed by a colony forming unit (CFU) assay. Here, for the first time, we demonstrated that S. cerevisiae CNCM I-3856 as well as L. rhamnosus GG were able (i) to significantly inhibit G. vaginalis biofilm formation, (ii) to markedly reduce G. vaginalis viability among the biomass constituting the biofilm, (iii) to induce disaggregation of preformed biofilm, and (iv) to kill a consistent amount of bacterial cells in a G. vaginalis preformed biofilm. Furthermore, S. cerevisiae CNCM I-3856 strongly potentiates the metronidazole effect on G. vaginalis biofilm viability. These results suggest that S. cerevisiae CNCM I-3856 as well as L. rhamnosus GG could be potential novel therapeutic agents against bacterial vaginosis.
Collapse
Affiliation(s)
- Samuele Sabbatini
- Department of Medicine, Medical Microbiology Section, University of Perugia, Polo Unico Sant’Andrea delle Fratte, 06132, Perugia, Italy; (S.S.); (S.B.); (B.C.); (S.P.); (A.V.)
| | - Claudia Monari
- Department of Medicine, Medical Microbiology Section, University of Perugia, Polo Unico Sant’Andrea delle Fratte, 06132, Perugia, Italy; (S.S.); (S.B.); (B.C.); (S.P.); (A.V.)
- Correspondence:
| | - Nathalie Ballet
- Lesaffre International, Lesaffre Group, Rue Gabriel Péri 137, Marcq-en-Baroeul, 59700, France;
| | | | - Silvia Bozza
- Department of Medicine, Medical Microbiology Section, University of Perugia, Polo Unico Sant’Andrea delle Fratte, 06132, Perugia, Italy; (S.S.); (S.B.); (B.C.); (S.P.); (A.V.)
| | - Barbara Camilloni
- Department of Medicine, Medical Microbiology Section, University of Perugia, Polo Unico Sant’Andrea delle Fratte, 06132, Perugia, Italy; (S.S.); (S.B.); (B.C.); (S.P.); (A.V.)
| | - Stefano Perito
- Department of Medicine, Medical Microbiology Section, University of Perugia, Polo Unico Sant’Andrea delle Fratte, 06132, Perugia, Italy; (S.S.); (S.B.); (B.C.); (S.P.); (A.V.)
| | - Anna Vecchiarelli
- Department of Medicine, Medical Microbiology Section, University of Perugia, Polo Unico Sant’Andrea delle Fratte, 06132, Perugia, Italy; (S.S.); (S.B.); (B.C.); (S.P.); (A.V.)
| |
Collapse
|
43
|
Virulence factors and antimicrobial resistance in Staphylococcus aureus isolated from bovine mastitis in Brazil. Braz J Microbiol 2020; 51:2111-2122. [PMID: 32815081 DOI: 10.1007/s42770-020-00363-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 08/08/2020] [Indexed: 02/07/2023] Open
Abstract
This study aimed to evaluate virulence factors and genetic markers of antimicrobial resistance in 400 Staphylococcus aureus strains isolated from bovine mastitis in four Brazilian states, as well as to assess the association between these characteristics and field information. Virulence factors and drug resistance genes were identified by PCR screening. Biofilm-forming and hemolytic phenotype were detected using Congo red Tryptic Soy Broth and defibrinated sheep blood agar, respectively. Of all isolates, 83.5% were biofilm-forming and 98.5% strains exhibited biofilm gene icaAD, and a significant association between phenotype and genotype for biofilm was observed (P = 0.0005). Hemolysin genes were observed in 82.85% (hla+hlb+), 16.5% (hla+) and 0.75% (hlb+) isolates, whereas the hemolytic phenotype exhibited was complete and incomplete hemolysis in 64.25%, complete in 28.25%, incomplete in 4.75%, and negative in 2.75% of the strains. Virulence factors genes luk, seb, sec, sed, and tst were observed in 3.5%, 0.5%, 1%, 0.25%, and 0.74% isolates, respectively. The gene blaZ was detected in 82.03% of penicillin-resistant isolates, whereas tetK and aac(6')-Ie-aph(2')-Ia were observed in 33.87% and 45.15% of the tetracycline and aminoglycosides-resistant isolates, respectively. Fluoroquinolone resistance gene mepA was detected for the first time in S. aureus from bovine mastitis. Resistance genes tetM (3.22%), tetL (1.61%), ermA (14.29%), ermB (14.29%), ermC (33.3%), ermT (9.52%), ermY (4.76%), msrA (9.52%), and mphC (9.52%) were also detected among resistant isolates. No association between virulence factors or antimicrobial-resistant genes and year of isolation, geographic origin, or antimicrobial resistance profile was observed. Our results showed that S. aureus strains isolated from bovine mastitis in the four Brazilian states sampled are mainly biofilm-forming and hemolytic, whereas virulence genes associated with enterotoxins, luk and tst, were less frequently observed. Moreover, a wide variety of resistance genes that confer resistance to almost all classes of antimicrobial agents approved for use in animals and humans were found. Overall, the data point to a great pathogenic potential of S. aureus associated with bovine mastitis and to the non-negligible risks to public health of staphylococcal infections from animal origin.
Collapse
|
44
|
Chen P, Sun F, Feng W, Hong H, Li B, Song J. Pathogenic characteristics of Staphylococcus aureus isolates from arthroplasty infections. Int J Artif Organs 2020; 44:208-214. [PMID: 32783484 DOI: 10.1177/0391398820948877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Staphylococcus aureus has a great ability to form biofilms on implant-related biomaterials. This study aimed to investigate the resistance, biofilm and molecular characteristics of S. aureus strains isolated from patients with postoperative infections after arthroplasty in two Chinese tertiary care hospitals during 2017 to 2018. METHODS Antimicrobial susceptibility was determined by the agar dilution method. Bacterial biofilm formation was determined by crystal violet staining. The genes related to biofilm formation and molecular typing were analyzed by PCR amplification. RESULTS A total of 33 isolates were collected, 21 of which were from Henan. The strains were completely sensitive to vancomycin, linezolid, and nitrofurantoin. All the isolates had adhesion ability and could produce biofilms. Of the isolates, 75.0% from Chongqing and 85.7% from Henan had stronger biofilm formation abilities. The strains from Henan had slightly higher resistance, adhesion and biofilm-forming abilities than those from Chongqing. The strains in both hospitals carried at least two genes related to biofilm formation, and the ica and fnb genes were the most frequently detected genes. Three SCCmec types and seven sequence types (STs) were found in Henan, and two SCCmec types and six STs were found in Chongqing. ST239-SCCmec III was the main epidemic clone in the two hospitals. CONCLUSION The resistance phenotype and molecular characteristics of S. aureus strains varied in different hospitals. The results reflect the potential risks of S. aureus infection in postoperative arthroplasty patients. Our study provides a powerful basis for the clinical treatment, infection control and monitoring of outbreaks of epidemic strains.
Collapse
Affiliation(s)
- Peng Chen
- Department of Orthopaedics, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province, China
| | - Fengjun Sun
- Department of Pharmacy, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Wei Feng
- Department of Pharmacy, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hai Hong
- Clinic of Zhengchangzhuang Sanatorium for Retired Cadres, Logistics Department of PLA, Beijing, China
| | - Bo Li
- Department of Orthopaedics, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province, China
| | - Jie Song
- Department of Prevention and Health Care, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
45
|
De Gregorio E, Esposito A, Vollaro A, De Fenza M, D’Alonzo D, Migliaccio A, Iula VD, Zarrilli R, Guaragna A. N-Nonyloxypentyl-l-Deoxynojirimycin Inhibits Growth, Biofilm Formation and Virulence Factors Expression of Staphylococcus aureus. Antibiotics (Basel) 2020; 9:E362. [PMID: 32604791 PMCID: PMC7344813 DOI: 10.3390/antibiotics9060362] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/15/2020] [Accepted: 06/24/2020] [Indexed: 12/14/2022] Open
Abstract
Staphylococcus aureus is one of the major causes of hospital- and community-associated bacterial infections throughout the world, which are difficult to treat due to the rising number of drug-resistant strains. New molecules displaying potent activity against this bacterium are urgently needed. In this study, d- and l-deoxynojirimycin (DNJ) and a small library of their N-alkyl derivatives were screened against S. aureus ATCC 29213, with the aim to identify novel candidates with inhibitory potential. Among them, N-nonyloxypentyl-l-DNJ (l-NPDNJ) proved to be the most active compound against S. aureus ATCC 29213 and its clinical isolates, with the minimum inhibitory concentration (MIC) value of 128 μg/mL. l-NPDNJ also displayed an additive effect with gentamicin and oxacillin against the gentamicin- and methicillin-resistant S. aureus isolate 00717. Sub-MIC values of l-NPDNJ affected S. aureus biofilm development in a dose-dependent manner, inducing a strong reduction in biofilm biomass. Moreover, real-time reverse transcriptase PCR analysis revealed that l-NPDNJ effectively inhibited at sub-MIC values the transcription of the spa, hla, hlb and sea virulence genes, as well as the agrA and saeR response regulator genes.
Collapse
Affiliation(s)
- Eliana De Gregorio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy;
| | - Anna Esposito
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia, 80126 Naples, Italy; (A.E.); (M.D.F.); (D.D.)
| | - Adriana Vollaro
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy;
| | - Maria De Fenza
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia, 80126 Naples, Italy; (A.E.); (M.D.F.); (D.D.)
| | - Daniele D’Alonzo
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia, 80126 Naples, Italy; (A.E.); (M.D.F.); (D.D.)
| | - Antonella Migliaccio
- Department of Public Health, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy; (A.M.); (R.Z.)
| | - Vita Dora Iula
- Complex Operative Unit of Clinical Pathology, “Ospedale del Mare-ASL NA1 Centro”, 80131 Naples, Italy;
| | - Raffaele Zarrilli
- Department of Public Health, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy; (A.M.); (R.Z.)
| | - Annalisa Guaragna
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia, 80126 Naples, Italy; (A.E.); (M.D.F.); (D.D.)
| |
Collapse
|
46
|
Common Plant-Derived Terpenoids Present Increased Anti-Biofilm Potential against Staphylococcus Bacteria Compared to a Quaternary Ammonium Biocide. Foods 2020; 9:foods9060697. [PMID: 32492772 PMCID: PMC7353659 DOI: 10.3390/foods9060697] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 12/15/2022] Open
Abstract
The antimicrobial actions of three common plant-derived terpenoids (i.e., carvacrol, thymol and eugenol) were compared to those of a typical quaternary ammonium biocide (i.e., benzalkonium chloride; BAC), against both planktonic and biofilm cells of two widespread Staphylococcus species (i.e., S. aureus and S. epidermidis). The minimum inhibitory and bactericidal concentrations (MICs, MBCs) of each compound against the planktonic cells of each species were initially determined, together with their minimum biofilm eradication concentrations (MBECs). Various concentrations of each compound were subsequently applied, for 6 min, against each type of cell, and survivors were enumerated by agar plating to calculate log reductions and determine the resistance coefficients (Rc) for each compound, as anti-biofilm effectiveness indicators. Sessile communities were always more resistant than planktonic ones, depending on the biocide and species. Although lower BAC concentrations were always needed to kill a specified population of either cell type compared to the terpenoids, for the latter, the required increases in their concentrations, to be equally effective against the biofilm cells with respect to the planktonic ones, were not as intense as those observed in the case of BAC, presenting thus significantly lower Rc. This indicates their significant anti-biofilm potential and advocate for their further promising use as anti-biofilm agents.
Collapse
|
47
|
Carothers KE, Liang Z, Mayfield J, Donahue DL, Lee M, Boggess B, Ploplis VA, Castellino FJ, Lee SW. The Streptococcal Protease SpeB Antagonizes the Biofilms of the Human Pathogen Staphylococcus aureus USA300 through Cleavage of the Staphylococcal SdrC Protein. J Bacteriol 2020; 202:e00008-20. [PMID: 32205460 PMCID: PMC7221255 DOI: 10.1128/jb.00008-20] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 03/06/2020] [Indexed: 01/11/2023] Open
Abstract
Streptococcus pyogenes, or group A Streptococcus (GAS), is both a pathogen and an asymptomatic colonizer of human hosts and produces a large number of surface-expressed and secreted factors that contribute to a variety of infection outcomes. The GAS-secreted cysteine protease SpeB has been well studied for its effects on the human host; however, despite its broad proteolytic activity, studies on how this factor is utilized in polymicrobial environments are lacking. Here, we utilized various forms of SpeB protease to evaluate its antimicrobial and antibiofilm properties against the clinically important human colonizer Staphylococcus aureus, which occupies niches similar to those of GAS. For our investigation, we used a skin-tropic GAS strain, AP53CovS+, and its isogenic ΔspeB mutant to compare the production and activity of native SpeB protease. We also generated active and inactive forms of recombinant purified SpeB for functional studies. We demonstrate that SpeB exhibits potent biofilm disruption activity at multiple stages of S. aureus biofilm formation. We hypothesized that the surface-expressed adhesin SdrC in S. aureus was cleaved by SpeB, which contributed to the observed biofilm disruption. Indeed, we found that SpeB cleaved recombinant SdrC in vitro and in the context of the full S. aureus biofilm. Our results suggest an understudied role for the broadly proteolytic SpeB as an important factor for GAS colonization and competition with other microorganisms in its niche.IMPORTANCEStreptococcus pyogenes (GAS) causes a range of diseases in humans, ranging from mild to severe, and produces many virulence factors in order to be a successful pathogen. One factor produced by many GAS strains is the protease SpeB, which has been studied for its ability to cleave and degrade human proteins, an important factor in GAS pathogenesis. An understudied aspect of SpeB is the manner in which its broad proteolytic activity affects other microorganisms that co-occupy niches similar to that of GAS. The significance of the research reported herein is the demonstration that SpeB can degrade the biofilms of the human pathogen Staphylococcus aureus, which has important implications for how SpeB may be utilized by GAS to successfully compete in a polymicrobial environment.
Collapse
Affiliation(s)
- Katelyn E Carothers
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, USA
| | - Zhong Liang
- W. M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, Indiana, USA
| | - Jeffrey Mayfield
- W. M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, Indiana, USA
| | - Deborah L Donahue
- W. M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, Indiana, USA
| | - Mijoon Lee
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Bill Boggess
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Victoria A Ploplis
- W. M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, Indiana, USA
| | - Francis J Castellino
- W. M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, Indiana, USA
| | - Shaun W Lee
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, USA
- W. M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, Indiana, USA
| |
Collapse
|
48
|
LuTheryn G, Glynne-Jones P, Webb JS, Carugo D. Ultrasound-mediated therapies for the treatment of biofilms in chronic wounds: a review of present knowledge. Microb Biotechnol 2020; 13:613-628. [PMID: 32237219 PMCID: PMC7111087 DOI: 10.1111/1751-7915.13471] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 07/21/2019] [Indexed: 12/11/2022] Open
Abstract
Bacterial biofilms are an ever-growing concern for public health, featuring both inherited genetic resistance and a conferred innate tolerance to traditional antibiotic therapies. Consequently, there is a growing interest in novel methods of drug delivery, in order to increase the efficacy of antimicrobial agents. One such method is the use of acoustically activated microbubbles, which undergo volumetric oscillations and collapse upon exposure to an ultrasound field. This facilitates physical perturbation of the biofilm and provides the means to control drug delivery both temporally and spatially. In line with current literature in this area, this review offers a rounded argument for why ultrasound-responsive agents could be an integral part of advancing wound care. To achieve this, we will outline the development and clinical significance of biofilms in the context of chronic infections. We will then discuss current practices used in combating biofilms in chronic wounds and then critically evaluate the use of acoustically activated gas microbubbles as an emerging treatment modality. Moreover, we will introduce the novel concept of microbubbles carrying biologically active gases that may facilitate biofilm dispersal.
Collapse
Affiliation(s)
- Gareth LuTheryn
- Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK
- National Biofilms Innovation Centre, University of Southampton, Southampton, UK
| | - Peter Glynne-Jones
- Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Jeremy S Webb
- National Biofilms Innovation Centre, University of Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
- Centre for Biological Sciences, University of Southampton, Southampton, UK
| | - Dario Carugo
- Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK
- National Biofilms Innovation Centre, University of Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
49
|
Zheng JX, Tu HP, Sun X, Xu GJ, Chen JW, Deng QW, Yu ZJ, Qu D. In vitro activities of telithromycin against Staphylococcus aureus biofilms compared with azithromycin, clindamycin, vancomycin and daptomycin. J Med Microbiol 2020; 69:120-131. [PMID: 31916929 DOI: 10.1099/jmm.0.001122] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Introduction. Staphylococcus aureus biofilms are difficult to treat and the effect of telithromycin treatment is still unclear.Aim. This study aimed to explore the effect of telithromycin against Staphylococcus aureus biofilms compared with azithromycin, clindamycin, vancomycin and daptomycin.Methodology. Eight methicillin-susceptible and eight methicillin-resistant S. aureus isolates (MSSA and MRSA, respectively) were used for this study. Biofilm biomasses were detected by crystal violet staining and the adherent cells in the established biofilms were quantified by determination of colony-forming units (c.f.u.). The RNA levels of biofilm formation-related genes were determined by RT-qPCR.Results. Telithromycin [8× minimum inhibitory concentration (MIC)] eradicated more established biofilms than azithromycin or clindamycin in the four MSSA isolates, and eliminated the established biofilms of six MRSA isolates more effectively than vancomycin or daptomycin. Telithromycin (8× MIC) killed more adherent cells in the established biofilms than azithromycin or clindamycin in the six MSSA isolates, and killed more adherent cells than vancomycin in all eight MRSA isolates. Daptomycin also showed an excellent effect on the adherent cells of MRSA isolates, with similarresults to telithromycin. The effect of a subinhibitory concentration of telithromycin (1/4× MIC) was significantly superior to that of azithromycin or clindamycin, inhibiting the biofilm formation of six MSSA isolates and seven MRSA isolates more effectively than vancomycin or daptomycin. The RNA levels of agrA, agrC, clfA, icaA and sigB decreased when treated with telithromycin (1/4× MIC).Conclusions. Telithromycin is more effective than azithromycin, clindamycin, vancomycin, or daptomycin against S. aureus biofilms.
Collapse
Affiliation(s)
- Jin-Xin Zheng
- Department of Infectious Diseases and the Key Laboratory of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, PR China.,Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, PR China
| | - Hao-Peng Tu
- Department of Infectious Diseases and the Key Laboratory of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, PR China
| | - Xiang Sun
- Department of Infectious Diseases and the Key Laboratory of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, PR China
| | - Guang-Jian Xu
- Department of Infectious Diseases and the Key Laboratory of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, PR China
| | - Jun-Wen Chen
- Department of Infectious Diseases and the Key Laboratory of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, PR China
| | - Qi-Wen Deng
- Department of Infectious Diseases and the Key Laboratory of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, PR China
| | - Zhi-Jian Yu
- Department of Infectious Diseases and the Key Laboratory of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, PR China
| | - Di Qu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, PR China
| |
Collapse
|
50
|
Castro RD, Pedroso SHSP, Sandes SHC, Silva GO, Luiz KCM, Dias RS, Filho RAT, Figueiredo HCP, Santos SG, Nunes AC, Souza MR. Virulence factors and antimicrobial resistance of Staphylococcus aureus isolated from the production process of Minas artisanal cheese from the region of Campo das Vertentes, Brazil. J Dairy Sci 2020; 103:2098-2110. [PMID: 31980224 DOI: 10.3168/jds.2019-17138] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/28/2019] [Indexed: 12/31/2022]
Abstract
Staphylococcus aureus is one of the main pathogens found in cheeses produced with raw milk, including Minas artisanal cheese from Brazil. However, information about S. aureus isolated from artisanal cheeses and its sources of production in small-scale dairies is very limited. We aimed to characterize the virulence factors of S. aureus isolated from raw milk, endogenous starter culture, Minas artisanal cheese, and cheese handlers from the region of Campo das Vertentes, Minas Gerais, Brazil. We identified the staphylococcal isolates by MALDI-TOF mass spectrometry. We evaluated biofilm production on Congo red agar and polystyrene plates. We used PCR to detect icaA, icaB, icaC, sea, seb, sec, sed, see, tsst-1, agr, and mecA. We evaluated the expression of staphylococcal toxin genes in PCR-positive staphylococcal isolates using quantitative reverse-transcription PCR, and we evaluated the production of these toxins and their hemolytic activity in vitro. We also evaluated the antimicrobial resistance profile of the staphylococcal isolates. For statistical analysis, we used cluster analysis, χ2 tests, and correspondence tests. We analyzed 76 staphylococcal isolates. According to PCR, 18.42, 18.42, 2.63, and 77.63% were positive for sea, tsst-1, sec, and agr, respectively. We found low expression of staphylococcal toxin genes according to quantitative reverse-transcription PCR, and only 2 staphylococcal isolates produced toxic shock syndrome toxins. A total of 43 staphylococcal isolates (56.58%) had hemolytic activity; 53 were biofilm-forming on Congo red agar (69.73%), and 62 on polystyrene plates (81.58%). None of the staphylococcal isolates expressed the mecA gene, and none presented a multi-drug resistance pattern. The highest resistance was observed for penicillin G (67.11%) in 51 isolates and for tetracycline (27.63%) in 21 isolates. The staphylococcal isolates we evaluated had toxigenic potential, with a higher prevalence of sea and tsst-1. Biofilm production was the main virulence factor of the studied bacteria. Six clusters were formed whose distribution frequencies differed for hemolytic activity, biofilm formation (qualitative and quantitative analyses), and resistance to penicillin, tetracycline, and erythromycin. These findings emphasize the need for effective measures to prevent staphylococcal food poisoning by limiting S. aureus growth and enterotoxin formation throughout the food production chain and the final product.
Collapse
Affiliation(s)
- R D Castro
- Departamento de Tecnologia e Inspeção de Produtos de Origem Animal, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil.
| | - S H S P Pedroso
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - S H C Sandes
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - G O Silva
- Departamento de Tecnologia e Inspeção de Produtos de Origem Animal, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - K C M Luiz
- Laboratório de Enterotoxinas de Alimentos, Fundação Ezequiel Dias, Belo Horizonte, 30510-010, Brazil
| | - R S Dias
- Laboratório de Enterotoxinas de Alimentos, Fundação Ezequiel Dias, Belo Horizonte, 30510-010, Brazil
| | - R A T Filho
- Departamento de Engenharia de Alimentos, Universidade Federal de Viçosa, Florestal, 35690-000, Brazil
| | - H C P Figueiredo
- Departamento de Medicina Veterinária Preventiva, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - S G Santos
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - A C Nunes
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - M R Souza
- Departamento de Tecnologia e Inspeção de Produtos de Origem Animal, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| |
Collapse
|