1
|
Pitton M, Valente LG, Oberhaensli S, Gözel B, Jakob SM, Sendi P, Fürholz M, Cameron DR, Que YA. Targeting Chronic Biofilm Infections With Patient-derived Phages: An In Vitro and Ex Vivo Proof-of-concept Study in Patients With Left Ventricular Assist Devices. Open Forum Infect Dis 2025; 12:ofaf158. [PMID: 40182131 PMCID: PMC11966103 DOI: 10.1093/ofid/ofaf158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 03/12/2025] [Indexed: 04/05/2025] Open
Abstract
Background Phage therapy is being reconsidered as a valuable approach to combat antimicrobial resistance. We recently established a personalized phage therapy pipeline in healthy volunteers, where therapeutic phages were isolated from individuals' skin microbiota. In this study, we aim to validate this pipeline in end-stage heart failure patients supported by left ventricular assist devices (LVADs), focusing on phages targeting Staphylococcus epidermidis, a common pathogen responsible for LVAD infections. Methods Over a 2.5-year period, 45 LVAD patients were consistently sampled at their driveline exit sites and foreheads. S epidermidis strains from patients' foreheads were used to amplify patient-specific phages. Newly isolated phages were characterized and tested against S epidermidis isolates (n = 42) from the patient cohort. The virulent phage vB_SepS_BE22, isolated from a patient with a driveline infection, was further tested for its bactericidal activity against S epidermidis biofilms ex vivo with rifampicin on driveline biofilms. Results S epidermidis was detected in 32 patients, 3 of whom had driveline infections. Phages were isolated from 8 patients, 6 of which were unique and exhibited narrow host ranges, infecting 19%-52% of S epidermidis strains. vB_SepS_BE22, isolated from patient ID25's microbiota, was the only phage that specifically killed S epidermidis clones linked to a patient's infection. vB_SepS_BE22 also reduced bacterial loads in exponential and stationary phase cultures, as well as in biofilms on drivelines when combined with rifampicin. Conclusions This study validated a personalized phage therapy approach, where phages from a patient's own microbiota can be used in chronic infection settings as therapeutic agents.
Collapse
Affiliation(s)
- Melissa Pitton
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
| | - Luca G Valente
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Simone Oberhaensli
- Interfaculty Bioinformatics Unit, University of Bern, Bern, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Bülent Gözel
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Stephan M Jakob
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Parham Sendi
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Monika Fürholz
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - David R Cameron
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Yok-Ai Que
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
2
|
Post V, Pascoe B, Hitchings MD, Erichsen C, Fischer J, Morgenstern M, Richards RG, Sheppard SK, Moriarty TF. Methicillin-sensitive Staphylococcus aureus lineages contribute towards poor patient outcomes in orthopaedic device-related infections. Microb Genom 2025; 11:001390. [PMID: 40238650 PMCID: PMC12068410 DOI: 10.1099/mgen.0.001390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 03/04/2025] [Indexed: 04/18/2025] Open
Abstract
Staphylococci are the most common cause of orthopaedic device-related infections (ODRIs), with Staphylococcus aureus responsible for a third or more of cases. This prospective clinical and laboratory study investigated the association of genomic and phenotypic variation with treatment outcomes in ODRI isolates. Eighty-six invasive S. aureus isolates were collected from patients with ODRI, and clinical outcome was assessed after a follow-up examination of 24 months. Each patient was then considered to have been 'cured' or 'not cured' based on predefined clinical criteria. Whole-genome sequencing and molecular characterization identified isolates belonging to globally circulating community- and hospital-acquired lineages. Most isolates were phenotypically susceptible to methicillin and lacked the staphylococcal cassette chromosome mec cassette [methicillin-susceptible S. aureus (MSSA); 94%] but contained several virulence genes, including toxins and biofilm genes. Whilst recognizing the role of the host immune response, we identified genetic variance, which could be associated with the infection severity or clinical outcome. Whilst this and several other studies reinforce the role antibiotic resistance [e.g. methicillin-resistant S. aureus (MRSA) infection] has on treatment failure, it is important not to overlook MSSA that can cause equally destructive infections and lead to poor patient outcomes.
Collapse
Affiliation(s)
| | - Ben Pascoe
- Ineos Oxford Institute for Antimicrobial Research, Department of Biology, University of Oxford, Oxford, UK
- Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona, USA
| | | | | | - Julian Fischer
- Centrum of Orthopedic Isartal, Pullach im Isartal, Germany
| | - Mario Morgenstern
- Department of Orthopedic and Trauma Surgery, University Hospital, Basel, Switzerland
| | | | - Samuel K. Sheppard
- Ineos Oxford Institute for Antimicrobial Research, Department of Biology, University of Oxford, Oxford, UK
| | - T. Fintan Moriarty
- AO Research Institute Davos, Davos, Switzerland
- Department of Orthopedic and Trauma Surgery, University Hospital, Basel, Switzerland
| |
Collapse
|
3
|
Li Y, Gao L, Fan S. The characteristics of surgical site infection with class I incision in neurosurgery. BMC Surg 2025; 25:97. [PMID: 40075338 PMCID: PMC11900087 DOI: 10.1186/s12893-025-02825-9] [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: 08/15/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
OBJECTIVE Surgical site infections (SSIs) were recognized to be the most common complication of neurological surgery, with substantial life quality threats to patients and additional cost burdens to healthcare facilities. This study sought to expound the infection characteristics of class I incision and provide clinical indication for the prevention and treatment of SSIs. METHODS A 2-year retrospective analysis was conducted according to patients who performed neurological surgery with class I incision in a tertiary comprehensive hospital in Shaanxi Province, China. Case mix index (CMI)-adjusted and national nosocomial infection surveillance (NNIS) risk index-adjusted SSI rate were utilized for analytical standardization. The SSIs were specifically analyzed according to various departments, surgeons, and surgical classifications. FINDINGS 6046 surgical cases were finally included in our study. The majority of the American Society of Aneshesiologists (ASA) score and NNIS risk index of surgeries were allocated in level 2 and score 1. Our study found 121 SSI cases, with the crude infection rate of 2.00%. 95.04% were organ/space infection. The most of the infection were found in the surgeries with score 1 (68.60%) of the NNIS risk index. The main surgical classification was resection of space occupying lesions (61.96%). The highest crude and NNIS risk index adjusted infection were individually found in hybrid operation (11.67%) and endoscopy-assisted resection of space occupying lesions (13.33%). 21 of 54 surgeons were found to have SSIs. We found the main pathogenic bacteria was Staphylococcus epidermidis (22.81%), and the commonly prophylactic used antibiotics was Cefazolin (51.95%). CONCLUSION Our study found the main infection was among surgeries with score 1 of NNIS risk index and the surgical classification of endoscopy-assisted resection of space occupying lesions. We indicated specific attention should be paid to the surgeon and surgical classification with highest infection rate to control and prevent SSIs.
Collapse
Affiliation(s)
- Yifei Li
- Department of Disease Control and Prevention, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710038, Shaanxi, China
| | - Ling Gao
- Department of Disease Control and Prevention, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710038, Shaanxi, China
| | - Shanhong Fan
- Department of Disease Control and Prevention, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710038, Shaanxi, China.
| |
Collapse
|
4
|
Shirmohammadpour M, Mehrasbi MR, Noshiranzadeh N, Afshar D, Mansori K, Mirzaei B. Investigation of the effect of anti-PIA/PNAG antibodies on biofilm formation in Escherichia coli. Front Microbiol 2025; 16:1552670. [PMID: 40115191 PMCID: PMC11922938 DOI: 10.3389/fmicb.2025.1552670] [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: 01/06/2025] [Accepted: 02/24/2025] [Indexed: 03/23/2025] Open
Abstract
Polysaccharide Intercellular Adhesin (PIA), a surface polysaccharide produced by Staphylococcus aureus and Staphylococcus epidermidis, is a compelling target for opsonic and protective antibodies against these bacteria. Escherichia coli has recently made an exopolysaccharide called poly-β(1,6)-N-acetylglucosamine (PNAG), biochemically indistinguishable from PIA. This study investigated the effect of antibodies generated against PNAG on biofilm formation and the opsonization activity of secreted antibodies in Escherichia coli. Following purification and structural confirmation of PIA polysaccharide from producing Staphylococcus epidermidis, the ability to inhibit biofilm and the function of secreted antibodies for the mentioned polysaccharide were evaluated using semi-quantitative methods in a mouse model. Subsequently, the opsonic activity of antibodies targeting Escherichia coli strain ATCC 25922 was evaluated. The extracted polysaccharide was confirmed using FTIR, NMR, and colorimetric methods, and the results showed that the purified PIA induced protective antibodies with 40.48% opsonization properties in E. coli. The sera of the PIA-immunized groups showed a significant increase in antibody production and protective IgG titer levels compared to the control group. Also, the antibodies produced showed a substantial difference in inhibiting biofilm production in vitro compared to non-immunized serum. Antibodies directed against PIA with a lethality of 40.48% showed a significant effect on the absence of biofilm formation in E. coli. Despite the opsonic properties of the antibodies for E. coli, the simultaneous impact of these antibodies on infections caused by S. epidermidis and E. coli may have a role that requires further investigation and studies in animal models.
Collapse
Affiliation(s)
- Mina Shirmohammadpour
- Department of Microbiology and Virology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
- Student Research Committee, Department of Medical Microbiology and Virology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohammad Reza Mehrasbi
- Department of Environmental Health Engineering, School of Public Health, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Nader Noshiranzadeh
- Department of Chemistry, Faculty of Sciences, University of Zanjan, Zanjan, Iran
| | - Davoud Afshar
- Department of Microbiology and Virology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Kamyar Mansori
- Department of Biostatistics and Epidemiology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Bahman Mirzaei
- Department of Microbiology and Virology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
5
|
Ichii O, Nakamura T, Hiraishi M, Namba T, Rubel MZU, Umeyama T, Asai M. Application of chlorous acid water for disinfection of surgical site in dairy cows. Front Vet Sci 2025; 12:1444674. [PMID: 40078216 PMCID: PMC11898742 DOI: 10.3389/fvets.2025.1444674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 01/31/2025] [Indexed: 03/14/2025] Open
Abstract
Disinfection is crucial for preventing surgical site infections. Recently, the effectiveness of sanitizers using chlorous acid (HClO2) under conditions rich in organic matter has been reported, and chlorous acid water (CAW) has been approved as a food additive. This study evaluated the potential of CAW as a new presurgical disinfectant for cattle. The experiments were performed on the paralumbar fossa of cattle in Sapporo during March (winter to spring) and August (summer). Colony-forming units (CFUs) of standard plate count bacteria (SPCB), Enterococcus faecalis (EF), Pseudomonas aeruginosa, Escherichia coli, and Staphylococcus spp. (SP) were analyzed as indicators of bacterial load. SPCB and SP were abundantly detected, exceeding 6 log10 CFU/100 g on clipped hair and 6 log10 CFU/100 cm2 on the skin immediately after clipping, with no significant seasonal differences. The bacterial load on the skin was evaluated at three time points: after clipping, cleansing, and disinfection. Clipping and cleansing with liquid soap were common procedures, following this, either the standard disinfection protocol using 7.5% iodine scrub for 1 min, 10% povidone-iodine for 5 min, and 70% alcohol for 5 min (SPA), or a modified protocol using CAW with contact times of 15, 10, or 5 min (CAW15, CAW10, CAW5) were performed separately. The cleansing procedure significantly reduced the SPCB, EF, and SP on the skin after clipping, and all disinfection methods significantly decreased the SP after cleansing. Draping significantly enhanced the disinfection efficiency of the SPA, CAW10, and CAW5 protocols. The CAW procedure did not alter skin histology in the paralumbar fossa or udder compared to 10% povidone-iodine or 70% alcohol. Our data suggest that the disinfection method using CAW is useful and comparable to routine disinfection methods and might reduce the time required for presurgical disinfection in farm fields.
Collapse
Affiliation(s)
- Osamu Ichii
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
| | - Teppei Nakamura
- Laboratory of Laboratory Animal Science and Medicine, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Masaya Hiraishi
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Takashi Namba
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Md. Zahir Uddin Rubel
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Takuya Umeyama
- Business Innovation Department, Furukawa Sangyo Kaisha, Ltd., Tokyo, Japan
| | - Megumi Asai
- Business Innovation Department, Furukawa Sangyo Kaisha, Ltd., Tokyo, Japan
| |
Collapse
|
6
|
Ghaznavi G, Vosough P, Ghasemian A, Tabar MMM, Tayebi L, Taghizadeh S, Savardashtaki A. Engineering bacteriophages for targeted superbug eradication. Mol Biol Rep 2025; 52:221. [PMID: 39934535 DOI: 10.1007/s11033-025-10332-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
The rise of antibiotic-resistant bacteria, termed "superbugs," presents a formidable challenge to global health. These pathogens, often responsible for persistent nosocomial infections, threaten the effectiveness of conventional antibiotic therapies. This review delves into the potential of bacteriophages, viruses specifically targeting bacteria, as a powerful tool to combat superbugs. We examined the latest developments in genetic engineering that improve the efficacy of bacteriophages, focusing on modifications in host range, lysis mechanisms, and their ability to overcome bacterial defense systems. This review article highlights the CRISPR-Cas system as a promising method for precisely manipulating phage genomes, enabling the development of novel phage therapies with enhanced efficacy and specificity. Furthermore, we discussed developing novel phage-based strategies, such as phage cocktails and phage-antibiotic combinations. We also analyzed the challenges and ethical considerations associated with phage engineering, emphasizing the need for responsible and rigorous research to ensure this technology's safe and effective deployment to combat the growing threat of antibiotic resistance.
Collapse
Affiliation(s)
- Ghazal Ghaznavi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parisa Vosough
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abdolmajid Ghasemian
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | | | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, 53233, USA
| | - Saeed Taghizadeh
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
- Pharmaceutical Science Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
7
|
Kariksiz M, Kirkik D. Antibacterial evaluation of antibiotic-coated titanium and stainless steel implants in orthopaedic application: a dip-coating approach. J Orthop Surg Res 2025; 20:37. [PMID: 39794818 PMCID: PMC11724499 DOI: 10.1186/s13018-025-05467-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/07/2025] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Despite their biocompatibility, metal implants are susceptible to infections, leading to implant failure and patient complications. The purpose of this study was to investigate the antibacterial potential of antibiotic-coated titanium and stainless steel implants. METHODS The study was designed as an experimental in vitro study, and it was conducted at the Department of Immunology of the University of Health Sciences, Istanbul/ Turkiye in January and February 2024. Pieces of titanium and stainless steel were coated with gentamicin, teicoplanin, rifampicin and a combination of rifampicin and gentamicin using a dipping method. Antibacterial activity against Staphylococcus epidermidis and Staphylococcus aureus was assessed using zone of inhibition (ZOI) measurements. RESULTS Both titanium and stainless steel implants displayed significant ZOI values for all antibiotic treatments, indicating effective inhibition of bacterial growth. The combination of gentamicin and rifampicin exhibited the largest ZOI for both implants. CONCLUSIONS Antibiotic-coated titanium and stainless steel implants demonstrate promising potential for reducing implant-associated infections. Further research is needed to optimise the coating method, explore combination therapies and evaluate the long-term efficacy of this approach in clinical settings.
Collapse
Affiliation(s)
- Mesut Kariksiz
- Department of Orthopedic and traumatology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Istanbul, Türkiye.
| | - Duygu Kirkik
- Hamidiye Medicine Faculty, Department of Medical Biology, University of Health Sciences, Istanbul, Türkiye
| |
Collapse
|
8
|
Zhang K, Li Q, Gong C, Mao H, Han D. Inhibitory effect of andrographolide on the expression of key regulatory genes in Staphylococcus epidermidis biofilm formation. J Mol Histol 2024; 56:53. [PMID: 39714542 DOI: 10.1007/s10735-024-10295-1] [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: 08/27/2024] [Accepted: 10/17/2024] [Indexed: 12/24/2024]
Abstract
The purpose of this study was to explore the inhibitory effect of andrographolide on the expression of key regulatory genes involved in the biofilm formation of Staphylococcus epidermidis (SE). Taking the film-producing strain Staphylococcus epidermidis SE1457 as the research object, the effect of andrographolide on the formation of Staphylococcus epidermidis biofilms was analyzed via crystal violet staining, and biofilm models of SE adhesion, aggregation and maturity were established in vitro. RT‒PCR was used to detect the effects of the expression of icaA-, atlE-, aap- and luxS-related genes of andrographolide on biofilm formation in SE. Congo red qualitative test to evaluate the ability of andrographolide to inhibit biofilm formation of Staphylococcus epidermidis. Compared with that of the control group, the light absorption value of the low- and high-concentration andrographolide groups was significantly lower, and the light absorption value of the high-concentration andrographolide group was significantly lower than that of the low-concentration andrographolide group. The levels of key genes involved in the adhesion, aggregation and maturation of icaA, atlE, aap and luxS in group C were greater than those in group B. The biofilm-forming ability of SE in group A was strong, and the colonies were obviously black. The colony in the direction of the arrow in group B was red, and the SE biofilm was inhibited. Most of the colonies in group C were red. SE biofilms were significantly inhibited. Andrographolide inhibits SE biofilm formation, and its mechanism may involve inhibition of the expression of the related genes icaA, atlE, aap and luxS.
Collapse
Affiliation(s)
- Kangjian Zhang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan City, 250000, Shandong Province, China
| | - Qing Li
- Department of Clinical Laboratory, The Third People's Hospital of Jinan, Jinan, 250000, Shandong Province, China
| | - Chengxia Gong
- Department of Emergency, The Second Hospital of Shandong University, Jinan, 250000, Shandong Province, China
| | - Huihui Mao
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan City, 250000, Shandong Province, China
| | - Daobin Han
- Department of Clinical Laboratory, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan City, 250000, Shandong Province, China.
| |
Collapse
|
9
|
Allen-Taylor D, Boro G, Cabato P, Mai C, Nguyen K, Rijal G. Staphylococcus epidermidis biofilm in inflammatory breast cancer and its treatment strategies. Biofilm 2024; 8:100220. [PMID: 39318870 PMCID: PMC11420492 DOI: 10.1016/j.bioflm.2024.100220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/26/2024] Open
Abstract
Bacterial biofilms represent a significant challenge in both clinical and industrial settings because of their robust nature and resistance to antimicrobials. Biofilms are formed by microorganisms that produce an exopolysaccharide matrix, protecting function and supporting for nutrients. Among the various bacterial species capable of forming biofilms, Staphylococcus epidermidis, a commensal organism found on human skin and mucous membranes, has emerged as a prominent opportunistic pathogen, when introduced into the body via medical devices, such as catheters, prosthetic joints, and heart valves. The formation of biofilms by S. epidermidis on these surfaces facilitates colonization and provides protection against host immune responses and antibiotic therapies, leading to persistent and difficult-to-treat infections. The possible involvement of biofilms for breast oncogenesis has recently created the curiosity. This paper therefore delves into S. epidermidis biofilm involvement in breast cancer. S. epidermidis biofilms can create a sustained inflammatory environment through their metabolites and can break DNA in breast tissue, promoting cellular proliferation, angiogenesis, and genetic instability. Preventing biofilm formation primarily involves preventing bacterial proliferation using prophylactic measures and sterilization of medical devices and equipment. In cancer treatment, common modalities include chemotherapy, surgery, immunotherapy, alkylating agents, and various anticancer drugs. Understanding the relationship between anticancer drugs and bacterial biofilms is crucial, especially for those undergoing cancer treatment who may be at increased risk of bacterial infections, for improving patient outcomes. By elucidating these interactions, strategies to prevent or disrupt biofilm formation, thereby reducing the incidence of infections associated with medical devices and implants, can be identified.
Collapse
Affiliation(s)
- D. Allen-Taylor
- Department of Medical Laboratory Sciences, Public Health, and Nutrition Science, College of Health Sciences, Tarleton State University, a Member of Texas A & M University System, Fort Worth, Texas, 76036, USA
| | - G. Boro
- Department of Medical Laboratory Sciences, Public Health, and Nutrition Science, College of Health Sciences, Tarleton State University, a Member of Texas A & M University System, Fort Worth, Texas, 76036, USA
| | - P.M. Cabato
- Department of Medical Laboratory Sciences, Public Health, and Nutrition Science, College of Health Sciences, Tarleton State University, a Member of Texas A & M University System, Fort Worth, Texas, 76036, USA
| | - C. Mai
- Department of Medical Laboratory Sciences, Public Health, and Nutrition Science, College of Health Sciences, Tarleton State University, a Member of Texas A & M University System, Fort Worth, Texas, 76036, USA
| | - K. Nguyen
- Department of Medical Laboratory Sciences, Public Health, and Nutrition Science, College of Health Sciences, Tarleton State University, a Member of Texas A & M University System, Fort Worth, Texas, 76036, USA
| | - G. Rijal
- Department of Medical Laboratory Sciences, Public Health, and Nutrition Science, College of Health Sciences, Tarleton State University, a Member of Texas A & M University System, Fort Worth, Texas, 76036, USA
| |
Collapse
|
10
|
Dinić M, Verpile R, Burgess JL, Ming J, Marjanovic J, Beliz CN, Plano L, Hower S, Thaller SR, Banerjee S, Lev‐Tov H, Tomic‐Canic M, Pastar I. Multi-drug resistant Staphylococcus epidermidis from chronic wounds impair healing in human wound model. Wound Repair Regen 2024; 32:799-810. [PMID: 39439244 PMCID: PMC11584363 DOI: 10.1111/wrr.13231] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/13/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024]
Abstract
Venous leg ulcers (VLUs) represent one of the most prevalent types of chronic wounds characterised by perturbed microbiome and biofilm-forming bacteria. As one of the most abundant skin-commensal, Staphylococcus epidermidis is known as beneficial for the host, however, some strains can form biofilms and hinder wound healing. In this study, S. epidermidis distribution in VLUs and associated resistome were analysed in ulcer tissue from patients. Virulence of S. epidermidis isolates from VLUs were evaluated by whole genome sequencing, antimicrobial susceptibility testing, in vitro biofilm and binding assays, and assessment of biofilm-forming capability and pro-inflammatory potential using human ex vivo wound model. We demonstrated that S. epidermidis isolates from VLUs inhibit re-epithelialization through biofilm-dependent induction of IL-1β, IL-8, and IL-6 which was in accordance with impaired healing outcomes observed in patients. High extracellular matrix binding ability of VLU isolates was associated with antimicrobial resistance and expression levels of the embp and sdrG, responsible for bacterial binding to fibrinogen and fibrin, respectively. Finally, we showed that S. epidermidis from VLUs demonstrate pathogenic features with ability to impair healing which underscores the emergence of treatment-resistant virulent lineages in patients with chronic ulcers.
Collapse
Affiliation(s)
- Miroslav Dinić
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Group for Probiotics and Microbiota‐Host Interaction, Institute of Molecular Genetics and Genetic EngineeringUniversity of BelgradeBelgradeSerbia
| | - Rebecca Verpile
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Jamie L. Burgess
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Jingjing Ming
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Jelena Marjanovic
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Carmen Nicole Beliz
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Lisa Plano
- Department of Microbiology and ImmunologyUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Suzanne Hower
- Department of Microbiology and ImmunologyUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Seth R. Thaller
- Department of SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Santanu Banerjee
- Department of SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Hadar Lev‐Tov
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Marjana Tomic‐Canic
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Irena Pastar
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Department of Microbiology and ImmunologyUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| |
Collapse
|
11
|
Schoberleitner I, Lackner M, Coraça-Huber DC, Augustin A, Imsirovic A, Sigl S, Wolfram D. SMI-Capsular Fibrosis and Biofilm Dynamics: Molecular Mechanisms, Clinical Implications, and Antimicrobial Approaches. Int J Mol Sci 2024; 25:11675. [PMID: 39519227 PMCID: PMC11546664 DOI: 10.3390/ijms252111675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Silicone mammary implants (SMIs) frequently result in capsular fibrosis, which is marked by the overproduction of fibrous tissue surrounding the implant. This review provides a detailed examination of the molecular and immunological mechanisms driving capsular fibrosis, focusing on the role of foreign body responses (FBRs) and microbial biofilm formation. We investigate how microbial adhesion to implant surfaces and biofilm development contribute to persistent inflammation and fibrotic responses. The review critically evaluates antimicrobial strategies, including preoperative antiseptic protocols and antimicrobial-impregnated materials, designed to mitigate infection and biofilm-related complications. Additionally, advancements in material science, such as surface modifications and antibiotic-impregnated meshes, are discussed for their potential to reduce capsular fibrosis and prevent contracture of the capsule. By integrating molecular insights with clinical applications, this review aims to elucidate the current understanding of SMI-related fibrotic responses and highlight knowledge gaps. The synthesis of these findings aims to guide future research directions of improved antimicrobial interventions and implant materials, ultimately advancing the management of capsular fibrosis and enhancing patient outcomes.
Collapse
Affiliation(s)
- Ines Schoberleitner
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Michaela Lackner
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Débora C. Coraça-Huber
- BIOFILM Lab, Department of Orthopedics and Traumatology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Angela Augustin
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Anja Imsirovic
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Stephan Sigl
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Dolores Wolfram
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
12
|
Virzì NF, Greco V, Stracquadanio S, Jasim A, Greish K, Diaz-Rodriguez P, Rotondo NP, Stefani S, Pittalà V, Giuffrida A. Berberine-styrene- co-maleic acid nanomicelles: unlocking opportunities for the treatment and prevention of bacterial infections. RSC Adv 2024; 14:34066-34080. [PMID: 39469023 PMCID: PMC11513620 DOI: 10.1039/d4ra04457f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/12/2024] [Indexed: 10/30/2024] Open
Abstract
The global spread of multi-drug-resistant (MDR) bacteria is rapidly increasing due to antibiotic overuse, posing a major public health threat and causing millions of deaths annually. The present study explored the potential of nanocarriers for delivering novel and alternative antibacterial agents using nanotechnology-based approaches to address the challenge of MDR bacteria. The purpose was to enhance the solubility, stability, and targeted delivery of berberine (BER) and its synthetic derivative NR16 using Styrene-co-Maleic Acid (SMA) nanoparticles. Characterization of the nanoparticles, including dynamic light scattering (DLS) analysis, TEM, and UV/Vis absorption spectroscopy, confirmed their suitability and high stability for passive drug delivery. Antibacterial and antifungal activities were evaluated against a panel of pathogens, revealing significant inhibitory effects on Gram-positive strains; particularly BER, SMA-BER, and NR16 were active against MRSA, MSSA, VR, and VS E. faecalis, and S. epidermidis. Additionally, SMA-BER and SMA-NR16 showed promising activity against biofilm formation of S. epidermidis; while the two free drugs contributed to S. epidermidis biofilm disruption activity. Hemolysis tests and in vitro studies on human embryonic kidney cells (HEK-293) confirmed the safety profiles of the nanoparticles and free drugs. Overall, this research highlighted the potential of nanotechnology in developing effective antibacterial agents with reduced toxicity, addressing the growing threat of MDR bacterial infections.
Collapse
Affiliation(s)
- Nicola F Virzì
- Department of Drug and Health Sciences, University of Catania 95125 Catania Italy
| | - Valentina Greco
- Department of Chemical Sciences, University of Catania 95125 Catania Italy
| | - Stefano Stracquadanio
- Section of Microbiology, Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania 95123 Catania Italy
| | - Anfal Jasim
- Department of Molecular Medicine, Arabian Gulf University Manama 329 Bahrain
| | - Khaled Greish
- Department of Molecular Medicine, Arabian Gulf University Manama 329 Bahrain
| | - Patricia Diaz-Rodriguez
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Faculty of Pharmacy, Instituto de Materiales (iMATUS), Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela 15782 Santiago de Compostela Spain
| | - Natalie P Rotondo
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro 70125 Bari Italy
| | - Stefania Stefani
- Section of Microbiology, Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania 95123 Catania Italy
| | - Valeria Pittalà
- Department of Drug and Health Sciences, University of Catania 95125 Catania Italy
- Department of Molecular Medicine, Arabian Gulf University Manama 329 Bahrain
| | | |
Collapse
|
13
|
Dhadwal S, Handa S, Chatterjee M, Banat IM. Sophorolipid: An Effective Biomolecule for Targeting Microbial Biofilms. Curr Microbiol 2024; 81:388. [PMID: 39367190 DOI: 10.1007/s00284-024-03892-6] [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: 01/28/2024] [Accepted: 09/09/2024] [Indexed: 10/06/2024]
Abstract
Biofilms are microbial aggregates encased in a matrix that is attached to biological or nonbiological surfaces and constitute serious problems in food, medical, and marine industries and can have major negative effects on both health and the economy. Biofilm's complex microbial community provides a resistant environment that is difficult to eradicate and is extremely resilient to antibiotics and sanitizers. There are various conventional techniques for combating biofilms, including, chemical removal, physical or mechanical removal, use of antibiotics and disinfectants to destroy biofilm producing organisms. In contrast to free living planktonic cells, biofilms are very resistant to these methods. Hence, new strategies that differ from traditional approaches are urgently required. Microbial world offers a wide range of effective "green" compounds such as biosurfactants. They outperform synthetic surfactants in terms of biodegradability, superior stabilization, and reduced toxicity concerns. They also have better antiadhesive and anti-biofilm capabilities which can be used to treat biofilm-related problems. Sophorolipids (SLs) are a major type of biosurfactants that have gained immense interest in the healthcare industries because of their antiadhesive and anti-biofilm properties. Sophorolipids may therefore prove to be attractive substances that can be used in biomedical applications as adjuvant to other antibiotics against some infections through growth inhibition and/or biofilm disruption.
Collapse
Affiliation(s)
- Sunidhi Dhadwal
- Biotechnology Branch, University Institute of Engineering and Technology, Sector-25, South Campus, Panjab University, Chandigarh, 160014, India
| | - Shristi Handa
- Biotechnology Branch, University Institute of Engineering and Technology, Sector-25, South Campus, Panjab University, Chandigarh, 160014, India
| | - Mary Chatterjee
- Biotechnology Branch, University Institute of Engineering and Technology, Sector-25, South Campus, Panjab University, Chandigarh, 160014, India.
| | - Ibrahim M Banat
- Faculty of Life & Health Sciences, University of Ulster, Coleraine, BT52 1SA, UK.
| |
Collapse
|
14
|
Pedretti N, Iseppi R, Condò C, Ghazanfar S, Messi P, Di Cerbo A, Sabia C. Characterization of virulence factors and antimicrobial resistance in Staphylococcus spp. isolated from clinical samples. Folia Microbiol (Praha) 2024; 69:1043-1052. [PMID: 38367164 DOI: 10.1007/s12223-024-01148-1] [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: 11/27/2023] [Accepted: 02/01/2024] [Indexed: 02/19/2024]
Abstract
The virulence factors, antibiotic resistance patterns, and the associated genetic elements have been investigated in Staphylococcus species. A total of 100 strains has been isolated from clinical samples in the Microbiology Laboratory of Hesperia Hospital, Modena, Italy, and identified as Staphylococcus aureus (65), Staphylococcus epidermidis (24), Staphylococcus hominis (3), Staphylococcus saprophyticus (3), and Staphylococcus warneri (5). All the strains were analyzed to determine phenotypic and genotypic characters, notably the virulence factors, the antibiotics susceptibility, and the genetic determinants. The highest percentage of resistance in Staphylococcus spp. was found for erythromycin and benzylpenicillin (87% and 85%, respectively). All S. aureus, two S. epidermidis (8.3%), and one S. saprophyticus (33.3%) strains were resistant to oxacillin. The methicillin resistance gene (mecA) was detected by polymerase chain reaction (PCR) amplification in 65 S. aureus strains and in 3 coagulase-negative staphylococci (CoNS) (8.6%). With regard to the virulence characteristics, all the S. aureus were positive to all virulence tests, except for slime test. Among the CoNS isolates, 19 (79.1%) S. epidermidis and one (33.3%) S. saprophyticus strains resulted positive for the slime test only. The results obtained are useful for a more in-depth understanding of the function and contribution of S. aureus and CoNS antibiotic resistance and virulence factors to staphylococcal infections. In particular, the production of slime is very important for CoNS, a virulence factor frequently found in infections caused by these strains. Further investigations on the genetic relatedness among strains of different sources will be useful for epidemiological and monitoring purposes and will enable us to develop new strategies to counteract the diffusion of methicillin-resistant S. aureus (MRSA) and CoNS strains not only in clinical field, but also in other related environments.
Collapse
Affiliation(s)
- Natalia Pedretti
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Ramona Iseppi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125, Modena, Italy
| | - Carla Condò
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125, Modena, Italy
| | - Shakira Ghazanfar
- National Institute for Genomics Advanced and Biotechnology (NIGAB), National Agricultural Research Centre, Park Road, 45500, Islamabad, Pakistan
| | - Patrizia Messi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125, Modena, Italy
| | - Alessandro Di Cerbo
- School of Biosciences and Veterinary Medicine, University of Camerino, 62024, Matelica, Italy
| | - Carla Sabia
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125, Modena, Italy.
| |
Collapse
|
15
|
Ronish LA, Biswas B, Bauer RM, Jacob ME, Piepenbrink KH. The role of extracellular structures in Clostridioides difficile biofilm formation. Anaerobe 2024; 88:102873. [PMID: 38844261 DOI: 10.1016/j.anaerobe.2024.102873] [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: 12/26/2023] [Revised: 04/27/2024] [Accepted: 06/03/2024] [Indexed: 07/08/2024]
Abstract
C. difficile infection (CDI) is a costly and increasing burden on the healthcare systems of many developed countries due to the high rates of nosocomial infections. Despite the availability of several antibiotics with high response rates, effective treatment is hampered by recurrent infections. One potential mechanism for recurrence is the existence of C. difficile biofilms in the gut which persist through the course of antibiotics. In this review, we describe current developments in understanding the molecular mechanisms by which C. difficile biofilms form and are stabilized through extracellular biomolecular interactions.
Collapse
Affiliation(s)
- Leslie A Ronish
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Baishakhi Biswas
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Robert M Bauer
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Mallory E Jacob
- Biochemistry Department, University of Geneva, Geneva, Switzerland
| | - Kurt H Piepenbrink
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA; Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA; Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA; Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA; Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA.
| |
Collapse
|
16
|
Zhang Y, Zhao X, Wang J, Liao L, Qin H, Zhang R, Li C, He Y, Huang S. VmsR, a LuxR-Type Regulator, Contributes to Virulence, Cell Motility, Extracellular Polysaccharide Production and Biofilm Formation in Xanthomonas oryzae pv. oryzicola. Int J Mol Sci 2024; 25:7595. [PMID: 39062838 PMCID: PMC11277528 DOI: 10.3390/ijms25147595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
LuxR-type regulators play pivotal roles in regulating numerous bacterial processes, including bacterial motility and virulence, thereby exerting a significant influence on bacterial behavior and pathogenicity. Xanthomonas oryzae pv. oryzicola, a rice pathogen, causes bacterial leaf streak. Our research has identified VmsR, which is a response regulator of the two-component system (TCS) that belongs to the LuxR family. These findings of the experiment reveal that VmsR plays a crucial role in regulating pathogenicity, motility, biofilm formation, and the production of extracellular polysaccharides (EPSs) in Xoc GX01. Notably, our study shows that the vmsR mutant exhibits a reduced swimming motility but an enhanced swarming motility. Furthermore, this mutant displays decreased virulence while significantly increasing EPS production and biofilm formation. We have uncovered that VmsR directly interacts with the promoter regions of fliC and fliS, promoting their expression. In contrast, VmsR specifically binds to the promoter of gumB, resulting in its downregulation. These findings indicate that the knockout of vmsR has profound effects on virulence, motility, biofilm formation, and EPS production in Xoc GX01, providing insights into the intricate regulatory network of Xoc.
Collapse
Affiliation(s)
- Yaqi Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, China; (Y.Z.); (X.Z.); (J.W.); (L.L.); (H.Q.); (R.Z.); (C.L.)
| | - Xiyao Zhao
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, China; (Y.Z.); (X.Z.); (J.W.); (L.L.); (H.Q.); (R.Z.); (C.L.)
| | - Jiuxiang Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, China; (Y.Z.); (X.Z.); (J.W.); (L.L.); (H.Q.); (R.Z.); (C.L.)
| | - Lindong Liao
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, China; (Y.Z.); (X.Z.); (J.W.); (L.L.); (H.Q.); (R.Z.); (C.L.)
| | - Huajun Qin
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, China; (Y.Z.); (X.Z.); (J.W.); (L.L.); (H.Q.); (R.Z.); (C.L.)
| | - Rongbo Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, China; (Y.Z.); (X.Z.); (J.W.); (L.L.); (H.Q.); (R.Z.); (C.L.)
| | - Changyu Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, China; (Y.Z.); (X.Z.); (J.W.); (L.L.); (H.Q.); (R.Z.); (C.L.)
| | - Yongqiang He
- College of Agronomy, Guangxi University, Nanning 530004, China
| | - Sheng Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, China; (Y.Z.); (X.Z.); (J.W.); (L.L.); (H.Q.); (R.Z.); (C.L.)
| |
Collapse
|
17
|
Xi H, Luo Z, Liu MF, Chen Q, Zhu Q, Yuan L, Sheng YY, Zhao R. Diclofenac sodium effectively inhibits the biofilm formation of Staphylococcus epidermidis. Arch Microbiol 2024; 206:289. [PMID: 38847838 DOI: 10.1007/s00203-024-04020-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 05/27/2024] [Indexed: 08/07/2024]
Abstract
Staphylococcus epidermidis is an opportunistic pathogen commonly implicated in medical device-related infections. Its propensity to form biofilms not only leads to chronic infections but also exacerbates the issue of antibiotic resistance, necessitating high-dose antimicrobial treatments. In this study, we explored the use of diclofenac sodium, a non-steroidal anti-inflammatory drug, as an anti-biofilm agent against S. epidermidis. In this study, crystal violet staining and confocal laser scanning microscope analysis showed that diclofenac sodium, at subinhibitory concentration (0.4 mM), significantly inhibited biofilm formation in both methicillin-susceptible and methicillin-resistant S. epidermidis isolates. MTT assays demonstrated that 0.4 mM diclofenac sodium reduced the metabolic activity of biofilms by 25.21-49.01% compared to untreated controls. Additionally, the treatment of diclofenac sodium resulted in a significant decrease (56.01-65.67%) in initial bacterial adhesion, a crucial early phase of biofilm development. Notably, diclofenac sodium decreased the production of polysaccharide intercellular adhesin (PIA), a key component of the S. epidermidis biofilm matrix, in a dose-dependent manner. Real-time quantitative PCR analysis revealed that diclofenac sodium treatment downregulated biofilm-associated genes icaA, fnbA, and sigB and upregulated negative regulatory genes icaR and luxS, providing potential mechanistic insights. These findings indicate that diclofenac sodium inhibits S. epidermidis biofilm formation by affecting initial bacterial adhesion and the PIA synthesis. This underscores the potential of diclofenac sodium as a supplementary antimicrobial agent in combating staphylococcal biofilm-associated infections.
Collapse
Affiliation(s)
- Huimin Xi
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, PR China
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, PR China
| | - Zhaoxia Luo
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, PR China
| | - Mei-Fang Liu
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, PR China
| | - Qiang Chen
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, PR China
| | - Qing Zhu
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, PR China
| | - Lei Yuan
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, PR China
| | - Yi-Yun Sheng
- Department of Pathology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, PR China.
| | - Rui Zhao
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, PR China.
| |
Collapse
|
18
|
Wongchai M, Wongkaewkhiaw S, Kanthawong S, Roytrakul S, Aunpad R. Dual-function antimicrobial-antibiofilm peptide hybrid to tackle biofilm-forming Staphylococcus epidermidis. Ann Clin Microbiol Antimicrob 2024; 23:44. [PMID: 38755634 PMCID: PMC11100219 DOI: 10.1186/s12941-024-00701-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/28/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Due to their resistance and difficulty in treatment, biofilm-associated infections are problematic among hospitalized patients globally and account for 60% of all bacterial infections in humans. Antibiofilm peptides have recently emerged as an alternative treatment since they can be effectively designed and exert a different mode of biofilm inhibition and eradication. METHODS A novel antibiofilm peptide, BiF, was designed from the conserved sequence of 18 α-helical antibiofilm peptides by template-assisted technique and its activity was improved by hybridization with a lipid binding motif (KILRR). Novel antibiofilm peptide derivatives were modified by substituting hydrophobic amino acids at positions 5 or 7, and both, with positively charged lysines (L5K, L7K). These peptide derivatives were tested for antibiofilm and antimicrobial activities against biofilm-forming Staphylococcus epidermidis and multiple other microbes using crystal violet and broth microdilution assays, respectively. To assess their impact on mammalian cells, the toxicity of peptides was determined through hemolysis and cytotoxicity assays. The stability of candidate peptide, BiF2_5K7K, was assessed in human serum and its secondary structure in bacterial membrane-like environments was analyzed using circular dichroism. The action of BiF2_5K7K on planktonic S. epidermidis and its effect on biofilm cell viability were assessed via viable counting assays. Its biofilm inhibition mechanism was investigated through confocal laser scanning microscopy and transcription analysis. Additionally, its ability to eradicate mature biofilms was examined using colony counting. Finally, a preliminary evaluation involved coating a catheter with BiF2_5K7K to assess its preventive efficacy against S. epidermidis biofilm formation on the catheter and its surrounding area. RESULTS BiF2_5K7K, the modified antibiofilm peptide, exhibited dose-dependent antibiofilm activity against S. epidermidis. It inhibited biofilm formation at subinhibitory concentrations by altering S. epidermidis extracellular polysaccharide production and quorum-sensing gene expression. Additionally, it exhibited broad-spectrum antimicrobial activity and no significant hemolysis or toxicity against mammalian cell lines was observed. Its activity is retained when exposed to human serum. In bacterial membrane-like environments, this peptide formed an α-helix amphipathic structure. Within 4 h, a reduction in the number of S. epidermidis colonies was observed, demonstrating the fast action of this peptide. As a preliminary test, a BiF2_5K7K-coated catheter was able to prevent the development of S. epidermidis biofilm both on the catheter surface and in its surrounding area. CONCLUSIONS Due to the safety and effectiveness of BiF2_5K7K, we suggest that this peptide be further developed to combat biofilm infections, particularly those of biofilm-forming S. epidermidis.
Collapse
Affiliation(s)
- Mathira Wongchai
- Graduate Program in Biomedical Sciences, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, Thailand
| | - Saharut Wongkaewkhiaw
- School of Dentistry, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand
| | - Sakawrat Kanthawong
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Ratchaneewan Aunpad
- Graduate Program in Biomedical Sciences, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, Thailand.
| |
Collapse
|
19
|
van Hal SJ, Jensen SO, Tong SYC, Bentley S, Holden MT. Unravelling the complex interplay between antibiotic consumption and adaptive changes in methicillin-resistant Staphylococcus aureus. J Antimicrob Chemother 2024; 79:891-896. [PMID: 38412336 DOI: 10.1093/jac/dkae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/29/2024] [Indexed: 02/29/2024] Open
Abstract
OBJECTIVES This study aims to elucidate the genomic dynamics driving the emergence of antimicrobial resistance (AMR), with a specific focus on the interplay between AMR and antimicrobial usage. METHODS We conducted a comprehensive analysis using a ST239 methicillin-resistant Staphylococcus aureus (MRSA) dataset over a continuous 12-year period from a single hospital. Genomic analyses were performed tracking the changes in MRSA populations, particularly the emergence of reduced vancomycin susceptibility, and assessing the impact of glycopeptide use on these emergence events. RESULTS Our findings reveal a significant correlation between hospital glycopeptide usage and the selection of MRSA strains with reduced vancomycin susceptibility. Genomic analyses provided insights into the molecular mechanisms driving resistance emergence, including the slowing of the molecular clock rate in response to heightened antimicrobial consumption. CONCLUSIONS In conclusion, this study the highlights the complex dynamics between AMR and antimicrobial use at the hospital level. The observed correlation between antimicrobial consumption and the development of less susceptible MRSA strains underscores the importance of antimicrobial stewardship programmes and the establishment of optimal consumption thresholds for mitigating AMR effectively.
Collapse
Affiliation(s)
- Sebastiaan J van Hal
- Department of Microbiology and Infectious Diseases, Royal Prince Alfred Hospital, Sydney, Australia
- Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
- Antimicrobial Resistance and Mobile Elements Group, Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
| | - Slade O Jensen
- Antimicrobial Resistance and Mobile Elements Group, Ingham Institute for Applied Medical Research, Sydney, NSW, Australia
- Microbiology and Infectious Diseases, School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | - Stephen Y C Tong
- Victorian Infectious Diseases Service, The Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Stephen Bentley
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Matthew T Holden
- School of Medicine, University of St Andrews, St Andrews, Fife KY16 9TF, UK
| |
Collapse
|
20
|
Schoberleitner I, Baier L, Lackner M, Zenz LM, Coraça-Huber DC, Ullmer W, Damerum A, Faserl K, Sigl S, Steinkellner T, Winkelmann S, Sarg B, Egle D, Brunner C, Wolfram D. Surface Topography, Microbial Adhesion, and Immune Responses in Silicone Mammary Implant-Associated Capsular Fibrosis. Int J Mol Sci 2024; 25:3163. [PMID: 38542137 PMCID: PMC10969816 DOI: 10.3390/ijms25063163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 11/11/2024] Open
Abstract
Breast cancer is the most common cancer in women globally, often necessitating mastectomy and subsequent breast reconstruction. Silicone mammary implants (SMIs) play a pivotal role in breast reconstruction, yet their interaction with the host immune system and microbiome remains poorly understood. This study investigates the impact of SMI surface topography on host antimicrobial responses, wound proteome dynamics, and microbial colonization. Biological samples were collected from ten human patients undergoing breast reconstruction with SMIs. Mass spectrometry profiles were analyzed for acute and chronic wound proteomes, revealing a nuanced interplay between topography and antimicrobial response proteins. 16S rRNA sequencing assessed microbiome dynamics, unveiling topography-specific variations in microbial composition. Surface topography alterations influenced wound proteome composition. Microbiome analysis revealed heightened diversity around rougher SMIs, emphasizing topography-dependent microbial invasion. In vitro experiments confirmed staphylococcal adhesion, growth, and biofilm formation on SMI surfaces, with increased texture correlating positively with bacterial colonization. This comprehensive investigation highlights the intricate interplay between SMI topography, wound proteome dynamics, and microbial transmission. The findings contribute to understanding host-microbe interactions on SMI surfaces, essential for optimizing clinical applications and minimizing complications in breast reconstruction.
Collapse
Affiliation(s)
- Ines Schoberleitner
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Leoni Baier
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Michaela Lackner
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Lisa-Maria Zenz
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Débora C. Coraça-Huber
- BIOFILM Lab, Department of Orthopedics and Traumatology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Wendy Ullmer
- Zymo Research Corp., Irvine, CA 92614, USA
- Pangea Laboratory, Tustin, CA 92614, USA
| | | | - Klaus Faserl
- Protein Core Facility, Institute of Medical Chemistry, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Stephan Sigl
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Theresia Steinkellner
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Selina Winkelmann
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Bettina Sarg
- Protein Core Facility, Institute of Medical Chemistry, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Daniel Egle
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Christine Brunner
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Dolores Wolfram
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
21
|
Almutleb ES, Ramachandran S, Khan AA, El-Hiti GA, Alanazi SA. Synergistic Effect of Nilavembu Choornam-Gold Nanoparticles on Antibiotic-Resistant Bacterial Susceptibility and Contact Lens Contamination-Associated Infectious Pathogenicity. Int J Mol Sci 2024; 25:2115. [PMID: 38396792 PMCID: PMC10889799 DOI: 10.3390/ijms25042115] [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: 11/30/2023] [Revised: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Antibiotic-resistant bacterial colonies mitigate rapid biofilm formation and have complex cell wall fabrications, making it challenging to penetrate drugs across their biofilm barriers. The objective of this study was to investigate the antibacterial susceptibility of antibiotic-resistant bacteria and contact lens barrenness. Nilavembu Choornam-Gold Nanoparticles (NC-GNPs) were synthesized using NC polyherbal extract and characterized by UV-visible spectrophotometer, SEM-EDX, XRD, Zeta sizer, FTIR, and TEM analysis. Contact lenses with overnight cultures of antibiotic-resistant bacteria K. pneumoniae and S. aureus showed significant differences in growth, biofilm formation, and infection pathogenicity. The NC-GNPs were observed in terms of size (average size is 57.6 nm) and surface chemistry. A zone of inhibition was calculated for K. pneumoniae 18.8 ± 1.06, S. aureus 23.6 ± 1.15, P. aeruginosa 24.16 ± 0.87, and E. faecalis 24.5 ± 1.54 mm at 24 h of NC-GNPs alone treatment. In electron microscopy studies, NC-GNP-treated groups showed nuclear shrinkage, nuclear disintegration, degeneration of cell walls, and inhibited chromosomal division. In contrast, normal bacterial colonies had a higher number of cell divisions and routinely migrated toward cell multiplications. NC-GNPs exhibited antibacterial efficacy against antibiotic-resistant bacteria when compared to NC extract alone. We suggest that NC-GNPs are highly valuable to the population of hospitalized patients and other people to reduce the primary complications of contact lens contamination-oriented microbial infection and the therapeutic efficiency of antibiotic-resistant bacterial pathogenicity.
Collapse
Affiliation(s)
| | - Samivel Ramachandran
- Cornea Research Chair, Department of Optometry, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia; (E.S.A.); (A.A.K.); (G.A.E.-H.); (S.A.A.)
| | | | | | | |
Collapse
|
22
|
Sekar A, Gil D, Tierney P, McCanne M, Daesety V, Trendafilova D, Muratoglu OK, Oral E. Synergistic use of anti-inflammatory ketorolac and gentamicin to target staphylococcal biofilms. J Transl Med 2024; 22:102. [PMID: 38273276 PMCID: PMC10809490 DOI: 10.1186/s12967-024-04871-y] [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: 10/19/2023] [Accepted: 01/08/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND While antibiotics remain our primary tools against microbial infection, increasing antibiotic resistance (inherent and acquired) is a major detriment to their efficacy. A practical approach to maintaining or reversing the efficacy of antibiotics is the use of other commonly used therapeutics, which show synergistic antibacterial action with antibiotics. Here, we investigated the extent of antibacterial synergy between the antibiotic gentamicin and the anti-inflammatory ketorolac regarding the dynamics of biofilm growth, the rate of acquired resistance, and the possible mechanism of synergy. METHODS Control (ATCC 12600, ATCC 35984) and clinical strains (L1101, L1116) of Staphylococcus aureus and Staphylococcus epidermidis with varying antibiotic susceptibility profiles were used in this study to simulate implant-material associated low-risk and high-risk biofilms in vitro. The synergistic action of gentamicin sulfate (GS) and ketorolac tromethamine (KT), against planktonic staphylococcal strains were determined using the fractional inhibitory concentration measurement assay. Nascent (6 h) and established (24 h) biofilms were grown on 316L stainless steel plates and the synergistic biofilm eradication activity was determined and characterized using adherent bacteria count, minimum biofilm eradication concentration (MBEC) measurement for GS, visualization by live/dead imaging, scanning electron microscopy, gene expression of biofilm-associated genes, and bacterial membrane fluidity assessment. RESULTS Gentamicin-ketorolac (GS-KT) combination demonstrated synergistic antibacterial action against planktonic Staphylococci. Control and clinical strains showed distinct biofilm growth dynamics and an increase in biofilm maturity was shown to confer further resistance to gentamicin for both 'low-risk' and 'high-risk' biofilms. The addition of ketorolac enhanced the antibiofilm activity of gentamicin against acquired resistance in staphylococcal biofilms. Mechanistic studies revealed that the synergistic action of gentamicin-ketorolac interferes with biofilm morphology and subverts bacterial stress response altering bacterial physiology, membrane dynamics, and biofilm properties. CONCLUSION The results of this study have a significant impact on the local administration of antibiotics and other therapeutic agents commonly used in the prevention and treatment of orthopaedic infections. Further, these results warrant the study of synergy for the concurrent or sequential administration of non-antibiotic drugs for antimicrobial effect.
Collapse
Affiliation(s)
- Amita Sekar
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, USA
- Department of Orthopaedic Surgery, Harvard Medical School, Harvard University, Boston, USA
| | - Dmitry Gil
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, USA
- Department of Orthopaedic Surgery, Harvard Medical School, Harvard University, Boston, USA
| | - Peyton Tierney
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, USA
| | - Madeline McCanne
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, USA
| | - Vikram Daesety
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, USA
| | | | - Orhun K Muratoglu
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, USA
- Department of Orthopaedic Surgery, Harvard Medical School, Harvard University, Boston, USA
| | - Ebru Oral
- Harris Orthopaedic Laboratory, Massachusetts General Hospital, Boston, USA.
- Department of Orthopaedic Surgery, Harvard Medical School, Harvard University, Boston, USA.
| |
Collapse
|
23
|
Gómez-Alonso IS, Betanzos-Cabrera G, Moreno-Lafont MC, Cancino-Diaz ME, García-Pérez BE, Cancino-Diaz JC. Non-biofilm-forming Staphylococcus epidermidis planktonic cell supernatant induces alterations in osteoblast biological function. Sci Rep 2024; 14:1807. [PMID: 38245549 PMCID: PMC10799936 DOI: 10.1038/s41598-024-51899-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/10/2024] [Indexed: 01/22/2024] Open
Abstract
Staphylococcal biofilms significantly contribute to prosthetic joint infection (PJI). However, 40% of S. epidermidis PJI isolates do not produce biofilms, which does not explain the role of biofilms in these cases. We studied whether the supernatant from planktonic S. epidermidis alters osteoblast function. Non-biofilm-forming S. epidermidis supernatants (PJI- clinical isolate, healthy skin isolate (HS), and ATCC12228 reference strain) and biofilm-forming supernatants (PJI+ clinical isolate, ATCC35984 reference strain, and Staphylococcus aureus USA300 reference strain) were included. Osteoblasts stimulated with supernatants from non-biofilm-forming isolates for 3, 7, and 14 days showed significantly reduced cellular DNA content compared with unstimulated osteoblasts, and apoptosis was induced in these osteoblasts. Similar results were obtained for biofilm-forming isolates, but with a greater reduction in DNA content and higher apoptosis. Alkaline phosphatase activity and mineralization were significantly reduced in osteoblasts treated with supernatants from non-biofilm-forming isolates compared to the control at the same time points. However, the supernatants from biofilm-forming isolates had a greater effect than those from non-biofilm-forming isolates. A significant decrease in the expression of ATF4, RUNX2, ALP, SPARC, and BGLAP, and a significant increase in RANK-L expression were observed in osteoblasts treated with both supernatants. These results demonstrate that the supernatants of the S. epidermidis isolate from the PJI- and HS (commensal) with a non-biofilm-forming phenotype alter the function of osteoblasts (apoptosis induction, failure of cell differentiation, activation of osteoblasts, and induction of bone resorption), similar to biofilm-forming isolates (PJI+, ATCC35984, and S. aureus USA300), suggesting that biofilm status contributes to impaired osteoblast function and that the planktonic state can do so independently of biofilm production.
Collapse
Affiliation(s)
- Itzia Sidney Gómez-Alonso
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Manuel Carpio, Plutarco Elías Calles, Miguel Hidalgo, 11350, Mexico City, Mexico
| | - Gabriel Betanzos-Cabrera
- Área Académica de Nutrición, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Carretera Pachuca-Actopan Camino a Tilcuautla S/N., Pueblo San Juan Tilcuautla, 42160, Pachuca Hidalgo, Mexico
| | - Martha Cecilia Moreno-Lafont
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Manuel Carpio, Plutarco Elías Calles, Miguel Hidalgo, 11350, Mexico City, Mexico
| | - Mario Eugenio Cancino-Diaz
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Manuel Carpio, Plutarco Elías Calles, Miguel Hidalgo, 11350, Mexico City, Mexico
| | - Blanca Estela García-Pérez
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Manuel Carpio, Plutarco Elías Calles, Miguel Hidalgo, 11350, Mexico City, Mexico
| | - Juan Carlos Cancino-Diaz
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Manuel Carpio, Plutarco Elías Calles, Miguel Hidalgo, 11350, Mexico City, Mexico.
| |
Collapse
|
24
|
Gustafsson L, Wikström C, Mueller RS, Bergvall K. Microbes on Clipper Blades after Use and Disinfection in Small Animal- and Equine Practice. Vet Sci 2024; 11:38. [PMID: 38250944 PMCID: PMC10819727 DOI: 10.3390/vetsci11010038] [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: 12/11/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/23/2024] Open
Abstract
Clipping hair on animals can produce microtraumas of the skin and the dislodgement of microorganisms to the clipper blade. This study evaluates if clipper blades in animal hospitals in Sweden are contaminated with bacteria and/or dermatophytes after disinfection. Eleven clipper blades from three veterinary referral hospitals, including one with a small animal department and an equine department, were sampled for bacteria and dermatophytes. All the hospitals had disinfection routines in accordance with the national recommendations for hygiene in veterinary medicine. The sampled clipper blades were supposed to be disinfected and they were considered to be ready for use by staff. Five sterilized clipper blades were used as controls. The results showed that 64-100% of the disinfected clipper blades, from all three hospitals, were contaminated with bacteria, whereas all the sterilized clipper blades were negative for bacterial growth (p < 0.05). One clipper blade from the equine department was contaminated with dermatophytes. The results indicate that the disinfection routines were not sufficient for removing bacteria from used clipper blades, and that sterilization would be a more reliable way to minimize the risk of contamination.
Collapse
Affiliation(s)
- Lina Gustafsson
- Evidensia Södra Djursjukhuset, Månskärsvägen 13, 141 75 Huddinge, Sweden;
| | | | - Ralf S. Mueller
- Centre for Clinical Veterinary Medicine, LMU Munich, Veterinärstrasse 13, 80539 Munich, Germany
| | - Kerstin Bergvall
- Evidensia Södra Djursjukhuset, Månskärsvägen 13, 141 75 Huddinge, Sweden;
- Department of Small Animal Clinical Sciences, Faculty of Veterinary Medicine and Animal Science Swedish, University of Agricultural Sciences, P.O. Box 7070, 750 07 Uppsala, Sweden
| |
Collapse
|
25
|
Hoyos C, Qian X, Matos CD, Gabr M, Hincapie D, Cadigan JB, Steiger N, Diaz JC, Sauer W, Romero JE. Measures to Prevent Infection in Cardiac Implantable Electronic Device Replacements or Upgrades. Rev Cardiovasc Med 2024; 25:19. [PMID: 39077641 PMCID: PMC11262353 DOI: 10.31083/j.rcm2501019] [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: 10/20/2023] [Revised: 11/03/2023] [Accepted: 11/21/2023] [Indexed: 07/31/2024] Open
Abstract
Cardiac implantable electronic device (CIED) infections represent one of the most threatening complications associated with device implantation, due to an increase in morbidity and mortality rates, as well as healthcare costs. Besides, it is important to highlight that when compared to the initial implantation of a device, the risks associated with procedures like generator changes, lead and pocket revisions, or device upgrades double. Consequently, to address this issue, various scoring systems, like the PADIT (Prior Procedures, Age, Depressed Renal Function, Immunocompromised Status, Type of Procedure), the RI-AIAC (Ricerca Sulle Infezioni Associate a ImpiAnto o Sostituzione di CIED), and the Shariff score, along with predictive models, have been developed to identify patients at a greater risk of infection. Moreover, several interventions have been assessed to evaluate their role in infection prevention ranging from improving skin preparation and surgical techniques to considering alternative strategies such as the subcutaneous Implantable Cardioverter-Defibrillator (ICD). Methods like antimicrobial prophylaxis, pocket irrigation, chlorhexidine gluconate pocket lavage, capsulectomy, and the use of antibacterial envelopes have been also explored as preventive measures. In this review, we provide a comprehensive assessment of CIED infections in patients undergoing repeat procedures and the strategies designed to reduce the risk of these infections.
Collapse
Affiliation(s)
- Carolina Hoyos
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xiaoxiao Qian
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Carlos D. Matos
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mohamed Gabr
- Cardiac Arrhythmia Center, Montefiore-Einstein Center for Heart and Vascular Care, Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | - Daniela Hincapie
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - John B. Cadigan
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Nathaniel Steiger
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Juan C. Diaz
- Electrophysiology and Cardiac Arrhythmia Service, Clinica Las Vegas, Universidad CES School of Medicine, 050022 Medellin, Colombia
| | - William Sauer
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jorge E. Romero
- Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
26
|
Radenkovs V, Valdovska A, Galina D, Cairns S, Jakovlevs D, Gaidukovs S, Cinkmanis I, Juhnevica-Radenkova K. Elaboration of Nanostructured Levan-Based Colloid System as a Biological Alternative with Antimicrobial Activity for Applications in the Management of Pathogenic Microorganisms. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2969. [PMID: 37999323 PMCID: PMC10674346 DOI: 10.3390/nano13222969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/05/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023]
Abstract
Considering the documented health benefits of bacterial exopolysaccharides (EPSs), specifically of bacterial levan (BL), including its intrinsic antimicrobial activity against certain pathogenic species, the current study concentrated on the development of active pharmaceutical ingredients (APIs) in the form of colloid systems (CoSs) containing silver nanoparticles (AgNPs) employing in-house biosynthesized BL as a reducing and capping agent. The established protocol of fermentation conditions implicating two species of lactic acid bacteria (LAB), i.e., Streptococcus salivarius K12 and Leuconostoc mesenteroides DSM 20343, ensured a yield of up to 25.7 and 13.7 g L-1 of BL within 72 h, respectively. An analytical approach accomplished by Fourier-transform infrared (FT-IR) spectroscopy allowed for the verification of structural features attributed to biosynthesized BL. Furthermore, scanning electron microscopy (SEM) revealed the crystalline morphology of biosynthesized BL with a smooth and glossy surface and highly porous structure. Molecular weight (Mw) estimated by multi-detector size-exclusion chromatography (SEC) indicated that BL biosynthesized using S. salivarius K12 has an impressively high Mw, corresponding to 15.435 × 104 kilodaltons (kDa). In turn, BL isolated from L. mesenteroides DSM 20343 was found to have an Mw of only 26.6 kDa. Polydispersity index estimation (PD = Mw/Mn) of produced BL displayed a monodispersed molecule isolated from S. salivarius K12, corresponding to 1.08, while this was 2.17 for L. mesenteroides DSM 20343 isolate. The presence of fructose as the main backbone and, to a lesser extent, glucose and galactose as side chain molecules in EPS hydrolysates was supported by HPLC-RID detection. In producing CoS-BL@AgNPs within green biosynthesis, the presence of nanostructured objects with a size distribution from 12.67 ± 5.56 nm to 46.97 ± 20.23 was confirmed by SEM and energy-dispersive X-ray spectroscopy (EDX). The prominent inhibitory potency of elaborated CoS-BL@AgNPs against both reference test cultures, i.e., Pseudomonas aeruginosa, Escherichia coli, Enterobacter aerogenes, and Staphylococcus aureus and those of clinical origin with multi-drug resistance (MDR), was confirmed by disc and well diffusion tests and supported by the values of the minimum inhibitory and bactericidal concentrations. CoS-BL@AgNPs can be treated as APIs suitable for designing new antimicrobial agents and modifying therapies in controlling MDR pathogens.
Collapse
Affiliation(s)
- Vitalijs Radenkovs
- Processing and Biochemistry Department, Institute of Horticulture, LV-3701 Dobele, Latvia;
- Research Laboratory of Biotechnology, Latvia University of Life Sciences and Technologies, LV-3004 Jelgava, Latvia; (A.V.); (D.G.); (D.J.)
| | - Anda Valdovska
- Research Laboratory of Biotechnology, Latvia University of Life Sciences and Technologies, LV-3004 Jelgava, Latvia; (A.V.); (D.G.); (D.J.)
- Faculty of Veterinary Medicine, Latvia University of Life Sciences and Technologies, LV-3004 Jelgava, Latvia
| | - Daiga Galina
- Research Laboratory of Biotechnology, Latvia University of Life Sciences and Technologies, LV-3004 Jelgava, Latvia; (A.V.); (D.G.); (D.J.)
- Faculty of Veterinary Medicine, Latvia University of Life Sciences and Technologies, LV-3004 Jelgava, Latvia
| | - Stefan Cairns
- Malvern Panalytical Ltd., Worcestershire, Malvern WR14 1XZ, UK
| | - Dmitrijs Jakovlevs
- Research Laboratory of Biotechnology, Latvia University of Life Sciences and Technologies, LV-3004 Jelgava, Latvia; (A.V.); (D.G.); (D.J.)
| | - Sergejs Gaidukovs
- Institute of Polymer Materials, Faculty of Materials Science and Applied Chemistry, Riga Technical University, LV-1048 Riga, Latvia;
| | - Ingmars Cinkmanis
- Faculty of Agriculture and Food Technology, Latvia University of Life Sciences and Technologies, LV-3004 Jelgava, Latvia;
| | | |
Collapse
|
27
|
Sekar A, Gil D, Tierney PA, McCanne M, Daesety V, Trendafilova D, Muratoglu OK, Oral E. Synergistic use of anti-inflammatory ketorolac and gentamicin to target staphylococcal biofilms. RESEARCH SQUARE 2023:rs.3.rs-3471646. [PMID: 37961705 PMCID: PMC10635368 DOI: 10.21203/rs.3.rs-3471646/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Background While antibiotics remain our primary tools against microbial infection, increasing antibiotic resistance (inherent and acquired) is a major detriment to their efficacy. A practical approach to maintaining or reversing the efficacy of antibiotics is the use of other commonly used therapeutics, which show synergistic antibacterial action with antibiotics. Here, we investigated the extent of antibacterial synergy between the antibiotic gentamicin and the anti-inflammatory ketorolac regarding the dynamics of biofilm growth, the rate of acquired resistance, and the possible mechanism of synergy. Methods Control (ATCC 12600, ATCC 35984) and clinical strains (L1101, L1116) of S. aureus and S. epidermidis with varying antibiotic susceptibility profiles were used in this study to simulate implant-material associated low-risk and high-risk biofilms in vitro. The synergistic action of gentamicin sulfate (GS) and ketorolac tromethamine (KT), against planktonic staphylococcal strains were determined using the fractional inhibitory concentration measurement assay. Nascent (6hr) and established (24hr) biofilms were grown on 316 stainless steel plates and the synergistic biofilm eradication activity was determined and characterized using adherent bacteria count, MBEC measurement for GS, gene expression of biofilm-associated genes, visualization by live/dead imaging, scanning electron microscopy, and bacterial membrane fluidity assessment. Results Gentamicin-ketorolac combination demonstrated synergistic antibacterial action against planktonic Staphylococci. Control and clinical strains showed distinct biofilm growth dynamics and an increase in biofilm maturity was shown to confer further resistance to gentamicin for both 'low-risk' and 'high-risk' biofilms. The addition of ketorolac enhanced the antibiofilm activity of gentamicin against acquired resistance in staphylococcal biofilms. Mechanistic studies revealed that the synergistic action of gentamicin-ketorolac interferes with biofilm morphology and subverts bacterial stress response altering bacterial physiology, membrane dynamics, and biofilm properties. Conclusion The results of this study have a significant impact on the local administration of antibiotics and other therapeutic agents commonly used in the prevention and treatment of orthopaedic infections. Further, these results warrant the study of synergy for the concurrent or sequential administration of non-antibiotic drugs for antimicrobial effect.
Collapse
Affiliation(s)
- Amita Sekar
- Harris Orthopaedic Laboratory, Massachusetts General Hospital; Boston, U.S.A
- Department of Orthopaedic Surgery, Harvard Medical School, Harvard University; Boston, U.S.A
| | - Dmitry Gil
- Harris Orthopaedic Laboratory, Massachusetts General Hospital; Boston, U.S.A
- Department of Orthopaedic Surgery, Harvard Medical School, Harvard University; Boston, U.S.A
| | - Peyton Anne Tierney
- Harris Orthopaedic Laboratory, Massachusetts General Hospital; Boston, U.S.A
| | - Madeline McCanne
- Harris Orthopaedic Laboratory, Massachusetts General Hospital; Boston, U.S.A
| | - Vikram Daesety
- Harris Orthopaedic Laboratory, Massachusetts General Hospital; Boston, U.S.A
| | - Darina Trendafilova
- Harris Orthopaedic Laboratory, Massachusetts General Hospital; Boston, U.S.A
| | - Orhun K Muratoglu
- Harris Orthopaedic Laboratory, Massachusetts General Hospital; Boston, U.S.A
- Department of Orthopaedic Surgery, Harvard Medical School, Harvard University; Boston, U.S.A
| | - Ebru Oral
- Harris Orthopaedic Laboratory, Massachusetts General Hospital; Boston, U.S.A
- Department of Orthopaedic Surgery, Harvard Medical School, Harvard University; Boston, U.S.A
| |
Collapse
|
28
|
Bogut A, Koper P, Marczak M, Całka P. The first genomic characterization of a stable, hemin-dependent small colony variant strain of Staphylococcus epidermidis isolated from a prosthetic-joint infection. Front Microbiol 2023; 14:1289844. [PMID: 37928677 PMCID: PMC10620731 DOI: 10.3389/fmicb.2023.1289844] [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: 09/09/2023] [Accepted: 10/05/2023] [Indexed: 11/07/2023] Open
Abstract
Phenotype switching from a wild type (WT) to a slow-growing subpopulation, referred to as small colony variants (SCVs), supports an infectious lifestyle of Staphylococcus epidermidis, the leading cause of medical device-related infections. Specific mechanisms underlying formation of SCVs and involved in the shaping of their pathogenic potential are of particular interest for stable strains as they have been only rarely cultured from clinical specimens. As the SCV phenotype stability implies the existence of genetic changes, the whole genome sequence of a stable, hemin-dependent S. epidermidis SCV strain (named 49SCV) involved in a late prosthetic joint infection was analyzed. The strain was isolated in a monoculture without a corresponding WT clone, therefore, its genome was compared against five reference S. epidermidis strains (ATCC12228, ATCC14990, NBRC113846, O47, and RP62A), both at the level of the genome structure and coding sequences. According to the Multilocus Sequence Typing analysis, the 49SCV strain represented the sequence type 2 (ST2) regarded as the most prominent infection-causing lineage with a worldwide dissemination. Genomic features unique to 49SCV included the absence of the Staphylococcal Cassette Chromosome (SCC), ~12 kb deletion with the loss of genes involved in the arginine deiminase pathway, and frameshift-generating mutations within the poly(A) and poly(T) homopolymeric tracts. Indels were identified in loci associated with adherence, metabolism, stress response, virulence, and cell wall synthesis. Of note, deletion in the poly(A) of the hemA gene has been considered a possible trigger factor for the phenotype transition and hemin auxotrophy in the strain. To our knowledge, the study represents the first genomic characterization of a clinical, stable and hemin-dependent S. epidermidis SCV strain. We propose that previously unreported indels in the homopolymeric tracts can constitute a background of the SCV phenotype due to a resulting truncation of the corresponding proteins and their possible biological dysfunction. Streamline of genetic content evidenced by the loss of the SCC and a large genomic deletion can represent a possible strategy associated both with the SCV phenotype and its adaptation to chronicity.
Collapse
Affiliation(s)
- Agnieszka Bogut
- Chair and Department of Medical Microbiology, Medical University of Lublin, Lublin, Poland
| | - Piotr Koper
- Department of Genetics and Microbiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland
| | - Małgorzata Marczak
- Department of Genetics and Microbiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland
| | - Paulina Całka
- Chair and Department of Forensic Medicine, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
29
|
Kalia VC, Patel SKS, Lee JK. Bacterial biofilm inhibitors: An overview. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 264:115389. [PMID: 37634478 DOI: 10.1016/j.ecoenv.2023.115389] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/05/2023] [Accepted: 08/17/2023] [Indexed: 08/29/2023]
Abstract
Bacteria that cause infectious diseases adopt biofilms as one of their most prevalent lifestyles. Biofilms enable bacteria to tolerate environmental stress and evade antibacterial agents. This bacterial defense mechanism has rendered the use of antibiotics ineffective for the treatment of infectious diseases. However, many highly drug-resistant microbes have rapidly emerged owing to such treatments. Different signaling mechanisms regulate bacterial biofilm formation, including cyclic dinucleotide (c-di-GMP), small non-coding RNAs, and quorum sensing (QS). A cell density-dependent phenomenon, QS is associated with c-di-GMP (a global messenger), which regulates gene expression related to adhesion, extracellular matrix production, the transition from the planktonic to biofilm stage, stability, pathogenicity, virulence, and acquisition of nutrients. The article aims to provide information on inhibiting biofilm formation and disintegrating mature/preformed biofilms. This treatment enables antimicrobials to target the free-living/exposed bacterial cells at lower concentrations than those needed to treat bacteria within the biofilm. Therefore, a complementary action of antibiofilm and antimicrobial agents can be a robust strategic approach to dealing with infectious diseases. Taken together, these molecules have broad implications for human health.
Collapse
Affiliation(s)
- Vipin Chandra Kalia
- Department of Chemical Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Sanjay K S Patel
- Department of Chemical Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jung-Kul Lee
- Department of Chemical Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
30
|
Qamer S, Che-Hamzah F, Misni N, Joseph NMS, Al-Haj NA, Amin-Nordin S. Deploying a Novel Approach to Prepare Silver Nanoparticle Bellamya bengalensis Extract Conjugate Coating on Orthopedic Implant Biomaterial Discs to Prevent Potential Biofilm Formation. Antibiotics (Basel) 2023; 12:1403. [PMID: 37760700 PMCID: PMC10526060 DOI: 10.3390/antibiotics12091403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/01/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
This study is based on the premise of investigating antibacterial activity through a novel conjugate of silver nanoparticles (AgNPs) and antimicrobial peptides (AMPs) in line with a green synthesis approach by developing antimicrobial-coated implants to prevent bacterial resistance. The AMPs were obtained from Bellamya Bengalensis (BB), a freshwater snail, to prepare the nanocomposite conjugate, e.g., AgNPs@BB extract, by making use of UV-Visible spectroscopy. The antimicrobial assessment of AgNPs@BB extract conjugate was performed using the Resazurin Microtiter Assay Method (REMA), followed by the use of three biocompatible implant materials (titanium alloys, Ti 6AL-4V stainless steel 316L, and polyethylene). Finally, the coating was analyzed under confocal microscopy. The results revealed a significant reduction of biofilm formation on the surfaces of implants coated with conjugate (AgNPs@BB extract) in comparison to uncoated implants. For the MTT assay, no significant changes were recorded for the cells grown on the AgNPs/AMP++ sample in high concentrations. Staphylococcus epidermidis, however, showed more prominent growth on all implants in comparison to Staphylococcus aureus. It is evident from the results that Staphylococcus epidermidis is more susceptible to AgNPs@BB extract, while the minimum inhibitory concentration (MIC) value of AgNPs@BB extract conjugates and biosynthesized AgNPs was also on the higher side. This study indicates that AgNPs@BB extract carries antibacterial activity, and concludes that an excessive concentration of AgNPs@BB extract may affect the improved biocompatibility. This study recommends using robust, retentive, and antimicrobial coatings of AgNPs@BB extract for implantable biocompatible materials in accordance with the novel strategy of biomaterial applications.
Collapse
Affiliation(s)
- Shafqat Qamer
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Malaysia 43400, Selangor, Malaysia; (S.Q.); (N.M.); (N.M.S.J.)
| | - Fahrudin Che-Hamzah
- Orthopedic Department, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Malaysia 43400, Selangor, Malaysia;
| | - Norashiqin Misni
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Malaysia 43400, Selangor, Malaysia; (S.Q.); (N.M.); (N.M.S.J.)
| | - Narcisse M. S. Joseph
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Malaysia 43400, Selangor, Malaysia; (S.Q.); (N.M.); (N.M.S.J.)
| | - Nagi A. Al-Haj
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Sana’a University, Sana’a 009671, Yemen;
| | - Syafinaz Amin-Nordin
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Malaysia 43400, Selangor, Malaysia; (S.Q.); (N.M.); (N.M.S.J.)
| |
Collapse
|
31
|
Fickenscher MC, Stewart M, Helber R, Quilligan EJ, Kreitenberg A, Prietto CA, Gardner VO. Operating room disinfection: operator-driven ultraviolet 'C' vs. chemical treatment. Infect Prev Pract 2023; 5:100301. [PMID: 37575675 PMCID: PMC10412461 DOI: 10.1016/j.infpip.2023.100301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/11/2023] [Indexed: 08/15/2023] Open
Abstract
Background In operating room (OR) surfaces, Nosocomial pathogens can persist on inanimate surfaces for long intervals and are highly resistant to traditional surface cleaning. Aim This study compares traditional chemical operating room terminal disinfection to a unique operator-driven device that emits germicidal UV light at short distance onto vertical and horizontal surfaces. Methods A randomized crossover analogous protocol assigned 40 end-of-day operating rooms into either group A (chemical then UVC treatments) or group B (UVC then chemical treatments). Initial Staphylococcal cultures were obtained prior to disinfection treatment, after the first treatment, and after the second treatment at 16 most commonly contaminated sites to represent overall room contamination. Success was defined as no growth and failure as 1 or more colony forming units. Thoroughness of chemical treatment vs UVC treatment was compared and used to determine if the second treatment was additive to the first treatment within each group. Findings The operator driven UVC device outperformed chemical treatment in reducing the number of contaminated sites in the OR by more than half (P<0.001). Operator-driven UVC reduced contaminated sites after chemical treatment by nearly half (P<0.001). In contrast, chemical treatment after operator-driven UVC did not significantly reduce the number of contaminated sites. The mean employee time of disinfection for chemical treatment was 49 minutes and for the operator-driven UVC emitter 7.9 minutes (P<0.001). Conclusions This study demonstrates that addition of an operator-driven UVC emitter to OR rooms between cases could be helpful in overall decreasing the number of contaminated sites.
Collapse
Affiliation(s)
| | - Madeline Stewart
- Hoag Orthopedics Education and Research, Hoag Orthopedic Institute, Irvine, CA, United States
| | - Ryan Helber
- Hoag Orthopedics Education and Research, Hoag Orthopedic Institute, Irvine, CA, United States
| | - Edward J. Quilligan
- Hoag Orthopedics Education and Research, Hoag Orthopedic Institute, Irvine, CA, United States
| | - Arthur Kreitenberg
- Department of Orthopedic Surgery, Center for Orthopedic & Sports Excellence, Los Angeles, CA, United States
| | - Carlos A. Prietto
- Hoag Orthopedics Education and Research, Hoag Orthopedic Institute, Irvine, CA, United States
| | - Vance O. Gardner
- Hoag Orthopedics Education and Research, Hoag Orthopedic Institute, Irvine, CA, United States
| |
Collapse
|
32
|
Vo N, Sidner BS, Yu Y, Piepenbrink KH. Type IV Pilus-Mediated Inhibition of Acinetobacter baumannii Biofilm Formation by Phenothiazine Compounds. Microbiol Spectr 2023; 11:e0102323. [PMID: 37341603 PMCID: PMC10433872 DOI: 10.1128/spectrum.01023-23] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/26/2023] [Indexed: 06/22/2023] Open
Abstract
Infections by pathogenic Acinetobacter species represent a significant burden on the health care system, despite their relative rarity, due to the difficulty of treating infections through oral antibiotics. Multidrug resistance is commonly observed in clinical Acinetobacter infections and multiple molecular mechanisms have been identified for this resistance, including multidrug efflux pumps, carbapenemase enzymes, and the formation of bacterial biofilm in persistent infections. Phenothiazine compounds have been identified as a potential inhibitor of type IV pilus production in multiple Gram-negative bacterial species. Here, we report the ability of two phenothiazines to inhibit type IV pilus-dependent surface (twitching) motility and biofilm formation in multiple Acinetobacter species. Biofilm formation was inhibited in both static and continuous flow models at micromolar concentrations without significant cytotoxicity, suggesting that type IV pilus biogenesis was the primary molecular target for these compounds. These results suggest that phenothiazines may be useful lead compounds for the development of biofilm dispersal agents against Gram-negative bacterial infections. IMPORTANCE Acinetobacter infections are a growing burden on health care systems worldwide due to increasing antimicrobial resistance through multiple mechanisms. Biofilm formation is an established mechanism of antimicrobial resistance, and its inhibition has the potential to potentiate the use of existing drugs against pathogenic Acinetobacter. Additionally, as discussed in the manuscript, anti-biofilm activity by phenothiazines has the potential to help to explain their known activity against other bacteria, including Staphylococcus aureus and Mycobacterium tuberculosis.
Collapse
Affiliation(s)
- Nam Vo
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Benjamin S. Sidner
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Yafan Yu
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Kurt H. Piepenbrink
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
- Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| |
Collapse
|
33
|
Choi E, Murray B, Choi S. Biofilm and Cancer: Interactions and Future Directions for Cancer Therapy. Int J Mol Sci 2023; 24:12836. [PMID: 37629016 PMCID: PMC10454087 DOI: 10.3390/ijms241612836] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
There is a growing body of evidence supporting the significant role of bacterial biofilms in the pathogenesis of various human diseases, including cancer. Biofilms are polymicrobial communities enclosed within an extracellular matrix composed of polysaccharides, proteins, extracellular DNA, and lipids. This complex matrix provides protection against antibiotics and host immune responses, enabling the microorganisms to establish persistent infections. Moreover, biofilms induce anti-inflammatory responses and metabolic changes in the host, further facilitating their survival. Many of these changes are comparable to those observed in cancer cells. This review will cover recent research on the role of bacterial biofilms in carcinogenesis, especially in colorectal (CRC) and gastric cancers, emphasizing the shared physical and chemical characteristics of biofilms and cancer. This review will also discuss the interactions between bacteria and the tumor microenvironment, which can facilitate oncogene expression and cancer progression. This information will provide insight into developing new therapies to identify and treat biofilm-associated cancers, such as utilizing bacteria as delivery vectors, using bacteria to upregulate immune function, or more selectively targeting biofilms and cancer for their shared traits.
Collapse
Affiliation(s)
- Euna Choi
- Department of Biology, Union University, Jackson, TN 38305, USA; (E.C.); (B.M.)
| | - Ben Murray
- Department of Biology, Union University, Jackson, TN 38305, USA; (E.C.); (B.M.)
| | - Sunga Choi
- Department of Bioinformatics and Biosystems, Seongnam Campus of Korea Polytechnics, Seongnam-si 13122, Republic of Korea
| |
Collapse
|
34
|
Busch L, Kröger M, Schleusener J, Klein AL, Lohan SB, Guttmann M, Keck CM, Meinke MC. Evaluation of DNA lesions and radicals generated by a 233 nm far-UVC LED in superficial ex vivo skin wounds. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2023; 245:112757. [PMID: 37481791 DOI: 10.1016/j.jphotobiol.2023.112757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/02/2023] [Accepted: 07/17/2023] [Indexed: 07/25/2023]
Abstract
The application of a far-ultraviolet C (UVC) light emitting diode (LED) of 233 nm showed significant bactericidal efficacy at an applied dose between 20 and 80 mJ cm-2 as reported recently. In addition, only minor epidermal DNA lesions were observed in ex vivo human skin and in vitro epidermal models <10% of the minimal erythema dose of UVB radiation. To broaden the potential range of applications of such systems, e.g. to include postoperative application on wounds for the purpose of decontamination, we assessed how a disruption of normal anatomic skin structure and function influences the skin damage induced by light from 233 nm far-UVC LEDs. Thus, we induced superficial skin wounds by mechanical detachment of the stratum corneum in ex vivo human skin. Barrier-disruption of the skin could be successfully determined by measuring an increase in the transepidermal water loss (TEWL) and the stratum corneum loss could be determined morphologically by 2-photon microscopy (2-PM). After far-UVC irradiation of the skin, we screened the tissue for the development of cyclobutane pyrimidine dimers (CPDs) and 6-4 photoproducts (6-4PPs). The abundance of DNA lesions was elevated in wound skin in comparison to intact skin after irradiation with far-UVC. However, no increase in DNA lesions was detected when artificial wound exudate consisting of cell culture medium and serum was applied to the disrupted skin surface prior to irradiation. This effect agrees with the results of ray tracing simulations of the absorption of far-UVC light incident on a superficial skin wound. Interestingly, no significant deviations in radical formation between intact skin and superficially wounded skin were detected after far-UVC irradiation as analyzed by electron paramagnetic resonance (EPR) spectroscopy. In conclusion, 233 nm LED light at a dose of 60 mJ/cm2 could be applied safely on superficial wounds for the purpose of skin antisepsis as long as the wounds are covered with wound fluid.
Collapse
Affiliation(s)
- Loris Busch
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Dermatology, Venereology and Allergology, Center of Experimental and Applied Cutaneous Physiology, Charitéplatz 1, 10117 Berlin, Germany; Philipps-Universität Marburg, Department of Pharmaceutics and Biopharmaceutics, Robert-Koch-Str. 4, 35032 Marburg, Germany.
| | - Marius Kröger
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Dermatology, Venereology and Allergology, Center of Experimental and Applied Cutaneous Physiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Johannes Schleusener
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Dermatology, Venereology and Allergology, Center of Experimental and Applied Cutaneous Physiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Anna Lena Klein
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Dermatology, Venereology and Allergology, Center of Experimental and Applied Cutaneous Physiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Silke B Lohan
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Dermatology, Venereology and Allergology, Center of Experimental and Applied Cutaneous Physiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Martin Guttmann
- Ferdinand-Braun-Institut (FBH), Gustav-Kirchhoff-Straße 4, 12489 Berlin, Germany
| | - Cornelia M Keck
- Philipps-Universität Marburg, Department of Pharmaceutics and Biopharmaceutics, Robert-Koch-Str. 4, 35032 Marburg, Germany
| | - Martina C Meinke
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Dermatology, Venereology and Allergology, Center of Experimental and Applied Cutaneous Physiology, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
35
|
Vargová M, Zigo F, Výrostková J, Farkašová Z, Rehan IF. Biofilm-Producing Ability of Staphylococcus aureus Obtained from Surfaces and Milk of Mastitic Cows. Vet Sci 2023; 10:386. [PMID: 37368772 DOI: 10.3390/vetsci10060386] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
This study was conducted to evaluate the incidence of mastitis in 153 dairy cows and to evaluate the kinetics of adhesion of isolates obtained from surfaces and milk in comparison with the reference strain (RS), CCM 4223. The surfaces of the floor, teat cup, and cow restraints were aseptically swabbed in three replicates (n = 27). Of the total number of infected cows (n = 43), 11 samples were found to be positive for Staphylococcus aureus, 12 samples tested positive for non-aureus staphylococci, 6 samples tested positive for Streptococcus spp., and 11 samples tested positive for other bacteria (Escherichia coli, Pseudomonas spp.) or a mixed infection. The most represented pathogen in milk (11/43) and on surfaces (14/27) was S. aureus. The kinetics of adhesion of the reference strain and isolates of S. aureus on stainless steel surfaces were determined after 3, 6, 9, 12, 24, and 48 h, and 3, 6, 9, 12, and 15 days of incubation. All strains reached counts higher than 5 Log10 CFU/cm2 needed for biofilm formation, except RS (4.40 Log10 CFU/cm2). The isolates of S. aureus revealed a higher capability to form biofilm in comparison with RS during the first 3 h (p < 0.001). Thus, there is a significant difference between the occurrence of S. aureus on monitored surfaces-floor, teat cup, and cow restraints-and the frequency with which mastitis is caused by S. aureus (p < 0.05). This finding raises the possibility that if various surfaces are contaminated by S. aureus, it can result in the formation of biofilm, which is a significant virulence factor.
Collapse
Affiliation(s)
- Mária Vargová
- Department of the Environment, Veterinary Legislation and Economy, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181 Košice, Slovakia
| | - František Zigo
- Department of Animal Nutrition and Husbandry, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181 Košice, Slovakia
| | - Jana Výrostková
- Department of Food Hygiene, Technology, and Safety, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181 Košice, Slovakia
| | - Zuzana Farkašová
- Department of Animal Nutrition and Husbandry, University of Veterinary Medicine and Pharmacy, Komenského 73, 04181 Košice, Slovakia
| | - Ibrahim F Rehan
- Department of Husbandry and Development of Animal Wealth, Faculty of Veterinary Medicine, Menoufia University, Shebin Alkom 32511, Egypt
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, Yagotoyama 150, Tempaku-Ku, Nagoya-Shi 468-8503, Japan
| |
Collapse
|
36
|
Landemaine L, Da Costa G, Fissier E, Francis C, Morand S, Verbeke J, Michel ML, Briandet R, Sokol H, Gueniche A, Bernard D, Chatel JM, Aguilar L, Langella P, Clavaud C, Richard ML. Staphylococcus epidermidis isolates from atopic or healthy skin have opposite effect on skin cells: potential implication of the AHR pathway modulation. Front Immunol 2023; 14:1098160. [PMID: 37304256 PMCID: PMC10250813 DOI: 10.3389/fimmu.2023.1098160] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 05/04/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction Staphylococcus epidermidis is a commensal bacterium ubiquitously present on human skin. This species is considered as a key member of the healthy skin microbiota, involved in the defense against pathogens, modulating the immune system, and involved in wound repair. Simultaneously, S. epidermidis is the second cause of nosocomial infections and an overgrowth of S. epidermidis has been described in skin disorders such as atopic dermatitis. Diverse isolates of S. epidermidis co-exist on the skin. Elucidating the genetic and phenotypic specificities of these species in skin health and disease is key to better understand their role in various skin conditions. Additionally, the exact mechanisms by which commensals interact with host cells is partially understood. We hypothesized that S. epidermidis isolates identified from different skin origins could play distinct roles on skin differentiation and that these effects could be mediated by the aryl hydrocarbon receptor (AhR) pathway. Methods For this purpose, a library of 12 strains originated from healthy skin (non-hyperseborrheic (NH) and hyperseborrheic (H) skin types) and disease skin (atopic (AD) skin type) was characterized at the genomic and phenotypic levels. Results and discussion Here we showed that strains from atopic lesional skin alter the epidermis structure of a 3D reconstructed skin model whereas strains from NH healthy skin do not. All strains from NH healthy skin induced AhR/OVOL1 path and produced high quantities of indole metabolites in co-culture with NHEK; especially indole-3-aldehyde (IAld) and indole-3-lactic acid (ILA); while AD strains did not induce AhR/OVOL1 path but its inhibitor STAT6 and produced the lowest levels of indoles as compared to the other strains. As a consequence, strains from AD skin altered the differentiation markers FLG and DSG1. The results presented here, on a library of 12 strains, showed that S. epidermidis originated from NH healthy skin and atopic skin have opposite effects on the epidermal cohesion and structure and that these differences could be linked to their capacity to produce metabolites, which in turn could activate AHR pathway. Our results on a specific library of strains provide new insights into how S. epidermidis may interact with the skin to promote health or disease.
Collapse
Affiliation(s)
- Leslie Landemaine
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- L’Oréal Research and Innovation, Aulnay-sous-Bois, France
| | - Gregory Da Costa
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| | - Elsa Fissier
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| | - Carine Francis
- L’Oréal Research and Innovation, Aulnay-sous-Bois, France
| | | | | | - Marie-Laure Michel
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| | - Romain Briandet
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Harry Sokol
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
- Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France
| | | | | | - Jean-Marc Chatel
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| | - Luc Aguilar
- L’Oréal Research and Innovation, Aulnay-sous-Bois, France
| | - Philippe Langella
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| | - Cecile Clavaud
- L’Oréal Research and Innovation, Aulnay-sous-Bois, France
| | - Mathias L. Richard
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| |
Collapse
|
37
|
Chen M, Chen L, Yuan D, Niu L, Hu J, Zhang X, Zhang X, Zhang Y, Zhang X, Ling P, Liu F, Zhang D. Preparation, function, and safety evaluation of a novel degradable dermal filler, the cross-linked poly-γ-glutamic acid hydrogel particles. J Biomed Mater Res B Appl Biomater 2023; 111:1407-1418. [PMID: 36930047 DOI: 10.1002/jbm.b.35245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 02/10/2023] [Accepted: 02/23/2023] [Indexed: 03/18/2023]
Abstract
Poly-γ-glutamic acid (PGA) is a naturally degradable hydrophilic linear microbial polymer with moisturizing, immunogenic, cross-linking, and hydrogel water absorption properties similar to hyaluronic acid, a biomaterial that is commonly used as a dermal filler. To explore the development feasibility of cross-linked PGA as a novel dermal filler, we studied the local skin response to PGA fillers and the effect of various cross-linking preparations on the average longevity of dermal injection. Injection site inflammation and the formation of collagen and elastin were also determined. PGA hydrogel particles prepared using 28% PGA and 10% 1,4-butanediol diglycidyl ether showed optimal filler properties, resistance to moist heat sterilization, and an average filling longevity of 94.7 ± 61.6 days in the dermis of rabbit ears. Local redness and swelling due to filler injection recovered within 14.2 ± 3.6 days. Local tissue necrosis or systemic allergic reactions were not observed, and local collagen formation was promoted. Preliminary results suggested that dermal injection of cross-linked PGA particles appeared safe and effective, suggesting that cross-linked PGA particles could be developed as a new hydrogel dermal filler.
Collapse
Affiliation(s)
- Mian Chen
- Shandong Academy of Pharmaceutical Sciences, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide drugs, National-Local Joint Engineering Laboratory of Polysaccharide drugs, Postdoctoral Scientific Research Workstation, Jinan, China
| | - Lei Chen
- Shandong Academy of Pharmaceutical Sciences, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide drugs, National-Local Joint Engineering Laboratory of Polysaccharide drugs, Postdoctoral Scientific Research Workstation, Jinan, China
| | - Dandan Yuan
- Shandong Academy of Pharmaceutical Sciences, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide drugs, National-Local Joint Engineering Laboratory of Polysaccharide drugs, Postdoctoral Scientific Research Workstation, Jinan, China
| | - Linlin Niu
- Shandong Academy of Pharmaceutical Sciences, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide drugs, National-Local Joint Engineering Laboratory of Polysaccharide drugs, Postdoctoral Scientific Research Workstation, Jinan, China
| | - Jianting Hu
- Shandong Academy of Pharmaceutical Sciences, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide drugs, National-Local Joint Engineering Laboratory of Polysaccharide drugs, Postdoctoral Scientific Research Workstation, Jinan, China
| | - Xiaoyuan Zhang
- Shandong Academy of Pharmaceutical Sciences, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide drugs, National-Local Joint Engineering Laboratory of Polysaccharide drugs, Postdoctoral Scientific Research Workstation, Jinan, China
| | - Xiuhua Zhang
- Shandong Academy of Pharmaceutical Sciences, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide drugs, National-Local Joint Engineering Laboratory of Polysaccharide drugs, Postdoctoral Scientific Research Workstation, Jinan, China
| | - Yanyan Zhang
- Shandong Academy of Pharmaceutical Sciences, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide drugs, National-Local Joint Engineering Laboratory of Polysaccharide drugs, Postdoctoral Scientific Research Workstation, Jinan, China
| | - Xiangjun Zhang
- Shandong Academy of Pharmaceutical Sciences, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide drugs, National-Local Joint Engineering Laboratory of Polysaccharide drugs, Postdoctoral Scientific Research Workstation, Jinan, China
| | - Peixue Ling
- Shandong Academy of Pharmaceutical Sciences, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide drugs, National-Local Joint Engineering Laboratory of Polysaccharide drugs, Postdoctoral Scientific Research Workstation, Jinan, China.,School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Fei Liu
- Shandong Academy of Pharmaceutical Sciences, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide drugs, National-Local Joint Engineering Laboratory of Polysaccharide drugs, Postdoctoral Scientific Research Workstation, Jinan, China.,School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Daizhou Zhang
- Shandong Academy of Pharmaceutical Sciences, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide drugs, National-Local Joint Engineering Laboratory of Polysaccharide drugs, Postdoctoral Scientific Research Workstation, Jinan, China
| |
Collapse
|
38
|
Caldwell M, Hughes M, Wei F, Ngo C, Pascua R, Pugazhendhi AS, Coathup MJ. Promising applications of D-amino acids in periprosthetic joint infection. Bone Res 2023; 11:14. [PMID: 36894568 PMCID: PMC9998894 DOI: 10.1038/s41413-023-00254-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 02/02/2023] [Accepted: 02/10/2023] [Indexed: 03/11/2023] Open
Abstract
Due to the rise in our aging population, a disproportionate demand for total joint arthroplasty (TJA) in the elderly is forecast. Periprosthetic joint infection (PJI) represents one of the most challenging complications that can occur following TJA, and as the number of primary and revision TJAs continues to rise, an increasing PJI burden is projected. Despite advances in operating room sterility, antiseptic protocols, and surgical techniques, approaches to prevent and treat PJI remain difficult, primarily due to the formation of microbial biofilms. This difficulty motivates researchers to continue searching for an effective antimicrobial strategy. The dextrorotatory-isoforms of amino acids (D-AAs) are essential components of peptidoglycan within the bacterial cell wall, providing strength and structural integrity in a diverse range of species. Among many tasks, D-AAs regulate cell morphology, spore germination, and bacterial survival, evasion, subversion, and adhesion in the host immune system. When administered exogenously, accumulating data have demonstrated that D-AAs play a pivotal role against bacterial adhesion to abiotic surfaces and subsequent biofilm formation; furthermore, D-AAs have substantial efficacy in promoting biofilm disassembly. This presents D-AAs as promising and novel targets for future therapeutic approaches. Despite their emerging antibacterial efficacy, their role in disrupting PJI biofilm formation, the disassembly of established TJA biofilm, and the host bone tissue response remains largely unexplored. This review aims to examine the role of D-AAs in the context of TJAs. Data to date suggest that D-AA bioengineering may serve as a promising future strategy in the prevention and treatment of PJI.
Collapse
Affiliation(s)
- Matthew Caldwell
- Biionix Cluster & College of Medicine, University of Central Florida, 6900 Lake Nona Blvd, Orlando, FL, 32827, USA
| | - Megan Hughes
- School of Biosciences, Cardiff University, CF10 3AT, Wales, UK
| | - Fei Wei
- Biionix Cluster & College of Medicine, University of Central Florida, 6900 Lake Nona Blvd, Orlando, FL, 32827, USA
| | - Christopher Ngo
- Biionix Cluster & College of Medicine, University of Central Florida, 6900 Lake Nona Blvd, Orlando, FL, 32827, USA
| | - Raven Pascua
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd, Orlando, FL, 32827, USA
| | - Abinaya Sindu Pugazhendhi
- Biionix Cluster & College of Medicine, University of Central Florida, 6900 Lake Nona Blvd, Orlando, FL, 32827, USA
| | - Melanie J Coathup
- Biionix Cluster & College of Medicine, University of Central Florida, 6900 Lake Nona Blvd, Orlando, FL, 32827, USA.
| |
Collapse
|
39
|
Memariani H, Memariani M. Antibiofilm properties of cathelicidin LL-37: an in-depth review. World J Microbiol Biotechnol 2023; 39:99. [PMID: 36781570 DOI: 10.1007/s11274-023-03545-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 02/08/2023] [Indexed: 02/15/2023]
Abstract
Notwithstanding ceaseless endeavors toward developing effective antibiofilm chemotherapeutics, biofilm-associated infections continue to be one of the most perplexing challenges confronting medicine today. Endogenous host defense peptides, such as the human cathelicidin LL-37, are being propounded as promising options for treating such infectious diseases. Over the past decennium, LL-37 has duly received tremendous research attention by virtue of its broad-spectrum antimicrobial activity and immunomodulatory properties. No attempt has hitherto been made, as far as we are aware, to comprehensively review the antibiofilm effects of LL-37. Accordingly, the intent in this paper is to provide a fairly all-embracing review of the literature available on the subject. Accumulating evidence suggests that LL-37 is able to prevent biofilm establishment by different bacterial pathogens such as Acinetobacter baumannii, Aggregatibacter actinomycetemcomitans, Bacteroides fragilis, Burkholderia thailandensis, Cutibacterium acnes, Escherichia coli, Francisella tularensis, Helicobacter pylori, Klebsiella pneumoniae, Pseudomonas aeruginosa, Staphylococcus aureus, and Streptococcus pyogenes. Inhibition of bacterial adhesion, downregulation of biofilm-associated genes, suppression of quorum-sensing pathways, degradation of biofilm matrix, and eradication of biofilm-residing cells are the major mechanisms responsible for antibiofilm properties of LL-37. In terms of its efficacy and safety in vivo, there are still many questions to be answered. Undoubtedly, LL-37 can open up new windows of opportunity to prevent and treat obstinate biofilm-mediated infections.
Collapse
Affiliation(s)
- Hamed Memariani
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mojtaba Memariani
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
40
|
Staphylococcus epidermidis and its dual lifestyle in skin health and infection. Nat Rev Microbiol 2023; 21:97-111. [PMID: 36042296 PMCID: PMC9903335 DOI: 10.1038/s41579-022-00780-3] [Citation(s) in RCA: 112] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2022] [Indexed: 01/20/2023]
Abstract
The coagulase-negative bacterium Staphylococcus epidermidis is a member of the human skin microbiota. S. epidermidis is not merely a passive resident on skin but actively primes the cutaneous immune response, maintains skin homeostasis and prevents opportunistic pathogens from causing disease via colonization resistance. However, it is now appreciated that S. epidermidis and its interactions with the host exist on a spectrum of potential pathogenicity derived from its high strain-level heterogeneity. S. epidermidis is the most common cause of implant-associated infections and is a canonical opportunistic biofilm former. Additional emerging evidence suggests that some strains of S. epidermidis may contribute to the pathogenesis of common skin diseases. Here, we highlight new developments in our understanding of S. epidermidis strain diversity, skin colonization dynamics and its multifaceted interactions with the host and other members of the skin microbiota.
Collapse
|
41
|
Molecular Characterizations of the Coagulase-Negative Staphylococci Species Causing Urinary Tract Infection in Tanzania: A Laboratory-Based Cross-Sectional Study. Pathogens 2023; 12:pathogens12020180. [PMID: 36839452 PMCID: PMC9967252 DOI: 10.3390/pathogens12020180] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 01/11/2023] [Accepted: 01/17/2023] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND There is a growing body of evidence on the potential involvement of coagulase-negative Staphylococci (CoNS) in causing urinary tract infections (UTIs). The aim of this study was to delineate virulence potential, antimicrobial resistance genes, and sequence types of CoNS isolated from patients with UTI symptoms and pyuria in Tanzania. METHODS CoNS from patients with UTI symptoms and more than 125 leucocytes/μL were retrieved, subcultured, and whole-genome sequenced. RESULTS Out of 65 CoNS isolates, 8 species of CoNS were identified; Staphylococcus haemolyticus, n = 27 (41.5%), and Staphylococcus epidermidis, n = 24 (36.9%), were predominant. The majority of S. haemolyticus were sequence type (ST) 30, with 8 new ST138-145 reported, while the majority of S. epidermidis were typed as ST490 with 7 new ST1184-1190 reported. Sixty isolates (92.3%) had either one or multiple antimicrobial resistance genes. The most frequently detected resistance genes were 53 (21%) dfrG, 32 (12.9%) blaZ, and 26 (10.5%) mecA genes conferring resistance to trimethoprim, penicillin, and methicillin, respectively. Out of 65 isolates, 59 (90.8%) had virulence genes associated with UTI, with a predominance of the icaC 47 (46.5%) and icaA 14 (13.9%) genes. Conclusion:S. haemolyticus and S. epidermidis harboring icaC, dfrG, blaZ, and mecA genes were the predominant CoNS causing UTI in Tanzania. Laboratories should carefully interpret the significant bacteriuria due to CoNS in relation to UTI symptoms and pyuria before labeling them as contaminants. Follow-up studies to document the outcome of the treated patients is needed to add more evidence that CoNS are UTI pathogens.
Collapse
|
42
|
Jenkins CL, Bean HD. Current Limitations of Staph Infection Diagnostics, and the Role for VOCs in Achieving Culture-Independent Detection. Pathogens 2023; 12:pathogens12020181. [PMID: 36839453 PMCID: PMC9963134 DOI: 10.3390/pathogens12020181] [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: 08/01/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Staphylococci are broadly adaptable and their ability to grow in unique environments has been widely established, but the most common and clinically relevant staphylococcal niche is the skin and mucous membranes of mammals and birds. S. aureus causes severe infections in mammalian tissues and organs, with high morbidities, mortalities, and treatment costs. S. epidermidis is an important human commensal but is also capable of deadly infections. Gold-standard diagnostic methods for staph infections currently rely upon retrieval and characterization of the infectious agent through various culture-based methods. Yet, obtaining a viable bacterial sample for in vitro identification of infection etiology remains a significant barrier in clinical diagnostics. The development of volatile organic compound (VOC) profiles for the detection and identification of pathogens is an area of intensive research, with significant efforts toward establishing breath tests for infections. This review describes the limitations of existing infection diagnostics, reviews the principles and advantages of VOC-based diagnostics, summarizes the analytical tools for VOC discovery and clinical detection, and highlights examples of how VOC biomarkers have been applied to diagnosing human and animal staph infections.
Collapse
Affiliation(s)
- Carrie L. Jenkins
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ 85287, USA
| | - Heather D. Bean
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
- Center for Fundamental and Applied Microbiomics, The Biodesign Institute, Tempe, AZ 85287, USA
- Correspondence:
| |
Collapse
|
43
|
Chen Y, Li X, Gao W, Zhang Y, Mo A, Jiang J, He D. Microfiber-loaded bacterial community in indoor fallout and air-conditioner filter dust. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 856:159211. [PMID: 36206901 DOI: 10.1016/j.scitotenv.2022.159211] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 06/16/2023]
Abstract
Microfibers (MFs) are widely existed in indoor air; however, characteristic of microbiota on MFs is largely unknown. In this study, air-borne MFs were collected from fallout or air-conditioner (AC) filter dust in three types of indoor space including living room, dormitory and office. Both plastic and natural MFs were identified by Fourier transform infrared spectroscopy. Ultramicroscopic observation showed dense biofilms adhering on surfaces of MFs. Fallout MFs contained more bacteria but fewer fungi than MFs from AC filter dust. MFs-loaded bacteria were of highest abundance in living rooms, following dormitories and offices. Bacterial community and its diversity were further analyzed by 16S rRNA High-throughput sequencing. Up to 4540 of bacterium OTUs were shared in these MFs samples, unique OTUs in fallout and AC filter samples accounting for 26.3 % and 25.7 % of the total. Compared to MFs fallout, AC filter MFs contained more species of pathogenic bacteria, such as Betaproteobacteriales and Ralstonia, with obviously different β-diversity between two groups. Phenotypic analysis showed that fallout and AC filter MFs bacteria presented high index values of film formation, oxidative stress tolerance and potential pathogenicity. Overall, these results suggest that abundant bacteria including pathogen can be loaded on MFs, and would pose health risks through delivery of indoor MFs.
Collapse
Affiliation(s)
- Yingxin Chen
- School of Ecological and Environmental Sciences, East China Normal University, Shanghai 200241, China
| | - Xinyu Li
- School of Ecological and Environmental Sciences, East China Normal University, Shanghai 200241, China
| | - Wei Gao
- School of Ecological and Environmental Sciences, East China Normal University, Shanghai 200241, China; Shanghai Engineering Research Center of Biotransformation of Organic Solid Waste, East China Normal University, Shanghai 200241, China
| | - Yalin Zhang
- School of Ecological and Environmental Sciences, East China Normal University, Shanghai 200241, China
| | - Aoyun Mo
- School of Ecological and Environmental Sciences, East China Normal University, Shanghai 200241, China
| | - Jie Jiang
- School of Ecological and Environmental Sciences, East China Normal University, Shanghai 200241, China; Shanghai Engineering Research Center of Biotransformation of Organic Solid Waste, East China Normal University, Shanghai 200241, China
| | - Defu He
- School of Ecological and Environmental Sciences, East China Normal University, Shanghai 200241, China; Shanghai Engineering Research Center of Biotransformation of Organic Solid Waste, East China Normal University, Shanghai 200241, China; Shanghai Key Laboratory for Urban Ecological Processes and Eco-Restoration, East China Normal University, Shanghai 200241, China; Technology Innovation Center for Land Spatial Eco-restoration in Metropolitan Area, Ministry of Natural Resources, Shanghai 200062, China.
| |
Collapse
|
44
|
Choi E, Wells B, Mirabella G, Atkins E, Choi S. Anti-biofilm activity of Pseudomonas fluorescens culture supernatants on biofilm formation of Staphylococcus epidermidis 1457. BMC Res Notes 2022; 15:370. [PMID: 36510276 PMCID: PMC9743590 DOI: 10.1186/s13104-022-06257-z] [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: 07/13/2022] [Accepted: 11/24/2022] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Staphylococcus epidermidis is a skin colonizer and a major cause of nosocomial infections that can lead to sepsis. It causes opportunistic infections by forming biofilms on medical devices, which are hard to control with conventional antibiotics. In an attempt to develop its biofilm inhibitors, the culture supernatant (CS) of Pseudomonas fluorescens was assessed. This study examined the effect of the CS on S. epidermidis 1457 biofilm formation, the characteristics of inhibitors in the CS, and the differential gene expression of S. epidermidis when treated with the CS. RESULTS P. fluorescens CS specifically targeted the maturation stage of S. epidermidis biofilm formation while not affecting planktonic growth. RT-qPCR analysis revealed that P. fluorescens CS significantly downregulated S. epidermidis ica genes and upregulated an ica repressor, tcaR. This indicates that the CS reduced polysaccharide intercellular adhesin synthesis, a major component of the S. epidermidis 1457 biofilm matrix. Further studies are required to elucidate the specific inhibitory components in the CS and their mechanism of action. Our results indicate that inhibitors in the P. fluorescens CS may have a significant value for inhibiting S. epidermidis biofilm. Combinations of specific inhibitors from the CS and antibiotics against staphylococci may provide an effective measure to control S. epidermidis biofilm formation while avoiding antibiotic resistance and compensating the attenuated effectiveness of antibiotics on biofilms.
Collapse
Affiliation(s)
- Euna Choi
- grid.441145.10000 0004 0414 0983Biology Department, Union University, 1050 Union University Drive, Jackson, TN 38305 USA
| | - Bethany Wells
- grid.441145.10000 0004 0414 0983Biology Department, Union University, 1050 Union University Drive, Jackson, TN 38305 USA
| | - Gabrielle Mirabella
- grid.441145.10000 0004 0414 0983Biology Department, Union University, 1050 Union University Drive, Jackson, TN 38305 USA
| | - Emilee Atkins
- grid.441145.10000 0004 0414 0983Biology Department, Union University, 1050 Union University Drive, Jackson, TN 38305 USA
| | - Sunga Choi
- Department of Bioinformatics and Biosystems, Seongnam-Campus of Korea Polytechnics, Seongnam, South Korea
| |
Collapse
|
45
|
Wang C, Chantraine C, Viljoen A, Herr AB, Fey PD, Horswill AR, Mathelié-Guinlet M, Dufrêne YF. The staphylococcal biofilm protein Aap mediates cell-cell adhesion through mechanically distinct homophilic and lectin interactions. PNAS NEXUS 2022; 1:pgac278. [PMID: 36712378 PMCID: PMC9802226 DOI: 10.1093/pnasnexus/pgac278] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 12/01/2022] [Indexed: 12/04/2022]
Abstract
The accumulation phase of staphylococcal biofilms relies on both the production of an extracellular polysaccharide matrix and the expression of bacterial surface proteins. A prototypical example of such adhesive proteins is the long multidomain protein Aap (accumulation-associated protein) from Staphylococcus epidermidis, which mediates zinc-dependent homophilic interactions between Aap B-repeat regions through molecular forces that have not been investigated yet. Here, we unravel the remarkable mechanical strength of single Aap-Aap homophilic bonds between living bacteria and we demonstrate that intercellular adhesion also involves sugar binding through the lectin domain of the Aap A region. We find that the mechanical force needed to unfold individual β-sheet-rich G5-E domains from the Aap B-repeat regions is very high, ranging from 300 up to 1,000 pN at high loading rates, indicating these are extremely stable. This high mechanostability provides a means to the cells to form highly adhesive and cohesive biofilms capable of sustaining high physiological shear stress. Importantly, we identify a previously undescribed role of Aap in bacterial-bacterial adhesion, that is, heterophilic sugar binding by a specific lectin domain located in the N-terminal A region, which might be important to establish initial contacts between cells before strong homophilic bonds come into play. This study emphasizes the remarkable mechanical and binding properties of Aap as well as its wide diversity of adhesive functions.
Collapse
Affiliation(s)
| | | | - Albertus Viljoen
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, bte L7.07.07, B-1348 Louvain-la-Neuve, Belgium
| | - Andrew B Herr
- Divisions of Immunobiology and Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Paul D Fey
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | | | | |
Collapse
|
46
|
Draft Genome Sequence of Staphylococcus epidermidis UMB7543, Isolated from a Female Patient with Recurrent Urinary Tract Infections. Microbiol Resour Announc 2022; 11:e0096222. [DOI: 10.1128/mra.00962-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Staphylococcus epidermidis
is a Gram-positive bacterium that is part of the normal human flora, found in multiple anatomical sites. Here, we present the 2.6-Mbp draft genome sequence of
S. epidermidis
UMB7543, isolated from a catheterized urine sample from a female patient with a documented diagnosis of recurrent urinary tract infection.
Collapse
|
47
|
Sung K, Park M, Chon J, Kweon O, Khan SA, Shen A, Paredes A. Concentration-Dependent Global Quantitative Proteome Response of Staphylococcus epidermidis RP62A Biofilms to Subinhibitory Tigecycline. Cells 2022; 11:3488. [PMID: 36359886 PMCID: PMC9655631 DOI: 10.3390/cells11213488] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 07/21/2023] Open
Abstract
Staphylococcus epidermidis is a leading cause of biofilm-associated infections on implanted medical devices. During the treatment of an infection, bacterial cells inside biofilms may be exposed to sublethal concentrations of the antimicrobial agents. In the present study, the effect of subinhibitory concentrations of tigecycline (TC) on biofilms formed by S. epidermidis strain RP62A was investigated using a quantitative global proteomic technique. Sublethal concentrations of TC [1/8 (T1) and 1/4 minimum inhibitory concentration (MIC) (T2)] promoted biofilm production in strain RP62A, but 1/2 MIC TC (T3) significantly inhibited biofilm production. Overall, 413, 429, and 518 proteins were differentially expressed in biofilms grown with 1/8 (T1), 1/4 (T2), and 1/2 (T3) MIC of TC, respectively. As the TC concentration increased, the number of induced proteins in each Cluster of Orthologous Groups (COG) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway increased. The TC concentration dependence of the proteome response highlights the diverse mechanisms of adaptive responses in strain RP62A biofilms. In both COG and KEGG functional analyses, most upregulated proteins belong to the metabolism pathway, suggesting that it may play an important role in the defense of strain RP62A biofilm cells against TC stress. Sub-MIC TC treatment of strain RP62A biofilms led to significant changes of protein expression related to biofilm formation, antimicrobial resistance, virulence, quorum sensing, ABC transporters, protein export, purine/pyrimidine biosynthesis, ribosomes, and essential proteins. Interestingly, in addition to tetracycline resistance, proteins involved in resistance of various antibiotics, including aminoglycosides, antimicrobial peptides, β-lactams, erythromycin, fluoroquinolones, fusidic acid, glycopeptides, lipopeptides, mupirocin, rifampicin and trimethoprim were differentially expressed. Our study demonstrates that global protein expression profiling of biofilm cells to antibiotic pressure may improve our understanding of the mechanisms of antibiotic resistance in biofilms.
Collapse
Affiliation(s)
- Kidon Sung
- Division of Microbiology, National Center for Toxicological Research, US FDA, Jefferson, AR 72079, USA
| | - Miseon Park
- Division of Microbiology, National Center for Toxicological Research, US FDA, Jefferson, AR 72079, USA
| | - Jungwhan Chon
- Companion Animal Health, Inje University, Gimhae 50834, Korea
| | - Ohgew Kweon
- Division of Microbiology, National Center for Toxicological Research, US FDA, Jefferson, AR 72079, USA
| | - Saeed A. Khan
- Division of Microbiology, National Center for Toxicological Research, US FDA, Jefferson, AR 72079, USA
| | - Andrew Shen
- Division of Neurotoxicology, National Center for Toxicological Research, US FDA, Jefferson, AR 72079, USA
| | - Angel Paredes
- Office of Scientific Coordination, National Center for Toxicological Research, US FDA, Jefferson, AR 72079, USA
| |
Collapse
|
48
|
Martínez-Santos VI, Torres-Añorve DA, Echániz-Aviles G, Parra-Rojas I, Ramírez-Peralta A, Castro-Alarcón N. Characterization of Staphylococcus epidermidis clinical isolates from hospitalized patients with bloodstream infection obtained in two time periods. PeerJ 2022; 10:e14030. [PMID: 36213498 PMCID: PMC9541613 DOI: 10.7717/peerj.14030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 08/16/2022] [Indexed: 01/19/2023] Open
Abstract
Background In recent years Staphylococcus epidermidis has been considered an important and frequent causative agent of health care-associated infections (HAIs), increasing the costs of hospitalization, morbidity, and mortality. Antibiotic resistance and biofilm formation are the most important obstacles in the treatment of infections caused by this microorganism. The aim of this work was to determine the most prevalent STs, as well as the antibiotic resistance profile and biofilm formation of S. epidermidis clinical isolates obtained from hospitalized patients in two hospitals in Acapulco, Guerrero in two time periods. Methods Twenty methicillin-resistant S. epidermidis strains isolated from patients with bacteremia in two hospitals in two time periods were analyzed. Identification and antibiotic susceptibility were performed using the Vitek automated system. Molecular confirmation of the identification and methicillin resistance was performed by duplex PCR of the mecA and nuc genes. Biofilm production was analyzed, and the clonal origin was determined by multilocus sequence typing (MLST). Results We identified 14 antibiotic resistance profiles as well as 13 sequence types (ST), including the new ST761. We also found that ST2 and ST23 were the most prevalent and, together with ST59, were found in both time periods. Seventeen of our clinical isolates were multidrug-resistant, but all of them were sensitive to linezolid and vancomycin, and this was not related to biofilm production. Additionally, we standardized a duplex PCR to identify methicillin-resistant S. epidermidis strains. In conclusion, S. epidermidis STs 2, 23, and 59 were found in both time periods. This study is the first report of S. epidermidis ST761. The clinical isolates obtained in this work showed a high multidrug resistance that is apparently not related to biofilm production.
Collapse
Affiliation(s)
| | - David A. Torres-Añorve
- Laboratorio de Investigación en Microbiología, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Gabriela Echániz-Aviles
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos, México
| | - Isela Parra-Rojas
- Labotatorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Arturo Ramírez-Peralta
- Laboratorio de Investigación en Patometabolismo Microbiano, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Natividad Castro-Alarcón
- Laboratorio de Investigación en Microbiología, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| |
Collapse
|
49
|
Mira P, Lozano‐Huntelman N, Johnson A, Savage VM, Yeh P. Evolution of antibiotic resistance impacts optimal temperature and growth rate in
Escherichia coli
and
Staphylococcus epidermidis. J Appl Microbiol 2022; 133:2655-2667. [DOI: 10.1111/jam.15736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Portia Mira
- Department of Ecology and Evolutionary Biology University of California Los Angeles U.S.A
| | | | - Adrienne Johnson
- Department of Ecology and Evolutionary Biology University of California Los Angeles U.S.A
| | - Van M. Savage
- Department of Ecology and Evolutionary Biology University of California Los Angeles U.S.A
- Department of Computational Medicine, David Geffen School of Medicine University of California Los Angeles U.S.A
- Santa Fe Institute Santa Fe New Mexico U.S.A
| | - Pamela Yeh
- Department of Ecology and Evolutionary Biology University of California Los Angeles U.S.A
- Santa Fe Institute Santa Fe New Mexico U.S.A
| |
Collapse
|
50
|
Effect of New 2-Thioxoimidazolidin-4-one Compounds against Staphylococcus aureus Clinical Strains and Immunological Markers’ Combinations. CANADIAN JOURNAL OF INFECTIOUS DISEASES AND MEDICAL MICROBIOLOGY 2022; 2022:6720241. [PMID: 35873361 PMCID: PMC9300335 DOI: 10.1155/2022/6720241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 04/03/2022] [Accepted: 04/07/2022] [Indexed: 12/03/2022]
Abstract
Although the structure-activity relationship indicates that the 4-thioxoimidazolidin ring is essential for antibacterial activities and pharmaceutical applications, there were no enough studies on the derivatives of this compound. Evaluating the new hydantoin compounds C5 (3-((2-bromobenzylidene) amino)-2- thioxoimidazolidin-4-one) and C6 (3-((4- methoxybenzylidene) amino)-2-thioxoimidazolidin-4-one) that were prepared against clinical Staphylococcus aureus isolates for antibacterial, antibiofilm, and antihemagglutination activities is the aim of this study. Therefore, the potential clinical resistance of the strains was evaluated by their ability to form biofilms, antibiotic resistance, and agglutinate erythrocytes macroscopically and microscopically; besides, the bacterial biofilm was screened for any association with the patient's serum immunoglobulin levels and complements. Despite the effective concentration for C5 and C6 compounds, which is ≤ 31.25 μg/ml, the reduction rate is not concentration-dependent; it depends on the molecular docking of the hydantoin compounds. Hence, the effect of the minimal inhibitory concentrations (MICs) is variable. In this study, the results for the compounds (with the concentration of 31.25–62.5 μg/mL for C5 and 62.5–125 μg/mL for C6) significantly manifest the antibacteria, antibiofilm, and antihemagglutination effects against the virulent strains of S. aureus due to the high percentage of biofilm inhibition that was caused by the new hydantoin compounds. Besides, time-kill kinetics studies showed that these compounds pose bactericidal action. Overall, this study revealed that the new hydantoin derivatives have an interesting potential as new antibacterial drugs through the inhibition of bacterial adhesion. The infections of these isolates activate the complement system through the lectin pathway. Nevertheless, these compounds can be improved in order to be used at even lower concentrations.
Collapse
|