1
|
Zaki RS, Abdelhaseib M, Mahmoud MMA. Optical interferometric: A novel approach for sensitive cost-effective measurement of multispecies biofilm from stress-tolerant field samples. Food Res Int 2025; 212:116378. [PMID: 40382065 DOI: 10.1016/j.foodres.2025.116378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/27/2025] [Accepted: 04/15/2025] [Indexed: 05/20/2025]
Abstract
Biofilms significantly impact public health and food safety within the meat industry. Detecting and quantifying biofilms are crucial tasks; however, their inherent complexity and heterogeneity, particularly in immature stages, present substantial challenges. Innovative techniques are necessary for accurate biofilm measurement, despite the presence of diverse interfering agents. An assessment of multispecies biofilms from cattle abattoir environmental samples (wall, basin, and door), equipment (tub, knives, and swivel), and offal (heart, lung, liver, spleen, rumen, and intestine). To elucidate the complexities of biofilm formation, a dual-pronged approach was employed, utilizing epifluorescence microscopy and optical interferometry to conduct a comprehensive analysis of biofilms that had developed on two distinct types of surfaces. Specifically, biotic surfaces that had undergone chilling and abiotic surfaces subjected to chlorination were examined to gain a deeper understanding of the structural and compositional characteristics of these microbial communities. Additionally, the presence of antibiotic-resistant bacteria within the submerged biofilm was evaluated. Despite chilling, the multispecies biofilm continued to grow from 0.2 μm to a maximum of 1.75 μm on the beef spleen surface, demonstrating the interplay of different bacterial species in biofilm formation on biotic surfaces. The highest antibacterial resistance was recorded for Staphylococcus simulans, followed by Staphylococcus saprophyticus and Staphylococcus aureus. In conclusion, the optical interferometer is a straightforward, fast, and sensitive technique for measuring biofilm thickness within the range of 0.1 to 4 μm. Progress in this field can revolutionize food safety, hygiene, biosecurity, and biomedical applications.
Collapse
Affiliation(s)
- Rania S Zaki
- Department of Food Hygiene, Faculty of Veterinary Medicine, New Valley University, El-Kharga 72511, Egypt; Food Science Department, Purdue University, West Lafayette, IN, USA, 47906
| | - Maha Abdelhaseib
- Department of Food Hygiene, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt; Animal Science Department, Purdue University, West Lafayette, IN 47906, USA.
| | - Manal M A Mahmoud
- Department of Animal Hygiene, Faculty of Veterinary Medicine, Assiut University, Assiut 71526, Egypt
| |
Collapse
|
2
|
Xu Z, Li Y, Xue L, Xu A, Yu G, Soteyome T, Yuan L, Li X, Liu J. Genomic-transcriptomic analysis of Staphylococcus aureus biofilm formation under sub-MIC antibiotic exposure. Food Res Int 2025; 211:116386. [PMID: 40356167 DOI: 10.1016/j.foodres.2025.116386] [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: 01/24/2025] [Revised: 03/23/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025]
Abstract
Antibiotics are widely used in animal husbandry to ensure the health of livestock, leading to the exposure of microorganisms to accumulated sub-lethal concentrations (sub-MICs) of antibiotics in meats. This study aimed to investigate the effects and mechanisms of sub-MICs of commonly used antibiotics on the biofilm formation of a S. aureus strain Guangzhou-SAU071 which displays weak biofilm formation despite harboring biofilm-associated genes. CV and MTS assays were used to determine biofilm biomass and cell viability, respectively. Dual-omics sequencing combining genomics and transcriptomics was used to study the global expression changes. Expression of biofilm and two-component system associated genes was further verified by RT-qPCR. Biofilm formation of Guangzhou-SAU071 was enhanced under sub-MIC of ciprofloxacin (2 μg/mL) and streptomycin (128 μg/mL). Nearly half of the genes associated with biofilm formation, cell wall anchoring, and two-component systems exhibited significant differential expression under sub-MIC of ciprofloxacin and streptomycin. As concluded, sub-MIC of ciprofloxacin and streptomycin enhanced biofilm formation of S. aureus, possibly due to its regulation on biofilm and two-component system associated genes.
Collapse
Affiliation(s)
- Zhenbo Xu
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou 510640, China; Department of Laboratory Medicine, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China..
| | - Yaqin Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou 510640, China
| | - Liang Xue
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou 510640, China; Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong, Guangzhou, China
| | - Aijuan Xu
- Guangzhou Hybribio Medical Laboratory, Guangzhou 510730, China
| | - Guangchao Yu
- Center of Clinical Laboratory Medicine, First Affiliated Hospital of Jinan University, Guangzhou 510620, China
| | - Thanapop Soteyome
- Home Economics Technology, Rajamangala University of Technology Phra Nakhon, Bangkok, Thailand
| | - Lei Yuan
- School of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu 225127, China
| | - Xuejie Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou 510640, China; Research Institute for Food Nutrition and Human Health, Guangzhou, China.
| | - Junyan Liu
- College of Light Industry and Food Science, Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Academy of Contemporary Agricultural Engineering Innovations, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China; Key Laboratory of Green Processing and Intelligent Manufacturing of Lingnan Specialty Food, Ministry of Agriculture, Guangzhou 510225, China.
| |
Collapse
|
3
|
He X, Zhang W, Liu J, Liu J, Chen Y, Luan C, Zhang J, Bao G, Lin X, Muh F, Lin T, Lu F. The global regulatory role of RsbUVW in virulence and biofilm formation in MRSA. Microb Pathog 2025; 203:107508. [PMID: 40158706 DOI: 10.1016/j.micpath.2025.107508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 03/22/2025] [Accepted: 03/23/2025] [Indexed: 04/02/2025]
Abstract
The widespread prevalence of methicillin-resistant Staphylococcus aureus (MRSA) has caused serious challenges to clinical treatment. This study was designed to explore effective targets for MRSA prevention and control. The key virulence regulator was screened through the correlation analysis between virulence and various regulatory factors in the main clinical epidemic MRSA. The potential key factors were inactivated to further evaluate the inhibitory effect on the virulence of MRSA standard strain S. aureus ATCC43300 and its influence on drug resistance and biofilm formation. Enterobacterial repetitive intergenic consensus-PCR was used to analyze the clinical epidemic genotypes of MRSA. The virulence of MRSA was evaluated mainly by measuring its adhesion and invasion ability to A549 cells, the lethality to Galleria mellonella larvae, and the transcription level of related genes. The biofilm formation was assessed by crystal violet staining on polystyrene microplates. The results showed that most virulence factors of clinical representative MRSA strain were significantly positively correlated with RsbUVW system. After knocking out the rsbV gene, a key component of the rsbUVW system, the virulence of S. aureus ATCC43300 was significantly reduced (P < 0.05), as indicated by a significant decrease in lethality against G. mellonella larvae and invasion against A549 cells, and a significant decrease in the expression of immune escape related virulence factors polysaccharide intercellular adhesin (PIA) and staphyloxanthin. The biomass and stability of protein-dependent biofilm by S. aureus ATCC43300 were significantly increased. This study will provide useful information for the effective prevention and control of MRSA.
Collapse
Affiliation(s)
- Xinlong He
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, 225001, China; The Key Laboratory of the Jiangsu Higher Education Institutions for Nucleic Acid & Cell Fate Regulation (Yangzhou University), Yangzhou, 225001, China; Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225001, China
| | - Wenwen Zhang
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; Department of Clinical Laboratory, Changning Maternity and Infant Health Hospital, Affiliated Hospital of East China Normal University, Shanghai, 200050, China
| | - Jie Liu
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Jiali Liu
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Yinsong Chen
- Department of Lung, Third People's Hospital of Yangzhou, Yangzhou, China
| | - Changjiao Luan
- Department of Lung, Third People's Hospital of Yangzhou, Yangzhou, China
| | - Jun Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China
| | - Guangyu Bao
- Department of Clinical Laboratory, First Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China
| | - Xiangfang Lin
- Department of Clinical Laboratory, First Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China
| | - Fauzi Muh
- Department of Epidemiology & Tropical Diseases, Faculty of Public Health, Universitas Diponegoro, Tembalang, Semarang, 50275, Indonesia
| | - Tao Lin
- Department of Clinical Laboratory, First Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China.
| | - Feng Lu
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China.
| |
Collapse
|
4
|
Zhang XH, Sun QJ, Zhao LC, Chen L, Li W. Traditional Chinese medicine in chronic rhinosinusitis: Mechanisms and postoperative recovery. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156658. [PMID: 40138776 DOI: 10.1016/j.phymed.2025.156658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/03/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Chronic rhinosinusitis (CRS) is inflammation of the sinuses and nasal passages that lasts for >3 months. Its pathogenesis is complex, treatment is difficult, and it has multiple effects on patients. Although surgical treatment can effectively relieve the symptoms, the recurrence rate is high, and there are postoperative complications such as infection. At present, nasal spray hormone, antibiotics and other western drugs are used in clinical treatment, but there are drug dependence and toxic side effects. However, traditional Chinese medicine (TCM) has made remarkable progress in the treatment and promotion of postoperative recovery, guided by its unique TCM theory, and has little toxic and side effects, providing more treatment options for patients. PURPOSE The review aims to elucidate the mechanism of CRS from the aspects of traditional medicine and modern medicine, and evaluate the influence of TCM compound, components of TCM, TCM nasal irrigation, TCM fumigation and other auxiliary treatment methods on CRS, providing a new perspective for the application of TCM in CRS. METHODS We conducted the literature retrieval with PubMed, Web of Science, Google Scholar and CNKI databases in a systematic manner (up to July 2024). The keywords included "sinusitis", "chronic rhinosinusitis", "nasal polyps", "herbal medicine", "medicinal plants", "traditional Chinese medicine", "oxidative stress", "pathogenic microbial", "anatomic structure" and so on. The obtained literatures were comprehensively sorted out. For image creation, Figdraw 2.0 was methodically employed. RESULTS The pathogenesis of CRS involves various interaction mechanisms such as bacterial biofilm formation, oxidative stress injury and impaired ciliary mucosa clearance. It is worth noting that TCM exerts therapeutic effects by targeting the above-mentioned pathological processes. Clinical studies have confirmed that TCM comprehensive therapy can significantly improve sinus symptom score, accelerate postoperative mucosal epithelialization, and promote postoperative rehabilitation of CRS. We also discussed the toxic side effects and clinical applications of related drugs. CONCLUSION In TCM, CRS is classified under the diagnostic category of Bi Yuan. Its pathogenesis is attributed to exogenous invasion of the six climatic pathogens (Liu Yin: wind, cold, summer heat, dampness, dryness, and fire), spleen-stomach qi deficiency, internal damp-heat accumulation, and qi-blood stasis. Guided by TCM principles, therapeutic strategies are individualized through syndrome differentiation, which tailors interventions to the patient's unique clinical manifestations. Therapeutic modalities include oral herbal formulations (e.g., decoctions or granules), acupuncture, and acupoint application. These approaches aim to restore physiological balance by harmonizing yin and yang, resolving meridian obstructions, and enhancing lung qi circulation to alleviate nasal congestion and improve ventilation.
Collapse
Affiliation(s)
- Xi-He Zhang
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Qing-Jia Sun
- Department of Otorhinolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Li-Chun Zhao
- College of Pharmaceutical Sciences, Guizhou University of Traditional Chinese Medicine. Guiyang 550025, China
| | - Long Chen
- College of Life Sciences, Jilin Agricultural University, Changchun 130118, China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun 130118, China; College of Life Sciences, Jilin Agricultural University, Changchun 130118, China.
| |
Collapse
|
5
|
García-García P, Évora C, Delgado A, Diaz-Rodriguez P. Chitosan-aloe vera scaffolds with tuned extracellular vesicles and histatin-5 display osteogenic and anti-biofilm activities. Int J Pharm 2025; 676:125592. [PMID: 40228611 DOI: 10.1016/j.ijpharm.2025.125592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 04/05/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
The use of extracellular vesicles (EVs) has garnered significant attention as an alternative to cell-based therapies due to their stability and biocompatibility. In this study, we stimulated mesenchymal stem cells (MSCs) with therapeutic agents affecting the bone regenerative cascade, including bone morphogenetic protein 2 (BMP-2), stromal-derived factor (SDF-1), interleukin 4 (IL-4), alendronate (ALD) and osteogenic differentiation media to obtain osteogenic EVs. The tuned EVs were tested on MSCs and fibroblasts, selecting EVs-BMP-2 as suitable systems. Chitosan-aloe vera (AV) scaffolds were designed to allow for the loading and release of these EVs while leveraging the antibacterial and anti-inflammatory properties of AV. To enhance the dual effect on regeneration and antibacterial activity, poly(lactic-co-glycolic acid) (PLGA) microspheres encapsulating Histatin 5 (Hist-5) were incorporated to the scaffolds. Hist-5 encapsulation was successful, and effectively prevented Staphylococcus aureus biofilm formation on the scaffolds surface. The optimized chitosan-AV scaffolds loaded with EVs-BMP-2 promoted MSCs adhesion and proliferation and exhibited a 2-fold increase in osteogenic differentiation compared to chitosan scaffolds. This study demonstrates the successful combination of bioengineered EVs and Hist-5-loaded microspheres within a chitosan-AV scaffold, providing a promising dual approach for enhancing bone regeneration while reducing the risk of infection. These systems show potential as effective implants for bone fractures, offering both antibacterial and regenerative capabilities.
Collapse
Affiliation(s)
- Patricia García-García
- Department of Chemical Engineering and Pharmaceutical Technology, Universidad de La Laguna 38206 La Laguna, Spain; Institute of Biomedical Technologies (ITB), Universidad de La Laguna 38320 La Laguna, Spain
| | - Carmen Évora
- Department of Chemical Engineering and Pharmaceutical Technology, Universidad de La Laguna 38206 La Laguna, Spain; Institute of Biomedical Technologies (ITB), Universidad de La Laguna 38320 La Laguna, Spain
| | - Araceli Delgado
- Department of Chemical Engineering and Pharmaceutical Technology, Universidad de La Laguna 38206 La Laguna, Spain; Institute of Biomedical Technologies (ITB), Universidad de La Laguna 38320 La Laguna, Spain.
| | - Patricia Diaz-Rodriguez
- Institute of Biomedical Technologies (ITB), Universidad de La Laguna 38320 La Laguna, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, I+D Farma Group (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela 15782 Santiago de Compostela, Spain.
| |
Collapse
|
6
|
Byun J, Jung M, Chung K, Jung SH, Jang H, Choi CH, Kim SH. Risk Factors for Failure to Eradicate Infection after Single Arthroscopic Debridement in Septic Arthritis of a Native Knee Joint. Yonsei Med J 2025; 66:295-301. [PMID: 40288901 PMCID: PMC12041402 DOI: 10.3349/ymj.2024.0190] [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: 07/10/2024] [Revised: 09/06/2024] [Accepted: 09/19/2024] [Indexed: 04/29/2025] Open
Abstract
PURPOSE To identify the risk factors and effect of empirical glycopeptide on the failure of single arthroscopic debridement for septic knee arthritis in a native knee joint. MATERIALS AND METHODS Patients who underwent arthroscopic debridement for septic knee arthritis from March 2005 to December 2022 at one institution were included in this study. Demographic data, comorbidities, preoperative factors including history of previous surgery, history of injection, laboratory data including preoperative C-reactive protein (CRP) and white blood cell (WBC) count, isolated pathogens from synovial fluid culture, and Gachter stage were analyzed. Statistical analyses using univariate and logistic regression were performed. RESULTS Out of 132 patients, 17 patients (12.9%) had more than one additional arthroscopic debridement. History of diabetes mellitus (DM) (p<0.001), previous injection (p=0.041), isolated Staphylococcus aureus in synovial fluid (p=0.010), and high Gachter stage (p=0.002) were identified as risk factors, whereas age, history of previous knee surgery at the affected knee, CRP level, preoperative WBC, and preoperative neutrophil count of synovial fluid had no significant relation. Logistic regression analysis showed significant increase of risk in patients with DM [odds ratio (OR) 12.002, 95% confidence interval (CI) 3.243-44.418, p<0.001], previous injection history (OR 4.812, 95% CI 1.367-16.939, p=0.017), and isolation of Staphylococcus aureus in synovial fluid (OR 4.804, 95% CI 1.282-18.001, p=0.031) as independent risk factors for failure of infection eradication after single arthroscopic debridement. CONCLUSION Comorbidity of DM, history of previous injection, isolated Staphylococcus aureus in synovial fluid, and high Gachter stage were associated with a higher risk of failure to eradicate infection with a single arthroscopic procedure. Empirical glycopeptide administration also showed no significant benefit in reducing the risk of additional surgical procedures for infection control, suggesting against the routine administration of glycopeptide.
Collapse
Affiliation(s)
- Junwoo Byun
- Arthroscopy and Joint Research Institute, Yonsei University College of Medicine, Seoul, Korea
- Department of Orthopedic Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Min Jung
- Arthroscopy and Joint Research Institute, Yonsei University College of Medicine, Seoul, Korea
- Department of Orthopedic Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Kwangho Chung
- Arthroscopy and Joint Research Institute, Yonsei University College of Medicine, Seoul, Korea
- Department of Orthopedic Surgery, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea
| | - Se-Han Jung
- Arthroscopy and Joint Research Institute, Yonsei University College of Medicine, Seoul, Korea
- Department of Orthopedic Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hyeokjoo Jang
- Department of Orthopedic Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Chong-Hyuk Choi
- Arthroscopy and Joint Research Institute, Yonsei University College of Medicine, Seoul, Korea
- Department of Orthopedic Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sung-Hwan Kim
- Arthroscopy and Joint Research Institute, Yonsei University College of Medicine, Seoul, Korea
- Department of Orthopedic Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
7
|
Gao M, Yu X, Yang C, Mi Z, Bai C, Liu C, Liu H. Characteristics and Antibacterial Activity of Staphylococcus aureus Phage-Derived Endolysin LysP4. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10543-0. [PMID: 40299201 DOI: 10.1007/s12602-025-10543-0] [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] [Accepted: 04/16/2025] [Indexed: 04/30/2025]
Abstract
The rise in multidrug resistance and strong biofilm-forming ability of Staphylococcus aureus has led to significant public health concerns. Phage or phage-derived components, such as depolymerase or endolysin, have been considered as potential alternatives to antibiotics for combating antibiotic-resistant bacterial infections. In this study, we cloned and expressed a Staphylococcus aureus phage endolysin, LysP4, and identified its lytic activity. The bactericidal effect of LysP4 was more pronounced against planktonic cells in the logarithmic phase compared to those in the stationary phase. LysP4 reduces bacterial counts by 3 log CFU/mL in 60 min and about 2 log CFU/mL during the stationary phase. LysP4 exhibited optimal lytic activity at pH 5.0-7.0 and remained stable across a temperature range of 16 to 40 °C, with maximal activity observed at 37 °C. LysP4 effectively targets 31 of 38 Staphylococcus strains and successfully eliminates biofilms, reducing bacterial counts by 4 log CFU/mL when combined with vancomycin. Notably, LysP4 demonstrated no hemolytic effects on human red blood cells and no toxic effects on embryonic kidney cells or lung cancer cells. Based on these findings, we believe that LysP4 holds promise as a biological control agent against Staphylococcus infections.
Collapse
Affiliation(s)
- Mingming Gao
- Chinese PLA General Hospital, Beijing, 100853, China
| | - Xinting Yu
- Mianyang Central Hospital, Mianyang, 621000, Sichuan, China
| | - Cuiping Yang
- College of Pulmonary and Critical Care Medicine, The 8th Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Zhiqiang Mi
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Changqing Bai
- Department of Respiratory and Critical Care Diseases, General Hospital of Shenzhen University, Shenzhen, 518055, China
| | - Chuanbin Liu
- Western Medical Branch of PLA General Hospital, Beijing, 100853, China.
| | - Huiying Liu
- College of Pulmonary and Critical Care Medicine, The 8th Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
8
|
Goldie SP, Lau LC, Jones HAS, Harries PG, Walls AF, Salib RJ. Identification of Novel Staphylococcus aureus Core and Accessory Virulence Patterns in Chronic Rhinosinusitis. Int J Mol Sci 2025; 26:3711. [PMID: 40332362 PMCID: PMC12027640 DOI: 10.3390/ijms26083711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 05/08/2025] Open
Abstract
Staphylococcus aureus (S. aureus) colonizes the nasal cavities of both healthy individuals and patients with chronic rhinosinusitis (CRS) with (CRSwNP) and without (CRSsNP) nasal polyps. Treatment-resistant S. aureus biofilms and intracellular persistence are common in CRS patients, requiring the expression of specific virulence factor genes to transition into these forms. We hypothesized that S. aureus isolates from non-diseased controls, CRSsNP patients, and CRSwNP patients would exhibit distinct virulence factor patterns contributing to persistence and intracellular survival in CRS patients. Nasal swabs from seventy-seven individuals yielded S. aureus cultures in eight non-diseased controls, eight CRSsNP patients, and five CRSwNP patients. Whole-genome sequencing analyzed stress, antimicrobial resistance, and virulence genes, including plasmids and prophages. Four virulence factor gene patterns emerged: a core set (hlgA, icaC, hlgB, hlgC, hld, and aur) present in all isolates, and accessory sets, including the enterotoxin gene cluster (seo, sem, seu, sei, and sen) and a partial/complete invasive virulence factor set (splE, splA, splB, lukE, and lukD) (p = 0.001). CRSwNP isolates exhibited incomplete carriage of the core set, with frequent loss of scn, icaC, and hlgA (p < 0.05). These findings suggest that S. aureus has clusters of virulence factors that may act in concert to support the survival and persistence of the bacteria, resulting in enhanced pathogenicity. This may manifest clinically with resistant disease and refractoriness to antibiotics.
Collapse
Affiliation(s)
- Simon P. Goldie
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (S.P.G.); (L.C.L.); (A.F.W.)
- Department of Otorhinolaryngology/Head & Neck Surgery, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Laurie C. Lau
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (S.P.G.); (L.C.L.); (A.F.W.)
| | - Huw A. S. Jones
- Department of Otorhinolaryngology/Head & Neck Surgery, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Philip G. Harries
- Department of Otorhinolaryngology/Head & Neck Surgery, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Andrew F. Walls
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (S.P.G.); (L.C.L.); (A.F.W.)
| | - Rami J. Salib
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (S.P.G.); (L.C.L.); (A.F.W.)
- Department of Otorhinolaryngology/Head & Neck Surgery, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| |
Collapse
|
9
|
Hayles A, Nguyen HN, Alemie M, Vongsvivut J, Ninan N, Bright R, Dabare PR, Gibson C, Truong VK, Vasilev K. Electrostatic charge at the biomaterial-pathogen interface influences antibiotic efficacy. ADVANCED BIOTECHNOLOGY 2025; 3:10. [PMID: 40175809 PMCID: PMC11965051 DOI: 10.1007/s44307-025-00061-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 04/04/2025]
Abstract
Implant-associated infections (IAI) are a considerable burden for healthcare systems globally. While novel anti-infective biomaterials are being pursued, prophylactic antibiotic treatment remains the most important intervention for mitigating IAI. The antibiotic tolerance of bacteria has been widely studied, but until recently, the contributions of biomaterial-pathogen interactions have been overlooked. In the present study, we investigate how material electrostatic charge influences the physiological state of the most clinically challenging pathogen-Staphylococcus aureus, and the implications on its antibiotic tolerance. We utilized a combination of techniques, including quantitative gene expression and synchrotron-sourced attenuated total reflectance Fourier-transform microspectroscopy, to characterize this phenomenon - elucidating how surface attachment to differently charged substrates drives the pathogen to modify its phenotype. Subsequently, we found a direct relationship between the activity of oppositely charged antibiotics (vancomycin and cefazolin) and the biomaterial-pathogen interface, which we determined to be governed by material electrostatic properties. The findings of the present study have the potential to inform the development of enhanced procedures of antibiotic prophylaxis by instructing personalized biomaterial-antibiotic pairing strategies. These new insights hold promise to contribute to reducing the rate of IAI by enabling clinicians and surgeons to maximize the efficacy of prophylactic antibiotic treatments during implant placement procedures.
Collapse
Affiliation(s)
- Andrew Hayles
- Biomedical Nanoengineering Laboratory, Flinders University, Bedford Park, SA, 5042, Australia.
| | - Huu Ngoc Nguyen
- Biomedical Nanoengineering Laboratory, Flinders University, Bedford Park, SA, 5042, Australia
- School of Biomedical Engineering, Faculty of Engineering, University of Sydney, Sydney, NSW, 2050, Australia
| | - Markos Alemie
- Biomedical Nanoengineering Laboratory, Flinders University, Bedford Park, SA, 5042, Australia
| | - Jitraporn Vongsvivut
- Infrared Microspectroscopy (IRM) Beamline, ANSTO ‒ Australian Synchrotron, 800 Blackburn Road, Clayton, VIC, 3168, Australia
| | - Neethu Ninan
- Biomedical Nanoengineering Laboratory, Flinders University, Bedford Park, SA, 5042, Australia
| | - Richard Bright
- Biomedical Nanoengineering Laboratory, Flinders University, Bedford Park, SA, 5042, Australia
| | | | - Christopher Gibson
- Flinders Microscopy and Microanalysis, Flinders University, Bedford Park, SA, 5042, Australia
- Adelaide Microscopy, the University of Adelaide, Adelaide, SA, 5000, Australia
| | - Vi Khanh Truong
- Biomedical Nanoengineering Laboratory, Flinders University, Bedford Park, SA, 5042, Australia
- Deaprtment of Biomedical Engineering, Healthcare Engineering Innovation Centre, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Krasimir Vasilev
- Biomedical Nanoengineering Laboratory, Flinders University, Bedford Park, SA, 5042, Australia.
| |
Collapse
|
10
|
Chen Z, Zhang R, Wang T, Peng Y, Zhou Q, Cao P, Xiao X, Li F, Wei Z, Wang Y, Xu D, Qiao B, Cheng S, Wu Q, Niu L. Nanosheet-shaped WS 2/ICG nanocomposite for photodynamic/photothermal synergistic bacterial clearance and cutaneous regeneration on infectious wounds. BIOMATERIALS ADVANCES 2025; 169:214192. [PMID: 39854997 DOI: 10.1016/j.bioadv.2025.214192] [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: 12/04/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025]
Abstract
Bacterial infections present a significant threat to human health, a challenge that is intensified by the slow pace of novel antibiotic development and the swift emergence of bacterial resistance. The development of novel antibacterial agents is crucial. Indocyanine green (ICG), a widely used imaging dye, efficiently generates reactive oxygen species (ROS) and heat for treating bacterial infections but suffers from aggregation and instability, limiting its efficacy. In this study, tungsten disulfide (WS₂) nanosheet with a high surface area was used to load ICG, creating a multifunctional nanocomposite, WS2/ICG, aimed at treating bacteria-infected wounds. The two-dimensional surface structure of WS₂ provides dispersible binding sites for ICG, and the synthesized nanocomposite exhibits excellent stability. Under near-infrared (NIR) laser excitation, the generated heat further synergistically enhances the yield of singlet oxygen. Additionally, the WS₂/ICG nanoplatform synergistically combines photothermal effect with photodynamic effect, achieving a "1 + 1 > 2" enhancement. Upon NIR laser excitation, the nanocomposite disrupts bacterial cell membranes through localized heating and ROS accumulation, leading to energy metabolism system disruption and subsequent bacterial lysis and death. The findings demonstrate WS₂/ICG's outstanding antibacterial properties and biocompatibility, effectively treating skin infections and promoting tissue regeneration, providing a simple and promising solution for bacteria-infected wounds.
Collapse
Affiliation(s)
- Zhiling Chen
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, School of Basic Medical Sciences and Life Sciences, Hainan Medical University, Haikou 571199, China
| | - Rui Zhang
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou 571199, China; Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital, Hainan Medical University, Haikou 570102, China
| | - Tao Wang
- Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital, Hainan Medical University, Haikou 570102, China; School of Public Health, Hainan Medical University, Haikou 571199, China
| | - Yanan Peng
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou 571199, China; Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital, Hainan Medical University, Haikou 570102, China
| | - Qionglin Zhou
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, School of Basic Medical Sciences and Life Sciences, Hainan Medical University, Haikou 571199, China
| | - Peipei Cao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, School of Basic Medical Sciences and Life Sciences, Hainan Medical University, Haikou 571199, China
| | - Xinxin Xiao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, School of Basic Medical Sciences and Life Sciences, Hainan Medical University, Haikou 571199, China
| | - Fengling Li
- Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education, International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, Hainan Provincial Key Laboratory of Research and Development on Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou 571199, China
| | - Ziming Wei
- Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital, Hainan Medical University, Haikou 570102, China
| | - Yuanyuan Wang
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou 571199, China
| | - Dan Xu
- Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education, International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province, Hainan Provincial Key Laboratory of Research and Development on Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou 571199, China
| | - Bin Qiao
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou 571199, China
| | - Shaowen Cheng
- Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital, Hainan Medical University, Haikou 570102, China; Department of Wound Repair, The First Affiliated Hospital, Hainan Medical University, Haikou 570102, China.
| | - Qiang Wu
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou 571199, China; Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital, Hainan Medical University, Haikou 570102, China.
| | - Lina Niu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, School of Basic Medical Sciences and Life Sciences, Hainan Medical University, Haikou 571199, China.
| |
Collapse
|
11
|
Mu WB, Yao LQ, Guo ZY, Ma YC, Wang F, Yang JH. Enhancing biofilm disruption and bactericidal efficiency using vancomycin-loaded microbubbles in sonodynamic therapy. JAC Antimicrob Resist 2025; 7:dlaf045. [PMID: 40110553 PMCID: PMC11920867 DOI: 10.1093/jacamr/dlaf045] [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: 11/21/2024] [Accepted: 03/06/2025] [Indexed: 03/22/2025] Open
Abstract
Background Periprosthetic joint infection (PJI) is a significant complication following arthroplasty, attributed to the biofilm formation. This study evaluates the effectiveness of vancomycin-loaded microbubbles (Van-MBs) in conjunction with ultrasound-targeted microbubble destruction (UTMD) on biofilm disruption and bactericidal efficiency. Methods Van-MBs were prepared using the thin-film hydration method and characterized using microscopy, dynamic light scattering analysis, and high-performance liquid chromatography (HPLC). Confocal laser scanning microscopy (CLSM) was used to assess the penetration of Van and Van-MBs into biofilms. Biofilms were treated with Van, Van-MBs, UTMD, and Van-MBs + UTMD. CLSM and crystal violet staining were utilized to assess the morphology, viability, and biomass of the biofilms. Bacterial activity was examined through scanning electron microscopy (SEM) and plate counting, while gene expression was analyzed using quantitative real-time polymerase chain reaction (qRT-PCR). Results The results demonstrated that Van-MBs penetrated deeper into methicillin-resistant Staphylococcus aureus (MRSA) biofilms compared with Van alone. The combination of Van-MBs and UTMD significantly reduced biofilm thickness, viability, and biomass. qRT-PCR analysis revealed that the Van-MBs + UTMD group exhibited lower transcription levels of the icaA gene, suggesting that the treatment disrupted biofilm formation by suppressing this key gene. SEM further confirmed the efficacy of the treatment, showing that Van-MBs + UTMD induced cytoplasmic shrinkage and separation of the outer and cytoplasmic membranes in MRSA cells, indicating substantial structural damage to the bacterial cells. Conclusion These findings demonstrate the potential of Van-MBs in combination with UTMD as an innovative approach to enhance antibiotic efficacy and eliminate biofilms in the treatment of PJI.
Collapse
Affiliation(s)
- Wen B Mu
- Department of Pharmacognosy, School of Pharmacy, Xinjiang Medical University, Urumqi, Xinjiang 830011, China
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Li Q Yao
- Department of Sports Medicine, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Zi Y Guo
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - You C Ma
- Department of Sports Medicine, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Fei Wang
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Jian H Yang
- Department of Pharmacognosy, School of Pharmacy, Xinjiang Medical University, Urumqi, Xinjiang 830011, China
- Department of Pharmacy, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, China
| |
Collapse
|
12
|
Zhang H, Qiao W, Liu Y, Yao X, Zhai Y, Du L. Addressing the challenges of infectious bone defects: a review of recent advances in bifunctional biomaterials. J Nanobiotechnology 2025; 23:257. [PMID: 40158189 PMCID: PMC11954225 DOI: 10.1186/s12951-025-03295-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/04/2025] [Indexed: 04/01/2025] Open
Abstract
Infectious bone defects present a substantial clinical challenge due to the complex interplay between infection control and bone regeneration. These defects often result from trauma, autoimmune diseases, infections, or tumors, requiring a nuanced approach that simultaneously addresses infection and promotes tissue repair. Recent advances in tissue engineering and materials science, particularly in nanomaterials and nano-drug formulations, have led to the development of bifunctional biomaterials with combined osteogenic and antibacterial properties. These materials offer an alternative to traditional bone grafts, minimizing complications such as multiple surgeries, high antibiotic dosages, and lengthy recovery periods. This review examines the repair mechanisms in the infectious microenvironment and highlights various bifunctional biomaterials that foster both anti-infective and osteogenic processes. Emerging design strategies are also discussed to provide a forward-looking perspective on treating infectious bone defects with clinically significant outcomes.
Collapse
Affiliation(s)
- Huaiyuan Zhang
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Wenyu Qiao
- Department of General Surgery, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Yu Liu
- Research Center for Clinical Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, 201508, China
| | - Xizhou Yao
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Yonghua Zhai
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
| | - Longhai Du
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, China.
| |
Collapse
|
13
|
Du B, Xue F, Xu H, Zhao R, Zhang T, Han S, Zhu T, Zhu Y, Zhao Y. Mechanism of antibacterial and antibiofilm of thiazolidinone derivative TD-H2-A against Staphylococcus aureus. Sci Rep 2025; 15:10380. [PMID: 40140486 PMCID: PMC11947284 DOI: 10.1038/s41598-025-94571-4] [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: 08/13/2024] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Staphylococcus aureus is one of the most common pathogens causing widespread infections. It has been demonstrated that thiazolidinone derivative (TD-H2-A), a small molecule compound that targets WalK protein through high-throughput screening, exerts antibacterial and anti-biofilm effects on S. aureus. In this study, we further ascertained the impact of TD-H2-A on biofilms at different stages. The phosphorylation assay and RNA sequencing were carried out to elucidate the underlying mechanism. The results revealed that TD-H2-A inhibited WalK autophosphorylation, implying that the antibacterial effect of TD-H2-A may be achieved by inhibiting the activity of WalK. The transcriptome analysis showed that TD-H2-A treatment induced 994 differentially expressed genes (DEGs), of which, 481 were upregulated and 513 were downregulated. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis revealed that 43 among 58 genes involved in ribosome synthesis were upregulated, and the transcript levels of the genes responsible for membrane transport were altered significantly. According to our research, TD-H2-A has an antibacterial mechanism with multitarget and multipathway. This study provided new ideas for the development of new drug target screening against S. aureus infections.
Collapse
Affiliation(s)
- Bingyu Du
- Laboratory Medicine Center, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Fen Xue
- Laboratory Medicine Center, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Hui Xu
- Laboratory Medicine Center, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Rui Zhao
- Department of Clinical Microbiology, Shanghai Centre for Clinical Laboratory, Shanghai, People's Republic of China
| | - Tiantian Zhang
- Laboratory Medicine Center, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Shiqing Han
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, People's Republic of China
| | - Tao Zhu
- Department of Medical Microbiology and Immunology, Wannan Medical College, Wuhu, People's Republic of China.
| | - Yefei Zhu
- Laboratory Medicine Center, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China.
| | - Yanfeng Zhao
- Laboratory Medicine Center, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China.
| |
Collapse
|
14
|
Lavoie T, Daffinee KE, Vicent ML, LaPlante KL. Staphylococcus biofilm dynamics and antibiotic resistance: insights into biofilm stages, zeta potential dynamics, and antibiotic susceptibility. Microbiol Spectr 2025; 13:e0291524. [PMID: 40135862 PMCID: PMC12054104 DOI: 10.1128/spectrum.02915-24] [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: 12/10/2024] [Accepted: 01/16/2025] [Indexed: 03/27/2025] Open
Abstract
Staphylococcus spp. infections often involve biofilms, but standard antibiotic minimum inhibitory concentration (MIC) testing used to determine treatment evaluates planktonic bacterial growth only and does not account for biofilm presence, strength, or growth stage. To aid in determining a cost-effective method to solve this issue, we built upon in vitro methods initially published by Stepanovic et al. used to determine weak and strong biofilm formations. First, we determined 115 unique S. aureus isolate biofilms at 2, 4, 6, 8, 16, and 24 h to classify the hourly stages of biofilm development based on statistically significant final growth results (P < 0.001): stages one (0-6 h), two (6-16 h), three (16-24 h), and four (>24 h). Next, to further evaluate in vitro biofilm strength, electrostatic differences were measured through zeta (ζ)-potential for strong and weak biofilm producers at early and late stage-formed biofilms. The early stages of weak biofilm formers had a greater negative electrostatic charge when compared to strong biofilm formers. Meanwhile, strong biofilm formers began early stages with less negative charges before increasing the negative electrostatic charge by stage-four biofilm. At all time points, weak biofilm-forming isolate mean ζ-potentials were significantly more negative than strong biofilm formers (P = ≤0.04). Finally, to elucidate minimum eradication concentrations for biofilms, we treated stage-four biofilms with progressively higher concentrations of either daptomycin, vancomycin, or levofloxacin. Daptomycin was the only antibiotic to achieve ≥75% reduction in biofilm viability, seen at 32-256 μg/mL (64-512× MIC), and significantly reduced residual biofilm across all strong and weak biofilms. Biofilm findings showed an unexpected initial biofilm decrease in response to lower concentrations of antibiotics, followed by an increase in biofilm biomass at higher antibiotic concentrations. While higher antibiotic concentrations can be used to overcome bacterial resistance and eliminate infections, our results suggest that antimicrobial resistance is observed, regardless of bacterial biofilm strength, and that there may be an optimal treatment concentration window for achieving maximum kill. Our data add to the increasing evidence of biofilms' role in recurrent infections and the importance of antibiotic concentration.IMPORTANCEThis work is significant, as it addresses a critical gap in standard antibiotic testing by focusing on the unique characteristics of biofilm-forming Staphylococcus aureus infections, which are major contributors to recurrent and chronic infections. Unlike traditional MIC testing that evaluates planktonic bacteria, this study emphasizes the importance of biofilm presence, growth stages, and electrostatic properties in determining treatment strategies. By classifying biofilm development into distinct stages in an easily reproducible assay and measuring the biofilm zeta-potential for key differences and overall biofilm response to multiple standard antibiotic concentrations, this research provides valuable insights for the future of biofilm in vitro work. Furthermore, it highlights the efficacy of daptomycin in eradicating biofilm while identifying possibilities of optimal antibiotic concentration windows, a critical consideration for mitigating resistance and achieving effective infection control. These findings underscore the necessity of tailoring treatment to biofilm-specific dynamics, offering a path toward more effective therapeutic approaches for biofilm-associated infections.
Collapse
Affiliation(s)
- T. Lavoie
- College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
- Providence Veterans Affairs Medical Center, Providence, Rhode Island, USA
| | - K. E. Daffinee
- Providence Veterans Affairs Medical Center, Providence, Rhode Island, USA
| | - M. L. Vicent
- College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
- Providence Veterans Affairs Medical Center, Providence, Rhode Island, USA
| | - K. L. LaPlante
- College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
- Providence Veterans Affairs Medical Center, Providence, Rhode Island, USA
- Division of Infectious Diseases, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| |
Collapse
|
15
|
Branca M, Silva TP, Lemos ASO, Campos LM, Souza TF, Palazzi C, Oliveira V, Coimbra ES, Silva FON, F B Pontes AC, M Apolônio AC, Melo RCN, de L Pontes D, Fabri RL. The Fe-Cyclam-Derived Compound [Fe(cyclam)sal]PF 6 Restrains Drug-Resistant Staphylococcus aureus Proliferation and Biofilm Formation. ACS OMEGA 2025; 10:11386-11396. [PMID: 40160729 PMCID: PMC11947776 DOI: 10.1021/acsomega.4c11347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/25/2025] [Accepted: 03/04/2025] [Indexed: 04/02/2025]
Abstract
Staphylococcus aureus is a bacterium found on the skin and mucous membranes of humans and animals. This micro-organism is classified as an opportunistic pathogen and causes infections in both hospital and community settings. The increase in antibiotic resistance, especially methicillin-resistant S. aureus (MRSA), is a major challenge for clinical and epidemiological practice. The present study aims to investigate the potential antibacterial and antibiofilm activities of the compound [Fe(cyclam)sal]PF6 against drug-resistant strains of S. aureus. The minimum inhibitory concentration (MIC) and the minimum bactericidal concentration (MBC) against S. aureus strains ATCC 25904, S. aureus ATCC 33591, and S. aureus 05-0052 were determined for [Fe(cyclam)sal]PF6. First, bacterial abundance, viability, and cell envelope damage in planktonic cultures were investigated in response to this compound. Second, its potential effect on biofilm proliferation and adhesion was evaluated using different approaches: optical density (OD), scanning electron microscopy (SEM), and biochemical analysis of the extracellular polymeric matrix. The complex [Fe(cyclam)sal]PF6 inhibited bacterial growth and induced an increase in cell death. The compound disrupted the integrity of the cell membrane, resulting in the release of cytoplasmic contents into the extracellular medium. Remarkably, the metal complex reduced the pre-established S. aureus biofilm and impaired its adhesion. Furthermore, it is not toxic to mammalian cells. The compound [Fe(cyclam)sal]PF6 affects both the proliferation and biofilm formation of drug-resistant strains of S. aureus, demonstrating strong potential for the design of novel antimicrobial agents.
Collapse
Affiliation(s)
- Matheus
T. Branca
- Laboratory
of Bioactive Natural Products, Department of Biochemistry, Institute
of Biological Sciences, Federal University
of Juiz de Fora, Campus, Juiz de Fora, Minas
Gerais 36036-900, Brazil
| | - Thiago P. Silva
- Laboratory
of Cellular Biology, Department of Biology, Institute of Biological
Sciences, Federal University of Juiz de
Fora, Campus, Juiz de Fora, Minas Gerais 36036-900, Brazil
| | - Ari S. O. Lemos
- Laboratory
of Bioactive Natural Products, Department of Biochemistry, Institute
of Biological Sciences, Federal University
of Juiz de Fora, Campus, Juiz de Fora, Minas
Gerais 36036-900, Brazil
| | - Lara M. Campos
- Laboratory
of Bioactive Natural Products, Department of Biochemistry, Institute
of Biological Sciences, Federal University
of Juiz de Fora, Campus, Juiz de Fora, Minas
Gerais 36036-900, Brazil
| | - Thalita F. Souza
- Laboratory
of Bioactive Natural Products, Department of Biochemistry, Institute
of Biological Sciences, Federal University
of Juiz de Fora, Campus, Juiz de Fora, Minas
Gerais 36036-900, Brazil
| | - Cinthia Palazzi
- Laboratory
of Cellular Biology, Department of Biology, Institute of Biological
Sciences, Federal University of Juiz de
Fora, Campus, Juiz de Fora, Minas Gerais 36036-900, Brazil
| | - Verônica
S. Oliveira
- Department
of Pharmacy, Health Sciences Center, Federal
University of Rio Grande do Norte, Campus, Natal, Rio Grande do Norte 59078-970, Brazil
| | - Elaine S. Coimbra
- Laboratory
of Parasitology, Department of Parasitology, Microbiology and Immunology,
Institute of Biological Sciences, Federal
University of Juiz de Fora, Juiz
de Fora 36036-900, Brazil
| | - Francisco O. N. Silva
- Laboratory
of Coordination Chemistry and Polymers, Institute of Chemistry, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| | - Ana Cristina F B Pontes
- Laboratory
of Coordination Chemistry and Polymers, Institute of Chemistry, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| | - Ana Carolina M Apolônio
- Laboratory
of Bacterial Physiology and Molecular Genetics, Department of Parasitology,
Microbiology and Immunology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Rossana C. N. Melo
- Laboratory
of Cellular Biology, Department of Biology, Institute of Biological
Sciences, Federal University of Juiz de
Fora, Campus, Juiz de Fora, Minas Gerais 36036-900, Brazil
| | - Daniel de L Pontes
- Laboratory
of Coordination Chemistry and Polymers, Institute of Chemistry, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| | - Rodrigo L. Fabri
- Laboratory
of Bioactive Natural Products, Department of Biochemistry, Institute
of Biological Sciences, Federal University
of Juiz de Fora, Campus, Juiz de Fora, Minas
Gerais 36036-900, Brazil
| |
Collapse
|
16
|
Xu Y, Wang L, Guo D, Wang Y, Liu X, Sun Y, Wang R, Sun L, Jiang P, Liu Q, Wang B, Yan M, Zhao Y. Baohuoside I targets SaeR as an antivirulence strategy to disrupt MRSA biofilm formation and pathogenicity. NPJ Biofilms Microbiomes 2025; 11:45. [PMID: 40102466 PMCID: PMC11920273 DOI: 10.1038/s41522-025-00681-2] [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: 09/29/2024] [Accepted: 03/08/2025] [Indexed: 03/20/2025] Open
Abstract
The emergence of methicillin-resistant Staphylococcus aureus (MRSA) represents a critical global health challenge, making the SaeRS two-component system (TCS), a key regulator of S. aureus virulence, an ideal target for novel therapeutic approaches. In this study, virtual screening and thermal shift assays identified Baohuoside I (BI), a flavonol glycoside, as a potent inhibitor of the SaeR response regulator. BI significantly attenuated S. aureus pathogenicity without bactericidal effects, suppressing the expression of key virulence factors, such as hemolysin A (Hla) and Panton-Valentine leukocidin (PVL), while modulating immune evasion pathways. Additionally, BI disrupted biofilm formation, promoting the development of porous, less structured biofilms. Biochemical assays, including EMSA, CETSA, fluorescence quenching, and SPR, confirmed strong binding interactions between SaeR and BI. In vivo, BI demonstrated therapeutic efficacy in Galleria mellonella and rat MRSA models. These findings establish BI as a promising lead for nonbactericidal therapies to combat MRSA infections and mitigate resistance.
Collapse
Affiliation(s)
- Yueshan Xu
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
- Department of Orthopedics, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Li Wang
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, China
| | - Dongbin Guo
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yueying Wang
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
- Department of Orthopedics, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Xinyao Liu
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yun Sun
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Rong Wang
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Luanbiao Sun
- China-Japan Union Hospital of Jilin University, Changchun, China
| | - Peitong Jiang
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Quan Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Jilin University, Changchun, China
| | - Bingmei Wang
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China.
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, China.
| | - Ming Yan
- Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China.
- Department of Orthopedics, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China.
| | - Yicheng Zhao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Jilin University, Changchun, China.
- Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, China.
| |
Collapse
|
17
|
Johnson CR, Schoenfisch MH. Photoinitiated Nitric Oxide Release as an Antibacterial Treatment for Chronic Wounds. ACS Biomater Sci Eng 2025; 11:1510-1522. [PMID: 39933146 DOI: 10.1021/acsbiomaterials.4c01955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
Taking advantage of their innate roles as antibacterial strategies, the dual activity of photobiomodulation (PBM) and nitric oxide (NO) was combined to provide a tunable, on-demand chronic wound therapeutic. S-nitrosothiol-modified mesoporous silica nanoparticles (RSNO-MSNs) were doped into polyurethane (PU) to demonstrate preliminary utility as an antibacterial wound dressing treatment for chronic wounds. Photoinitiated and resultant NO-release kinetics and payloads were evaluated at 405, 430, and 530 nm for multiple irradiances. The use of photons and the NO-releasing MSNs against common chronic wound pathogens, such as Pseudomonas aeruginosa and Staphylococcus aureus, proved to be highly bactericidal. Cytocompatibility of the treatment was confirmed using human epidermal keratinocytes, a representative skin cell line.
Collapse
Affiliation(s)
- Courtney R Johnson
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Mark H Schoenfisch
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
18
|
Isaac P, Breser ML, De Lillo MF, Bohl LP, Calvinho LF, Porporatto C. Understanding the bovine mastitis co-infections: Coexistence with Enterobacter alters S. aureus antibiotic susceptibility and virulence phenotype. Res Vet Sci 2025; 185:105547. [PMID: 39855057 DOI: 10.1016/j.rvsc.2025.105547] [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: 10/22/2024] [Revised: 01/14/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
The World Health Organization recently reported an alarming evolution and spread of antibiotic resistance, a global risk factor recognized as a One Health challenge. In veterinary, the general lack of clear treatment guidelines often leads to antibiotic misuse. Bovine mastitis is responsible for major economic losses and the main cause of antibiotic administration in the dairy industry, favoring the emergence of multi-resistant phenotypes. The complexity of inter-microbial and host-pathogen interactions in the mammary gland, demonstrated by culture-independent techniques, not only complicates the prediction of antibiotic treatment outcomes but also underscores the urgent need for further research in this field. This work evaluated the interactions between S. aureus L33 and Enterobacter sp. L34 obtained from an intramammary co-infection. The behavior of the dual-species culture resembled that of the Enterobacter monoculture in all the evaluated contexts. Most of the selected S. aureus virulence factors and the antibiotic susceptibility were altered by coexisting with Enterobacter. Under the protection of Enterobacter, S. aureus was able to survive upon exposure to concentrations of cloxacillin and other antibiotics that would be bactericidal for the monoculture. This could have serious implications for bacterial clearance of mastitis originating from the underestimated co-infections. These findings highlight the importance of broadening our knowledge of how microbial interactions in intramammary infections could contribute to antibiotic treatments failures. Moreover, they open new perspectives for the design of bovine mastitis therapies that consider the ecological context in order to optimize the antibiotic usage, improve the success rates and reduce antibiotic resistance.
Collapse
Affiliation(s)
- Paula Isaac
- Instituto Multidisciplinario de Investigación y Transferencia Agroalimentaria y Biotecnológica (IMITAB), CONICET-UNVM, Villa María, Córdoba, Argentina; Instituto Académico Pedagógico de Ciencias Básicas y Aplicadas de la Universidad Nacional Villa María (IAPCByA-UNVM), Villa María, Córdoba, Argentina.
| | - María Laura Breser
- Instituto Multidisciplinario de Investigación y Transferencia Agroalimentaria y Biotecnológica (IMITAB), CONICET-UNVM, Villa María, Córdoba, Argentina; Instituto Académico Pedagógico de Ciencias Básicas y Aplicadas de la Universidad Nacional Villa María (IAPCByA-UNVM), Villa María, Córdoba, Argentina
| | - María Florencia De Lillo
- Instituto Multidisciplinario de Investigación y Transferencia Agroalimentaria y Biotecnológica (IMITAB), CONICET-UNVM, Villa María, Córdoba, Argentina
| | - Luciana Paola Bohl
- Instituto Multidisciplinario de Investigación y Transferencia Agroalimentaria y Biotecnológica (IMITAB), CONICET-UNVM, Villa María, Córdoba, Argentina; Instituto Académico Pedagógico de Ciencias Básicas y Aplicadas de la Universidad Nacional Villa María (IAPCByA-UNVM), Villa María, Córdoba, Argentina
| | - Luis Fernando Calvinho
- Departamento de Clínicas, Facultad de Ciencias Veterinarias, Universidad Nacional del Litoral (UNL), Esperanza, Santa Fe, Argentina
| | - Carina Porporatto
- Instituto Multidisciplinario de Investigación y Transferencia Agroalimentaria y Biotecnológica (IMITAB), CONICET-UNVM, Villa María, Córdoba, Argentina; Instituto Académico Pedagógico de Ciencias Básicas y Aplicadas de la Universidad Nacional Villa María (IAPCByA-UNVM), Villa María, Córdoba, Argentina.
| |
Collapse
|
19
|
Pontanayodsakorn C, Eurtivong C, Jiamboonsri P. Antibacterial and antibiofilm activities of kaffir lime essential oils and their active constituents against Staphylococcus aureus focusing on sortase A. Heliyon 2025; 11:e41977. [PMID: 40013263 PMCID: PMC11862495 DOI: 10.1016/j.heliyon.2025.e41977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 02/28/2025] Open
Abstract
Staphylococcus aureus biofilm has become a global medical concern due to rising antibiotic resistance. This study aimed to evaluate the potential activities of kaffir lime essential oils and their active compounds as alternative anti-S. aureus biofilm agents. The compositions of the essential oils were identified by gas chromatography/mass spectrometry (GC/MS), and their antibacterial activity was determined through broth-microdilution and time-kill assays. Antibiofilm activities were assessed using Congo red agar (CRA) well diffusion method, pre-biofilm inhibition resazurin assay, and post-biofilm inhibition assay. Sortase A (SrtA) inhibition was also investigated using in silico and in vitro approaches. This is followed by morphological studies to observe change in biofilm formations using light and scanning electron microscopy (SEM). Phytochemical analysis revealed that the kaffir lime essential oils from leaves (KLL) and peels (KLP) were primarily composed of the monoterpene aldehyde citronellal (59.13 %) and the monoterpene hydrocarbon (-)-limonene (25.69 %). However, the monoterpenoid alcohols, β-citronellol and terpinen-4-ol, which were the third most abundant compounds in KLL (5.35 %) and KLP (10.87 %), respectively, were selected for further study. All test compounds exhibited anti-Staphylococcal activity with a minimum inhibitory concentration (MIC) range of 0.1-0.3 % v/v. Their inhibition above the MIC levels showed time- and concentration-independence. Among the test compounds, terpinen-4-ol revealed good antibiofilm activity by inhibiting biofilm formation rather than eradicating the established biofilm. However, terpinen-4-ol exhibited weak SrtA inhibition with docking score of 32.58 and in vitro SrtA inhibition of 46.14 ± 3.58 % at 1 % v/v. Interestingly, terpinen-4-ol caused visible damage to the bacterial cell barrier, as revealed by SEM micrographs. These findings suggest the potential use of kaffir lime oils and their active compounds to combat biofilm-forming S. aureus infection.
Collapse
Affiliation(s)
- Chayapol Pontanayodsakorn
- Faculty of Medicine, King Mongkut's Institute of Technology Ladkrabang, 1 Chalongkrung Road, Ladkrabang, Bangkok, 10520, Thailand
| | - Chatchakorn Eurtivong
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mahidol University, 447 Si Ayutthaya Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Pimsumon Jiamboonsri
- Faculty of Medicine, King Mongkut's Institute of Technology Ladkrabang, 1 Chalongkrung Road, Ladkrabang, Bangkok, 10520, Thailand
| |
Collapse
|
20
|
Obořilová R, Kučerová E, Botka T, Vaisocherová-Lísalová H, Skládal P, Farka Z. Piezoelectric biosensor with dissipation monitoring enables the analysis of bacterial lytic agent activity. Sci Rep 2025; 15:3419. [PMID: 39870739 PMCID: PMC11772602 DOI: 10.1038/s41598-024-85064-x] [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: 09/11/2024] [Accepted: 12/30/2024] [Indexed: 01/29/2025] Open
Abstract
Antibiotic-resistant strains of Staphylococcus aureus pose a significant threat in healthcare, demanding urgent therapeutic solutions. Combining bacteriophages with conventional antibiotics, an innovative approach termed phage-antibiotic synergy, presents a promising treatment avenue. However, to enable new treatment strategies, there is a pressing need for methods to assess their efficacy reliably and rapidly. Here, we introduce a novel approach for real-time monitoring of pathogen lysis dynamics employing the piezoelectric quartz crystal microbalance (QCM) with dissipation (QCM-D) technique. The sensor, a QCM chip modified with the bacterium S. aureus RN4220 ΔtarM, was utilized to monitor the activity of the enzyme lysostaphin and the phage P68 as model lytic agents. Unlike conventional QCM solely measuring resonance frequency changes, our study demonstrates that dissipation monitoring enables differentiation of bacterial growth and lysis caused by cell-attached lytic agents. Compared to reference turbidimetry measurements, our results reveal distinct alterations in the growth curve of the bacteria adhered to the sensor, characterized by a delayed lag phase. Furthermore, the dissipation signal analysis facilitated the precise real-time monitoring of phage-mediated lysis. Finally, the QCM-D biosensor was employed to evaluate the synergistic effect of subinhibitory concentrations of the antibiotic amoxicillin with the bacteriophage P68, enabling monitoring of the lysis of P68-resistant wild-type strain S. aureus RN4220. Our findings suggest that this synergy also impedes the formation of bacterial aggregates, the precursors of biofilm formation. Overall, this method brings new insights into phage-antibiotic synergy, underpinning it as a promising strategy against antibiotic-resistant bacterial strains with broad implications for treatment and prevention.
Collapse
Affiliation(s)
- Radka Obořilová
- Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic.
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic.
| | - Eliška Kučerová
- Department of Experimental Biology, Section of Genetics and Molecular Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Tibor Botka
- Department of Experimental Biology, Section of Genetics and Molecular Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Hana Vaisocherová-Lísalová
- FZU - Institute of Physics of the Czech Academy of Sciences, Na Slovance 1999/2, 182 00, Prague, Czech Republic
| | - Petr Skládal
- Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Zdeněk Farka
- Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic.
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic.
| |
Collapse
|
21
|
Xia Y, Hu Z, Jin Q, Chen Q, Zhao C, Qiang R, Xie Z, Li L, Zhang H. Structural characteristics, functions, and counteracting strategies of biofilms in Staphylococcus aureus. Comput Struct Biotechnol J 2025; 27:488-500. [PMID: 39916696 PMCID: PMC11799891 DOI: 10.1016/j.csbj.2025.01.021] [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: 11/16/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/09/2025] Open
Abstract
Background Staphylococcus aureus (S. aureus) is a prevalent pathogen associated with a wide range of infections, exhibiting significant antibiotic resistance and posing therapeutic challenges in clinical settings. The formation of biofilms contributes to the emergence of resistant strains, further diminishing the efficacy of antibiotics. This, in turn, leads to chronic and recurrent infections, ultimately increasing the healthcare burden. Consequently, preventing and eliminating biofilms has become a critical focus in clinical management and research. Aim of review This review systematically examines the mechanisms underlying biofilm formation in S. aureus and its contribution to antibiotic resistance, emphasizing the essential roles biofilms play in maintaining structural integrity and enhancing resistance. It also analyses the protective mechanisms that fortify S. aureus biofilms against antimicrobial treatments. Furthermore, the review provides a comprehensive overview of recent therapeutic innovations, including enzymatic therapy, nanotechnology, gene editing, and phage therapy. Key scientific concepts of review Emerging therapeutic strategies present novel approaches to combat S. aureus biofilm-associated infections through various mechanisms. This review discusses recent advancements in these therapies, their practical challenges in clinical application, and provides an in-depth analysis of each strategy's mechanisms and therapeutic potential. By mapping future research directions, this review aims to refine anti-biofilm strategies to control infection progression and effectively mitigate recurrence.
Collapse
Affiliation(s)
- Yanze Xia
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhenghui Hu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qiyuan Jin
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qi Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Chenhao Zhao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Rui Qiang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zonggang Xie
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liubing Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Haifang Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, China
| |
Collapse
|
22
|
Swandiny GF, Filaila E, Priyanto JA, Lotulung PDN, Permatasari V, Okselni T, Prastya ME, Yuliani T, Haryadi W, Darmawan A, Primahana G. Potent antibacterial and cytotoxic bioactive compounds from endophytic fungi Diaporthe sp. associated with Salacia intermedia. Arch Microbiol 2025; 207:40. [PMID: 39841194 DOI: 10.1007/s00203-025-04236-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/30/2024] [Accepted: 01/05/2025] [Indexed: 01/23/2025]
Abstract
Antibacterial screening of endophytic fungi from Salacia intermedia identified Diaporthe longicolla as a potent strain exhibiting good activity against multidrug-resistant Staphylococcus aureus and Pseudomonas aeruginosa, with an MIC of 39.1 µg/mL. Scale-up fermentation and chromatographic purification of this strain yielded three known compounds, which were cytochalasin J (1), cytochalasin H (2), and dicerandrol C (3), as identified by liquid chromatography - high mass resolution mass spectrometry (LC-HRMS) and nuclear magnetic resonance (NMR) spectroscopy. Among those compounds, dicerandrol C exhibited broad-spectrum antibacterial activity against ATCC and multidrug-resistant strains of Bacillus subtilis, S. aureus, and P. aeruginosa, and multidrug-resistant strains of Klebsiella pneumoniae and Escherichia coli, with MIC values ranging from 1.04 to 33.30 µM. Furthermore, dicerandrol C outperformed tetracycline in antibacterial efficacy against S. aureus ATCC 6538 and methicillin-resistant S. aureus (MRSA) strains (MIC of 1.04 µM). Further antibacterial evaluation showed that cytochalasin J (221.43 µM), cytochalasin H (202.59 µM), and dicerandrol C (tested at its MIC values of 1.04 µM for S. aureus ATCC 6538 and 16.65 µM for P. aeruginosa ATCC 15442) significantly inhibited bacterial biofilm formation. The biofilm inhibition percentages ranged from 61.09 to 78.17% for S. aureus and 41.22-56.83% for P. aeruginosa. In cytotoxicity assays against MCF-7 cells, all three compounds reduced cell viability (48.68-74.50%), with dicerandrol C demonstrating the highest potency. These findings highlight the potential of dicerandrol C as a powerful antibacterial and cytotoxic agent, facilitating further investigations into its therapeutic applications.
Collapse
Affiliation(s)
| | - Euis Filaila
- Research Center for Chemistry, National Research and Innovation Agency (BRIN), KST B.J. Habibie, Serpong, South Tangerang, 15314, Indonesia
| | - Jepri Agung Priyanto
- Division of Microbiology, Department of Biology, Faculty of Mathematics and Natural Sciences, IPB University, Bogor, 16680, Indonesia
| | - Puspa Dewi Narrij Lotulung
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), KST B.J. Habibie, Serpong, South Tangerang, 15314, Indonesia
| | - Vera Permatasari
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), KST B.J. Habibie, Serpong, South Tangerang, 15314, Indonesia
| | - Tia Okselni
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), KST B.J. Habibie, Serpong, South Tangerang, 15314, Indonesia
| | - Muhammad Eka Prastya
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), KST B.J. Habibie, Serpong, South Tangerang, 15314, Indonesia
| | - Tri Yuliani
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), KST B.J. Habibie, Serpong, South Tangerang, 15314, Indonesia
| | - Winarto Haryadi
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Gadjah Mada, Sekip Utara, Bulaksumur, Yogyakarta, 55281, Indonesia
| | - Akhmad Darmawan
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), KST B.J. Habibie, Serpong, South Tangerang, 15314, Indonesia
| | - Gian Primahana
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), KST B.J. Habibie, Serpong, South Tangerang, 15314, Indonesia.
| |
Collapse
|
23
|
Leong PY, Tan WQ, Choo WS. Biofilm destruction activity of α-tocopherol against Staphylococcus aureus, Proteus mirabilis, and Pseudomonas aeruginosa. FEMS Microbiol Lett 2025; 372:fnaf020. [PMID: 39904546 PMCID: PMC11992689 DOI: 10.1093/femsle/fnaf020] [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: 08/17/2024] [Revised: 01/27/2025] [Accepted: 02/03/2025] [Indexed: 02/06/2025] Open
Abstract
Antibiotic resistance and the persistence of sessile cells within biofilms complicate the eradication of biofilm-related infections using conventional antibiotics. This highlights the necessity for alternate therapy methods. The objective of this study was to investigate the biofilm destruction activity of α-tocopherol against Staphylococcus aureus, Proteus mirabilis, and Pseudomonas aeruginosa on polystyrene. α-Tocopherol showed significant biofilm destruction activity on the pre-formed biofilms of S. aureus (45%-46%), Pr. mirabilis (42%-54%), and Ps. aeruginosa (28%). Resazurin assay showed that α-tocopherol disrupted all bacterial biofilms without interfering with their cell viability. Scanning electron microscope images showed lower bacterial cell count and less compacted cell aggregates on polystyrene surfaces after treatment with α-tocopherol. This study demonstrated the biofilm destruction activity of α-tocopherol against S. aureus, Pr. mirabilis, and Ps. aeruginosa. α-Tocopherol could potentially be used to decrease biofilm-associated infections of these bacteria.
Collapse
Affiliation(s)
- Pui Yee Leong
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| | - Wei Qi Tan
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| | - Wee Sim Choo
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| |
Collapse
|
24
|
Ran W, Yi P, Jiang L, Yu Y, Zhong K, Wu Y, Gao H. Antibiofilm mechanism of 2R,3R-dihydromyricetin by targeting sortase A and its application against Staphylococcus aureus adhesion on eggshell. Int J Food Microbiol 2025; 426:110925. [PMID: 39366090 DOI: 10.1016/j.ijfoodmicro.2024.110925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/14/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
Biofilm formation of Staphylococcus aureus in food processing environments raises significant safety concerns, necessitating the development of new antibiofilm approaches for controlling S. aureus contamination. This study aimed to elucidate the antibiofilm mechanism of 2R,3R-dihydromyricetin (DMY), a natural flavonoid, against S. aureus and evaluate its efficacy in reducing bacterial adhesion to eggshell. The results revealed that DMY was a potent inhibitor of S. aureus sortase A (SrtA) with an IC50 of 73.43 μM, preventing bacterial adhesion to fibrinogen and subsequent biofilm formation. Fluorescence quenching assay and surface plasmon resonance analysis confirmed that DMY could directly bind to S. aureus SrtA. Notably, circular dichroism spectra demonstrated a conformational change in SrtA from α-helical to β-sheet structure upon DMY binding. Molecular dynamics simulation suggested that DMY bound to the catalytic pocket of S. aureus SrtA via hydrophobic interactions and hydrogen bonds. Furthermore, fluorescence microscopic observations further revealed that DMY attenuated the biofilm-related phenotype of SrtA by decreasing the anchoring of S. aureus protein A (SpA) onto cell wall. Importantly, pretreatment with 125 μg/mL DMY significantly reduced 1.14-1.75 log CFU/cm2 of S. aureus adhered on eggshells. Overall, these findings highlight how specific targeting of SrtA by DMY inhibits the attachment stages of biofilm development in S. aureus, making it a promising candidate for a novel disinfectant against this pathogen in the food industry.
Collapse
Affiliation(s)
- Wenyi Ran
- College of Biomass Science and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, China
| | - Peirui Yi
- College of Biomass Science and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, China
| | - Ling Jiang
- College of Biomass Science and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, China
| | - Yang Yu
- College of Biomass Science and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, China
| | - Kai Zhong
- College of Biomass Science and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, China
| | - Yanping Wu
- College of Biomass Science and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, China.
| | - Hong Gao
- College of Biomass Science and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, China
| |
Collapse
|
25
|
Li Y, Sung Min H, Chen C, Shan H, Lin Y, Yin F, Chen Y, Lu L, Yu X. A chitosan/gelatin/aldehyde hyaluronic acid hydrogel coating releasing calcium ions and vancomycin in pH response to prevent the formation of bacterial biofilm. Carbohydr Polym 2025; 347:122723. [PMID: 39486953 DOI: 10.1016/j.carbpol.2024.122723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/26/2024] [Accepted: 09/04/2024] [Indexed: 11/04/2024]
Abstract
Osteomyelitis is a refractory disease of orthopedics, part of which is caused by medical implants. The main difficulties in treatment are the barrier effect after the formation of bacterial biofilm, and the difficulty in achieving sustained antibiotic intervention. In view of this situation, we studied a hydrogel coating that can release CaCl2 and vancomycin in pH-responsive manner. We used nano-TiO2 to modify Chitosan/ Gelatin/Aldehyde Hyaluronic Acid (CS/Gel/AHA) hydrogel, and combined with the dip-coating technique, prepared a coating with good mechanical strength. The hydrogel-loaded zeolitic imidazolate framework (ZIF) decomposes under acidic conditions, and the released Ca2+ act on the bacterial Bap protein to inhibit the formation of biofilm, and the released vancomycin kills free bacteria. The antibacterial coating achieved good bactericidal effect in both in vitro experiments and rat subcutaneous implant model. These results not only provide a new way to enhance the strength of hydrogels to prepare coatings, but also utilize a new approach to responsively inhibit the formation of biofilms, showing the promising application prospects of the coating in antibacterial treatment of medical implants.
Collapse
Affiliation(s)
- Yuange Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Hong Sung Min
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Chen Chen
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Haojie Shan
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Yiwei Lin
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Fuli Yin
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Yixian Chen
- Department of Surgery of Chinese Medicine, Jiangxi University of Chinese Medicine, Jiangxi 330004, PR China
| | - Liheng Lu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Xiaowei Yu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China.
| |
Collapse
|
26
|
Salikin NH, Keong LC, Azemin WA, Philip N, Yusuf N, Daud SA, Rashid SA. Combating multidrug-resistant (MDR) Staphylococcus aureus infection using terpene and its derivative. World J Microbiol Biotechnol 2024; 40:402. [PMID: 39627623 DOI: 10.1007/s11274-024-04190-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/01/2024] [Indexed: 12/13/2024]
Abstract
Multidrug-resistant (MDR) Staphylococcus aureus represents a major global health issue resulting in a wide range of debilitating infections and fatalities. The slow progression of new antibiotics, limited choices for treatment, and scarcity of new drug approvals create immense obstacles in new drug line development. S. aureus poses a significant public health risk, due to the emergence of methicillin-resistant (MRSA) and vancomycin-resistant strains (VRSA), necessitating novel antibiotics for effective control management. Current studies are delving into the terpenes' potential as an antimicrobial agent, indicating positive prospects as promising substitutes or complementary to conventional antibiotics. Concurrent reactions of terpenes with conventional antibiotics create synergistic effects that significantly enhance antibiotic efficacy. Accumulated evidence has shown that while efflux pump (e.g., NorA, TetK, and MepA) is revealed as an essential defense of S. aureus against antibiotics, terpene and its derivative act as its potent inhibitor, suggesting the promising potential of terpenes in combating those infectious pathogens. Furthermore, pronounced cell membrane disruptive activity and antibiofilm properties by terpenes have been exerted, signifying their significance as promising prevention against microbial pathogenesis and antimicrobial resistance. This review provides an overview of the potential of terpenes and their derivatives in combating S. aureus infections, highlighting their potential mechanisms of action (MOA), synergistic effects with conventional antibiotics, and challenges in clinical translation. The unique properties of terpenes offer an opportunity for their use in developing an exceptional defense strategy against antibiotic-resistant S. aureus.
Collapse
Affiliation(s)
- Nor Hawani Salikin
- School of Industrial Technology, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Lee Chee Keong
- School of Industrial Technology, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Wan-Atirah Azemin
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Noraini Philip
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Nurhaida Yusuf
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Syiah Kuala University, Aceh, Indonesia
| | - Siti Aisyah Daud
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Syarifah Ab Rashid
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia.
| |
Collapse
|
27
|
Singh S, Kumar S, Dhanasingh I. Overexpression, purification and biochemical studies of Sortase A from Enterococcus faecalis (Ef) and its inhibition studies with Aloenin. Acta Trop 2024; 260:107419. [PMID: 39353540 DOI: 10.1016/j.actatropica.2024.107419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/13/2024] [Accepted: 09/28/2024] [Indexed: 10/04/2024]
Abstract
Sortase A (SrtA) is a bacterial transpeptidase that garnishes the bacterial surface by adding various virulent factors or proteins by cleaving the LPXTG-specific motif between T and G amino acids. These virulence factors assist in the attachment of host cells, which are essential for bacterial virulence. Enterococcus species are among the multidrug-resistant bacteria that cause nosocomial infections; they have drawn a lot of attention recently. SrtA from E. faecalis (Ef) plays a critical role in pathogenesis, making it a suitable target for the development of antibacterial agents. Since SrtA is not involved in bacterial growth and is present on the surface of bacteria, the probability of developing antibiotic resistance is minimal. In this work, we have done cloning, expression and purification of Ef-SrtA using IMAC (Immobilised Metal Affinity Chromatography) followed by Gel filtration chromatography. Purified Ef-SrtA showed maximum activity at pH-8 and temperature between 45 and 55 °C. The fluorescent assay for kinetic studies of Ef-SrtA showed Vmax 3.852 µM.min-1 and kcat 7.7 × 10-2s-1 for the hydrolysis of substrate using Abz-LPETG-K(Dnp)-NH2. We have selected fifteen Aloe vera extracted compounds and performed virtual screening and docking experiments to identify potential inhibitors against Ef-SrtA. Among fifteen molecules, Aloenin-a which was bound to the active site with a binding energy of -6.1 kcal/mol, interacted with the active site residues, Arg139, Pro105, Leu39, Ala46, and Cys126. Aloenin-a showed a significant inhibitory effect against Ef-SrtA, with an IC50 value of 20.68 µM. Aloenin-a inhibits biofilm formation at concentrations of 20-250 µg/mL. The fibrinogen assay showed adherence to fibrinogen was reduced in the presence of Aloenin-a for E. faecalis. The results demonstrated that Aloe vera extracts containing Aloenin-a can be a significant antagonist of Ef-SrtA.
Collapse
Affiliation(s)
- Suraj Singh
- Centre for Bio-Separation and Technology (CBST), Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Sanjit Kumar
- Centre for Bio-Separation and Technology (CBST), Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India; Department of Biotechnology, School of Interdisciplinary Education and Research Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh 495009, India.
| | - Immanuel Dhanasingh
- Centre for Bio-Separation and Technology (CBST), Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India.
| |
Collapse
|
28
|
Barman S, Kurnaz LB, Leighton R, Hossain MW, Decho AW, Tang C. Intrinsic antimicrobial resistance: Molecular biomaterials to combat microbial biofilms and bacterial persisters. Biomaterials 2024; 311:122690. [PMID: 38976935 PMCID: PMC11298303 DOI: 10.1016/j.biomaterials.2024.122690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 05/13/2024] [Accepted: 06/26/2024] [Indexed: 07/10/2024]
Abstract
The escalating rise in antimicrobial resistance (AMR) coupled with a declining arsenal of new antibiotics is imposing serious threats to global public health. A pervasive aspect of many acquired AMR infections is that the pathogenic microorganisms exist as biofilms, which are equipped with superior survival strategies. In addition, persistent and recalcitrant infections are seeded with bacterial persister cells at infection sites. Together, conventional antibiotic therapeutics often fail in the complete treatment of infections associated with bacterial persisters and biofilms. Novel therapeutics have been attempted to tackle AMR, biofilms, and persister-associated complex infections. This review focuses on the progress in designing molecular biomaterials and therapeutics to address acquired and intrinsic AMR, and the fundamental microbiology behind biofilms and persisters. Starting with a brief introduction of AMR basics and approaches to tackling acquired AMR, the emphasis is placed on various biomaterial approaches to combating intrinsic AMR, including (1) semi-synthetic antibiotics; (2) macromolecular or polymeric biomaterials mimicking antimicrobial peptides; (3) adjuvant effects in synergy; (4) nano-therapeutics; (5) nitric oxide-releasing antimicrobials; (6) antimicrobial hydrogels; (7) antimicrobial coatings. Particularly, the structure-activity relationship is elucidated in each category of these biomaterials. Finally, illuminating perspectives are provided for the future design of molecular biomaterials to bypass AMR and cure chronic multi-drug resistant (MDR) infections.
Collapse
Affiliation(s)
- Swagatam Barman
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, United States; Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, 29208, United States
| | - Leman Buzoglu Kurnaz
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, United States
| | - Ryan Leighton
- Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, 29208, United States
| | - Md Waliullah Hossain
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, United States
| | - Alan W Decho
- Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, 29208, United States.
| | - Chuanbing Tang
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, United States.
| |
Collapse
|
29
|
Cuellar-Gaviria TZ, Rincon-Benavides MA, Halipci Topsakal HN, Salazar-Puerta AI, Jaramillo-Garrido S, Kordowski M, Vasquez-Martinez CA, Nguyen KT, Rima XY, Rana PSJB, Combita-Heredia O, Deng B, Dathathreya K, McComb DW, Reategui E, Wozniak D, Higuita-Castro N, Gallego-Perez D. Tissue nano-transfection of antimicrobial genes drives bacterial biofilm killing in wounds and is potentially mediated by extracellular vesicles. J Control Release 2024; 376:1300-1315. [PMID: 39491627 PMCID: PMC11780627 DOI: 10.1016/j.jconrel.2024.10.071] [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: 04/13/2024] [Revised: 10/06/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
The emergence of bacteria that are resistant to antibiotics is on track to become a major global health crisis. Therefore, there is an urgent need for new treatment options. Here, we studied the implementation of tissue-nanotransfection (TNT) to treat Staphylococcus aureus-infected wounds by delivering gene cargos that boost the levels of naturally produced antimicrobial peptides. The Cathelicidin Antimicrobial Peptide gene (CAMP), which produces the antimicrobial peptide LL-37, was used as model gene cargo. In vitro evaluation showed successful transfection and an increase in the transcription and translation of CAMP-coding plasmid in mouse primary epithelial cells. Moreover, we found that the extracellular vesicles (EVs) derived from the transfected cells (in vitro and in vivo) carried significantly higher concentrations of CAMP transcripts and LL-37 peptide compared to control EVs, possibly mediating the trafficking of the antimicrobial contents to other neighboring cells. The TNT platform was then used in vivo on an excisional wound model in mice to nanotransfect the CAMP-coding plasmid on the edge of infected wounds. After 4 days of daily treatment, we observed a significant decrease in the bacterial load in the CAMP-treated group compared to the sham group. Moreover, histological analysis and bacterial load quantification also revealed that TNT of CAMP on S. aureus-infected wounds was effective in treating biofilm progression by reducing the bacterial load. Lastly, we observed a significant increase in macrophage recruitment to the infected tissue, a robust increase in vascularization, as well as and an increased expression of IL10 and Fli1. Our results demonstrate that TNT-based delivery of gene cargos coding for antimicrobial compounds to the wound is a promising approach for combating biofilm infections in wounds.
Collapse
Affiliation(s)
- Tatiana Z Cuellar-Gaviria
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Gene Therapy Institute, The Ohio State University, Columbus, OH 43210, USA; Infectious Disease Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Maria Angelica Rincon-Benavides
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
| | - Hatice Nur Halipci Topsakal
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Istanbul Atlas University, Istanbul 34408, Turkiye
| | | | | | - Mia Kordowski
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
| | - Carlos A Vasquez-Martinez
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; CONACYT - Faculty of Medicine, Benito Juárez Autonomous University of Oaxaca, Oaxaca 68020, Mexico
| | - Kim Truc Nguyen
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Xilal Y Rima
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Pranav S J B Rana
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA; Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | | | - Binbin Deng
- Center for Electron Microscopy and Analysis (CEMAS), The Ohio State University, Columbus, OH 43210, USA
| | - Kavya Dathathreya
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - David W McComb
- Center for Electron Microscopy and Analysis (CEMAS), The Ohio State University, Columbus, OH 43210, USA; Department of Materials Science and Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Eduardo Reategui
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA; Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel Wozniak
- Infectious Disease Institute, The Ohio State University, Columbus, OH 43210, USA; Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA; Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | - Natalia Higuita-Castro
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Gene Therapy Institute, The Ohio State University, Columbus, OH 43210, USA; Infectious Disease Institute, The Ohio State University, Columbus, OH 43210, USA; Biophysics Program, The Ohio State University, Columbus, OH 43210, USA; Department of Neurological Surgery, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel Gallego-Perez
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Gene Therapy Institute, The Ohio State University, Columbus, OH 43210, USA; Infectious Disease Institute, The Ohio State University, Columbus, OH 43210, USA; Biophysics Program, The Ohio State University, Columbus, OH 43210, USA; Department of Surgery, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
30
|
Zhang Z, Chen G, Hussain W, Pan Y, Yang Z, Liu Y, Li E. Machine learning and network analysis with focus on the biofilm in Staphylococcus aureus. Comput Struct Biotechnol J 2024; 23:4148-4160. [PMID: 39640530 PMCID: PMC11617897 DOI: 10.1016/j.csbj.2024.11.011] [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: 05/15/2024] [Revised: 11/04/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Research on biofilm formation in Staphylococcus aureus has greatly benefited from the generation of high-throughput sequencing data to drive molecular analysis. The accumulation of high-throughput sequencing data, particularly transcriptomic data, offers a unique opportunity to unearth the network and constituent genes involved in biofilm formation using machine learning strategies and co-expression analysis. Herein, the available RNA sequencing data related to Staphylococcus aureus biofilm studies and identified influenced functional pathways and corresponding genes in the process of the transition of bacteria from planktonic to biofilm state by employing machine learning and differential expression analysis. Using weighted gene co-expression analysis and previously developed online prediction platform, important functional modules, potential biofilm-associated proteins, and subnetworks of the biofilm-formation pathway were uncovered. Additionally, several novel protein interactions within these functional modules were identified by constructing a protein-protein interaction (PPI) network. To make this data more straightforward for experimental biologists, an online database named SAdb was developed (http://sadb.biownmcli.info/), which integrates gene annotations, transcriptomics, and proteomics data. Thus, the current study will be of interest to researchers in the field of bacteriology, particularly those studying biofilms, which play a crucial role in bacterial growth, pathogenicity, and drug resistance.
Collapse
Affiliation(s)
- Zhiyuan Zhang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Department of Medical Information Engineering, School of Medical Information, Wannan Medical College, Wuhu 241000, China
| | - Guozhong Chen
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong, China
| | - Wajid Hussain
- Advanced Biomaterials and Tissue Engineering Center, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yuanyuan Pan
- Department of Medical Information Engineering, School of Medical Information, Wannan Medical College, Wuhu 241000, China
| | - Zhu Yang
- Department of Medical Microbiology and Immunology, Wannan Medical College, Wuhu, Anhui, China
| | - Yin Liu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Erguang Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| |
Collapse
|
31
|
Dewan D, Basu A, Dolai D, Pal S. Biological and Biophysical Methods for Evaluation of Inhibitors of Sortase A in Staphylococcus aureus: An Overview. Cell Biochem Funct 2024; 42:e70002. [PMID: 39470102 DOI: 10.1002/cbf.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 09/01/2024] [Accepted: 10/02/2024] [Indexed: 10/30/2024]
Abstract
Staphylococcus aureus, one of the most notorious pathogens, develops antibiotic resistance by the formation of a thick layer of exopolysaccharides known as biofilms. Sortase A, a transpeptidase responsible for biofilm formation and attachment to the host surface, has emerged as an important drug target for development of anti-virulence agents. A number of sortase A inhibitors, both peptide and non-peptides are reported which involved the use of several experiments which may provide insights regarding binding affinity, specificity, safety, and efficacy of ligands. In this review, we focus on the principles, pros and cons, and the type of information obtained from biophysical (FRET assay, Microscale Thermophoresis, Surface Plasmon Resonance, CD spectroscopy etc.) and biological (cell viability assay, biofilm formation assay, CLSM, western blot analysis, in vivo characterization on mice etc.) methods for estimation of probable sortase A inhibitors, which might be helpful to the researchers who might be interested to delve into the development of sortase A inhibitors as a drug, to address the burning question of antimicrobial resistance (AMR).
Collapse
|
32
|
Sunnerhagen T, Bjarnsholt T, Qvortrup K, Bundgaard H, Moser C. Transcatheter aortic valve implantation (TAVI) prostheses in vitro - biofilm formation and antibiotic effects. Biofilm 2024; 8:100236. [PMID: 39555138 PMCID: PMC11565431 DOI: 10.1016/j.bioflm.2024.100236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 11/19/2024] Open
Abstract
Background Transcatheter aortic valve implantation (TAVI) is a percutaneous catheter-based treatment of aortic stenosis as an alternative to open heart valve surgery. In cases of TAVI endocarditis, the treatment possibilities may be limited as surgical removal of the infected valve may be associated with a high risk in elderly, comorbid or frail patients. The propensity of bacteria to form a biofilm on foreign material is assumed to be of importance part of the disease process in TAVI endocarditis, but no studies on biofilm formation on TAVI valves have been conducted. We hypothesize that Staphylococcus aureus and Enterococcus faecalis biofilm formation on TAVI valves may have an impact on antibiotic tolerance and non-surgical cure rates. Methods TAVI valves (pieces including part of the metal frame, approximately 1 cm wide) were exposed to either species in vitro in LB-Krebs Ringer medium at 37 °C, with the bacterial count being assessed by culturing of sonicated TAVI pieces and broth at 0, 4, 18 and 24 h after bacterial exposure. Scanning electron microscopy (SEM) was performed. Effects of ampicillin, gentamicin, moxifloxacin, rifampicin (for S. aureus), and ceftriaxone (for E. faecalis) at 5 times minimal inhibitory concentration were tested alone and in combination with ampicillin. Antibiotics were added to biofilm aged 0 or 24 h and the effects assessed. Results Exposure for 15 min established attachment to all of valve pieces. SEM findings were consistent with biofilm formation and suggested lower amounts of bacteria on the metal compared to the tissue part of the TAVI valves. The number of bacteria attached to the TAVI valves increased until 24 h of incubation from less than 10^1 to a level of approximately 10^9 CFU/g. The bacteria became more tolerant to antibiotics on the TAVI valves over time, with the bactericidal effect against 24-h old biofilm being significantly less effective than against 0-h old biofilm depending on antibiotic. Conclusions The results indicate that bacteria can adhere to metal and tissue parts of the TAVI valves within minutes after an exposure which is comparable to transient bacteremia in vivo, and that the bacteria rapidly gain biofilm properties, associated with significantly reduced antibiotic effect.
Collapse
Affiliation(s)
- Torgny Sunnerhagen
- Department of Clinical Microbiology, Rigshospitalet Copenhagen University Hospital, Copenhagen, Denmark
- Division for Infection Medicine, Department for Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
- Clinical Microbiology and Infection Control, Office for Medical Services, Region Skåne, Lund, Sweden
| | - Thomas Bjarnsholt
- Department of Clinical Microbiology, Rigshospitalet Copenhagen University Hospital, Copenhagen, Denmark
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Klaus Qvortrup
- Department of Biomedical Sciences, Core Facility for Integrated Microscopy, University of Copenhagen, Copenhagen, Denmark
| | - Henning Bundgaard
- Department of Cardiology, Rigshospitalet Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Claus Moser
- Department of Clinical Microbiology, Rigshospitalet Copenhagen University Hospital, Copenhagen, Denmark
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
33
|
Wang D, Liu N, Qiao M, Xu C. Gallic acid as biofilm inhibitor can improve transformation efficiency of Ruminiclostridium papyrosolvens. Biotechnol Lett 2024; 46:1143-1153. [PMID: 39162860 DOI: 10.1007/s10529-024-03522-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/26/2024] [Accepted: 08/03/2024] [Indexed: 08/21/2024]
Abstract
Ruminiclostridium papyrosolvens is an anaerobic, mesophilic, and cellulolytic clostridia, promising consolidated bioprocessing (CBP) candidate for producing renewable green chemicals from cellulose, but its genetic transformation has been severely impeded by extracellular biofilm. Here, we analyzed the effects of five different inhibitors with gradient concentrations on R. papyrosolvens growth and biofilm formation. Gallic acid was proved to be a potent inhibitor of biofilm synthesis of R. papyrosolvens. Furthermore, the transformation efficiency of R. papyrosolvens was significantly increased when the cells were treated by the gallic acid, and the mutant strain was successfully obtained by the improved transformation method. Thus, inhibition of biofilm formation of R. papyrosolvens by using gallic acid will contribute to its genetic transformation and efficient metabolic engineering.
Collapse
Affiliation(s)
- Duodong Wang
- School of Life Science, Shanxi University, Taiyuan, 030006, Shanxi, China
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, 311300, Zhejiang Province, China
| | - Na Liu
- School of Life Science, Shanxi University, Taiyuan, 030006, Shanxi, China
| | - Mingqiang Qiao
- School of Life Science, Shanxi University, Taiyuan, 030006, Shanxi, China.
| | - Chenggang Xu
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, 311300, Zhejiang Province, China.
| |
Collapse
|
34
|
Sim M, Kim YG, Lee JH, Lee J. Antibiofilm Activities of Multiple Halogenated Pyrimidines Against Staphylococcus aureus. Int J Mol Sci 2024; 25:12830. [PMID: 39684543 DOI: 10.3390/ijms252312830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Staphylococcus aureus, prevalent in hospital and community settings, forms biofilms that are highly resistant to antibiotics and immune responses, complicating treatment and contributing to chronic infections. These challenges underscore the need for novel treatments that target biofilm formation and effectively reduce bacterial virulence. This study investigates the antibiofilm and antimicrobial efficacy of novel halogenated pyrimidine derivatives against S. aureus, focusing on three compounds identified as potent biofilm inhibitors: 2,4-dichloro-5-fluoropyrimidine (24DC5FP), 5-bromo-2,4-dichloro-7H-pyrrolo[2,3-d]pyrimidine (24DC5BPP), and 2,4-dichloro-5-iodo-7H-pyrrolo[2,3-d]pyrimidine (24DC5IPP). The three active compounds are bacteriostatic. In particular, 24DC5FP at 5 µg/mL achieved a 95% reduction in hemolysis with a minimum inhibitory concentration (MIC) of 50 µg/mL. Interestingly, 24DC5FP increased cell size and produced wrinkled colonies. qRT-PCR analysis showed that 24DC5FP suppressed the gene expressions of agrA and RNAIII (quorum sensing regulator and effector), hla (α-hemolysin), nuc1 (nucleases nuc1), and saeR (S. aureus virulence regulator). These findings suggest that extensive halogenation enhances the antibiofilm and antivirulence activities of pyrimidine derivatives, offering a promising strategy for combatting S. aureus infections, including those resistant to conventional treatments.
Collapse
Affiliation(s)
- MinHwi Sim
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Yong-Guy Kim
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Jin-Hyung Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
35
|
Korshoj LE, Kielian T. Bacterial single-cell RNA sequencing captures biofilm transcriptional heterogeneity and differential responses to immune pressure. Nat Commun 2024; 15:10184. [PMID: 39580490 PMCID: PMC11585574 DOI: 10.1038/s41467-024-54581-8] [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: 09/18/2024] [Accepted: 11/14/2024] [Indexed: 11/25/2024] Open
Abstract
Biofilm formation is an important mechanism of survival and persistence for many bacterial pathogens. These multicellular communities contain subpopulations of cells that display metabolic and transcriptional diversity along with recalcitrance to antibiotics and host immune defenses. Here, we present an optimized bacterial single-cell RNA sequencing method, BaSSSh-seq, to study Staphylococcus aureus diversity during biofilm growth and transcriptional adaptations following immune cell exposure. BaSSSh-seq captures extensive transcriptional heterogeneity during biofilm compared to planktonic growth. We quantify and visualize transcriptional regulatory networks across heterogeneous biofilm subpopulations and identify gene sets that are associated with a trajectory from planktonic to biofilm growth. BaSSSh-seq also detects alterations in biofilm metabolism, stress response, and virulence induced by distinct immune cell populations. This work facilitates the exploration of biofilm dynamics at single-cell resolution, unlocking the potential for identifying biofilm adaptations to environmental signals and immune pressure.
Collapse
Affiliation(s)
- Lee E Korshoj
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Tammy Kielian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
36
|
Sinha S, Aggarwal S, Singh DV. Efflux pumps: gatekeepers of antibiotic resistance in Staphylococcus aureus biofilms. MICROBIAL CELL (GRAZ, AUSTRIA) 2024; 11:368-377. [PMID: 39568862 PMCID: PMC11576857 DOI: 10.15698/mic2024.11.839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/01/2024] [Accepted: 10/08/2024] [Indexed: 11/22/2024]
Abstract
Staphylococcus aureus, a versatile human pathogen, poses a significant challenge in healthcare settings due to its ability to develop antibiotic resistance and form robust biofilms. Understanding the intricate mechanisms underlying the antibiotic resistance is crucial for effective infection treatment and control. This comprehensive review delves into the multifaceted roles of efflux pumps in S. aureus, with a focus on their contribution to antibiotic resistance and biofilm formation. Efflux pumps, integral components of the bacterial cell membrane, are responsible for expelling a wide range of toxic substances, including antibiotics, from bacterial cells. By actively extruding antibiotics, these pumps reduce intracellular drug concentrations, rendering antibiotics less effective. Moreover, efflux pumps have emerged as significant contributors to both antibiotic resistance and biofilm formation in S. aureus. Biofilms, structured communities of bacterial cells embedded in a protective matrix, enable S. aureus to adhere to surfaces, evade host immune responses, and resist antibiotic therapy. Efflux pumps play a pivotal role in the development and maintenance of S. aureus biofilms. However, the interplay between efflux pumps, antibiotic resistance and biofilm formation remains unexplored in S. aureus. This review aims to elucidate the complex relationship between efflux pumps, antibiotic resistance and biofilm formation in S. aureus with the aim to aid in the development of potential therapeutic targets for combating S. aureus infections, especially those associated with biofilms. The insights provided herein may contribute to the advancement of novel strategies to overcome antibiotic resistance and disrupt biofilm formation in this clinically significant pathogen.
Collapse
Affiliation(s)
- Shweta Sinha
- Department of Biotechnology, School of Earth, Biological and Environmental Sciences, Central University of South Bihar Gaya, 824236 India
| | - Shifu Aggarwal
- Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023 India
- Current Address: Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts USA
| | - Durg Vijai Singh
- Department of Biotechnology, School of Earth, Biological and Environmental Sciences, Central University of South Bihar Gaya, 824236 India
| |
Collapse
|
37
|
Tooke AK, Hodges RE, Pyrah JF, Bayles KW, Renshaw SA, Foster SJ. Tetracycline and Oxacillin Act Synergistically on Biofilms and Display Increased Efficacy In Vivo Against Staphylococcus aureus. Curr Microbiol 2024; 81:447. [PMID: 39505760 PMCID: PMC11541413 DOI: 10.1007/s00284-024-03959-4] [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: 07/04/2024] [Accepted: 10/18/2024] [Indexed: 11/08/2024]
Abstract
Oxacillin (bactericidal) and tetracycline (bacteriostatic) are clinically relevant antibiotics that are routinely prescribed to treat Staphylococcus aureus infections but not conventionally used in combination. There is an urgent need for treatment regimens that can act upon biofilms during infection, associated with chronic infections on indwelling devices, as well as acute planktonic (systemic) infection. Here we show that in an in vitro model oxacillin and tetracycline act synergistically against S. aureus UAMS-1 biofilms, reducing the concentration of both antibiotics necessary to eradicate an established biofilm. Using an in vivo zebrafish larval infection model with S. aureus NewHG, they display improved bacterial clearance compared to each drug alone and can counteract a loss of host phagocytes, an important innate defence against S. aureus. In these cases, the bacteriostatic nature of tetracycline enhances rather than dampens the bactericidal action of oxacillin, although an exact mechanism remains to be elucidated. We suggest a dual therapy could be of clinical use against biofilm-forming S. aureus and has a potential use in patients with a compromised immune system.
Collapse
Affiliation(s)
- Amy K Tooke
- School of Biosciences, University of Sheffield, Sheffield, S10 2TH, UK
- Microbes in Health and Disease Theme, Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Rebecca E Hodges
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Josie F Pyrah
- School of Biosciences, University of Sheffield, Sheffield, S10 2TH, UK
| | - Kenneth W Bayles
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Stephen A Renshaw
- The Bateson Centre, University of Sheffield, Western Bank, Sheffield, S10 2TH, UK
- School of Medicine and Population Health, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Simon J Foster
- School of Biosciences, University of Sheffield, Sheffield, S10 2TH, UK.
- Florey Institute, University of Sheffield, Sheffield, S10 2TH, UK.
| |
Collapse
|
38
|
Ma Y, Kang X, Wang G, Luo S, Luo X, Wang G. Inhibition of Staphylococcus aureus biofilm by quercetin combined with antibiotics. BIOFOULING 2024; 40:996-1011. [PMID: 39639551 DOI: 10.1080/08927014.2024.2435027] [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: 12/12/2023] [Revised: 11/13/2024] [Accepted: 11/20/2024] [Indexed: 12/07/2024]
Abstract
This study aimed to investigate the effects of combined quercetin and antibiotics on the bacteriostatic activity and biofilm formation of Staphylococcus aureus. Optimal concentrations of quercetin and antibiotics (tetracycline and doxycycline) for inhibiting biofilm formation were determined using the Fractional Inhibitory Concentration Index and Minimum Biofilm Inhibitory Concentration assays. The impact of the drug combinations on biofilm clearance at various formation stages was determined using crystal violet staining, scanning electron microscopy and confocal laser microscopy. The results indicated that quercetin enhanced the bactericidal effect of tetracycline antibiotics against S. aureus. The combination significantly reduced both the metabolic activity within S. aureus biofilms and the production of biofilm matrix components. Scanning electron microscopy and confocal laser microscopy confirmed that the combination treatment significantly reduced bacterial cell counts within the biofilm. Quercetin treatment significantly increased the sensitivity of biofilms to antibiotics, supporting its potential application as a novel antibiotic synergist.
Collapse
Affiliation(s)
- Yanjun Ma
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Xinyun Kang
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Guiqin Wang
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Shuangyan Luo
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Xiaofeng Luo
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Guilai Wang
- Yinchuan Hospital of Traditional Chinese Medicine, Yinchuan, China
| |
Collapse
|
39
|
Zou S, Ma Y, Zhao L, Chen X, Gao H, Chen J, Xue Y, Zheng Y. Revealing the regulatory impact of nutrient on the production of (R)-2-(4-Hydroxyphenoxy)propanoic acid by Beauveria bassiana biofilms through comparative transcriptomics analyse. Bioprocess Biosyst Eng 2024; 47:1803-1814. [PMID: 39080012 DOI: 10.1007/s00449-024-03070-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/22/2024] [Indexed: 09/29/2024]
Abstract
Carbon and nitrogen play a fundamental role in the architecture of fungal biofilm morphology and metabolite production. However, the regulatory mechanism of nutrients remains to be fully understood. In this study, the formation of Beauveria bassiana biofilm and the production of (R)-2-(4-Hydroxyphenoxy)propanoic acid in two media with different carbon and nitrogen sources (GY: Glucose as a carbon source and yeast extract as a nitrogen source, MT: Mannitol as a carbon source and tryptone as a nitrogen source) were compared. R-HPPA production increased 2.85-fold in media MT than in media GY. Different fungal biofilm morphology and architecture were discovered in media GY and MT. Comparative transcriptomics revealed up-regulation of mitogen-activated protein kinase (MAPK) pathway and polysaccharides degradation genes affecting mycelial morphology and polysaccharides yield of the extracellular polymeric substances (EPS) in MT medium biofilms. Upregulation of genes related to NADH synthesis (carbon metabolism, amino acid metabolism, glutamate cycle) causes NADH accumulation and triggers an increase in R-HPPA production. These data provide a valuable basis for future studies on regulating fungal biofilm morphology and improving the production of high-value compounds.
Collapse
Affiliation(s)
- Shuping Zou
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
- Engineering Research Center of Bioconversion and Biopurification of Ministry of Education, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
| | - Yizhi Ma
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
- Engineering Research Center of Bioconversion and Biopurification of Ministry of Education, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
| | - Lixiang Zhao
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
- Engineering Research Center of Bioconversion and Biopurification of Ministry of Education, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
| | - Xiaomin Chen
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
- Engineering Research Center of Bioconversion and Biopurification of Ministry of Education, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
| | - Hailing Gao
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
- Engineering Research Center of Bioconversion and Biopurification of Ministry of Education, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
| | - Juan Chen
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
- Engineering Research Center of Bioconversion and Biopurification of Ministry of Education, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
| | - Yaping Xue
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China.
- Engineering Research Center of Bioconversion and Biopurification of Ministry of Education, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China.
| | - Yuguo Zheng
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
- Engineering Research Center of Bioconversion and Biopurification of Ministry of Education, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
| |
Collapse
|
40
|
Oo T, Saiboonjan B, Mongmonsin U, Srijampa S, Srisrattakarn A, Tavichakorntrakool R, Chanawong A, Lulitanond A, Roytrakul S, Sutthanut K, Tippayawat P. Effectiveness of co-cultured Myristica fragrans Houtt. seed extracts with commensal Staphylococcus epidermidis and its metabolites in antimicrobial activity and biofilm formation of skin pathogenic bacteria. BMC Complement Med Ther 2024; 24:380. [PMID: 39482677 PMCID: PMC11526599 DOI: 10.1186/s12906-024-04675-z] [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: 06/13/2024] [Accepted: 10/07/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Skin commensal bacteria (Staphylococcus epidermidis) can help defend against skin infections, and they are increasingly being recognized for their role in benefiting skin health. This study aims to demonstrate the activities that Myristica fragrans Houtt. seed extracts, crude extract (CE) and essential oil (EO), have in terms of promoting the growth of the skin commensal bacterium S. epidermidis and providing metabolites under culture conditions to disrupt the biofilm formation of the common pathogen Staphylococcus aureus. METHODS The culture supernatant obtained from a co-culture of S. epidermidis with M. fragrans Houtt. seed extracts in either CE or EO forms were analyzed using gas chromatography-mass spectrometry (GC-MS) and liquid chromatography tandem mass spectrometry (LC-MS/MS), in silico investigations, and applied to assess the survival and biofilm formation of S. aureus. RESULTS The combination of commensal bacteria with M. fragrans Houtt. seed extract either CE or EO produced metabolic compounds such as short-chain fatty acids and antimicrobial peptides, contributing to the antimicrobial activity. This antimicrobial activity was related to downregulating key genes involved in bacterial adherence and biofilm development in S. aureus, including cna, agr, and fnbA. CONCLUSION These findings suggest that using the culture supernatant of the commensal bacteria in combination with CE or EO may provide a potential approach to combat biofilm formation and control the bacterial proliferation of S. aureus. This may be a putative non-invasive therapeutic strategy for maintaining a healthy skin microbiota and preventing skin infections.
Collapse
Affiliation(s)
- Thidar Oo
- Medical Technology Program, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Bhanubong Saiboonjan
- Center for Innovation and Standard for Medical Technology and Physical Therapy, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Urairat Mongmonsin
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Sukanya Srijampa
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Arpasiri Srisrattakarn
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
- Department of Medical Technology, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Ratree Tavichakorntrakool
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
- Department of Medical Technology, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Aroonwadee Chanawong
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
- Department of Medical Technology, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Aroonlug Lulitanond
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
- Department of Medical Technology, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Khaetthareeya Sutthanut
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Patcharaporn Tippayawat
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand.
- Department of Medical Technology, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand.
| |
Collapse
|
41
|
Faleye OO, Lee JH, Kim YG, Faleye OS, Lee J. Antibiofilm and antivirulence potentials of iodinated fmoc-phenylalanine against Staphylococcus aureus. Microb Pathog 2024; 197:107080. [PMID: 39454802 DOI: 10.1016/j.micpath.2024.107080] [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: 05/08/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
Staphylococcus aureus poses significant risks to public health due to its ability to form biofilm and produce virulence factors, contributing to the increase in antibiotic resistance and treatment complications. This emphasizes the urgent need for novel antimicrobial controls. Based on the premise that halogenation improves antimicrobial efficacy, this study investigated the ability of halogenated phenylalanine to effectively inhibit S. aureus biofilm formation and virulence activities. Among 29 halogenated compounds, Fmoc-4-iodo-phenylalanine (Fmoc-Iodo-Phe) displayed the highest antibiofilm effect against S. aureus, achieving 94.3 % reduction at 50 μg/mL. Microscopic studies confirmed its ability to prevent and disrupt mature biofilms. At 10 μg/mL, Fmoc-Iodo-Phe markedly inhibited virulence factors, such as cell surface hydrophobicity, hemolysin and slime production. It showed low propensity for resistance development and effectively inhibited biofilms formed by methicillin-resistant S. aureus (MRSA) and S. epidermidis, but was inactive against Gram-negative bacteria. Gene expression analysis complemented by molecular docking suggest that Fmoc-Iodo-Phe could target the AgrA quorum sensing cascade due to strong interactions with key residues at its DNA binding sites. Notably, it was non-cytotoxic in Caenorhabditis elegans model and satisfied drug-likeliness criteria based on ADMET prediction. Therefore, our findings position Fmoc-Iodo-Phe as a promising antimicrobial candidate against S. aureus infections, underscoring its potential as an alternative to traditional antibiotics.
Collapse
Affiliation(s)
| | - Jin-Hyung Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan, 38541, Republic of Korea
| | - Yong-Guy Kim
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan, 38541, Republic of Korea
| | - Olajide Sunday Faleye
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan, 38541, Republic of Korea
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan, 38541, Republic of Korea.
| |
Collapse
|
42
|
Carmona-Orozco ML, Echeverri F. Induction of biofilm in extended-spectrum beta-lactamase Staphylococcus aureus with drugs commonly used in pharmacotherapy. Microb Pathog 2024; 195:106863. [PMID: 39159772 DOI: 10.1016/j.micpath.2024.106863] [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: 01/18/2024] [Revised: 07/15/2024] [Accepted: 08/14/2024] [Indexed: 08/21/2024]
Abstract
Staphylococcus aureus is a bacterial pathogen that causes bloodstream infections, pneumonia, and skin abscesses and is the primary pathogen responsible for medical devices associated with biofilm infections, accounting for approximately 70 % of cases. Therefore, the World Health Organization (WHO) has designated this microorganism as a top priority due to its role in causing over 20,000 bacteremia-related deaths in the US each year. The issue of pathogen resistance to antibiotics, mainly by a biofilm, further complicates these infections since biofilms render the bacterial colony impervious to antibiotics. However, many natural and synthetic substances also induce bacterial biofilm formation. Therefore, we investigated whether the most common active pharmaceutical ingredients (APIs) could induce biofilm formation in two clinical isolates of extended-spectrum beta-lactamase Staphylococcus aureus, one of them also methicillin-resistant (A2M) and two medical devices. We detected biofilm inducers, inhibitors, and destabilizers. Microbial strain, medical devices, API structure, and concentration influenced the modulatory effects of biofilm. In all devices tested, including microplates, FR18 duodenal probe, and respiratory probe, the clinic isolate methicillin-resistant S. aureus A2M exhibited lower susceptibility to biofilm formation than S. aureus A1. The anti-inflammatory acetaminophen, the hypocholesterolemic lovastatin, and the diuretic hydrochlorothiazide all induced biofilm. However, verapamil, an antihypertensive, and cetirizine, an antihistamine, inhibited biofilm on S. aureus A2M, while propranolol, another antihypertensive, inhibited biofilm on S. aureus A1. Additionally, diclofenac, an analgesic, and cetirizine destabilized the biofilm, resulting in more free bacteria and possibly making them more susceptible to external agents such as antibiotics. Nonetheless, further epidemiologic analyses and in vivo assays are needed to confirm these findings and to establish a correlation between drug use, the onset of bacterial infections in patients, and the use of medical devices. This work provides information about the probable clinical implications of drugs in patients using medical devices or undergoing surgical procedures. Inhibitory APIs could also be used as drug repurposing or templates to design new, more potent biofilm inhibitors.
Collapse
Affiliation(s)
- Maria L Carmona-Orozco
- Química Orgánica de Productos Naturales, Instituto de Química, Universidad de Antioquia, Medellín, Colombia
| | - Fernando Echeverri
- Química Orgánica de Productos Naturales, Instituto de Química, Universidad de Antioquia, Medellín, Colombia.
| |
Collapse
|
43
|
Map Schuh C, Ezquer F, Mamani S, Campodónico PR, Cárcamo C, Martinez-Gómez F, Aburto I, Ezquer M, Morales B, Olivares B. A Natural deep eutectic solvent as an effective material for dual debridement and antibiofilm effects in chronic wound treatment. Int J Pharm 2024; 663:124553. [PMID: 39103063 DOI: 10.1016/j.ijpharm.2024.124553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 07/28/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
In chronic wound treatment, the debridement of devitalized tissue and the eradication of the biofilm must balance aggressiveness with care to protect regenerating tissues. In this study, urea, a potent chaotropic molecule, was modulated through the formation of a Natural Deep Eutectic Solvent (NADES) with betaine to develop a new debriding material (BU) suitable for application into injured dermal tissues. To evaluate BU's debriding capacity, along with its antibiofilm effect and biocompatibility, pre-clinical to clinical methods were employed. In vitro determinations using artificial and clinical slough samples indicate that BU has a high debriding capacity. Additionally, BU's de-structuring effects lead to a strong antibiofilm capability, demonstrated by a reduced bacterial load compared to the antiseptic PHMB-Betaine or medical honey, evaluated in artificial slough and ex vivo human skin. Furthermore, BU's efficacy was evaluated in a murine model of diabetic wound, demonstrating significant effects on debriding and antibiofilm capacity, similar to those observed in PHMB-Betaine and medical honey-treated animals. Finally, BU was clinically evaluated in leg ulcers, showing superiority in reduction of bacterial load and wound area compared to honey, with no adverse effects. Thus, BU represents a simple and non-biocidal option that could contributes to chronic wound care.
Collapse
Affiliation(s)
- Christina Map Schuh
- Centro de Medicina Regenerativa, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Fernando Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Sigde Mamani
- Centro de Química Medica, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Paola R Campodónico
- Centro de Química Medica, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Constanza Cárcamo
- Centro de Química Medica, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Fabián Martinez-Gómez
- Laboratorio de Resonancia Magnética Nuclear, Universidad de Santiago de Chile, Santiago, Chile
| | - Isabel Aburto
- Fundación Instituto Nacional de Heridas, Santiago, Chile
| | - Marcelo Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | | | - Belén Olivares
- Centro de Química Medica, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile.
| |
Collapse
|
44
|
Giordano V, Giannoudis PV. Biofilm Formation, Antibiotic Resistance, and Infection (BARI): The Triangle of Death. J Clin Med 2024; 13:5779. [PMID: 39407838 PMCID: PMC11476620 DOI: 10.3390/jcm13195779] [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: 08/29/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Fracture-related infection (FRI) is a devastating event, directly affecting fracture healing, impairing patient function, prolonging treatment, and increasing healthcare costs. Time plays a decisive role in prognosis, as biofilm maturation leads to the development of antibiotic resistance, potentially contributing to infection chronicity and increasing morbidity and mortality. Research exploring the association between biofilm maturation and antibiotic resistance in orthopaedics primarily addresses aspects related to quality of life and physical function; however, little exists on life-threatening conditions and mortality. Understanding the intrinsic relationship between biofilm maturation, bacterial resistance, and mortality is critical in all fields of medicine. In the herein narrative review, we summarize recent evidence regarding biofilm formation, antibiotic resistance, and infection chronicity (BARI), the three basic components of the "triangle of death" of FRI, and its implications. Preoperative, perioperative, and postoperative prevention strategies to avoid the "triangle of death" of FRI are presented and discussed. Additionally, the importance of the orthopaedic trauma surgeon in understanding new tools to combat infections related to orthopaedic devices is highlighted.
Collapse
Affiliation(s)
- Vincenzo Giordano
- Serviço de Ortopedia e Traumatologia Prof. Nova Monteiro, Hospital Municipal Miguel Couto, Rua Mário Ribeiro 117/2º Andar, Gávea, Rio de Janeiro 22430-160, RJ, Brazil
| | - Peter V. Giannoudis
- Academic Department of Trauma and Orthopaedics, School of Medicine, University of Leeds, Leeds LS2 9LU, UK
- NIHR Leeds Biomedical Research Center, Chapel Allerton Hospital, Leeds LS7 4SA, UK
| |
Collapse
|
45
|
Canchola J, Donkor GYB, Tawiah PO, Fasawe A, Ayim E, Engelke MF, Dahl JU. Alkyl Pyridinol Compounds Exhibit Antimicrobial Effects against Gram-Positive Bacteria. Antibiotics (Basel) 2024; 13:897. [PMID: 39335070 PMCID: PMC11428593 DOI: 10.3390/antibiotics13090897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Background/Objectives. The rise of antibiotic-resistant pathogens represents a significant global challenge in infectious disease control, which is amplified by the decline in the discovery of novel antibiotics. Staphylococcus aureus continues to be a highly significant pathogen, causing infections in multiple organs and tissues in both healthcare institutions and community settings. The bacterium has become increasingly resistant to all available antibiotics. Consequently, there is an urgent need for novel small molecules that inhibit the growth or impair the survival of bacterial pathogens. Given their large structural and chemical diversity, as well as often unique mechanisms of action, natural products represent an excellent avenue for the discovery and development of novel antimicrobial treatments. Anaephene A and B are two such naturally occurring compounds with significant antimicrobial activity against Gram-positive bacteria. Here, we report the rapid syntheses and biological characterization of five novel anaephene derivatives, which display low cytotoxicity against mammalian cells but potent antibacterial activity against various S. aureus strains, including methicillin-resistant S. aureus (MRSA) and the multi-drug-resistant community-acquired strain USA300LAC. Methods. A Sonogashira cross-coupling reaction served as the key step for the synthesis of the alkyl pyridinol products. Results/Conclusions. Using the compound JC-01-074, which displays bactericidal activity already at low concentrations (MIC: 16 μg/mL), we provide evidence that alkyl pyridinols target actively growing and biofilm-forming cells and show that these compounds cause disruption and deformation of the staphylococcal membrane, indicating a membrane-associated mechanism of action.
Collapse
Affiliation(s)
- Juan Canchola
- Department of Chemistry, Illinois State University, Normal, IL 61761, USA
| | | | - Patrick Ofori Tawiah
- School of Biological Sciences, Microbiology, Illinois State University, Normal, IL 61761, USA
| | - Ayoola Fasawe
- School of Biological Sciences, Cell Physiology, Illinois State University, Normal, IL 61761, USA
| | - Emmanuel Ayim
- Department of Chemistry, Illinois State University, Normal, IL 61761, USA
| | - Martin F. Engelke
- School of Biological Sciences, Cell Physiology, Illinois State University, Normal, IL 61761, USA
| | - Jan-Ulrik Dahl
- School of Biological Sciences, Microbiology, Illinois State University, Normal, IL 61761, USA
| |
Collapse
|
46
|
Reigh EL. STAPH: The Biofilm. Ann Allergy Asthma Immunol 2024; 133:284-285. [PMID: 39179305 DOI: 10.1016/j.anai.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 08/26/2024]
|
47
|
Qian Y, Wang X, Wang P, Wu J, Shen Y, Cai K, Bai J, Lu M, Tang C. Biodegradable implant of magnesium/polylactic acid composite with enhanced antibacterial and anti-inflammatory properties. J Biomater Appl 2024; 39:165-178. [PMID: 38816339 DOI: 10.1177/08853282241257183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
Addressing fracture-related infections (FRI) and impaired bone healing remains a significant challenge in orthopedics and stomatology. Researchers aim to address this issue by utilizing biodegradable biomaterials, such as magnesium/poly lactic acid (Mg/PLA) composites, to offer antibacterial properties during the degradation of biodegradable implants. Existing Mg/PLA composites often lack sufficient Mg content, hindering their ability to achieve the desired antibacterial effect. Additionally, research on the anti-inflammatory effects of these composites during late-stage degradation is limited. To strengthen mechanical properties, bolster antibacterial efficacy, and enhance anti-inflammatory capabilities during degradation, we incorporated elevated Mg content into PLA to yield Mg/PLA composites. These composites underwent in vitro degradation studies, cellular assays, bacterial tests, and simulation of the PLA degradation microenvironment. 20 wt% and 40 wt% Mg/PLA composites displayed significant antibacterial properties, with three composites exhibiting notable anti-inflammatory effects. In contrast, elevated Mg content detrimentally impacted mechanical properties. The findings suggest that Mg/PLA composites hold promise in augmenting antibacterial and anti-inflammatory attributes within polymers, potentially serving as temporary regenerative materials for treating bone tissue defects complicated by infections.
Collapse
Affiliation(s)
- Yuxin Qian
- Department of Implantology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Xianli Wang
- Jiangsu Key Laboratory for Advanced Metallic Materials, Southeast University, Nanjing, China
| | - Ping Wang
- Department of Implantology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Jin Wu
- Department of Implantology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Yue Shen
- Department of Implantology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Kunzhan Cai
- Department of Implantology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Jing Bai
- Jiangsu Key Laboratory for Advanced Metallic Materials, Southeast University, Nanjing, China
| | - Mengmeng Lu
- Department of Implantology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Chunbo Tang
- Department of Implantology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| |
Collapse
|
48
|
Duarte AC, Fernández L, Jurado A, Campelo AB, Shen Y, Rodríguez A, García P. Synergistic removal of Staphylococcus aureus biofilms by using a combination of phage Kayvirus rodi with the exopolysaccharide depolymerase Dpo7. Front Microbiol 2024; 15:1438022. [PMID: 39171257 PMCID: PMC11335607 DOI: 10.3389/fmicb.2024.1438022] [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: 05/24/2024] [Accepted: 07/29/2024] [Indexed: 08/23/2024] Open
Abstract
Introduction Bacteriophages have been shown to penetrate biofilms and replicate if they find suitable host cells. Therefore, these viruses appear to be a good option to tackle the biofilm problem and complement or even substitute more conventional antimicrobials. However, in order to successfully remove biofilms, in particular mature biofilms, phages may need to be administered along with other compounds. Phage-derived proteins, such as endolysins or depolymerases, offer a safer alternative to other compounds in the era of antibiotic resistance. Methods This study examined the interactions between phage Kayvirus rodi with a polysaccharide depolymerase (Dpo7) from another phage (Rockefellervirus IPLA7) against biofilms formed by different Staphylococcus aureus strains, as determined by crystal violet staining, viable cell counts and microscopy analysis. Results and discussion Our results demonstrated that there was synergy between the two antimicrobials, with a more significant decreased in biomass and viable cell number with the combination treatment compared to the phage and enzyme alone. This observation was confirmed by microscopy analysis, which also showed that polysaccharide depolymerase treatment reduced, but did not eliminate extracellular matrix polysaccharides. Activity assays on mutant strains did not identify teichoic acids or PNAG/PIA as the exclusive target of Dpo7, suggesting that may be both are degraded by this enzyme. Phage adsorption to S. aureus cells was not significantly altered by incubation with Dpo7, indicating that the mechanism of the observed synergistic interaction is likely through loosening of the biofilm structure. This would allow easier access of the phage particles to their host cells and facilitate infection progression within the bacterial population.
Collapse
Affiliation(s)
- Ana Catarina Duarte
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Spain
- DairySafe Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Lucía Fernández
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Spain
- DairySafe Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Andrea Jurado
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Spain
- DairySafe Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Ana Belén Campelo
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Spain
| | - Yang Shen
- Laboratory of Food Microbiology, Institute of Food, Nutrition and Health, Zurich, Switzerland
| | - Ana Rodríguez
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Spain
- DairySafe Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Pilar García
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Spain
- DairySafe Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| |
Collapse
|
49
|
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
|
50
|
Xu Z, Li Y, Xu A, Soteyome T, Yuan L, Ma Q, Seneviratne G, Li X, Liu J. Cell-wall-anchored proteins affect invasive host colonization and biofilm formation in Staphylococcus aureus. Microbiol Res 2024; 285:127782. [PMID: 38833832 DOI: 10.1016/j.micres.2024.127782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 06/06/2024]
Abstract
As a major human and animal pathogen, Staphylococcus aureus can attach to medical implants (abiotic surface) or host tissues (biotic surface), and further establish robust biofilms which enhances resistance and persistence to host immune system and antibiotics. Cell-wall-anchored proteins (CWAPs) covalently link to peptidoglycan, and largely facilitate the colonization of S. aureus on various surfaces (including adhesion and biofilm formation) and invasion into host cells (including adhesion, immune evasion, iron acquisition and biofilm formation). During biofilm formation, CWAPs function in adhesion, aggregation, collagen-like fiber network formation, and consortia formation. In this review, we firstly focus on the structural features of CWAPs, including their intracellular function and interactions with host cells, as well as the functions and ligand binding of CWAPs in different stages of S. aureus biofilm formation. Then, the roles of CWAPs in different biofilm processes with regards in development of therapeutic approaches are clarified, followed by the association between CWAPs genes and clonal lineages. By touching upon these aspects, we hope to provide comprehensive knowledge and clearer understanding on the CWAPs of S. aureus and their roles in biofilm formation, which may further aid in prevention and treatment infection and vaccine development.
Collapse
Affiliation(s)
- Zhenbo Xu
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou 510640, China; Department of Laboratory Medicine, the Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China.
| | - Yaqin Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou 510640, China
| | - Aijuan Xu
- Guangzhou Hybribio Medical Laboratory, Guangzhou 510730, China
| | - Thanapop Soteyome
- Home Economics Technology, Rajamangala University of Technology Phra Nakhon, Bangkok, Thailand
| | - Lei Yuan
- School of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu 225127, China
| | - Qin Ma
- Sericultural & Agri-Food Research Institute Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture /Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, China
| | - Gamini Seneviratne
- National Institute of Fundamental Studies, Hantana road, Kandy, Sri Lanka
| | - Xuejie Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou 510640, China.
| | - Junyan Liu
- College of Light Industry and Food Science, Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Academy of Contemporary Agricultural Engineering Innovations, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China; Key Laboratory of Green Processing and Intelligent Manufacturing of Lingnan Specialty Food, Ministry of Agriculture, Guangzhou 510225, China.
| |
Collapse
|