51
|
Godart GA, Elwasila SM, Durvasula RV. A rare case of candida osteomyelitis of the mandible associated with osteoradionecrosis and biofilm formation. IDCases 2024; 37:e02029. [PMID: 39155946 PMCID: PMC11327805 DOI: 10.1016/j.idcr.2024.e02029] [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: 09/05/2023] [Revised: 07/08/2024] [Accepted: 07/14/2024] [Indexed: 08/20/2024] Open
Abstract
Candida osteomyelitis, in general, is a relatively rare manifestation compared to its bacterial counterparts. The mandible's involvement is rarer, lacking established management and fewer guidelines. Herein, we aim to illustrate the significant challenge in treatment, namely due to the persistent and resistant nature of Candida albicans-associated biofilm. A multidisciplinary approach involving adjunctive use of antifungals with surgical interventions is typically necessary and feasible in this case. However, surgical interventions may not always be possible in challenging instances in which the patient may be structurally (including osteoradionecrosis) and vascularly compromised, raising questions about the feasibility of standard-of-care as well as the success of alternative therapies aimed at disrupting biofilm formation. Clinicians should maintain a high index of suspicion for complicating, deep-seated Candidiasis in at-risk populations and endeavor to treat as aggressively as possible to limit recurrent disease owing to persistence.
Collapse
Affiliation(s)
- Gabriel A. Godart
- Mayo Clinic, Department of Medicine, Division of Infectious Diseases and Travel Medicine, Jacksonville, FL, USA
| | - Sammer M. Elwasila
- Mayo Clinic, Department of Medicine, Division of Infectious Diseases and Travel Medicine, Jacksonville, FL, USA
| | - Ravindra V. Durvasula
- Mayo Clinic, Department of Medicine, Division of Infectious Diseases and Travel Medicine, Jacksonville, FL, USA
| |
Collapse
|
52
|
Yang S, Meng X, Zhen Y, Baima Q, Wang Y, Jiang X, Xu Z. Strategies and mechanisms targeting Enterococcus faecalis biofilms associated with endodontic infections: a comprehensive review. Front Cell Infect Microbiol 2024; 14:1433313. [PMID: 39091674 PMCID: PMC11291369 DOI: 10.3389/fcimb.2024.1433313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024] Open
Abstract
Enterococcus faecalis is one of the main microorganisms that infects root canals, ranking among the most prevalent microorganisms associated with endodontic treatment failure. Given its pervasive presence in persistent endodontic infections, the successful elimination of Enterococcus faecalis is crucial for effective endodontic treatment and retreatment. Furthermore, Enterococcus faecalis can form biofilms - defense structures that microbes use to fight environmental threats. These biofilms confer resistance against host immune system attacks and antibiotic interventions. Consequently, the presence of biofilms poses a significant challenge in the complete eradication of Enterococcus faecalis and its associated disease. In response, numerous scholars have discovered promising outcomes in addressing Enterococcus faecalis biofilms within root canals and undertaken endeavors to explore more efficacious approaches in combating these biofilms. This study provides a comprehensive review of strategies and mechanisms for the removal of Enterococcus faecalis biofilms.
Collapse
Affiliation(s)
- Shipeng Yang
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Xiuping Meng
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yuqi Zhen
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Quzhen Baima
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yu Wang
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Xinmiao Jiang
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Zhibo Xu
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
53
|
Tang D, Lin Y, Yao H, Liu Y, Xi Y, Li M, Mao A. Effect of L-HSL on biofilm and motility of Pseudomonas aeruginosa and its mechanism. Appl Microbiol Biotechnol 2024; 108:418. [PMID: 39012538 PMCID: PMC11252199 DOI: 10.1007/s00253-024-13247-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 06/24/2024] [Accepted: 06/28/2024] [Indexed: 07/17/2024]
Abstract
Pseudomonas aeruginosa (P. aeruginosa) biofilm formation is a crucial cause of enhanced antibiotic resistance. Quorum sensing (QS) is involved in regulating biofilm formation; QS inhibitors block the QS signaling pathway as a new strategy to address bacterial resistance. This study investigated the potential and mechanism of L-HSL (N-(3-cyclic butyrolactone)-4-trifluorophenylacetamide) as a QS inhibitor for P. aeruginosa. The results showed that L-HSL effectively inhibited the biofilm formation and dispersed the pre-formed biofilm of P. aeruginosa. The production of extracellular polysaccharides and the motility ability of P. aeruginosa were suppressed by L-HSL. C. elegans infection experiment showed that L-HSL was non-toxic and provided protection to C. elegans against P. aeruginosa infection. Transcriptomic analysis revealed that L-HSL downregulated genes related to QS pathways and biofilm formation. L-HSL exhibits a promising potential as a therapeutic drug for P. aeruginosa infection. KEY POINTS: • Chemical synthesis of N-(3-cyclic butyrolactone)-4-trifluorophenylacetamide, named L-HSL. • L-HSL does not generate survival pressure on the growth of P. aeruginosa and can inhibit the QS system. • KEGG enrichment analysis found that after L-HSL treatment, QS-related genes were downregulated.
Collapse
Affiliation(s)
- Deping Tang
- School of Biological & Pharmaceutical Engineering, Lanzhou Jiaotong University, Lanzhou, 730070, Gansu, China
| | - Yanyan Lin
- School of Biological & Pharmaceutical Engineering, Lanzhou Jiaotong University, Lanzhou, 730070, Gansu, China
| | - Huihui Yao
- School of Biological & Pharmaceutical Engineering, Lanzhou Jiaotong University, Lanzhou, 730070, Gansu, China
| | - Yali Liu
- School of Biological & Pharmaceutical Engineering, Lanzhou Jiaotong University, Lanzhou, 730070, Gansu, China
| | - Yanpeng Xi
- School of Biological & Pharmaceutical Engineering, Lanzhou Jiaotong University, Lanzhou, 730070, Gansu, China
| | - Mengjiao Li
- School of Biological & Pharmaceutical Engineering, Lanzhou Jiaotong University, Lanzhou, 730070, Gansu, China
| | - Aihong Mao
- Gansu Provincial Academic Institute for Medical Research, Lanzhou, 730050, Gansu, China.
| |
Collapse
|
54
|
Li XM, Gao Y, Wang SH, Huang YG, Long GQ, Wang DD, Zhang R, Wang AH, Huang SH, Jia JM. Natural Prenylflavonoids from Sophora flavescens Root Bark against Multidrug-Resistant Methicillin-Sensitive Staphylococcus aureus Targeting the Membrane Permeability. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:14684-14700. [PMID: 38905352 DOI: 10.1021/acs.jafc.4c01430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/23/2024]
Abstract
The overuse of antibiotics in animal farming and aquaculture has led to multidrug-resistant methicillin-sensitive Staphylococcus aureus (MR-MSSA) becoming a common pathogen in foodborne diseases. Sophora flavescens Ait. serves as a traditional plant antibacterial agent and functional food ingredient. A total of 30 compounds (1-30) were isolated from the root bark of S. flavescens, consisting of 20 new compounds (1-20). In the biological activity assay, compound 1 demonstrated a remarkable inhibitory effect on MR-MSSA, with an MIC of 2 μg/mL. Furthermore, 1 was found to rapidly eliminate bacteria, inhibit biofilm growth, and exhibit exceptionally low cytotoxicity. Mechanistic studies have revealed that 1 possesses an enhanced membrane-targeting ability, binding to the bacterial cell membrane components phosphatidylglycerol (PG), phosphatidylethanolamine (PE), and cardiolipin (CL). This disruption of bacterial cell membrane integrity increases intracellular reactive oxygen species, protein and DNA leakage, reduced bacterial metabolism, and ultimately bacterial death. In summary, these findings suggest that compound 1 holds promise as a lead compound against MR-MSSA.
Collapse
Affiliation(s)
- Xin-Min Li
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Yun Gao
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Si-Han Wang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Yao-Guang Huang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Guo-Qing Long
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Dong-Dong Wang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Rui Zhang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - An-Hua Wang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Shao-Hui Huang
- Department of Head and Neck Surgery, Stomatological Hospital of China Medical University, Shenyang 110002, People's Republic of China
| | - Jing-Ming Jia
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| |
Collapse
|
55
|
D’Aquila P, De Rose E, Sena G, Scorza A, Cretella B, Passarino G, Bellizzi D. Quorum Quenching Approaches against Bacterial-Biofilm-Induced Antibiotic Resistance. Antibiotics (Basel) 2024; 13:619. [PMID: 39061301 PMCID: PMC11273524 DOI: 10.3390/antibiotics13070619] [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: 06/10/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
With the widespread phenomenon of antibiotic resistance and the diffusion of multiple drug-resistant bacterial strains, enormous efforts are being conducted to identify suitable alternative agents against pathogenic microorganisms. Since an association between biofilm formation and antibiotic resistance phenotype has been observed, a promising strategy pursued in recent years focuses on controlling and preventing this formation by targeting and inhibiting the Quorum Sensing (QS) system, whose central role in biofilm has been extensively demonstrated. Therefore, the research and development of Quorum Quenching (QQ) compounds, which inhibit QS, has gradually attracted the attention of researchers and has become a new strategy for controlling harmful microorganisms. Among these, a number of both natural and synthetic compounds have been progressively identified as able to interrupt the intercellular communication within a microbial community and the adhesion to a surface, thus disintegrating mature/preformed biofilms. This review describes the role played by QS in the formation of bacterial biofilms and then focuses on the mechanisms of different natural and synthetic QS inhibitors (QSIs) exhibiting promising antibiofilm ability against Gram-positive and Gram-negative bacterial pathogens and on their applications as biocontrol strategies in various fields.
Collapse
Affiliation(s)
- Patrizia D’Aquila
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy; (P.D.); (E.D.R.); (G.S.); (G.P.)
| | - Elisabetta De Rose
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy; (P.D.); (E.D.R.); (G.S.); (G.P.)
| | - Giada Sena
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy; (P.D.); (E.D.R.); (G.S.); (G.P.)
| | - Angelo Scorza
- Villa Ermelinda, Progetto Terza Età, 88842 Cutro, Italy; (A.S.); (B.C.)
| | | | - Giuseppe Passarino
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy; (P.D.); (E.D.R.); (G.S.); (G.P.)
| | - Dina Bellizzi
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy; (P.D.); (E.D.R.); (G.S.); (G.P.)
| |
Collapse
|
56
|
Aherne O, Mørch M, Ortiz R, Shannon O, Davies JR. A novel multiplex fluorescent-labeling method for the visualization of mixed-species biofilms in vitro. Microbiol Spectr 2024; 12:e0025324. [PMID: 38785429 PMCID: PMC11218471 DOI: 10.1128/spectrum.00253-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/17/2024] [Indexed: 05/25/2024] Open
Abstract
In nature, bacteria usually exist as mixed-species biofilms, where they engage in a range of synergistic and antagonistic interactions that increase their resistance to environmental challenges. Biofilms are a major cause of persistent infections, and dispersal from initial foci can cause new infections at distal sites thus warranting further investigation. Studies of development and spatial interactions in mixed-species biofilms can be challenging due to difficulties in identifying the different bacterial species in situ. Here, we apply CellTrace dyes to studies of biofilm bacteria and present a novel application for multiplex labeling, allowing identification of different bacteria in mixed-species, in vitro biofilm models. Oral bacteria labeled with CellTrace dyes (far red, yellow, violet, and CFSE [green]) were used to create single- and mixed-species biofilms, which were analyzed with confocal spinning disk microscopy (CSDM). Biofilm supernatants were studied with flow cytometry (FC). Both Gram-positive and Gram-negative bacteria were well labeled and CSDM revealed biofilms with clear morphology and stable staining for up to 4 days. Analysis of CellTrace labeled cells in supernatants using FC showed differences in the biofilm dispersal between bacterial species. Multiplexing with different colored dyes allowed visualization of spatial relationships between bacteria in mixed-species biofilms and relative coverage by the different species was revealed through segmentation of the CSDM images. This novel application, thus, offers a powerful tool for studying structure and composition of mixed-species biofilms in vitro.IMPORTANCEAlthough most chronic infections are caused by mixed-species biofilms, much of our knowledge still comes from planktonic cultures of single bacterial species. Studies of formation and development of mixed-species biofilms are, therefore, required. This work describes a method applicable to labeling of bacteria for in vitro studies of biofilm structure and dispersal. Critically, labeled bacteria can be multiplexed for identification of different species in mixed-species biofilms using confocal spinning disk microscopy, facilitating investigation of biofilm development and spatial interactions under different environmental conditions. The study is an important step in increasing the tools available for such complex and challenging studies.
Collapse
Affiliation(s)
- Olivia Aherne
- Section for Oral Biology and Pathology, Faculty of Odontology and Biofilms Research Center for Biointerfaces, Malmö University, Malmö, Sweden
- CR Competence, Lund, Sweden
| | - Martina Mørch
- Section for Oral Biology and Pathology, Faculty of Odontology and Biofilms Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | | | - Oonagh Shannon
- Section for Oral Biology and Pathology, Faculty of Odontology and Biofilms Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | - Julia R Davies
- Section for Oral Biology and Pathology, Faculty of Odontology and Biofilms Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| |
Collapse
|
57
|
Rajangam SL, Narasimhan MK. Current treatment strategies for targeting virulence factors and biofilm formation in Acinetobacter baumannii. Future Microbiol 2024; 19:941-961. [PMID: 38683166 PMCID: PMC11290764 DOI: 10.2217/fmb-2023-0263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 03/20/2024] [Indexed: 05/01/2024] Open
Abstract
A higher prevalence of Acinetobacter baumannii infections and mortality rate has been reported recently in hospital-acquired infections (HAI). The biofilm-forming capability of A. baumannii makes it an extremely dangerous pathogen, especially in device-associated hospital-acquired infections (DA-HAI), thereby it resists the penetration of antibiotics. Further, the transmission of the SARS-CoV-2 virus was exacerbated in DA-HAI during the epidemic. This review specifically examines the complex interconnections between several components and genes that play a role in the biofilm formation and the development of infections. The current review provides insights into innovative treatments and therapeutic approaches to combat A. baumannii biofilm-related infections, thereby ultimately improving patient outcomes and reducing the burden of HAI.
Collapse
Affiliation(s)
- Seetha Lakshmi Rajangam
- Department of Genetic Engineering, School of Bioengineering, College of Engineering & Technology, SRM Institute of Science & Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| | - Manoj Kumar Narasimhan
- Department of Genetic Engineering, School of Bioengineering, College of Engineering & Technology, SRM Institute of Science & Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India
| |
Collapse
|
58
|
Li C, Gao D, Li C, Cheng G, Zhang L. Fighting against biofilm: The antifouling and antimicrobial material. Biointerphases 2024; 19:040802. [PMID: 39023091 DOI: 10.1116/6.0003695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/07/2024] [Indexed: 07/20/2024] Open
Abstract
Biofilms are groups of microorganisms protected by self-secreted extracellular substances. Biofilm formation on the surface of biomaterial or engineering materials becomes a severe challenge. It has caused significant health, environmental, and societal concerns. It is believed that biofilms lead to life-threatening infection, medical implant failure, foodborne disease, and marine biofouling. To address these issues, tremendous effort has been made to inhibit biofilm formation on materials. Biofilms are extremely difficult to treat once formed, so designing material and coating bearing functional groups that are capable of resisting biofilm formation has attracted increasing attention for the last two decades. Many types of antibiofilm strategies have been designed to target different stages of biofilm formation. Development of the antibiofilm material can be classified into antifouling material, antimicrobial material, fouling release material, and integrated antifouling/antimicrobial material. This review summarizes relevant research utilizing these four approaches and comments on their antibiofilm properties. The feature of each method was compared to reveal the research trend. Antibiofilm strategies in fundamental research and industrial applications were summarized.
Collapse
Affiliation(s)
- Chao Li
- School of Engineering, Westlake University, Hangzhou, Zhejiang 310030, China
- Department of Pharmaceutical Sciences, State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning 116023, China
| | - Dongdong Gao
- School of Engineering, Westlake University, Hangzhou, Zhejiang 310030, China
- Department of Pharmaceutical Sciences, State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning 116023, China
| | - Chunmei Li
- Tsinglan School, Songshan Lake, Dongguan 523000, China
| | - Gang Cheng
- School of Engineering, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Lijun Zhang
- Liaoning Provincial Key Laboratory of Cornea and Ocular Surface Diseases, Liaoning Provincial Optometry Technology Engineering Research Center, The Third People's Hospital of Dalian, Dalian, Liaoning 116033, China
| |
Collapse
|
59
|
Wang Z, Zeng S, Hao Y, Cai W, Sun W, Du J, Long S, Fan J, Wang J, Chen X, Peng X. Gram-negative bacteria recognition and photodynamic elimination by Zn-DPA based sensitizers. Biomaterials 2024; 308:122571. [PMID: 38636132 DOI: 10.1016/j.biomaterials.2024.122571] [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: 03/07/2024] [Revised: 04/07/2024] [Accepted: 04/08/2024] [Indexed: 04/20/2024]
Abstract
The abuse and overuse of antibiotics let drug-resistant bacteria emerges. Antibacterial photodynamic therapy (APDT) has shown outstanding merits to eliminate the drug-resistant bacteria via cytotoxic reactive oxygen species produced by irradiating photosensitizer. However, most of photosensitizers are not effective for Gram-negative bacteria elimination. Herein conjugates of NBS, a photosensitizer, linked with one (NBS-DPA-Zn) or two (NBS-2DPA-Zn) equivalents of zinc-dipicolylamine (Zn-DPA) have been designed to achieve the functional recognition of different bacteria. Due to the cationic character of NBS and metal transfer channel effect of Zn-DPA, NBS-DPA-Zn exhibited the first regent to distinguish P. aeruginosa from other Gram-negative bacteria. Whereas NBS-2DPA-Zn showed broad-spectrum antibacterial effect because the two arm of double Zn-DPA enhanced interactions with anionic membranes of bacteria, led the bacteria aggregation and thus provided the efficacy of APDT to bacteria and corresponding biofilm. In combination with a hydrogel of Pluronic, NBS-2DPA-Zn@gel shows promising clinical application in mixed bacterial diabetic mouse model infection. This might propose a new method that can realize functional identification and elimination of bacteria through intelligent regulation of Zn-DPA, and shows excellent potential for antibacterial application.
Collapse
Affiliation(s)
- Zuokai Wang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, Dalian University of Technology, Dalian, 116024, PR China
| | - Shuang Zeng
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Hi-tech Zone, Dalian, 116024, PR China
| | - Yifu Hao
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Hi-tech Zone, Dalian, 116024, PR China
| | - Wenlin Cai
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, Dalian University of Technology, Dalian, 116024, PR China
| | - Wen Sun
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, Dalian University of Technology, Dalian, 116024, PR China
| | - Jianjun Du
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, Dalian University of Technology, Dalian, 116024, PR China
| | - Saran Long
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, Dalian University of Technology, Dalian, 116024, PR China
| | - Jiangli Fan
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, Dalian University of Technology, Dalian, 116024, PR China
| | - Jingyun Wang
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Hi-tech Zone, Dalian, 116024, PR China
| | - Xiaoqiang Chen
- State Key Laboratory of Fine Chemicals, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518060, PR China
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials, Dalian University of Technology, Dalian, 116024, PR China; State Key Laboratory of Fine Chemicals, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518060, PR China.
| |
Collapse
|
60
|
Thompson E, Qureshi A. Pathogens in FRI - Do bugs matter? - An analysis of FRI studies to assess your enemy. J Orthop 2024; 53:59-72. [PMID: 38476676 PMCID: PMC10925936 DOI: 10.1016/j.jor.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 02/09/2024] [Indexed: 03/14/2024] Open
Abstract
Fracture-related infection (FRI) is a devasting complication for both patients and their treating Orthopaedic surgeon that can lead to loss of limb function or even amputation. The unique and unpredictable features of FRI make its diagnosis and treatment a significant challenge. It has substantial morbidity and financial implications for patients, their families and healthcare providers. In this article, we perform an in-depth and comprehensive review of FRI through recent and seminal literature to highlight evolving definitions, diagnostic and treatment approaches, focusing on common pathogens such as Staphylococcus aureus, polymicrobial infections and multi-drug-resistant organisms (MDRO). Furthermore, multiple resistance mechanisms and adaptations for microbial survival are discussed, as well as modern evidence-based medical and surgical advancements in treatment strategies in combating FRI.
Collapse
Affiliation(s)
- Emmet Thompson
- Limb Reconstruction Service, Trauma & Orthopaedic Department, University Hospital Southampton, Southampton, UK
| | - Amir Qureshi
- Limb Reconstruction Service, Trauma & Orthopaedic Department, University Hospital Southampton, Southampton, UK
| |
Collapse
|
61
|
Tungare K, Gupta J, Bhori M, Garse S, Kadam A, Jha P, Jobby R, Amanullah M, Vijayakumar S. Nanomaterial in controlling biofilms and virulence of microbial pathogens. Microb Pathog 2024; 192:106722. [PMID: 38815775 DOI: 10.1016/j.micpath.2024.106722] [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: 01/28/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
The escalating threat of antimicrobial resistance (AMR) poses a grave concern to global public health, exacerbated by the alarming shortage of effective antibiotics in the pipeline. Biofilms, intricate populations of bacteria encased in self-produced matrices, pose a significant challenge to treatment, as they enhance resistance to antibiotics and contribute to the persistence of organisms. Amid these challenges, nanotechnology emerges as a promising domain in the fight against biofilms. Nanomaterials, with their unique properties at the nanoscale, offer innovative antibacterial modalities not present in traditional defensive mechanisms. This comprehensive review focuses on the potential of nanotechnology in combating biofilms, focusing on green-synthesized nanoparticles and their associated anti-biofilm potential. The review encompasses various aspects of nanoparticle-mediated biofilm inhibition, including mechanisms of action. The diverse mechanisms of action of green-synthesized nanoparticles offer valuable insights into their potential applications in addressing AMR and improving treatment outcomes, highlighting novel strategies in the ongoing battle against infectious diseases.
Collapse
Affiliation(s)
- Kanchanlata Tungare
- School of Biotechnology and Bioinformatics, D Y Patil Deemed to be University, Navi Mumbai, Plot no 50, Sector 15, CBD Belapur, 400614, Maharashtra, India.
| | - Juhi Gupta
- School of Biotechnology and Bioinformatics, D Y Patil Deemed to be University, Navi Mumbai, Plot no 50, Sector 15, CBD Belapur, 400614, Maharashtra, India
| | - Mustansir Bhori
- Inveniolife Technology PVT LTD, Office No.118, Grow More Tower, Plot No.5, Sector 2, Kharghar, Navi Mumbai, Maharashtra, 410210, India
| | - Samiksha Garse
- School of Biotechnology and Bioinformatics, D Y Patil Deemed to be University, Navi Mumbai, Plot no 50, Sector 15, CBD Belapur, 400614, Maharashtra, India
| | - Aayushi Kadam
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, R3T 2N2, Canada; Anatek Services PVT LTD, 10, Sai Chamber, Near Santacruz Railway Bridge, Sen Nagar, Santacruz East, Mumbai, Maharashtra, 400055, India
| | - Pamela Jha
- Department of Biological Sciences, Sunandan Divatia School of Science, NMIMS Deemed to be University, Mumbai, Maharashtra, India
| | - Renitta Jobby
- Amity Institute of Biotechnology, Amity University, Maharashtra, Mumbai-Pune Expressway, Bhatan, Panvel, Navi Mumbai, Maharashtra, 410206, India; Amity Centre of Excellence in Astrobiology, Amity University Maharashtra, Mumbai-Pune Expressway, Bhatan, Panvel, Navi Mumbai, Maharashtra, 410206, India
| | - Mohammed Amanullah
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia, 61421
| | - Sekar Vijayakumar
- Center for Global Health Research (CGHR), Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, India; Marine College, Shandong University, Weihai, 264209, PR China
| |
Collapse
|
62
|
Nosair N, Elzayat S, Elsharaby R, Abdulghaffar IA, Elfarargy HH, Sharaf NA. The association of bacterial biofilm and middle ear mucosa in patients with mucosal chronic suppurative otitis media. ACTA OTORRINOLARINGOLOGICA ESPANOLA 2024; 75:244-251. [PMID: 38729238 DOI: 10.1016/j.otoeng.2024.02.004] [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: 10/27/2023] [Accepted: 02/17/2024] [Indexed: 05/12/2024]
Abstract
OBJECTIVES To evaluate the bacterial biofilm's role in mucosal chronic suppurative otitis media (CSOM) utilizing scanning electron microscopy (SEM). METHODS This study involved 123 participating patients with active and inactive mucosal CSOM who underwent tympanomastoid surgery. SEM was used to examine middle ear mucosa biopsies for the development of biofilms. Middle ear discharge or mucosal swabs from patients were cultured to detect any bacterial growth. The biofilm formation was correlated to the culture results. RESULTS The biofilm was present in 69.9 % of patients (59% of them were with active mucosal CSOM) and absent in 30.1% of the patients (70% of them were with inactive mucosal CSOM), being more statistically significant in active mucosal CSOM (p-value = 0.003). A correlation that was statistically significant was found between active mucosal CSOM and higher grades (3 and 4) of biofilms (p-value <0.05). The mucosal CSOM type and the results of the culture had a relationship that was statistically significant (p-value <0.001). 60% of patients had positive culture (70% of them were with active mucosal CSOM). There was a statistically significant relation between Pseudomonas aeruginosa bacterial growth and active mucosal CSOM (p-value = 0.004) as well as higher grades of biofilms in mucosal CSOM. CONCLUSION Mucosal CSOM, especially the active type, is a biofilm-related disease. There is a significant relation between the state of mucosal CSOM (active or inactive) and culture results with predominance of Pseudomonas aeruginosa bacterial growth in active mucosal CSOM and in higher grades of biofilms in mucosal CSOM.
Collapse
Affiliation(s)
- Nahla Nosair
- Clinical Pathology, Faculty of Medicine, Kafrelsheikh University, Egypt
| | - Saad Elzayat
- Otorhinolaryngology, Faculty of Medicine, Kafrelsheikh University, Egypt
| | - Radwa Elsharaby
- Clinical Pathology, Faculty of Medicine, Tanta University, Egypt
| | | | | | - Nehal A Sharaf
- Clinical Pathology, Faculty of Medicine, Kafrelsheikh University, Egypt
| |
Collapse
|
63
|
Zhang B, Lan W, Yan P, Xie J. The antibacterial and inhibition effect of chitosan grafted gentisate acid derivatives against Pseudomonas fluorescens: Attacking multiple targets on structure, metabolism system, antioxidant system, and biofilm. Int J Biol Macromol 2024; 273:133225. [PMID: 38897501 DOI: 10.1016/j.ijbiomac.2024.133225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 06/08/2024] [Accepted: 06/15/2024] [Indexed: 06/21/2024]
Abstract
This work aimed to investigate the antibacterial ability and potential mechanism of chitosan grafted gentisate acid derivatives (CS-g-GA) against Pseudomonas fluorescens. The results showed that CS-g-GA had a significant suppressive impact on the growth of Pseudomonas fluorescens, the minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) were 0.64 mg/mL and 1.28 mg/mL, respectively. Results of scanning electron microscopy (SEM) and alkaline phosphatase (AKPase) confirmed that CS-g-GA destroyed the cell structure thereby causing the leakage of intracellular components. In addition, 1 × MIC of CS-g-GA could significantly inhibit the formation of biofilms, and 74.78 % mature biofilm and 86.21 % extracellular polysaccharide of Pseudomonas fluorescens were eradicated by CS-g-GA at 2 × MIC. The results on the respiratory energy metabolism system and antioxidant system demonstrated that CS-g-GA caused respiratory disturbance and energy limitation by influencing the key enzyme activities. It could also bind to DNA and affect genetic metabolism. From this, it could be seen that CS-g-GA had the potential to control foodborne contamination of Pseudomonas fluorescens by attacking multiple targets.
Collapse
Affiliation(s)
- Bingjie Zhang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Weiqing Lan
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; Shanghai Aquatic Products Processing and Storage Engineering Technology Research Center, Shanghai 201306, China; National Experimental Teaching Demonstration Center for Food Science and Engineering (Shanghai Ocean University), Shanghai 201306, China.
| | - Peiling Yan
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Jing Xie
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; Shanghai Aquatic Products Processing and Storage Engineering Technology Research Center, Shanghai 201306, China; National Experimental Teaching Demonstration Center for Food Science and Engineering (Shanghai Ocean University), Shanghai 201306, China.
| |
Collapse
|
64
|
Takahara M, Hirayama S, Futamata H, Nakao R, Tashiro Y. Biofilm-derived membrane vesicles exhibit potent immunomodulatory activity in Pseudomonas aeruginosa PAO1. Microbiol Immunol 2024; 68:224-236. [PMID: 38797913 DOI: 10.1111/1348-0421.13156] [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: 12/05/2023] [Revised: 02/15/2024] [Accepted: 05/09/2024] [Indexed: 05/29/2024]
Abstract
Pathogenic bacteria form biofilms on epithelial cells, and most bacterial biofilms show increased production of membrane vesicles (MVs), also known as outer membrane vesicles in Gram-negative bacteria. Numerous studies have investigated the MVs released under planktonic conditions; however, the impact of MVs released from biofilms on immune responses remains unclear. This study aimed to investigate the characteristics and immunomodulatory activity of MVs obtained from both planktonic and biofilm cultures of Pseudomonas aeruginosa PAO1. The innate immune responses of macrophages to planktonic-derived MVs (p-MVs) and biofilm-derived MVs (b-MVs) were investigated by measuring the mRNA expression of proinflammatory cytokines. Our results showed that b-MVs induced a higher expression of inflammatory cytokines, including Il1b, Il6, and Il12p40, than p-MVs. The mRNA expression levels of Toll-like receptor 4 (Tlr4) differed between the two types of MVs, but not Tlr2. Polymyxin B significantly neutralized b-MV-mediated cytokine induction, suggesting that lipopolysaccharide of native b-MVs is the origin of the immune response. In addition, heat-treated or homogenized b-MVs induced the mRNA expression of cytokines, including Tnfa, Il1b, Il6, and Il12p40. Heat treatment of MVs led to increased expression of Tlr2 but not Tlr4, suggesting that TLR2 ligands play a role in detecting the pathogen-associated molecular patterns in lysed MVs. Taken together, our data indicate that potent immunomodulatory MVs are produced in P. aeruginosa biofilms and that this behavior could be a strategy for the bacteria to infect host cells. Furthermore, our findings would contribute to developing novel vaccines using MVs.
Collapse
Affiliation(s)
- Minato Takahara
- Department of Engineering, Graduate School of Integrated Science and Technology, Shizuoka University, Hamamatsu, Japan
| | - Satoru Hirayama
- Department of Bacteriology I, National Institute of Infectious Diseases, Tokyo, Japan
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hiroyuki Futamata
- Department of Engineering, Graduate School of Integrated Science and Technology, Shizuoka University, Hamamatsu, Japan
- Graduate School of Science and Technology, Shizuoka University, Hamamatsu, Japan
- Research Institute of Green Science and Technology, Shizuoka University, Shizuoka, Japan
| | - Ryoma Nakao
- Department of Bacteriology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yosuke Tashiro
- Department of Engineering, Graduate School of Integrated Science and Technology, Shizuoka University, Hamamatsu, Japan
- Graduate School of Science and Technology, Shizuoka University, Hamamatsu, Japan
- JST PRESTO, Kawaguchi, Japan
| |
Collapse
|
65
|
Khursheed A, Xu LC, Siedlecki CA. The effects of submicron-textured surface topography on antibiotic efficacy against biofilms. J Biomed Mater Res B Appl Biomater 2024; 112:e35436. [PMID: 38961592 PMCID: PMC11239140 DOI: 10.1002/jbm.b.35436] [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: 02/29/2024] [Revised: 04/23/2024] [Accepted: 05/14/2024] [Indexed: 07/05/2024]
Abstract
Submicron-textured surfaces have been a promising approach to mitigate biofilm development and control microbial infection. However, the use of the single surface texturing approach is still far from ideal for achieving complete control of microbial infections on implanted biomedical devices. The use of a surface topographic modification that might improve the utility of standard antibiotic therapy could alleviate the complications of biofilms on devices. In this study, we characterized the biofilms of Staphylococcus aureus and Pseudomonas aeruginosa on smooth and submicron-textured polyurethane surfaces after 1, 2, 3, and 7 days, and measured the efficacy of common antibiotics against these biofilms. Results show that the submicron-textured surfaces significantly reduced biofilm formation and growth, and that the efficacy of antibiotics against biofilms grown on textured surfaces was improved compared with smooth surfaces. The antibiotic efficacy appears to be related to the degree of biofilm development. At early time points in biofilm formation, antibiotic treatment reveals reasonably good antibiotic efficacy against biofilms on both smooth and textured surfaces, but as biofilms mature, the efficacy of antibiotics drops dramatically on smooth surfaces, with lesser decreases seen for the textured surfaces. The results demonstrate that surface texturing with submicron patterns is able to improve the use of standard antibiotic therapy to treat device-centered biofilms by slowing the development of the biofilm, thereby offering less resistance to antibiotic delivery to the bacteria within the biofilm community.
Collapse
Affiliation(s)
- Asma Khursheed
- Department of Surgery, The Pennsylvania State University, College of Medicine, Hershey, PA 17033
| | - Li-Chong Xu
- Department of Surgery, The Pennsylvania State University, College of Medicine, Hershey, PA 17033
| | - Christopher A. Siedlecki
- Department of Surgery, The Pennsylvania State University, College of Medicine, Hershey, PA 17033
- Department of Biomedical Engineering, The Pennsylvania State University, College of Medicine, Hershey, PA 17033
| |
Collapse
|
66
|
Arrigoni R, Ballini A, Jirillo E, Santacroce L. Current View on Major Natural Compounds Endowed with Antibacterial and Antiviral Effects. Antibiotics (Basel) 2024; 13:603. [PMID: 39061285 PMCID: PMC11274329 DOI: 10.3390/antibiotics13070603] [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: 05/23/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Nowadays, infectious diseases of bacterial and viral origins represent a serious medical problem worldwide. In fact, the development of antibiotic resistance is responsible for the emergence of bacterial strains that are refractory even to new classes of antibiotics. Furthermore, the recent COVID-19 pandemic suggests that new viruses can emerge and spread all over the world. The increase in infectious diseases depends on multiple factors, including malnutrition, massive migration of population from developing to industrialized areas, and alteration of the human microbiota. Alternative treatments to conventional antibiotics and antiviral drugs have intensively been explored. In this regard, plants and marine organisms represent an immense source of products, such as polyphenols, alkaloids, lanthipeptides, and terpenoids, which possess antibacterial and antiviral activities. Their main mechanisms of action involve modifications of bacterial cell membranes, with the formation of pores, the release of cellular content, and the inhibition of bacterial adherence to host cells, as well as of the efflux pump. Natural antivirals can interfere with viral replication and spreading, protecting the host with the enhanced production of interferon. Of note, these antivirals are not free of side effects, and their administration to humans needs more research in terms of safety. Preclinical research with natural antibacterial and antiviral compounds confirms their effects against bacteria and viruses, but there are still only a few clinical trials. Therefore, their full exploitation and more intensive clinical studies represent the next steps to be pursued in this area of medicine.
Collapse
Affiliation(s)
- Roberto Arrigoni
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), 70124 Bari, Italy
| | - Andrea Ballini
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
| | - Emilio Jirillo
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Luigi Santacroce
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| |
Collapse
|
67
|
Recktenwald M, Kaur M, Benmassaoud MM, Copling A, Khanna T, Curry M, Cortes D, Fleischer G, Carabetta VJ, Vega SL. Antimicrobial Peptide Screening for Designing Custom Bactericidal Hydrogels. Pharmaceutics 2024; 16:860. [PMID: 39065557 PMCID: PMC11279943 DOI: 10.3390/pharmaceutics16070860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Staphylococcus aureus (S. aureus) is an opportunistic pathogen that lives on surfaces and skin and can cause serious infections inside the body. Antimicrobial peptides (AMPs) are part of the innate immune system and can eliminate pathogens, including bacteria and viruses, and are a promising alternative to antibiotics. Although studies have reported that AMP-functionalized hydrogels can prevent bacterial adhesion and biofilm formation, AMP dosing and the combined effects of multiple AMPs are not well understood. Here, three AMPs with different antibacterial properties were synthesized and the soluble minimum inhibitory concentrations (MICs) of each AMP against methicillin-susceptible S. aureus (MSSA) and methicillin-resistant S. aureus (MRSA) were determined. Hydrogels with immobilized AMPs at their MIC (DD13-RIP 27.5 µM; indolicidin 43.8 µM; P10 120 µM) were effective in preventing MRSA adhesion and biofilm formation. Checkerboard AMP screens identified synergy between indolicidin (3.1 µM) and P10 (12.5 µM) based on soluble fractional inhibitory concentration indices (FICIs) against MRSA, and hydrogels formed with these AMPs at half of their synergistic concentrations (total peptide concentration, 7.8 µM) were highly efficacious in killing MRSA. Mammalian cells cultured atop these hydrogels were highly viable, demonstrating that these AMP hydrogels are biocompatible and selectively eradicate bacteria, based on soluble checkerboard-screening data.
Collapse
Affiliation(s)
- Matthias Recktenwald
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA; (M.R.); (M.M.B.)
| | - Muskanjot Kaur
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (M.K.); (M.C.); (D.C.); (G.F.)
| | - Mohammed M. Benmassaoud
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA; (M.R.); (M.M.B.)
| | - Aryanna Copling
- Department of Translational Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA;
| | - Tulika Khanna
- Department of Biological Sciences, Rowan University, Glassboro, NJ 08028, USA;
| | - Michael Curry
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (M.K.); (M.C.); (D.C.); (G.F.)
| | - Dennise Cortes
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (M.K.); (M.C.); (D.C.); (G.F.)
| | - Gilbert Fleischer
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (M.K.); (M.C.); (D.C.); (G.F.)
| | - Valerie J. Carabetta
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (M.K.); (M.C.); (D.C.); (G.F.)
| | - Sebastián L. Vega
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA; (M.R.); (M.M.B.)
- Department of Orthopedic Surgery, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| |
Collapse
|
68
|
Guo T, Zhou N, Yang L, Wang Z, Huan C, Lin T, Bao G, Hu J, Li G. Acinetobacter baumannii biofilm was inhibited by tryptanthrin through disrupting its different stages and genes expression. iScience 2024; 27:109942. [PMID: 38812547 PMCID: PMC11134903 DOI: 10.1016/j.isci.2024.109942] [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: 02/22/2024] [Revised: 03/25/2024] [Accepted: 05/06/2024] [Indexed: 05/31/2024] Open
Abstract
Biofilm formation plays a significant role in antibiotic resistance, necessitating the search for alternative therapies against biofilm-associated infections. This study demonstrates that 20 μg/mL tryptanthrin can hinder biofilm formation above 50% in various A. baumannii strains. Tryptanthrin impacts various stages of biofilm formation, including the inhibition of surface motility and eDNA release in A. baumannii, as well as an increase in its sensitivity to H202. RT-qPCR analysis reveals that tryptanthrin significantly decreases the expression of the following genes: abaI (19.07%), abaR (33.47%), bfmR (43.41%), csuA/B (64.16%), csuE (50.20%), ompA (67.93%), and katE (72.53%), which are related to biofilm formation and quorum sensing. Furthermore, tryptanthrin is relatively safe and can reduce the virulence of A. baumannii in a Galleria mellonella infection model. Overall, our study demonstrates the potential of tryptanthrin in controlling biofilm formation and virulence of A. baumannii by disrupting different stages of biofilm formation and intercellular signaling communication.
Collapse
Affiliation(s)
- Tingting Guo
- Department of Microbiology, Medical College, Yangzhou University, Yangzhou 225001, China
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225001, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou 225001, China
| | - Na Zhou
- Department of Microbiology, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Liying Yang
- Department of Microbiology, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Zichen Wang
- Department of Microbiology, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Changchao Huan
- Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou 225001, China
| | - Tao Lin
- Department of Laboratory Medicine, Affiliated Hospital, Yangzhou University, Yangzhou 225009, China
| | - Guangyu Bao
- Department of Laboratory Medicine, Affiliated Hospital, Yangzhou University, Yangzhou 225009, China
| | - Jian Hu
- Department of Laboratory Medicine, Yixing Hospital of Traditional Chinese Medicine/Clinical Medical College, Guangling College, Yangzhou University, Yangzhou 214200, China
| | - Guocai Li
- Department of Microbiology, Medical College, Yangzhou University, Yangzhou 225001, China
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225001, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou 225001, China
| |
Collapse
|
69
|
Beltrán-Martínez ME, Tapia-Rodríguez MR, Ayala-Zavala JF, Gómez-Álvarez A, Robles-Zepeda RE, Torres-Moreno H, de Rodríguez DJ, López-Romero JC. Antimicrobial and Antibiofilm Potential of Flourensia retinophylla against Staphylococcus aureus. PLANTS (BASEL, SWITZERLAND) 2024; 13:1671. [PMID: 38931103 PMCID: PMC11207523 DOI: 10.3390/plants13121671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024]
Abstract
Staphylococcus aureus is a Gram-positive bacteria with the greatest impact in the clinical area, due to the high rate of infections and deaths reaching every year. A previous scenario is associated with the bacteria's ability to develop resistance against conventional antibiotic therapies as well as biofilm formation. The above situation exhibits the necessity to reach new effective strategies against this pathogen. Flourensia retinophylla is a medicinal plant commonly used for bacterial infections treatments and has demonstrated antimicrobial effect, although its effect against S. aureus and bacterial biofilms has not been investigated. The purpose of this work was to analyze the antimicrobial and antibiofilm potential of F. retinophylla against S. aureus. The antimicrobial effect was determined using an ethanolic extract of F. retinophylla. The surface charge of the bacterial membrane, the K+ leakage and the effect on motility were determined. The ability to prevent and remove bacterial biofilms was analyzed in terms of bacterial biomass, metabolic activity and viability. The results showed that F. retinophylla presents inhibitory (MIC: 250 µg/mL) and bactericidal (MBC: 500 µg/mL) activity against S. aureus. The MIC extract increased the bacterial surface charge by 1.4 times and the K+ concentration in the extracellular medium by 60%. The MIC extract inhibited the motility process by 100%, 61% and 40% after 24, 48 and 72 h, respectively. The MIC extract prevented the formation of biofilms by more than 80% in terms of biomass production and metabolic activity. An extract at 10 × MIC reduced the metabolic activity by 82% and the viability by ≈50% in preformed biofilms. The results suggest that F. retinophylla affects S. areus membrane and the process of biofilm formation and removal. This effect could set a precedent to use this plant as alternative for antimicrobial and disinfectant therapies to control infections caused by this pathogen. In addition, this shrub could be considered for carrying out a purification process in order to identify the compounds responsible for the antimicrobial and antibiofilm effect.
Collapse
Affiliation(s)
- Minerva Edith Beltrán-Martínez
- Coordinación de Tecnología de Alimentos de Origen Vegetal, Centro de Investigación en Alimentación y Desarrollo, A.C. Carretera Gustavo Astiazarán Rosas No. 46, Colonia la Victoria, Hermosillo 83304, Mexico; (M.E.B.-M.); (J.F.A.-Z.)
| | - Melvin Roberto Tapia-Rodríguez
- Departamento de Biotecnología y Ciencias Alimentarias, Instituto Tecnológico de Sonora, 5 de Febrero 818 sur, Col. Centro, Ciudad Obregón 85000, Mexico;
| | - Jesús Fernando Ayala-Zavala
- Coordinación de Tecnología de Alimentos de Origen Vegetal, Centro de Investigación en Alimentación y Desarrollo, A.C. Carretera Gustavo Astiazarán Rosas No. 46, Colonia la Victoria, Hermosillo 83304, Mexico; (M.E.B.-M.); (J.F.A.-Z.)
| | - Agustín Gómez-Álvarez
- Departamento de Ingeniería Química y Metalurgia, Universidad de Sonora, Hermosillo 83000, Mexico;
| | | | - Heriberto Torres-Moreno
- Departamento de Ciencias Químico Biológicas y Agropecuarias, Universidad de Sonora, Caborca 83600, Mexico;
| | | | - Julio César López-Romero
- Departamento de Ciencias Químico Biológicas y Agropecuarias, Universidad de Sonora, Caborca 83600, Mexico;
| |
Collapse
|
70
|
Mikziński P, Kraus K, Widelski J, Paluch E. Modern Microbiological Methods to Detect Biofilm Formation in Orthopedy and Suggestions for Antibiotic Therapy, with Particular Emphasis on Prosthetic Joint Infection (PJI). Microorganisms 2024; 12:1198. [PMID: 38930580 PMCID: PMC11205407 DOI: 10.3390/microorganisms12061198] [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: 05/12/2024] [Revised: 06/05/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Biofilm formation is a serious problem that relatively often causes complications in orthopedic surgery. Biofilm-forming pathogens invade implanted foreign bodies and surrounding tissues. Such a condition, if not limited at the appropriate time, often requires reoperation. This can be partially prevented by selecting an appropriate prosthesis material that prevents the development of biofilm. There are many modern techniques available to detect the formed biofilm. By applying them we can identify and visualize biofilm-forming microorganisms. The most common etiological factors associated with biofilms in orthopedics are: Staphylococcus aureus, coagulase-negative Staphylococci (CoNS), and Enterococcus spp., whereas Gram-negative bacilli and Candida spp. also deserve attention. It seems crucial, for therapeutic success, to eradicate the microorganisms able to form biofilm after the implantation of endoprostheses. Planning the effective targeted antimicrobial treatment of postoperative infections requires accurate identification of the microorganism responsible for the complications of the procedure. The modern microbiological testing techniques described in this article show the diagnostic options that can be followed to enable the implementation of effective treatment.
Collapse
Affiliation(s)
- Paweł Mikziński
- Faculty of Medicine, Wroclaw Medical University, Wyb. Pasteura 1, 50-376 Wroclaw, Poland; (P.M.); (K.K.)
| | - Karolina Kraus
- Faculty of Medicine, Wroclaw Medical University, Wyb. Pasteura 1, 50-376 Wroclaw, Poland; (P.M.); (K.K.)
| | - Jarosław Widelski
- Department of Pharmacognosy with Medicinal Plants Garden, Lublin Medical University, 20-093 Lublin, Poland;
| | - Emil Paluch
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, Tytusa Chalubinskiego 4, 50-376 Wroclaw, Poland
| |
Collapse
|
71
|
Verma J, Devi S, Narang A, Kaur S, Manhas RK. Probiotic potential of Streptomyces levis strain HFM-2 isolated from human gut and its antibiofilm properties against pathogenic bacteria. BMC Microbiol 2024; 24:208. [PMID: 38862894 PMCID: PMC11165917 DOI: 10.1186/s12866-024-03353-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: 02/20/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Antimicrobial resistance (AMR) is a serious worldwide public health concern that needs immediate action. Probiotics could be a promising alternative for fighting antibiotic resistance, displaying beneficial effects to the host by combating diseases, improving growth, and stimulating the host immune responses against infection. This study was conducted to evaluate the probiotic, antibacterial, and antibiofilm potential of Streptomyces levis strain HFM-2 isolated from the healthy human gut. RESULTS In vitro antibacterial activity in the cell-free supernatant of S. levis strain HFM-2 was evaluated against different pathogens viz. K. pneumoniae sub sp. pneumoniae, S. aureus, B. subtilis, VRE, S. typhi, S. epidermidis, MRSA, V. cholerae, M. smegmatis, E. coli, P. aeruginosa and E. aerogenes. Further, the ethyl acetate extract from S. levis strain HFM-2 showed strong biofilm inhibition against S. typhi, K. pneumoniae sub sp. pneumoniae, P. aeruginosa and E. coli. Fluorescence microscopy was used to detect biofilm inhibition properties. MIC and MBC values of EtOAc extract were determined at 500 and 1000 µg/mL, respectively. Further, strain HFM-2 showed high tolerance in gastric juice, pancreatin, bile, and at low pH. It exhibited efficient adhesion properties, displaying auto-aggregation (97.0%), hydrophobicity (95.71%, 88.96%, and 81.15% for ethyl acetate, chloroform and xylene, respectively), and showed 89.75%, 86.53%, 83.06% and 76.13% co-aggregation with S. typhi, MRSA, S. pyogenes and E. coli, respectively after 60 min of incubation. The S. levis strain HFM-2 was susceptible to different antibiotics such as tetracycline, streptomycin, kanamycin, ciprofloxacin, erythromycin, linezolid, meropenem, amikacin, gentamycin, clindamycin, moxifloxacin and vancomycin, but resistant to ampicillin and penicillin G. CONCLUSION The study shows that S. levis strain HFM-2 has significant probiotic properties such as good viability in bile, gastric juice, pancreatin environment, and at low pH; proficient adhesion properties, and antibiotic susceptibility. Further, the EtOAc extract of Streptomyces levis strain HFM-2 has a potent antibiofilm and antibacterial activity against antibacterial-resistant clinical pathogens.
Collapse
Affiliation(s)
- Jaya Verma
- Department of Microbiology, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Sapna Devi
- Department of Microbiology, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Anmol Narang
- Department of Microbiology, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Sukhraj Kaur
- Department of Microbiology, Guru Nanak Dev University, Amritsar, Punjab, India
| | | |
Collapse
|
72
|
Wang S, Courreges MC, Xu L, Gurung B, Berryman M, Gu T. Revealing roles of S-layer protein (SlpA) in Clostridioides difficile pathogenicity by generating the first slpA gene deletion mutant. Microbiol Spectr 2024; 12:e0400523. [PMID: 38709045 PMCID: PMC11237437 DOI: 10.1128/spectrum.04005-23] [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: 11/21/2023] [Accepted: 04/16/2024] [Indexed: 05/07/2024] Open
Abstract
Clostridioides difficile infection (CDI) with high morbidity and high mortality is an urgent threat to public health, and C. difficile pathogenesis studies are eagerly required for CDI therapy. The major surface layer protein, SlpA, was supposed to play a key role in C. difficile pathogenesis; however, a lack of isogenic slpA mutants has greatly hampered analysis of SlpA functions. In this study, the whole slpA gene was successfully deleted for the first time via CRISPR-Cas9 system. Deletion of slpA in C. difficile resulted in smaller, smother-edged colonies, shorter bacterial cell size, and aggregation in suspension. For life cycle, the mutant demonstrated lower growth (changes of optical density at 600 nm, OD600) but higher cell density (colony-forming unit, CFU), decreased toxins production, and inhibited sporulation. Moreover, the mutant was more impaired in motility, more sensitive to vancomycin and Triton X-100-induced autolysis, releasing more lactate dehydrogenase. In addition, SlpA deficiency led to robust biofilm formation but weak adhesion to human host cells.IMPORTANCEClostridioides difficile infection (CDI) has been the most common hospital-acquired infection, with a high rate of antibiotic resistance and recurrence incidences, become a debilitating public health threat. It is urgently needed to study C. difficile pathogenesis for developing efficient strategies as CDI therapy. SlpA was indicated to play a key role in C. difficile pathogenesis. However, analysis of SlpA functions was hampered due to lack of isogenic slpA mutants. Surprisingly, the first slpA deletion C. difficile strain was generated in this study via CRISPR-Cas9, further negating the previous thought about slpA being essential. Results in this study will provide direct proof for roles of SlpA in C. difficile pathogenesis, which will facilitate future investigations for new targets as vaccines, new therapeutic agents, and intervention strategies in combating CDI.
Collapse
Affiliation(s)
- Shaohua Wang
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
- Infectious and Tropical Disease Institute, Ohio University, Athens, Ohio, USA
| | - Maria C. Courreges
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| | - Lingjun Xu
- Department of Chemical and Biomolecular Engineering, Institute for Corrosion and Multiphase Technology, Ohio University, Athens, Ohio, USA
| | - Bijay Gurung
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| | - Mark Berryman
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| | - Tingyue Gu
- Department of Chemical and Biomolecular Engineering, Institute for Corrosion and Multiphase Technology, Ohio University, Athens, Ohio, USA
| |
Collapse
|
73
|
Ben-Amram H, Azrad M, Cohen-Assodi J, Sharabi-Nov A, Edelstein S, Agay-Shay K, Peretz A. Biofilm Formation by Hospital-Acquired Resistant Bacteria Isolated from Respiratory Samples. J Epidemiol Glob Health 2024; 14:291-297. [PMID: 38564110 PMCID: PMC11176280 DOI: 10.1007/s44197-024-00215-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Hospital-acquired resistant infections (HARI) are infections, which develop 48 h or more after admission to a healthcare facility. HARI pose a considerably acute challenge, due to limited treatment options. These infections are associated bacterial biofilms, which act as a physical barrier to diverse external stresses, such as desiccation, antimicrobials and biocides. We assessed the influence of multiple factors on biofilm production by HARI -associated bacteria. METHODS Bacteria were isolated from samples of patients with respiratory HARI who were hospitalized during 2020-2022 in north Israel. Following antibiotic susceptibility testing by disc diffusion or broth microdilution, biofilm formation capacities of resistant bacteria (methicillin-resistant staphylococcus aureus, extended spectrum beta-lactamase-producing Escherichia coli and Klebsiela pneumonia, and multidrug-resistant Pseudomonas aeruginosa and Acinetobacter baumannii) was assessed using the crystalline violet staining method. Data regarding season, time to infection, bacterial species, patient age and gender, year, and medical department were collected from the patient medical records. RESULTS Among the 226 study isolates, K. pneumonia was the most prevalent (35.4%) bacteria, followed by P. aeruginosa (23.5%), and methicillin-resistant staphylococcus aureus (MRSA) (21.7%). A significantly higher rate of HARI was documented in 2022 compared to 2020-2021. The majority of isolates (63.3%) were strong biofilm producers, with K. pneumonia (50.3%) being most dominant, followed by P. aeruginosa (29.4%). Biofilm production strength was significantly affected by seasonality and hospitalization length, with strong biofilm production in autumn and in cases where hospitalization length exceeded 30 days. CONCLUSION Biofilm production by HARI bacteria is influenced by bacterial species, season and hospitalization length.
Collapse
Affiliation(s)
- Hila Ben-Amram
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
- The Clinical Microbiology Laboratory, Ziv Medical Center, Affiliated with Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Maya Azrad
- The Clinical Microbiology Laboratory, Tzafon Medical Center, Affiliated with Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Jackie Cohen-Assodi
- The Clinical Microbiology Laboratory, Ziv Medical Center, Affiliated with Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | | | - Shimon Edelstein
- The Infectious Diseases, Ziv Medical Center, Affiliated with Azrieli Faculty of Medicine, Bar Ilan University, 1311502, Safed, Israel
| | - Keren Agay-Shay
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Avi Peretz
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel.
- The Clinical Microbiology Laboratory, Tzafon Medical Center, Affiliated with Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel.
| |
Collapse
|
74
|
da Cruz Nizer WS, Adams ME, Allison KN, Montgomery MC, Mosher H, Cassol E, Overhage J. Oxidative stress responses in biofilms. Biofilm 2024; 7:100203. [PMID: 38827632 PMCID: PMC11139773 DOI: 10.1016/j.bioflm.2024.100203] [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: 02/28/2024] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 06/04/2024] Open
Abstract
Oxidizing agents are low-molecular-weight molecules that oxidize other substances by accepting electrons from them. They include reactive oxygen species (ROS), such as superoxide anions (O2-), hydrogen peroxide (H2O2), and hydroxyl radicals (HO-), and reactive chlorine species (RCS) including sodium hypochlorite (NaOCl) and its active ingredient hypochlorous acid (HOCl), and chloramines. Bacteria encounter oxidizing agents in many different environments and from diverse sources. Among them, they can be produced endogenously by aerobic respiration or exogenously by the use of disinfectants and cleaning agents, as well as by the mammalian immune system. Furthermore, human activities like industrial effluent pollution, agricultural runoff, and environmental activities like volcanic eruptions and photosynthesis are also sources of oxidants. Despite their antimicrobial effects, bacteria have developed many mechanisms to resist the damage caused by these toxic molecules. Previous research has demonstrated that growing as a biofilm particularly enhances bacterial survival against oxidizing agents. This review aims to summarize the current knowledge on the resistance mechanisms employed by bacterial biofilms against ROS and RCS, focussing on the most important mechanisms, including the formation of biofilms in response to oxidative stressors, the biofilm matrix as a protective barrier, the importance of detoxifying enzymes, and increased protection within multi-species biofilm communities. Understanding the complexity of bacterial responses against oxidative stress will provide valuable insights for potential therapeutic interventions and biofilm control strategies in diverse bacterial species.
Collapse
Affiliation(s)
| | - Madison Elisabeth Adams
- Department of Health Sciences, Carleton University, 1125 Colonel By Drive, Ottawa, K1S 5B6, ON, Canada
| | - Kira Noelle Allison
- Department of Health Sciences, Carleton University, 1125 Colonel By Drive, Ottawa, K1S 5B6, ON, Canada
| | | | - Hailey Mosher
- Department of Health Sciences, Carleton University, 1125 Colonel By Drive, Ottawa, K1S 5B6, ON, Canada
| | - Edana Cassol
- Department of Health Sciences, Carleton University, 1125 Colonel By Drive, Ottawa, K1S 5B6, ON, Canada
| | - Joerg Overhage
- Department of Health Sciences, Carleton University, 1125 Colonel By Drive, Ottawa, K1S 5B6, ON, Canada
| |
Collapse
|
75
|
Deng W, Zhou C, Qin J, Jiang Y, Li D, Tang X, Luo J, Kong J, Wang K. Molecular mechanisms of DNase inhibition of early biofilm formation Pseudomonas aeruginosa or Staphylococcus aureus: A transcriptome analysis. Biofilm 2024; 7:100174. [PMID: 38292330 PMCID: PMC10826141 DOI: 10.1016/j.bioflm.2023.100174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 11/14/2023] [Accepted: 12/18/2023] [Indexed: 02/01/2024] Open
Abstract
In vitro studies show that DNase can inhibit Pseudomonas aeruginosa and Staphylococcus aureus biofilm formation. However, the underlying molecular mechanisms remain poorly understood. This study used an RNA-sequencing transcriptomic approach to investigate the mechanism by which DNase I inhibits early P. aeruginosa and S. aureus biofilm formation on a transcriptional level, respectively. A total of 1171 differentially expressed genes (DEGs) in P. aeruginosa and 1016 DEGs in S. aureus enriched in a variety of biological processes and pathways were identified, respectively. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses revealed that the DEGs were primarily involved in P. aeruginosa two-component system, biofilm formation, and flagellar assembly and in S. aureus biosynthesis of secondary metabolites, microbial metabolism in diverse environments, and biosynthesis of amino acids, respectively. The transcriptional data were validated using quantitative real-time polymerase chain reaction (RT-qPCR), and the expression profiles of 22 major genes remained consistent. These findings suggested that DNase I may inhibit early biofilm formation by downregulating the expression of P. aeruginosa genes associated with flagellar assembly and the type VI secretion system, and by downregulating S. aureus capsular polysaccharide and amino acids metabolism gene expression, respectively. This study offers insights into the mechanisms of DNase treatment-based inhibition of early P. aeruginosa and S. aureus biofilm formation.
Collapse
Affiliation(s)
- Wusheng Deng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Respiratory and Critical Care Medicine, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Chuanlin Zhou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jiaoxia Qin
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yun Jiang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Shaoyang University, Shaoyang, Hunan, China
| | - Dingbin Li
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xiujia Tang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jing Luo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jinliang Kong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Ke Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
76
|
Puca V, Marinacci B, Pellegrini B, Campanile F, Santagati M, Grande R. Biofilm and bacterial membrane vesicles: recent advances. Expert Opin Ther Pat 2024; 34:475-491. [PMID: 38578180 DOI: 10.1080/13543776.2024.2338101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/21/2024] [Indexed: 04/06/2024]
Abstract
INTRODUCTION Bacterial Membrane Vesicles (MVs) play important roles in cell-to-cell communication and transport of several molecules. Such structures are essential components of Extracellular Polymeric Substances (EPS) biofilm matrix of many bacterial species displaying a structural function and a role in virulence and pathogenesis. AREAS COVERED In this review were included original articles from the last ten years by searching the keywords 'biofilm' and 'vesicles' on PUBMED and Scopus databases. The articles available in literature mainly describe a positive correlation between bacterial MVs and biofilms formation. The research on Espacenet and Google Patent databases underlines the available patents related to the application of both biofilm MVs and planktonic MVs in inhibiting biofilm formation. EXPERT OPINION This review covers and analyzes recent advances in the study of the relationship between bacterial vesicles and biofilm. The huge number of papers discussing the role of MVs confirms the interest aimed at developing new applications in the medical field. The study of the MVs composition and biogenesis may contribute to the identification of components which could be (i) the target for the development of new drugs inhibiting the biofilm establishment; (ii) candidates for the development of vaccines; (iii) biomarkers for the diagnosis of bacterial infections.
Collapse
Affiliation(s)
- Valentina Puca
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Beatrice Marinacci
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine & Dentistry, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Benedetta Pellegrini
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Floriana Campanile
- Department of Biomedical and Biotechnological Sciences (BIOMETEC) - Microbiology Section, University of Catania, Catania, Italy
| | - Maria Santagati
- Department of Biomedical and Biotechnological Sciences (BIOMETEC) - Microbiology Section, University of Catania, Catania, Italy
| | - Rossella Grande
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
77
|
Simmons JSC, Bourgault AM, Sole ML, Peach BC. A Review of Chlorhexidine Oral Care in Patients Receiving Mechanical Ventilation. Crit Care Nurse 2024; 44:45-53. [PMID: 38821530 DOI: 10.4037/ccn2024995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2024]
Abstract
BACKGROUND Chlorhexidine gluconate has been considered the criterion standard of oral care for patients receiving mechanical ventilation because of its ability to reduce the incidence of ventilator-associated events. Optimal concentrations and frequencies remain unclear, as do adverse events related to mortality in various intensive care unit populations. OBJECTIVE To examine the current evidence for the efficacy of chlorhexidine gluconate in reducing the incidence of ventilator-associated events, mortality, intensive care unit length of stay, and duration of mechanical ventilation in patients receiving ventilator support. METHODS In this integrative review, CINAHL (Cumulative Index to Nursing and Allied Health Literature), MEDLINE, and Health Source: Nursing/Academic Edition were searched using terms related to mechanical ventilation and chlorhexidine gluconate oral care with dates ranging from 2012 to 2023. RESULTS Seventeen articles were included in this review: 8 systematic reviews, 8 randomized clinical trials (3 of which were not included in any systematic review), and 1 quasi-experimental study. Chlorhexidine gluconate oral care was associated with a reduced incidence of ventilator-associated events, but efficacy depended on concentration and frequency of administration. With stratification by intensive care unit population type, a nonsignificant trend toward increased mortality was found among non-cardiac surgical patients who received this care. CONCLUSION The evidence regarding the efficacy of chlorhexidine gluconate oral care in reducing ventilator-associated events in specific intensive care unit populations is contradictory. Recently published guidelines recommend de-implementation of chlorhexidine gluconate oral care in all patients receiving mechanical ventilation. Such care may be beneficial only in the cardiac surgical population.
Collapse
Affiliation(s)
| | - Annette M Bourgault
- Annette M. Bourgault is an associate professor, University of Central Florida College of Nursing. She is the Editor of Critical Care Nurse journal
| | - Mary Lou Sole
- Mary Lou Sole is the Dean and a professor, University of Central Florida College of Nursing
| | - Brian C Peach
- Brian C. Peach is an assistant professor, University of Central Florida College of Nursing
| |
Collapse
|
78
|
Flores P, Luo J, Mueller DW, Muecklich F, Zea L. Space biofilms - An overview of the morphology of Pseudomonas aeruginosa biofilms grown on silicone and cellulose membranes on board the international space station. Biofilm 2024; 7:100182. [PMID: 38370151 PMCID: PMC10869243 DOI: 10.1016/j.bioflm.2024.100182] [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: 11/20/2023] [Revised: 01/22/2024] [Accepted: 02/04/2024] [Indexed: 02/20/2024] Open
Abstract
Microorganisms' natural ability to live as organized multicellular communities - also known as biofilms - provides them with unique survival advantages. For instance, bacterial biofilms are protected against environmental stresses thanks to their extracellular matrix, which could contribute to persistent infections after treatment with antibiotics. Bacterial biofilms are also capable of strongly attaching to surfaces, where their metabolic by-products could lead to surface material degradation. Furthermore, microgravity can alter biofilm behavior in unexpected ways, making the presence of biofilms in space a risk for both astronauts and spaceflight hardware. Despite the efforts to eliminate microorganism contamination from spacecraft surfaces, it is impossible to prevent human-associated bacteria from eventually establishing biofilm surface colonization. Nevertheless, by understanding the changes that bacterial biofilms undergo in microgravity, it is possible to identify key differences and pathways that could be targeted to significantly reduce biofilm formation. The bacterial component of Space Biofilms project, performed on the International Space Station in early 2020, contributes to such understanding by characterizing the morphology and gene expression of bacterial biofilms formed in microgravity with respect to ground controls. Pseudomonas aeruginosa was used as model organism due to its relevance in biofilm studies and its ability to cause urinary tract infections as an opportunistic pathogen. Biofilm formation was characterized at one, two, and three days of incubation (37 °C) over six different materials. Materials reported in this manuscript include catheter grade silicone, selected due to its medical relevance in hospital acquired infections, catheter grade silicone with ultrashort pulsed direct laser interference patterning, included to test microtopographies as a potential biofilm control strategy, and cellulose membrane to replicate the column and canopy structure previously reported from a microgravity study. We here present an overview of the biofilm morphology, including 3D images of the biofilms to represent the distinctive morphology observed in each material tested, and some of the key differences in biofilm thickness, mass, and surface area coverage. We also present the impact of the surface microtopography in biofilm formation across materials, incubation time, and gravitational conditions. The Space Biofilms project (bacterial side) is supported by the National Aeronautics and Space Administration under Grant No. 80NSSC17K0036 and 80NSSC21K1950.
Collapse
Affiliation(s)
- Pamela Flores
- BioServe Space Technologies, Aerospace Engineering Sciences Department, University of Colorado, 3775 Discovery Drive, Boulder, CO, USA, 80309
| | - Jiaqi Luo
- Saarland University, 66123, Saarbrücken, Saarland, Germany
| | | | | | - Luis Zea
- BioServe Space Technologies, Aerospace Engineering Sciences Department, University of Colorado, 3775 Discovery Drive, Boulder, CO, USA, 80309
| |
Collapse
|
79
|
Bahatheg G, Kuppusamy R, Yasir M, Bridge S, Mishra SK, Cranfield CG, StC Black D, Willcox M, Kumar N. Dimeric peptoids as antibacterial agents. Bioorg Chem 2024; 147:107334. [PMID: 38583251 DOI: 10.1016/j.bioorg.2024.107334] [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: 01/29/2024] [Revised: 03/31/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024]
Abstract
Building upon our previous study on peptoid-based antibacterials which showed good activity against Gram-positive bacteria only, herein we report the synthesis of 34 dimeric peptoid compounds and the investigation of their activity against Gram-positive and Gram-negative pathogens. The newly designed peptoids feature a di-hydrophobic moiety incorporating phenyl, bromo-phenyl, and naphthyl groups, combined with variable lengths of cationic units such as amino and guanidine groups. The study also underscores the pivotal interplay between hydrophobicity and cationicity in optimizing efficacy against specific bacteria. The bromophenyl dimeric guanidinium peptoid compound 10j showed excellent activity against S. aureus 38 and E. coli K12 with MIC of 0.8 μg mL-1 and 6.2 μg mL-1, respectively. Further investigation into the mechanism of action revealed that the antibacterial effect might be attributed to the disruption of bacterial cell membranes, as suggested by tethered bilayer lipid membranes (tBLMs) and cytoplasmic membrane permeability studies. Notably, these promising antibacterial agents exhibited negligible toxicity against mammalian red blood cells. Additionally, the study explored the potential of 12 active compounds to disrupt established biofilms of S. aureus 38. The most effective biofilm disruptors were ethyl and octyl-naphthyl guanidinium peptoids (10c and 10 k). These compounds 10c and 10 k disrupted the established biofilms of S. aureus 38 with 51 % at 4x MIC (MIC = 17.6 μg mL-1 and 11.2 μg mL-1) and 56 %-58 % at 8x MIC (MIC = 35.2 μg mL-1 and 22.4 μg mL-1) respectively. Overall, this research contributes insights into the design principles of cationic dimeric peptoids and their antibacterial activity, with implications for the development of new antibacterial compounds.
Collapse
Affiliation(s)
- Ghayah Bahatheg
- School of Chemistry, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia; Department of Chemistry, Faculty of Science, University of Jeddah, Jeddah 21589, Saudi Arabia
| | - Rajesh Kuppusamy
- School of Chemistry, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia; School of Optometry and Vision Science, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia.
| | - Muhammad Yasir
- School of Optometry and Vision Science, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Samara Bridge
- School of Life Sciences, University of Technology Sydney, PO Box 123, Ultimo 2007, Australia
| | - Shyam K Mishra
- School of Optometry and Vision Science, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Charles G Cranfield
- School of Life Sciences, University of Technology Sydney, PO Box 123, Ultimo 2007, Australia
| | - David StC Black
- School of Chemistry, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Mark Willcox
- School of Optometry and Vision Science, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Naresh Kumar
- School of Chemistry, The University of New South Wales (UNSW), Sydney, NSW 2052, Australia.
| |
Collapse
|
80
|
Beyer BR, Sheppard C, Mullins J, Igbadumhe A. Campylobacter Infection Introduced Following Fecal Microbiota Transplantation. Cureus 2024; 16:e62541. [PMID: 39022481 PMCID: PMC11254095 DOI: 10.7759/cureus.62541] [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] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Fecal microbiota transplantation is an evidence-based therapeutic option for recurrent Clostridium difficile infection, involving the transfer of healthy donor fecal material to restore gut microbial balance. Despite meticulous donor screening, Campylobacter jejuni, a prevalent cause of bacterial gastroenteritis, is not routinely tested, potentially impacting fecal microbiota transplant safety. We present a case of a female with recurrent C. difficile infection treated with fecal microbiota transplantation, complicated by a subsequent C. jejuni infection. The emergence of Campylobacter post fecal microbiota transplantation underscores the importance of comprehensive donor screening protocols. Our case prompts a reevaluation of fecal microbiota transplantation safety measures and advocates for inclusive screening to enhance patient outcomes.
Collapse
Affiliation(s)
- Brian R Beyer
- Medicine, Burrell College of Osteopathic Medicine, Las Cruces, USA
| | - Cody Sheppard
- Medicine, Burrell College of Osteopathic Medicine, Las Cruces, USA
| | - Jordyn Mullins
- Medicine, Burrell College of Osteopathic Medicine, Las Cruces, USA
| | - Anthony Igbadumhe
- Family Medicine, Burrell College of Osteopathic Medicine, Las Cruces, USA
| |
Collapse
|
81
|
Moussa AY. The limitless endophytes: their role as antifungal agents against top priority pathogens. Microb Cell Fact 2024; 23:161. [PMID: 38822407 PMCID: PMC11140875 DOI: 10.1186/s12934-024-02411-3] [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: 03/19/2024] [Accepted: 04/29/2024] [Indexed: 06/03/2024] Open
Abstract
Multi resistant fungi are on the rise, and our arsenal compounds are limited to few choices in the market such as polyenes, pyrimidine analogs, azoles, allylamines, and echinocandins. Although each of these drugs featured a unique mechanism, antifungal resistant strains did emerge and continued to arise against them worldwide. Moreover, the genetic variation between fungi and their host humans is small, which leads to significant challenges in new antifungal drug discovery. Endophytes are still an underexplored source of bioactive secondary metabolites. Many studies were conducted to isolate and screen endophytic pure compounds with efficacy against resistant yeasts and fungi; especially, Candida albicans, C. auris, Cryptococcus neoformans and Aspergillus fumigatus, which encouraged writing this review to critically analyze the chemical nature, potency, and fungal source of the isolated endophytic compounds as well as their novelty features and SAR when possible. Herein, we report a comprehensive list of around 320 assayed antifungal compounds against Candida albicans, C. auris, Cryptococcus neoformans and Aspergillus fumigatus in the period 1980-2024, the majority of which were isolated from fungi of orders Eurotiales and Hypocreales associated with terrestrial plants, probably due to the ease of laboratory cultivation of these strains. 46% of the reviewed compounds were active against C. albicans, 23% against C. neoformans, 29% against A. fumigatus and only 2% against C. auris. Coculturing was proved to be an effective technique to induce cryptic metabolites absent in other axenic cultures or host extract cultures, with Irperide as the most promising compounds MIC value 1 μg/mL. C. auris was susceptible to only persephacin and rubiginosin C. The latter showed potent inhibition against this recalcitrant strain in a non-fungicide way, which unveils the potential of fungal biofilm inhibition. Further development of culturing techniques and activation of silent metabolic pathways would be favorable to inspire the search for novel bioactive antifungals.
Collapse
Affiliation(s)
- Ashaimaa Y Moussa
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, African Union Organization Street, Abbassia, Cairo, 11566, Egypt.
| |
Collapse
|
82
|
Kosmeri C, Giapros V, Serbis A, Balomenou F, Baltogianni M. Antibiofilm Strategies in Neonatal and Pediatric Infections. Antibiotics (Basel) 2024; 13:509. [PMID: 38927176 PMCID: PMC11200539 DOI: 10.3390/antibiotics13060509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Biofilm-related infections pose significant challenges in neonatal and pediatric care, contributing to increased morbidity and mortality rates. These complex microbial communities, comprising bacteria and fungi, exhibit resilience against antibiotics and host immune responses. Bacterial species such as Enterococcus faecalis, Pseudomonas aeruginosa, Staphylococcus aureus, and Staphylococcus epidermidis commonly form biofilms on medical devices, exacerbating infection risks. Neonates and children, particularly those in intensive care units, are highly susceptible to biofilm-associated infections due to the prolonged use of invasive devices, such as central lines and endotracheal tubes. Enteral feeding tubes, crucial for neonatal nutritional support, also serve as potential sites for biofilm formation, contributing to recurrent microbial contamination. Moreover, Candida species, including Candida pelliculosa, present emerging challenges in neonatal care, with multi-drug resistant strains posing treatment complexities. Current antimicrobial therapies, while important in managing infections, often fall short in eradicating biofilms, necessitating alternative strategies. The aim of this review is to summarize current knowledge regarding antibiofilm strategies in neonates and in children. Novel approaches focusing on biofilm inhibition and dispersal show promise, including surface modifications, matrix-degrading enzymes, and quorum-sensing inhibitors. Prudent use of medical devices and exploration of innovative antibiofilm therapies are imperative in mitigating neonatal and pediatric biofilm infections.
Collapse
Affiliation(s)
- Chrysoula Kosmeri
- Department of Pediatrics, University Hospital of Ioannina, 45500 Ioannina, Greece; (C.K.); (A.S.)
| | - Vasileios Giapros
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 45110 Ioannina, Greece; (F.B.); (M.B.)
| | - Anastasios Serbis
- Department of Pediatrics, University Hospital of Ioannina, 45500 Ioannina, Greece; (C.K.); (A.S.)
| | - Foteini Balomenou
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 45110 Ioannina, Greece; (F.B.); (M.B.)
| | - Maria Baltogianni
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 45110 Ioannina, Greece; (F.B.); (M.B.)
| |
Collapse
|
83
|
Vestweber PK, Wächter J, Planz V, Jung N, Windbergs M. The interplay of Pseudomonas aeruginosa and Staphylococcus aureus in dual-species biofilms impacts development, antibiotic resistance and virulence of biofilms in in vitro wound infection models. PLoS One 2024; 19:e0304491. [PMID: 38805522 PMCID: PMC11132468 DOI: 10.1371/journal.pone.0304491] [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: 03/12/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024] Open
Abstract
Due to high tolerance to antibiotics and pronounced virulence, bacterial biofilms are considered a key factor and major clinical challenge in persistent wound infections. They are typically composed of multiple species, whose interactions determine the biofilm's structural development, functional properties and thus the progression of wound infections. However, most attempts to study bacterial biofilms in vitro solely rely on mono-species populations, since cultivating multi-species biofilms, especially for prolonged periods of time, poses significant challenges. To address this, the present study examined the influence of bacterial composition on structural biofilm development, morphology and spatial organization, as well as antibiotic tolerance and virulence on human skin cells in the context of persistent wound infections. By creating a wound-mimetic microenvironment, the successful cultivation of dual-species biofilms of two of the most prevalent wound pathogens, Pseudomonas aeruginosa and Staphylococcus aureus, was realized over a period of 72 h. Combining quantitative analysis with electron microscopy and label-free imaging enabled a comprehensive evaluation of the dynamics of biofilm formation and matrix secretion, revealing a twofold increased maturation of dual-species biofilms. Antibiotic tolerance was comparable for both mono-species cultures, however, dual-species communities showed a 50% increase in tolerance, mediated by a significantly reduced penetration of the applied antibiotic into the biofilm matrix. Further synergistic effects were observed, where dual-species biofilms exacerbated wound healing beyond the effects observed from either Pseudomonas or Staphylococcus. Consequently, predicting biofilm development, antimicrobial tolerance and virulence for multi-species biofilms based solely on the results from mono-species biofilms is unreliable. This study underscores the substantial impact of a multi-species composition on biofilm functional properties and emphasizes the need to tailor future studies reflecting the bacterial composition of the respective in vivo situation, leading to a more comprehensive understanding of microbial communities in the context of basic microbiology and the development of effective treatments.
Collapse
Affiliation(s)
- Pia Katharina Vestweber
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Jana Wächter
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Viktoria Planz
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Nathalie Jung
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Maike Windbergs
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
84
|
Wang S, Zhao S, Jin S, Ye T, Xinling P. Sepsis risk in diabetic patients with urinary tract infection. PLoS One 2024; 19:e0303557. [PMID: 38771840 PMCID: PMC11108167 DOI: 10.1371/journal.pone.0303557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/27/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND Urinary tract infections (UTI) is a prevalent condition in those with diabetes, and in severe cases, it may escalate to sepsis. Therefore, it is important to analyze the risk variables associated with sepsis in diabetes individuals with UTI. METHODS This research was a retrospective cross-sectional analysis. From January 2011 to June 2022, a group of individuals with diabetes were identified as having UTI at a tertiary hospital situated in Southeastern China. Patient data, including information on urine culture, was collected retrospectively from a clinical record database. The participants were categorized into the sepsis and non-sepsis groups. The risk variables were derived using both uni-and multiple- variable regression analysis. RESULTS The research included 1919 patients, of whom 1106 cases (57.63%) had positive urine cultures. In total, 445 blood culture samples were tested, identifying 186 positive cases (41.80%). The prevalence of bacteria in urine and blood samples was highest for Escherichia coli and Klebsiella pneumoniae, respectively. Moreover, 268 individuals (13.97%) exhibited sepsis. The regression analysis indicated a positive correlation between sepsis and albumin (ALB)<34.35 g/L, C-reactive protein (CRP)>55.84 mg/L and white blood cell count (WBC) >8.485 X 109/L in diabetic cases with UTIs. By integrating the three aforementioned parameters, the area under the receiver operating characteristic curve was 0.809. CONCLUSIONS The early detection of sepsis in diabetic individuals with UTI may be achieved using a comprehensive analysis of CRP, WBC, and ALB test findings.
Collapse
Affiliation(s)
- Sipei Wang
- Department of Clinical Laboratory, Wenzhou Medical University Affiliated Dongyang Hospital, Dongyang, Zhejiang, China
| | - Sheng Zhao
- Department of Clinical Laboratory, Wenzhou Medical University Affiliated Dongyang Hospital, Dongyang, Zhejiang, China
| | - Shanshan Jin
- Department of Clinical Laboratory, Wenzhou Medical University Affiliated Dongyang Hospital, Dongyang, Zhejiang, China
| | - Tinghua Ye
- Department of Clinical Laboratory, Wenzhou Medical University Affiliated Dongyang Hospital, Dongyang, Zhejiang, China
| | - Pan Xinling
- Department of Biomedical Sciences Laboratory, Wenzhou Medical University Affiliated Dongyang Hospital, Dongyang, Zhejiang, China
| |
Collapse
|
85
|
Wang B, Du L, Dong B, Kou E, Wang L, Zhu Y. Current Knowledge and Perspectives of Phage Therapy for Combating Refractory Wound Infections. Int J Mol Sci 2024; 25:5465. [PMID: 38791502 PMCID: PMC11122179 DOI: 10.3390/ijms25105465] [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: 03/22/2024] [Revised: 05/09/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Wound infection is one of the most important factors affecting wound healing, so its effective control is critical to promote the process of wound healing. However, with the increasing prevalence of multi-drug-resistant (MDR) bacterial strains, the prevention and treatment of wound infections are now more challenging, imposing heavy medical and financial burdens on patients. Furthermore, the diminishing effectiveness of conventional antimicrobials and the declining research on new antibiotics necessitate the urgent exploration of alternative treatments for wound infections. Recently, phage therapy has been revitalized as a promising strategy to address the challenges posed by bacterial infections in the era of antibiotic resistance. The use of phage therapy in treating infectious diseases has demonstrated positive results. This review provides an overview of the mechanisms, characteristics, and delivery methods of phage therapy for combating pathogenic bacteria. Then, we focus on the clinical application of various phage therapies in managing refractory wound infections, such as diabetic foot infections, as well as traumatic, surgical, and burn wound infections. Additionally, an analysis of the potential obstacles and challenges of phage therapy in clinical practice is presented, along with corresponding strategies for addressing these issues. This review serves to enhance our understanding of phage therapy and provides innovative avenues for addressing refractory infections in wound healing.
Collapse
Affiliation(s)
- Bo Wang
- Department of Dermatology, Naval Medical Center, Naval Medical University, Shanghai 200052, China
| | - Lin Du
- Department of Dermatology, Naval Medical Center, Naval Medical University, Shanghai 200052, China
| | - Baiping Dong
- Department of Dermatology, Naval Medical Center, Naval Medical University, Shanghai 200052, China
| | - Erwen Kou
- Department of Dermatology, Naval Medical Center, Naval Medical University, Shanghai 200052, China
| | - Liangzhe Wang
- Department of Dermatology, Naval Medical Center, Naval Medical University, Shanghai 200052, China
| | - Yuanjie Zhu
- Department of Dermatology, Naval Medical Center, Naval Medical University, Shanghai 200052, China
| |
Collapse
|
86
|
Wongchai M, Wongkaewkhiaw S, Kanthawong S, Roytrakul S, Aunpad R. Dual-function antimicrobial-antibiofilm peptide hybrid to tackle biofilm-forming Staphylococcus epidermidis. Ann Clin Microbiol Antimicrob 2024; 23:44. [PMID: 38755634 PMCID: PMC11100219 DOI: 10.1186/s12941-024-00701-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/28/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Due to their resistance and difficulty in treatment, biofilm-associated infections are problematic among hospitalized patients globally and account for 60% of all bacterial infections in humans. Antibiofilm peptides have recently emerged as an alternative treatment since they can be effectively designed and exert a different mode of biofilm inhibition and eradication. METHODS A novel antibiofilm peptide, BiF, was designed from the conserved sequence of 18 α-helical antibiofilm peptides by template-assisted technique and its activity was improved by hybridization with a lipid binding motif (KILRR). Novel antibiofilm peptide derivatives were modified by substituting hydrophobic amino acids at positions 5 or 7, and both, with positively charged lysines (L5K, L7K). These peptide derivatives were tested for antibiofilm and antimicrobial activities against biofilm-forming Staphylococcus epidermidis and multiple other microbes using crystal violet and broth microdilution assays, respectively. To assess their impact on mammalian cells, the toxicity of peptides was determined through hemolysis and cytotoxicity assays. The stability of candidate peptide, BiF2_5K7K, was assessed in human serum and its secondary structure in bacterial membrane-like environments was analyzed using circular dichroism. The action of BiF2_5K7K on planktonic S. epidermidis and its effect on biofilm cell viability were assessed via viable counting assays. Its biofilm inhibition mechanism was investigated through confocal laser scanning microscopy and transcription analysis. Additionally, its ability to eradicate mature biofilms was examined using colony counting. Finally, a preliminary evaluation involved coating a catheter with BiF2_5K7K to assess its preventive efficacy against S. epidermidis biofilm formation on the catheter and its surrounding area. RESULTS BiF2_5K7K, the modified antibiofilm peptide, exhibited dose-dependent antibiofilm activity against S. epidermidis. It inhibited biofilm formation at subinhibitory concentrations by altering S. epidermidis extracellular polysaccharide production and quorum-sensing gene expression. Additionally, it exhibited broad-spectrum antimicrobial activity and no significant hemolysis or toxicity against mammalian cell lines was observed. Its activity is retained when exposed to human serum. In bacterial membrane-like environments, this peptide formed an α-helix amphipathic structure. Within 4 h, a reduction in the number of S. epidermidis colonies was observed, demonstrating the fast action of this peptide. As a preliminary test, a BiF2_5K7K-coated catheter was able to prevent the development of S. epidermidis biofilm both on the catheter surface and in its surrounding area. CONCLUSIONS Due to the safety and effectiveness of BiF2_5K7K, we suggest that this peptide be further developed to combat biofilm infections, particularly those of biofilm-forming S. epidermidis.
Collapse
Affiliation(s)
- Mathira Wongchai
- Graduate Program in Biomedical Sciences, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, Thailand
| | - Saharut Wongkaewkhiaw
- School of Dentistry, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand
| | - Sakawrat Kanthawong
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Ratchaneewan Aunpad
- Graduate Program in Biomedical Sciences, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, Thailand.
| |
Collapse
|
87
|
Aonofriesei F. Surfactants' Interplay with Biofilm Development in Staphylococcus and Candida. Pharmaceutics 2024; 16:657. [PMID: 38794319 PMCID: PMC11125353 DOI: 10.3390/pharmaceutics16050657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/09/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
The capacity of micro-organisms to form biofilms is a pervasive trait in the microbial realm. For pathogens, biofilm formation serves as a virulence factor facilitating successful host colonization. Simultaneously, infections stemming from biofilm-forming micro-organisms pose significant treatment challenges due to their heightened resistance to antimicrobial agents. Hence, the quest for active compounds capable of impeding microbial biofilm development stands as a pivotal pursuit in biomedical research. This study presents findings concerning the impact of three surfactants, namely, polysorbate 20 (T20), polysorbate 80 (T80), and sodium dodecyl sulfate (SDS), on the initial stage of biofilm development in both Staphylococcus aureus and Candida dubliniensis. In contrast to previous investigations, we conducted a comparative assessment of the biofilm development capacity of these two taxonomically distant groups, predicated on their shared ability to reduce TTC. The common metabolic trait shared by S. aureus and C. dubliniensis in reducing TTC to formazan facilitated a simultaneous evaluation of biofilm development under the influence of surfactants across both groups. Our results revealed that surfactants could impede the development of biofilms in both species by disrupting the initial cell attachment step. The observed effect was contingent upon the concentration and type of compound, with a higher inhibition observed in culture media supplemented with SDS. At maximum concentrations (5%), T20 and T80 significantly curtailed the formation and viability of S. aureus and C. dubliniensis biofilms. Specifically, T20 inhibited biofilm development by 75.36% in S. aureus and 71.18% in C. dubliniensis, while T80 exhibited a slightly lower inhibitory effect, with values ranging between 66.68% (C. dubliniensis) and 65.54% (S. aureus) compared to the controls. Incorporating these two non-toxic surfactants into pharmaceutical formulations could potentially enhance the inhibitory efficacy of selected antimicrobial agents, particularly in external topical applications.
Collapse
Affiliation(s)
- Florin Aonofriesei
- Department of Natural Sciences, Faculty of Natural and Agricultural Sciences, Ovidius University of Constanta, 1, University Street, 900470 Constanța, Romania
| |
Collapse
|
88
|
Qin L, Yang S, Zhao C, Yang J, Li F, Xu Z, Yang Y, Zhou H, Li K, Xiong C, Huang W, Hu N, Hu X. Prospects and challenges for the application of tissue engineering technologies in the treatment of bone infections. Bone Res 2024; 12:28. [PMID: 38744863 PMCID: PMC11094017 DOI: 10.1038/s41413-024-00332-w] [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: 12/06/2023] [Revised: 03/08/2024] [Accepted: 04/01/2024] [Indexed: 05/16/2024] Open
Abstract
Osteomyelitis is a devastating disease caused by microbial infection in deep bone tissue. Its high recurrence rate and impaired restoration of bone deficiencies are major challenges in treatment. Microbes have evolved numerous mechanisms to effectively evade host intrinsic and adaptive immune attacks to persistently localize in the host, such as drug-resistant bacteria, biofilms, persister cells, intracellular bacteria, and small colony variants (SCVs). Moreover, microbial-mediated dysregulation of the bone immune microenvironment impedes the bone regeneration process, leading to impaired bone defect repair. Despite advances in surgical strategies and drug applications for the treatment of bone infections within the last decade, challenges remain in clinical management. The development and application of tissue engineering materials have provided new strategies for the treatment of bone infections, but a comprehensive review of their research progress is lacking. This review discusses the critical pathogenic mechanisms of microbes in the skeletal system and their immunomodulatory effects on bone regeneration, and highlights the prospects and challenges for the application of tissue engineering technologies in the treatment of bone infections. It will inform the development and translation of antimicrobial and bone repair tissue engineering materials for the management of bone infections.
Collapse
Affiliation(s)
- Leilei Qin
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Shuhao Yang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Chen Zhao
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Jianye Yang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Feilong Li
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Zhenghao Xu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Yaji Yang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Haotian Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Kainan Li
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan, 610081, China
| | - Chengdong Xiong
- University of Chinese Academy of Sciences, Bei Jing, 101408, China
| | - Wei Huang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Ning Hu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China.
| | - Xulin Hu
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan, 610081, China.
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
89
|
Perasoli FB, B Silva LS, C Figueiredo BI, Pinto IC, F Amaro LJ, S Almeida Bastos JC, Carneiro SP, R Araújo VP, G Beato FR, M Barboza AP, M Teixeira LF, Gallagher MP, Bradley M, Venkateswaran S, H dos Santos OD. Poly(methylmethacrylate-co-dimethyl acrylamide)-silver nanocomposite prevents biofilm formation in medical devices. Nanomedicine (Lond) 2024; 19:1285-1296. [PMID: 38722243 PMCID: PMC11285241 DOI: 10.1080/17435889.2024.2345044] [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: 02/02/2024] [Accepted: 04/16/2024] [Indexed: 07/25/2024] Open
Abstract
Aim: To investigate whether medical devices coated with a synthesized nanocomposite of poly(methylmethacrylate-co-dimethyl acrylamide) (PMMDMA) and silver nanoparticles (AgNPs) could improve their antibiofilm and antimicrobial activities. We also investigated the nanocomposite's safety. Materials & methods: The nanocomposite was synthesized and characterized using analytical techniques. Medical devices coated with the nanocomposite were evaluated for bacterial adhesion and hemolytic activity in vitro. Results: The nanocomposite formation was demonstrated with the incorporation of AgNPs into the polymer matrix. The nanocomposite proved to be nonhemolytic and significantly inhibited bacterial biofilm formation. Conclusion: The PMMDMA-AgNPs nanocomposite was more effective in preventing biofilm formation than PMMDMA alone and is a promising strategy for coating medical devices and reducing mortality due to hospital-acquired infections.
Collapse
Affiliation(s)
- Fernanda B Perasoli
- Laboratório de Fitotecnologia, Departamento de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Luan S B Silva
- Laboratório de Fitotecnologia, Departamento de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Bruna I C Figueiredo
- Laboratório de Fitotecnologia, Departamento de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Isabelle C Pinto
- Laboratório de Fitotecnologia, Departamento de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Lorrane J F Amaro
- Laboratório de Fitotecnologia, Departamento de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Juliana C S Almeida Bastos
- Laboratório de Fitotecnologia, Departamento de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Simone P Carneiro
- Laboratório de Fitotecnologia, Departamento de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Vânia P R Araújo
- Nano Lab, Departamento de Engenharia Metalúrgica e de Materiais, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Felipe R G Beato
- Laboratório de Microscopia, Departamento de Física, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Ana P M Barboza
- Laboratório de Microscopia, Departamento de Física, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Luiz F M Teixeira
- Laboratório de Fitotecnologia, Departamento de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Maurice P Gallagher
- School of Biological Sciences, University of Edinburgh, King's Buildings, David Brewster Road, Edinburgh, EH9 3FJ, UK
| | - Mark Bradley
- Precision Healthcare University Research Institute, Queen Mary University of London, Empire House, London, E1 1HH, UK
| | - Seshasailam Venkateswaran
- Precision Healthcare University Research Institute, Queen Mary University of London, Empire House, London, E1 1HH, UK
| | - Orlando D H dos Santos
- Laboratório de Fitotecnologia, Departamento de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| |
Collapse
|
90
|
Bouhrour N, Nibbering PH, Bendali F. Medical Device-Associated Biofilm Infections and Multidrug-Resistant Pathogens. Pathogens 2024; 13:393. [PMID: 38787246 PMCID: PMC11124157 DOI: 10.3390/pathogens13050393] [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: 03/27/2024] [Revised: 04/29/2024] [Accepted: 05/04/2024] [Indexed: 05/25/2024] Open
Abstract
Medical devices such as venous catheters (VCs) and urinary catheters (UCs) are widely used in the hospital setting. However, the implantation of these devices is often accompanied by complications. About 60 to 70% of nosocomial infections (NIs) are linked to biofilms. The main complication is the ability of microorganisms to adhere to surfaces and form biofilms which protect them and help them to persist in the host. Indeed, by crossing the skin barrier, the insertion of VC inevitably allows skin flora or accidental environmental contaminants to access the underlying tissues and cause fatal complications like bloodstream infections (BSIs). In fact, 80,000 central venous catheters-BSIs (CVC-BSIs)-mainly occur in intensive care units (ICUs) with a death rate of 12 to 25%. Similarly, catheter-associated urinary tract infections (CA-UTIs) are the most commonlyhospital-acquired infections (HAIs) worldwide.These infections represent up to 40% of NIs.In this review, we present a summary of biofilm formation steps. We provide an overview of two main and important infections in clinical settings linked to medical devices, namely the catheter-asociated bloodstream infections (CA-BSIs) and catheter-associated urinary tract infections (CA-UTIs), and highlight also the most multidrug resistant bacteria implicated in these infections. Furthermore, we draw attention toseveral useful prevention strategies, and advanced antimicrobial and antifouling approaches developed to reduce bacterial colonization on catheter surfaces and the incidence of the catheter-related infections.
Collapse
Affiliation(s)
- Nesrine Bouhrour
- Laboratoire de Microbiologie Appliquée, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia 06000, Algeria;
| | - Peter H. Nibbering
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Farida Bendali
- Laboratoire de Microbiologie Appliquée, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia 06000, Algeria;
| |
Collapse
|
91
|
Zhu R, Stone T, Wang Y. The role of shear rates on amyloid formation from biofilm peptide phenol-soluble modulins. Biophys J 2024; 123:1106-1115. [PMID: 38549371 PMCID: PMC11079943 DOI: 10.1016/j.bpj.2024.03.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 01/28/2024] [Accepted: 03/26/2024] [Indexed: 04/15/2024] Open
Abstract
Biofilms, microbial communities enclosed in the self-produced extracellular matrix, have a significant impact on human health, environment, and industry. The pathogen Staphylococcus aureus (S. aureus) is recognized as one of the most frequent causes of biofilm-related infections. Phenol-soluble modulins (PSMs) serve as a crucial component, fortifying S. aureus biofilm matrix through self-assembly into amyloid fibrils, which enhances S. aureus colonization and resistance to antibiotics. However, the role of shear rate, one of the critical physiological factors within blood vessels, on the formation of PSM amyloids remains poorly understood. In this work, using a combination of thioflavin T fluorescence kinetic studies, circular dichroism spectrometry, and electron microscopy, we demonstrated that shear rates ranging from 150 to 300 s-1 accelerate fibrillation of PSMα1, α3, and α4 into amyloids, resulting in elongated amyloid structures. Furthermore, PSMα1, α3, and α4 predominantly self-assembled into amyloid fibers with a cross-α structure under shear conditions, deviating from the typical β-sheet configuration of PSM amyloids. These findings imply the role of shear rates within the bloodstream on enhancing PSM self-assembly that is associated with S. aureus biofilm formation.
Collapse
Affiliation(s)
- Runyao Zhu
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Trevor Stone
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana; Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia
| | - Yichun Wang
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana.
| |
Collapse
|
92
|
Di Pietro M, Filardo S, Mattioli R, Bozzuto G, Raponi G, Mosca L, Sessa R. Anti-Biofilm Activity of Oleacein and Oleocanthal from Extra-Virgin Olive Oil toward Pseudomonas aeruginosa. Int J Mol Sci 2024; 25:5051. [PMID: 38732269 PMCID: PMC11084407 DOI: 10.3390/ijms25095051] [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: 03/19/2024] [Revised: 04/24/2024] [Accepted: 05/04/2024] [Indexed: 05/13/2024] Open
Abstract
New antimicrobial molecules effective against Pseudomonas aeruginosa, known as an antibiotic-resistant "high-priority pathogen", are urgently required because of its ability to develop biofilms related to healthcare-acquired infections. In this study, for the first time, the anti-biofilm and anti-virulence activities of a polyphenolic extract of extra-virgin olive oil as well as purified oleocanthal and oleacein, toward P. aeruginosa clinical isolates were investigated. The main result of our study was the anti-virulence activity of the mixture of oleacein and oleocanthal toward multidrug-resistant and intermediately resistant strains of P. aeruginosa isolated from patients with ventilator-associated pneumonia or surgical site infection. Specifically, the mixture of oleacein (2.5 mM)/oleocanthal (2.5 mM) significantly inhibited biofilm formation, alginate and pyocyanin production, and motility in both P. aeruginosa strains (p < 0.05); scanning electron microscopy analysis further evidenced its ability to inhibit bacterial cell adhesion as well as the production of the extracellular matrix. In conclusion, our results suggest the potential application of the oleacein/oleocanthal mixture in the management of healthcare-associated P. aeruginosa infections, particularly in the era of increasing antimicrobial resistance.
Collapse
Affiliation(s)
- Marisa Di Pietro
- Department of Public Health and Infectious Diseases, “Sapienza” University, p.le Aldo Moro, 5, 00185 Rome, Italy; (M.D.P.); (S.F.); (G.R.)
| | - Simone Filardo
- Department of Public Health and Infectious Diseases, “Sapienza” University, p.le Aldo Moro, 5, 00185 Rome, Italy; (M.D.P.); (S.F.); (G.R.)
| | - Roberto Mattioli
- Department of Biochemical Sciences, Faculty of Pharmacy and Medicine, “Sapienza” University, p.le Aldo Moro, 5, 00185 Rome, Italy; (R.M.); (L.M.)
| | - Giuseppina Bozzuto
- National Centre for Drug Research and Evaluation, Italian National Institute of Health, 00161 Rome, Italy;
| | - Giammarco Raponi
- Department of Public Health and Infectious Diseases, “Sapienza” University, p.le Aldo Moro, 5, 00185 Rome, Italy; (M.D.P.); (S.F.); (G.R.)
| | - Luciana Mosca
- Department of Biochemical Sciences, Faculty of Pharmacy and Medicine, “Sapienza” University, p.le Aldo Moro, 5, 00185 Rome, Italy; (R.M.); (L.M.)
| | - Rosa Sessa
- Department of Public Health and Infectious Diseases, “Sapienza” University, p.le Aldo Moro, 5, 00185 Rome, Italy; (M.D.P.); (S.F.); (G.R.)
| |
Collapse
|
93
|
Jarman E, Burgess J, Sharma A, Hayashigatani K, Singh A, Fox P. Human-Derived collagen hydrogel as an antibiotic vehicle for topical treatment of bacterial biofilms. PLoS One 2024; 19:e0303039. [PMID: 38701045 PMCID: PMC11068178 DOI: 10.1371/journal.pone.0303039] [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: 02/11/2024] [Accepted: 04/13/2024] [Indexed: 05/05/2024] Open
Abstract
The complexity of chronic wounds creates difficulty in effective treatments, leading to prolonged care and significant morbidity. Additionally, these wounds are incredibly prone to bacterial biofilm development, further complicating treatment. The current standard treatment of colonized superficial wounds, debridement with intermittent systemic antibiotics, can lead to systemic side-effects and often fails to directly target the bacterial biofilm. Furthermore, standard of care dressings do not directly provide adequate antimicrobial properties. This study aims to assess the capacity of human-derived collagen hydrogel to provide sustained antibiotic release to disrupt bacterial biofilms and decrease bacterial load while maintaining host cell viability and scaffold integrity. Human collagen harvested from flexor tendons underwent processing to yield a gellable liquid, and subsequently was combined with varying concentrations of gentamicin (50-500 mg/L) or clindamycin (10-100 mg/L). The elution kinetics of antibiotics from the hydrogel were analyzed using liquid chromatography-mass spectrometry. The gel was used to topically treat Methicillin-resistant Staphylococcus aureus (MRSA) and Clostridium perfringens in established Kirby-Bauer and Crystal Violet models to assess the efficacy of bacterial inhibition. 2D mammalian cell monolayers were topically treated, and cell death was quantified to assess cytotoxicity. Bacteria-enhanced in vitro scratch assays were treated with antibiotic-embedded hydrogel and imaged over time to assess cell death and mobility. Collagen hydrogel embedded with antibiotics (cHG+abx) demonstrated sustained antibiotic release for up to 48 hours with successful inhibition of both MRSA and C. perfringens biofilms, while remaining bioactive up to 72 hours. Administration of cHG+abx with antibiotic concentrations up to 100X minimum inhibitory concentration was found to be non-toxic and facilitated mammalian cell migration in an in vitro scratch model. Collagen hydrogel is a promising pharmaceutical delivery vehicle that allows for safe, precise bacterial targeting for effective bacterial inhibition in a pro-regenerative scaffold.
Collapse
Affiliation(s)
- Evan Jarman
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Division of Plastic & Reconstructive Surgery, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Jordan Burgess
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Division of Plastic & Reconstructive Surgery, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Ayushi Sharma
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Division of Plastic & Reconstructive Surgery, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Kate Hayashigatani
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Division of Plastic & Reconstructive Surgery, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Amar Singh
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Division of Plastic & Reconstructive Surgery, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Paige Fox
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Division of Plastic & Reconstructive Surgery, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| |
Collapse
|
94
|
Tang Y, Zhang Z, Tao C, Wang X. The mechanism of biofilm detachment in porous medium under flow field. BIOMICROFLUIDICS 2024; 18:034103. [PMID: 38737754 PMCID: PMC11080962 DOI: 10.1063/5.0203061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/25/2024] [Indexed: 05/14/2024]
Abstract
Biofilms are communities formed by bacteria adhering to surfaces, widely present in porous medium, and their growth can lead to clogging. Our experiment finds that under certain flow conditions, biofilms detach in pores and form a dynamically changing flow path. We define detachment that occurs far from the boundary of the flow path (with a distance greater than 200 μm) as internal detachment and detachment that occurs at the boundary of the flow path as external detachment. To understand the mechanism of biofilm detachment, we study the detachment behaviors of the Bacillus subtilis biofilm in a porous medium in a microfluidic device, where Bacillus subtilis strain is triple fluorescent labeled, which can represent three main phenotypes during the biofilm formation: motile cells, matrix-producing cells, and spores. We find that slow small-scale internal detachment occurs in regions with very few motile cells and matrix-producing cells, and bacterial movement in these areas is disordered. The increase in the number of matrix-producing cells induces clogging, and after clogging, the rapid detachment of the bulk internal biofilm occurs due to the increased pressure difference at the inlet and outlet. When both internal and external detachments occur simultaneously, the number of matrix-producing cells in the internal detachment area is 2.5 times that in the external detachment area. The results indicate that biofilm detachment occurs in areas with fewer matrix-producing cells, as matrix-producing cells can help resist detachment by secreting extracellular polymeric substances (EPSs).
Collapse
Affiliation(s)
- Yangyang Tang
- School of Mechanical Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Zheng Zhang
- School of Mechanical Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Cong Tao
- School of Mechanical Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | | |
Collapse
|
95
|
Decollogny M, Rottenberg S. Persisting cancer cells are different from bacterial persisters. Trends Cancer 2024; 10:393-406. [PMID: 38429144 DOI: 10.1016/j.trecan.2024.02.002] [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: 10/20/2023] [Revised: 01/22/2024] [Accepted: 02/02/2024] [Indexed: 03/03/2024]
Abstract
The persistence of drug-sensitive tumors poses a significant challenge in cancer treatment. The concept of bacterial persisters, which are a subpopulation of bacteria that survive lethal antibiotic doses, is frequently used to compare to residual disease in cancer. Here, we explore drug tolerance of cancer cells and bacteria. We highlight the fact that bacteria, in contrast to cancer cells, have been selected for survival at the population level and may therefore possess contingency mechanisms that cancer cells lack. The precise mechanisms of drug-tolerant cancer cells and bacterial persisters are still being investigated. Undoubtedly, by understanding common features as well as differences, we, in the cancer field, can learn from microbiology to find strategies to eradicate persisting cancer cells.
Collapse
Affiliation(s)
- Morgane Decollogny
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland; Bern Center for Precision Medicine and Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Sven Rottenberg
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland; Bern Center for Precision Medicine and Department for BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
96
|
Khan MAS, Islam Z, Shah ST, Rahman SR. Characterization of biofilm formation and multi-drug resistance among Pseudomonas aeruginosa isolated from hospital wastewater in Dhaka, Bangladesh. JOURNAL OF WATER AND HEALTH 2024; 22:825-834. [PMID: 38822462 DOI: 10.2166/wh.2024.294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 04/09/2024] [Indexed: 06/03/2024]
Abstract
Hospital wastewater has been identified as a hotspot for the emergence and transmission of multidrug-resistant (MDR) pathogens that present a serious threat to public health. Therefore, we investigated the current status of antibiotic resistance as well as the phenotypic and genotypic basis of biofilm formation in Pseudomonas aeruginosa from hospital wastewater in Dhaka, Bangladesh. The disc diffusion method and the crystal violet assay were performed to characterize antimicrobial resistance and biofilm formation, respectively. Biofilm and integron-associated genes were amplified by the polymerase chain reaction. Isolates exhibited varying degrees of resistance to different antibiotics, in which >80% of isolates showed sensitivity to meropenem, amikacin, and gentamicin. The results indicated that 93.82% of isolates were MDR and 71 out of 76 MDR isolates showed biofilm formation activities. We observed the high prevalence of biofilm-related genes, in which algD+pelF+pslD+ (82.7%) was found to be the prevalent biofilm genotypic pattern. Sixteen isolates (19.75%) possessed class 1 integron (int1) genes. However, statistical analysis revealed no significant association between biofilm formation and multidrug resistance (χ2 = 0.35, P = 0.55). Taken together, hospital wastewater in Dhaka city may act as a reservoir for MDR and biofilm-forming P. aeruginosa, and therefore, the adequate treatment of wastewater is recommended to reduce the occurrence of outbreaks.
Collapse
Affiliation(s)
- Md Abu Sayem Khan
- Department of Microbiology, University of Dhaka, Dhaka 1000, Bangladesh; Md Abu Sayem Khan and Zahidul Islam contributed equally to this study
| | - Zahidul Islam
- Department of Microbiology, University of Dhaka, Dhaka 1000, Bangladesh; Md Abu Sayem Khan and Zahidul Islam contributed equally to this study
| | - Sm Tanjil Shah
- Department of Microbiology, University of Dhaka, Dhaka 1000, Bangladesh
| | | |
Collapse
|
97
|
Castagnini D, Palma K, Jara-Wilde J, Navarro N, González MJ, Toledo J, Canales-Huerta N, Scavone P, Härtel S. Proteus mirabilis biofilm expansion microscopy yields over 4-fold magnification for super-resolution of biofilm structure and subcellular DNA organization. J Microbiol Methods 2024; 220:106927. [PMID: 38561125 DOI: 10.1016/j.mimet.2024.106927] [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: 02/09/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
Bacterial biofilms form when bacteria attach to surfaces and generate an extracellular matrix that embeds and stabilizes a growing community. Detailed visualization and quantitative analysis of biofilm architecture by optical microscopy are limited by the law of diffraction. Expansion Microscopy (ExM) is a novel Super-Resolution technique where specimens are physically enlarged by a factor of ∼4, prior to observation by conventional fluorescence microscopy. ExM requires homogenization of rigid constituents of biological components by enzymatic digestion. We developed an ExM approach capable of expanding 48-h old Proteus mirabilis biofilms 4.3-fold (termed PmbExM), close to the theoretic maximum expansion factor without gross shape distortions. Our protocol, based on lytic and glycoside-hydrolase enzymatic treatments, degrades rigid components in bacteria and extracellular matrix. Our results prove PmbExM to be a versatile and easy-to-use Super-Resolution approach for enabling studies of P. mirabilis biofilm architecture, assembly, and even intracellular features, such as DNA organization.
Collapse
Affiliation(s)
- Dante Castagnini
- Laboratory for Scientific Image Analysis SCIAN-Lab, Integrative Biology Program, Institute of Biomedical Sciences ICBM, Faculty of Medicine, University of Chile, Santiago, Chile; Biomedical Neuroscience Institute BNI, Independencia, Santiago, Chile
| | - Karina Palma
- Laboratory for Scientific Image Analysis SCIAN-Lab, Integrative Biology Program, Institute of Biomedical Sciences ICBM, Faculty of Medicine, University of Chile, Santiago, Chile; Biomedical Neuroscience Institute BNI, Independencia, Santiago, Chile; Centro de Informática Médica y Telemedicina CIMT, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Jorge Jara-Wilde
- Laboratory for Scientific Image Analysis SCIAN-Lab, Integrative Biology Program, Institute of Biomedical Sciences ICBM, Faculty of Medicine, University of Chile, Santiago, Chile; Biomedical Neuroscience Institute BNI, Independencia, Santiago, Chile; Centro de Informática Médica y Telemedicina CIMT, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Nicolás Navarro
- Advanced Center for Chronic Diseases ACCDiS, Santiago, Chile.; Laboratorio de Biofilms Microbianos, Departamento de Microbiología, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - María José González
- Laboratorio de Biofilms Microbianos, Departamento de Microbiología, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Jorge Toledo
- Red de Equipamiento Científico Avanzado REDECA, Institute of Biomedical Sciences ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Nicole Canales-Huerta
- Laboratory for Scientific Image Analysis SCIAN-Lab, Integrative Biology Program, Institute of Biomedical Sciences ICBM, Faculty of Medicine, University of Chile, Santiago, Chile; Biomedical Neuroscience Institute BNI, Independencia, Santiago, Chile
| | - Paola Scavone
- Laboratorio de Biofilms Microbianos, Departamento de Microbiología, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Steffen Härtel
- Laboratory for Scientific Image Analysis SCIAN-Lab, Integrative Biology Program, Institute of Biomedical Sciences ICBM, Faculty of Medicine, University of Chile, Santiago, Chile; Biomedical Neuroscience Institute BNI, Independencia, Santiago, Chile; Centro de Informática Médica y Telemedicina CIMT, Faculty of Medicine, University of Chile, Santiago, Chile; National Center for Health Information Systems CENS, Santiago, Chile.; Red de Equipamiento Científico Avanzado REDECA, Institute of Biomedical Sciences ICBM, Faculty of Medicine, University of Chile, Santiago, Chile; Centro de Modelamiento Matemático, Universidad de Chile, Beauchef 851, Casilla 170-3, Santiago, Chile.
| |
Collapse
|
98
|
Hajfathalian M, Mossburg KJ, Radaic A, Woo KE, Jonnalagadda P, Kapila Y, Bollyky PL, Cormode DP. A review of recent advances in the use of complex metal nanostructures for biomedical applications from diagnosis to treatment. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1959. [PMID: 38711134 PMCID: PMC11114100 DOI: 10.1002/wnan.1959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/31/2024] [Accepted: 04/01/2024] [Indexed: 05/08/2024]
Abstract
Complex metal nanostructures represent an exceptional category of materials characterized by distinct morphologies and physicochemical properties. Nanostructures with shape anisotropies, such as nanorods, nanostars, nanocages, and nanoprisms, are particularly appealing due to their tunable surface plasmon resonances, controllable surface chemistries, and effective targeting capabilities. These complex nanostructures can absorb light in the near-infrared, enabling noteworthy applications in nanomedicine, molecular imaging, and biology. The engineering of targeting abilities through surface modifications involving ligands, antibodies, peptides, and other agents potentiates their effects. Recent years have witnessed the development of innovative structures with diverse compositions, expanding their applications in biomedicine. These applications encompass targeted imaging, surface-enhanced Raman spectroscopy, near-infrared II imaging, catalytic therapy, photothermal therapy, and cancer treatment. This review seeks to provide the nanomedicine community with a thorough and informative overview of the evolving landscape of complex metal nanoparticle research, with a specific emphasis on their roles in imaging, cancer therapy, infectious diseases, and biofilm treatment. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Diagnostic Tools > Diagnostic Nanodevices.
Collapse
Affiliation(s)
- Maryam Hajfathalian
- Department of Biomedical Engineering, New Jersey Institute of Technology, University Heights, Newark, NJ 07102
- Division of Infectious Diseases, School of Medicine, Stanford University, Stanford, CA 94305
| | - Katherine J. Mossburg
- Department of Radiology, University of Pennsylvania, 3400 Spruce Street, 1 Silverstein, Philadelphia, Pennsylvania 19104, United States
| | - Allan Radaic
- School of Dentistry, University of California Los Angeles
| | - Katherine E. Woo
- Division of Infectious Diseases, School of Medicine, Stanford University, Stanford, CA 94305
| | - Pallavi Jonnalagadda
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Yvonne Kapila
- School of Dentistry, University of California Los Angeles
| | - Paul L. Bollyky
- Division of Infectious Diseases, Department of Medicine, Stanford University
| | - David P. Cormode
- Department of Radiology, Department of Bioengineering, University of Pennsylvania
| |
Collapse
|
99
|
Bereanu AS, Bereanu R, Mohor C, Vintilă BI, Codru IR, Olteanu C, Sava M. Prevalence of Infections and Antimicrobial Resistance of ESKAPE Group Bacteria Isolated from Patients Admitted to the Intensive Care Unit of a County Emergency Hospital in Romania. Antibiotics (Basel) 2024; 13:400. [PMID: 38786129 PMCID: PMC11117271 DOI: 10.3390/antibiotics13050400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
The ESKAPE group (Enterococcus faecium, Staphylococcus aureus, Klebsiella Pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacter spp.) is a group of bacteria very difficult to treat due to their high ability to acquire resistance to antibiotics and are the main cause of nosocomial infections worldwide, posing a threat to global public health. Nosocomial infections with MDR bacteria are found mainly in Intensive Care Units, due to the multitude of maneuvers and invasive medical devices used, the prolonged antibiotic treatments, the serious general condition of these critical patients, and the prolonged duration of hospitalization. MATERIALS AND METHODS During a period of one year, from January 2023 to December 2023, this cross-sectional study was conducted on patients diagnosed with sepsis admitted to the Intensive Care Unit of the Sibiu County Emergency Clinical Hospital. Samples taken were tracheal aspirate, catheter tip, pharyngeal exudate, wound secretion, urine culture, blood culture, and peritoneal fluid. RESULTS The most common bacteria isolated from patients admitted to our Intensive Care Unit was Klebsiella pneumoniae, followed by Acinetobacter baumanii and Pseudomonas aeruginosa. Gram-positive cocci (Enterococcus faecium and Staphilococcus aureus) were rarely isolated. Most of the bacteria isolated were MDR bacteria. CONCLUSIONS The rise of antibiotic and antimicrobial resistance among strains in the nosocomial environment and especially in Intensive Care Units raises serious concerns about limited treatment options.
Collapse
Affiliation(s)
- Alina-Simona Bereanu
- Faculty of Medicine, Lucian Blaga University of Sibiu, Lucian Blaga Street 2A, 550169 Sibiu, Romania; (A.-S.B.); (B.I.V.); (I.R.C.); (M.S.)
- County Clinical Emergency Hospital, Bld. Corneliu Coposu, nr. 2-4, 550245 Sibiu, Romania;
| | - Rareș Bereanu
- Faculty of Medicine, Lucian Blaga University of Sibiu, Lucian Blaga Street 2A, 550169 Sibiu, Romania; (A.-S.B.); (B.I.V.); (I.R.C.); (M.S.)
| | - Cosmin Mohor
- Faculty of Medicine, Lucian Blaga University of Sibiu, Lucian Blaga Street 2A, 550169 Sibiu, Romania; (A.-S.B.); (B.I.V.); (I.R.C.); (M.S.)
- County Clinical Emergency Hospital, Bld. Corneliu Coposu, nr. 2-4, 550245 Sibiu, Romania;
| | - Bogdan Ioan Vintilă
- Faculty of Medicine, Lucian Blaga University of Sibiu, Lucian Blaga Street 2A, 550169 Sibiu, Romania; (A.-S.B.); (B.I.V.); (I.R.C.); (M.S.)
- County Clinical Emergency Hospital, Bld. Corneliu Coposu, nr. 2-4, 550245 Sibiu, Romania;
| | - Ioana Roxana Codru
- Faculty of Medicine, Lucian Blaga University of Sibiu, Lucian Blaga Street 2A, 550169 Sibiu, Romania; (A.-S.B.); (B.I.V.); (I.R.C.); (M.S.)
- County Clinical Emergency Hospital, Bld. Corneliu Coposu, nr. 2-4, 550245 Sibiu, Romania;
| | - Ciprian Olteanu
- County Clinical Emergency Hospital, Bld. Corneliu Coposu, nr. 2-4, 550245 Sibiu, Romania;
| | - Mihai Sava
- Faculty of Medicine, Lucian Blaga University of Sibiu, Lucian Blaga Street 2A, 550169 Sibiu, Romania; (A.-S.B.); (B.I.V.); (I.R.C.); (M.S.)
- County Clinical Emergency Hospital, Bld. Corneliu Coposu, nr. 2-4, 550245 Sibiu, Romania;
| |
Collapse
|
100
|
Jin E, Lv Z, Zhu Y, Zhang H, Li H. Nature-Inspired Micro/Nano-Structured Antibacterial Surfaces. Molecules 2024; 29:1906. [PMID: 38731407 PMCID: PMC11085384 DOI: 10.3390/molecules29091906] [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: 04/09/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
The problem of bacterial resistance has become more and more common with improvements in health care. Worryingly, the misuse of antibiotics leads to an increase in bacterial multidrug resistance and the development of new antibiotics has virtually stalled. These challenges have prompted the need to combat bacterial infections with the use of radically different approaches. Taking lessons from the exciting properties of micro-/nano-natural-patterned surfaces, which can destroy cellular integrity, the construction of artificial surfaces to mimic natural functions provides new opportunities for the innovation and development of biomedicine. Due to the diversity of natural surfaces, functional surfaces inspired by natural surfaces have a wide range of applications in healthcare. Nature-inspired surface structures have emerged as an effective and durable strategy to prevent bacterial infection, opening a new way to alleviate the problem of bacterial drug resistance. The present situation of bactericidal and antifouling surfaces with natural and biomimetic micro-/nano-structures is briefly reviewed. In addition, these innovative nature-inspired methods are used to manufacture a variety of artificial surfaces to achieve extraordinary antibacterial properties. In particular, the physical antibacterial effect of nature-inspired surfaces and the functional mechanisms of chemical groups, small molecules, and ions are discussed, as well as the wide current and future applications of artificial biomimetic micro-/nano-surfaces. Current challenges and future development directions are also discussed at the end. In the future, controlling the use of micro-/nano-structures and their subsequent functions will lead to biomimetic surfaces offering great potential applications in biomedicine.
Collapse
Affiliation(s)
| | | | | | | | - He Li
- School of Mechanical and Electrical Engineering, Henan Agricultural University, Zhengzhou 450002, China; (E.J.); (Z.L.); (Y.Z.); (H.Z.)
| |
Collapse
|