1
|
Kanthenga HT, Banicod RJS, Ntege W, Njiru MN, Javaid A, Tabassum N, Kim YM, Khan F. Functional diversity of AI-2/LuxS system in lactic acid bacteria: Impacts on biofilm formation and environmental resilience. Res Microbiol 2025:104296. [PMID: 40122434 DOI: 10.1016/j.resmic.2025.104296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 03/25/2025]
Abstract
A key component of microbial communication, autoinducer-2 (AI-2) signaling, affects several physiological processes, including environmental adaptation and biofilm formation in lactic acid bacteria (LAB). The multifarious contribution of AI-2, synthesized by LuxS, in improving biofilms and tolerance to hostile conditions in LAB has been investigated in this review. The evolutionary conservation and diversity of AI-2 are shown by a phylogenetic analysis of luxS gene among several LAB species. Furthermore, AI-2 signaling in LAB improves resistance to unfavorable environmental factors, including pH fluctuations, temperature extremes, and antimicrobial agents. Lactic acid bacteria could set off defenses against harmful impacts from environmental stresses.
Collapse
Affiliation(s)
- Hopeful Tusalifye Kanthenga
- KOICA-PKNU International Graduate Program of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea; Department of Fisheries, Malawi College of Fisheries, Mangochi, 301401, Malawi
| | - Riza Jane S Banicod
- KOICA-PKNU International Graduate Program of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea; Fisheries Postharvest Research and Development Division, National Fisheries Research and Development Institute, Quezon City, 1103, Philippines
| | - Wilson Ntege
- KOICA-PKNU International Graduate Program of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea; Fisheries Control Regulation and Quality Assurance, Ministry of Agriculture, Animal Industry and Fisheries, Entebbe, 10101, Uganda
| | - Moses Njeru Njiru
- KOICA-PKNU International Graduate Program of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea; Department of Fisheries and Aquaculture, Turkana County Government, Lodwar, 30500, Kenya
| | - Aqib Javaid
- Interdisciplinary Program of Marine and Fisheries Sciences and Convergent Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Young-Mog Kim
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea; Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Fazlurrahman Khan
- Interdisciplinary Program of Marine and Fisheries Sciences and Convergent Technology, Pukyong National University, Busan, 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea; Ocean and Fisheries Development International Cooperation Institute, Pukyong National University, Busan, 48513, Republic of Korea; International Graduate Program of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
2
|
Gruszecka J, Filip R. Bacterial Biofilms-A Threat to Biliary Stents, Understanding Their Formation, Clinical Consequences and Management. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:512. [PMID: 40142322 PMCID: PMC11943510 DOI: 10.3390/medicina61030512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/09/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025]
Abstract
A biofilm is a community of microbial cells which are enclosed in an external matrix and separated by a network of water channels attached to natural or artificial surfaces. Biofilms formed inside biliary stents consist of a mixed spectrum of bacterial communities, most of which usually originate from the intestines. The patency of biliary stents is the most important problem. Stent occlusion can threaten the health and even life of patients. The main cause of this phenomenon is bile sludge, which is an excellent environment for the multiplication and existence of microorganisms. Due to the great clinical importance of maintaining the patency of biliary stents, several methods have been developed to prevent the accumulation of sludge and the subsequent formation of biofilm; these include, among others, the use of anti-adhesive materials, coating the inner surface of stents with metal cations (silver, copper) or other antimicrobial substances, the implementation of biodegradable drug-eluting biliary stents and the development of a new stent design with an anti-reflux effect. This article presents the latest information on the formation of biofilms in biliary stents, as well as historical and future methods of prevention.
Collapse
Affiliation(s)
- Jolanta Gruszecka
- Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
- Department of Clinical Microbiology, Clinical Hospital No. 2, 35-301 Rzeszow, Poland
| | - Rafał Filip
- Faculty of Medicine, University of Rzeszow, 35-959 Rzeszow, Poland
- Department of Gastroenterology with IBD Unit, Clinical Hospital No. 2, 35-301 Rzeszow, Poland
| |
Collapse
|
3
|
Xue Y, Kang X. Time-resolved compositional and dynamics analysis of biofilm maturation and dispersal via solid-state NMR spectroscopy. NPJ Biofilms Microbiomes 2025; 11:21. [PMID: 39880834 PMCID: PMC11779841 DOI: 10.1038/s41522-025-00655-4] [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: 09/02/2024] [Accepted: 01/20/2025] [Indexed: 01/31/2025] Open
Abstract
Dispersal plays a crucial role in the development and ecology of biofilms. While extensive studies focused on elucidating the molecular mechanisms governing this process, few have characterized the associated temporal changes in composition and structure. Here, we employed solid-state nuclear magnetic resonance (NMR) techniques to achieve time-resolved characterization of Bacillus subtilis biofilms over a 5-day period. The mature biofilm, established within 48 h, undergoes significant degradation in following 72 h. The steepest decline of proteins precedes that of exopolysaccharides, likely reflecting their distinct spatial distribution. Exopolysaccharide sugar units display clustered temporal patterns, suggesting the presence of distinct polysaccharide types. A sharp rise in aliphatic carbon signals on day 4 probably corresponds to a surge in biosurfactant production. Different dynamic regimes respond differently to dispersal: the mobile domain exhibits increased rigidity, while the rigid domain remains stable. These findings provide novel insights and perspectives on the complex process of biofilm dispersal.
Collapse
Affiliation(s)
- Yi Xue
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, 315211, Zhejiang, China
| | - Xue Kang
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, 315211, Zhejiang, China.
| |
Collapse
|
4
|
Ji J, Hu F, Qin J, Zhao Y, Dong Y, Yang H, Bai Z, Wu G, Wang Q, Jin B. Comparation on the responses and resilience of single-Anammox system and synergistic partial-denitrification/anammox system to long-term nutrient starvation: Performance and metagenomic insights. BIORESOURCE TECHNOLOGY 2025; 415:131694. [PMID: 39447919 DOI: 10.1016/j.biortech.2024.131694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/26/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024]
Abstract
Starvation disturbance was a common problem in biological sewage treatment processes. However, understanding about the responses and resilience of different active anammox biomass in autotrophic and heterotrophic systems to long-term nutrient starvation remains limited. This study compared responses and potential recovery mechanisms of autotrophic single-Anammox and heterotrophic synergistic partial-denitrification/anammox (PD/anammox) systems to prolonged starvation (31-40 days). After starvation, total inorganic nitrogen (TIN) removal efficiency of single-Anammox and synergistic PD/anammox systems decreased to 62.16 % and 78.52 %, respectively, of their original level. After the nutrient resupply, the performance of both systems gradually recovered to a similar-to-pre-starvation level at the rate of 1.26 %/day and 1.89 %/day, respectively. Compared with single-Anammox system, complex synergistic relationship of microorganisms and effective quorum sensing (QS) regulation strategies might mitigate the negative influences were caused by starvation and ensure the performance quickly return of synergistic PD/anammox system. This study would contribute to promote the application of Anammox technology.
Collapse
Affiliation(s)
- Jiantao Ji
- College of Ecology and Environment, Zhengzhou University, Zhengzhou 450001, PR China.
| | - Feiyue Hu
- College of Ecology and Environment, Zhengzhou University, Zhengzhou 450001, PR China
| | - Jing Qin
- College of Ecology and Environment, Zhengzhou University, Zhengzhou 450001, PR China
| | - Ying Zhao
- College of Ecology and Environment, Zhengzhou University, Zhengzhou 450001, PR China
| | - Yongen Dong
- College of Ecology and Environment, Zhengzhou University, Zhengzhou 450001, PR China
| | - Haosen Yang
- College of Ecology and Environment, Zhengzhou University, Zhengzhou 450001, PR China
| | - Zhixuan Bai
- College of Ecology and Environment, Zhengzhou University, Zhengzhou 450001, PR China
| | - Guanqi Wu
- College of Ecology and Environment, Zhengzhou University, Zhengzhou 450001, PR China
| | - Qiyue Wang
- College of Ecology and Environment, Zhengzhou University, Zhengzhou 450001, PR China
| | - Baodan Jin
- College of Material and Chemical Engineering, Zhengzhou University of Light Industry, Zhengzhou 450001, PR China.
| |
Collapse
|
5
|
Agarwal H, Gurnani B, Pippal B, Jain N. Capturing the micro-communities: Insights into biogenesis and architecture of bacterial biofilms. BBA ADVANCES 2024; 7:100133. [PMID: 39839441 PMCID: PMC11750278 DOI: 10.1016/j.bbadva.2024.100133] [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] [Indexed: 01/23/2025] Open
Abstract
Biofilm is an assemblage of microorganisms embedded within the extracellular matrix that provides mechanical stability, nutrient absorption, antimicrobial resistance, cell-cell interactions, and defence against host immune system. Various biomolecules such as lipids, carbohydrates, protein polymers (amyloid), and eDNA are present in the matrix playing significant role in determining the distinctive properties of biofilm. The formation of biofilms contributes to resistance against antimicrobial therapy in most of the human infections and exacerbates existing diseases. Therefore, this field requires several state-of-the-art techniques to fully understand the 3-D organization of biofilms, their cell behaviour and responses to pharmaceutical treatments. Here, we explore the assembly and regulation of biofilm biogenesis in the context of matrix components and highlight the significance of high-resolution imaging and analysing techniques for monitoring complex biofilm architecture. Our review also emphasizes the novelty and advancements in techniques to visualise biofilm structure and composition, providing valuable insights to understand biofilm-related infections.
Collapse
Affiliation(s)
- Harshita Agarwal
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, NH 65, Nagaur Road, Karwar, Rajasthan 342037, India
| | - Bharat Gurnani
- Centre of Excellence-AyurTech, Indian Institute of Technology Jodhpur, NH 65, Nagaur Road, Karwar, Rajasthan 342037, India
| | - Bhumika Pippal
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, NH 65, Nagaur Road, Karwar, Rajasthan 342037, India
| | - Neha Jain
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, NH 65, Nagaur Road, Karwar, Rajasthan 342037, India
| |
Collapse
|
6
|
Roberts JM, Milo S, Metcalf DG. Harnessing the Power of Our Immune System: The Antimicrobial and Antibiofilm Properties of Nitric Oxide. Microorganisms 2024; 12:2543. [PMID: 39770746 PMCID: PMC11677572 DOI: 10.3390/microorganisms12122543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Nitric oxide (NO) is a free radical of the human innate immune response to invading pathogens. NO, produced by nitric oxide synthases (NOSs), is used by the immune system to kill microorganisms encapsulated within phagosomes via protein and DNA disruption. Owing to its ability to disperse biofilm-bound microorganisms, penetrate the biofilm matrix, and act as a signal molecule, NO may also be effective as an antibiofilm agent. NO can be considered an underappreciated antimicrobial that could be levied against infected, at-risk, and hard-to-heal wounds due to the inherent lack of bacterial resistance, and tolerance by human tissues. NO produced within a wound dressing may be an effective method of disrupting biofilms and killing microorganisms in hard-to-heal wounds such as diabetic foot ulcers, venous leg ulcers, and pressure injuries. We have conducted a narrative review of the evidence underlying the key antimicrobial and antibiofilm mechanisms of action of NO for it to serve as an exogenously-produced antimicrobial agent in dressings used in the treatment of hard-to-heal wounds.
Collapse
Affiliation(s)
| | | | - Daniel Gary Metcalf
- Advanced Wound Care Research & Development, Convatec, Deeside Industrial Park, Deeside CH5 2NU, UK; (J.M.R.); (S.M.)
| |
Collapse
|
7
|
Hu Y, Ding M, Lv X, Jiang J, Zhang J, Yang D. Stimuli-Responsive NO Delivery Platforms for Bacterial Infection Treatment. Adv Healthc Mater 2024; 13:e2402240. [PMID: 39171769 DOI: 10.1002/adhm.202402240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/11/2024] [Indexed: 08/23/2024]
Abstract
The prevalence of drug-resistant bacterial infections has emerged as a grave threat to clinical treatment and global human health, presenting one of the foremost challenges in medical care. Thus, there is an urgent imperative to develop safe and efficacious novel antimicrobial strategies. Nitric oxide (NO) is a recognized endogenous signaling molecule, which plays a pivotal role in numerous pathological processes. Currently, NO has garnered significant interest as an antibacterial agent due to its capability to eradicate bacteria, disrupt biofilms, and facilitate wound healing, all while circumventing the emergence of drug resistance. However, the inherently unstable characteristic of NO therapeutic gas renders the controlled administration of NO gases exceedingly challenging. Hence, in this review, the current challenge of bacterial infection is discussed; then it is briefly elucidated the antibacterial mechanism of NO and comprehensively delineate the recent advancements in stimulus-responsive NO delivery platforms, along with their merits, obstacles, and prospective avenues for clinical application. This review offers guidance for future advancements in NO-medicated anti-infection therapy is hoped.
Collapse
Affiliation(s)
- Yanling Hu
- College of Life and Health, Nanjing Polytechnic Institute, Nanjing, 210048, P. R. China
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing, 211816, P. R. China
| | - Meng Ding
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu, 210008, P. R. China
| | - Xinyi Lv
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing, 211816, P. R. China
| | - Jingai Jiang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing, 211816, P. R. China
| | - Junjie Zhang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, 233030, P. R. China
| | - Dongliang Yang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing, 211816, P. R. China
| |
Collapse
|
8
|
Stubbusch AKM, Keegstra JM, Schwartzman J, Pontrelli S, Clerc EE, Charlton S, Stocker R, Magnabosco C, Schubert OT, Ackermann M, D'Souza GG. Polysaccharide breakdown products drive degradation-dispersal cycles of foraging bacteria through changes in metabolism and motility. eLife 2024; 13:RP93855. [PMID: 39429128 PMCID: PMC11493405 DOI: 10.7554/elife.93855] [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] [Indexed: 10/22/2024] Open
Abstract
Most of Earth's biomass is composed of polysaccharides. During biomass decomposition, polysaccharides are degraded by heterotrophic bacteria as a nutrient and energy source and are thereby partly remineralized into CO2. As polysaccharides are heterogeneously distributed in nature, following the colonization and degradation of a polysaccharide hotspot the cells need to reach new polysaccharide hotspots. Even though many studies indicate that these degradation-dispersal cycles contribute to the carbon flow in marine systems, we know little about how cells alternate between polysaccharide degradation and motility, and which environmental factors trigger this behavioral switch. Here, we studied the growth of the marine bacterium Vibrio cyclitrophicus ZF270 on the abundant marine polysaccharide alginate, both in its soluble polymeric form as well as on its breakdown products. We used microfluidics coupled to time-lapse microscopy to analyze motility and growth of individual cells, and RNA sequencing to study associated changes in gene expression. We found that single cells grow at reduced rate on alginate until they form large groups that cooperatively break down the polymer. Exposing cell groups to digested alginate accelerates cell growth and changes the expression of genes involved in alginate degradation and catabolism, central metabolism, ribosomal biosynthesis, and transport. However, exposure to digested alginate also triggers cells to become motile and disperse from cell groups, proportionally increasing with the group size before the nutrient switch, and this is accompanied by high expression of genes involved in flagellar assembly, chemotaxis, and quorum sensing. The motile cells chemotax toward polymeric but not digested alginate, likely enabling them to find new polysaccharide hotspots. Overall, our findings reveal cellular mechanisms that might also underlie bacterial degradation-dispersal cycles, which influence the remineralization of biomass in marine environments.
Collapse
Affiliation(s)
- Astrid Katharina Maria Stubbusch
- Institute of Biogeochemistry and Pollutant Dynamics, Department of Environmental Systems Science, ETH ZurichZurichSwitzerland
- Department of Environmental Microbiology, Eawag: Swiss Federal Institute of Aquatic Science and TechnologyDübendorfSwitzerland
- Geological Institute, Department of Earth Sciences, ETH ZurichZurichSwitzerland
| | - Johannes M Keegstra
- Institute of Environmental Engineering, Department of Civil, Environmental and Geomatic Engineering, ETH ZurichZurichSwitzerland
| | - Julia Schwartzman
- Department of Civil and Environmental Engineering, MITCambridgeUnited States
- Department of Biology, University of Southern CaliforniaLos AngelesUnited States
| | - Sammy Pontrelli
- Institute of Molecular Systems Biology, Department of Biology, ETH ZurichZurichSwitzerland
| | - Estelle E Clerc
- Institute of Environmental Engineering, Department of Civil, Environmental and Geomatic Engineering, ETH ZurichZurichSwitzerland
| | - Samuel Charlton
- Institute of Environmental Engineering, Department of Civil, Environmental and Geomatic Engineering, ETH ZurichZurichSwitzerland
| | - Roman Stocker
- Institute of Environmental Engineering, Department of Civil, Environmental and Geomatic Engineering, ETH ZurichZurichSwitzerland
| | - Cara Magnabosco
- Geological Institute, Department of Earth Sciences, ETH ZurichZurichSwitzerland
| | - Olga T Schubert
- Institute of Biogeochemistry and Pollutant Dynamics, Department of Environmental Systems Science, ETH ZurichZurichSwitzerland
- Department of Environmental Microbiology, Eawag: Swiss Federal Institute of Aquatic Science and TechnologyDübendorfSwitzerland
| | - Martin Ackermann
- Institute of Biogeochemistry and Pollutant Dynamics, Department of Environmental Systems Science, ETH ZurichZurichSwitzerland
- Department of Environmental Microbiology, Eawag: Swiss Federal Institute of Aquatic Science and TechnologyDübendorfSwitzerland
- Laboratory of Microbial Systems Ecology, School of Architecture, Civil and Environmental Engineering (ENAC), École Polytechnique Fédéral de Lausanne (EPFL)LausanneSwitzerland
| | - Glen G D'Souza
- Institute of Biogeochemistry and Pollutant Dynamics, Department of Environmental Systems Science, ETH ZurichZurichSwitzerland
- Department of Environmental Microbiology, Eawag: Swiss Federal Institute of Aquatic Science and TechnologyDübendorfSwitzerland
| |
Collapse
|
9
|
Ghahari A, Khosravi‐Darani K. Hurdle technology using enzymes and essential oil to remove biofilm and increase the effectiveness of this process with the microencapsulation method. Food Sci Nutr 2024; 12:8483-8492. [PMID: 39479686 PMCID: PMC11521719 DOI: 10.1002/fsn3.4377] [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: 04/28/2024] [Revised: 07/13/2024] [Accepted: 07/16/2024] [Indexed: 11/02/2024] Open
Abstract
The formation of biofilm in different places and the failure to effectively remove it by the usual disinfection methods is due to its structure and the rich genetic resource available in it to deal with disinfectants. These impenetrable structures and diverse microbial genetics have caused biofilm pollution in different industries like the food industry, the medicine industry, the hospitals and the water distribution system, resulting in pathogenicity and reduction of industrial quality. An efficient way to deal with the resistant population of biofilm-forming microbes is the use of hurdle technology including enzymes and essential oils. Enzymes reduce the resistance of the biofilm structure due to degradation of its extracellular polymer matrix (EPS) by their abilities to break down the organic molecules, and then the essential oils weaken the cells by penetrating the lipid membrane of the cell and destroying its integrity; as a result, the biofilm will be destroyed. The advantage of this hurdle technology is the environmental friendly of both methods, which reduces concerns about the use of chemical disinfection methods, but on the other hand, due to the sensitivity of enzymes as biological agents also the expensiveness of this technique and the considerations of working with essential oils as volatile and unstable liquids should abandon the routine methods of applying this disinfectant to biofilm and go for the microencapsulation method, which as a protective system increases the effectiveness of enzymes and essential oils as antibiofilm agents.
Collapse
Affiliation(s)
- Ayda Ghahari
- Bioprocess Engineering DepartmentInstitute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and BiotechnologyTehranIran
| | - Kianoush Khosravi‐Darani
- Research Department of Food Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Sciences and Food TechnologyShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
10
|
Giordano V, Giannoudis PV. Biofilm Formation, Antibiotic Resistance, and Infection (BARI): The Triangle of Death. J Clin Med 2024; 13:5779. [PMID: 39407838 PMCID: PMC11476620 DOI: 10.3390/jcm13195779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Fracture-related infection (FRI) is a devastating event, directly affecting fracture healing, impairing patient function, prolonging treatment, and increasing healthcare costs. Time plays a decisive role in prognosis, as biofilm maturation leads to the development of antibiotic resistance, potentially contributing to infection chronicity and increasing morbidity and mortality. Research exploring the association between biofilm maturation and antibiotic resistance in orthopaedics primarily addresses aspects related to quality of life and physical function; however, little exists on life-threatening conditions and mortality. Understanding the intrinsic relationship between biofilm maturation, bacterial resistance, and mortality is critical in all fields of medicine. In the herein narrative review, we summarize recent evidence regarding biofilm formation, antibiotic resistance, and infection chronicity (BARI), the three basic components of the "triangle of death" of FRI, and its implications. Preoperative, perioperative, and postoperative prevention strategies to avoid the "triangle of death" of FRI are presented and discussed. Additionally, the importance of the orthopaedic trauma surgeon in understanding new tools to combat infections related to orthopaedic devices is highlighted.
Collapse
Affiliation(s)
- Vincenzo Giordano
- Serviço de Ortopedia e Traumatologia Prof. Nova Monteiro, Hospital Municipal Miguel Couto, Rua Mário Ribeiro 117/2º Andar, Gávea, Rio de Janeiro 22430-160, RJ, Brazil
| | - Peter V. Giannoudis
- Academic Department of Trauma and Orthopaedics, School of Medicine, University of Leeds, Leeds LS2 9LU, UK
- NIHR Leeds Biomedical Research Center, Chapel Allerton Hospital, Leeds LS7 4SA, UK
| |
Collapse
|
11
|
Holt JD, Schultz D, Nadell CD. Dispersal of a dominant competitor can drive multispecies coexistence in biofilms. Curr Biol 2024; 34:4129-4142.e4. [PMID: 39163856 PMCID: PMC11686572 DOI: 10.1016/j.cub.2024.07.078] [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: 12/11/2023] [Revised: 04/25/2024] [Accepted: 07/22/2024] [Indexed: 08/22/2024]
Abstract
Despite competition for both space and nutrients, bacterial species often coexist within structured, surface-attached communities termed biofilms. While these communities play important, widespread roles in ecosystems and are agents of human infection, understanding how multiple bacterial species assemble to form these communities and what physical processes underpin the composition of multispecies biofilms remains an active area of research. Using a model three-species community composed of Pseudomonas aeruginosa, Escherichia coli, and Enterococcus faecalis, we show with cellular-scale resolution that biased dispersal of the dominant community member, P. aeruginosa, prevents competitive exclusion from occurring, leading to the coexistence of the three species. A P. aeruginosa bqsS deletion mutant no longer undergoes periodic mass dispersal, leading to the local competitive exclusion of E. coli. Introducing periodic, asymmetric dispersal behavior into minimal models, parameterized by only maximal growth rate and local density, supports the intuition that biased dispersal of an otherwise dominant competitor can permit coexistence generally. Colonization experiments show that WT P. aeruginosa is superior at colonizing new areas, in comparison to ΔbqsS P. aeruginosa, but at the cost of decreased local competitive ability against E. coli and E. faecalis. Overall, our experiments document how one species' modulation of a competition-dispersal-colonization trade-off can go on to influence the stability of multispecies coexistence in spatially structured ecosystems.
Collapse
Affiliation(s)
- Jacob D Holt
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA; Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Hanover, NH 03755, USA
| | - Daniel Schultz
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Hanover, NH 03755, USA
| | - Carey D Nadell
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA; Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Hanover, NH 03755, USA.
| |
Collapse
|
12
|
Gong F, Xin S, Liu X, He C, Yu X, Pan L, Zhang S, Gao H, Xu J. Multiple biological characteristics and functions of intestinal biofilm extracellular polymers: friend or foe? Front Microbiol 2024; 15:1445630. [PMID: 39224216 PMCID: PMC11367570 DOI: 10.3389/fmicb.2024.1445630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
The gut microbiota is vital to human health, and their biofilms significantly impact intestinal immunity and the maintenance of microbial balance. Certain pathogens, however, can employ biofilms to elude identification by the immune system and medical therapy, resulting in intestinal diseases. The biofilm is formed by extracellular polymorphic substances (EPS), which shield microbial pathogens from the host immune system and enhance its antimicrobial resistance. Therefore, investigating the impact of extracellular polysaccharides released by pathogens that form biofilms on virulence and defence mechanisms is crucial. In this review, we provide a comprehensive overview of current pathogenic biofilm research, deal with the role of extracellular polymers in the formation and maintenance of pathogenic biofilm, and elaborate different prevention and treatment strategies to provide an innovative approach to the treatment of intestinal pathogen-based diseases.
Collapse
Affiliation(s)
- Fengrong Gong
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shuzi Xin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaohui Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chengwei He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xinyi Yu
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Luming Pan
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Sitian Zhang
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Han Gao
- Department of Clinical Laboratory, Aerospace Center Hospital, Beijing, China
| | - Jingdong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Concha-Eloko R, Stock M, De Baets B, Briers Y, Sanjuán R, Domingo-Calap P, Boeckaerts D. DepoScope: Accurate phage depolymerase annotation and domain delineation using large language models. PLoS Comput Biol 2024; 20:e1011831. [PMID: 39102416 PMCID: PMC11326577 DOI: 10.1371/journal.pcbi.1011831] [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: 01/15/2024] [Revised: 08/15/2024] [Accepted: 07/20/2024] [Indexed: 08/07/2024] Open
Abstract
Bacteriophages (phages) are viruses that infect bacteria. Many of them produce specific enzymes called depolymerases to break down external polysaccharide structures. Accurate annotation and domain identification of these depolymerases are challenging due to their inherent sequence diversity. Hence, we present DepoScope, a machine learning tool that combines a fine-tuned ESM-2 model with a convolutional neural network to identify depolymerase sequences and their enzymatic domains precisely. To accomplish this, we curated a dataset from the INPHARED phage genome database, created a polysaccharide-degrading domain database, and applied sequential filters to construct a high-quality dataset, which is subsequently used to train DepoScope. Our work is the first approach that combines sequence-level predictions with amino-acid-level predictions for accurate depolymerase detection and functional domain identification. In that way, we believe that DepoScope can greatly enhance our understanding of phage-host interactions at the level of depolymerases.
Collapse
Affiliation(s)
- Robby Concha-Eloko
- Institute for Integrative Systems Biology (I2SysBio), Universitat de Valencia-CSIC, Paterna, Spain
| | - Michiel Stock
- KERMIT, Department of Data Analysis and Mathematical Modelling, Ghent University, Ghent, Belgium
| | - Bernard De Baets
- KERMIT, Department of Data Analysis and Mathematical Modelling, Ghent University, Ghent, Belgium
| | - Yves Briers
- Laboratory of Applied Biotechnology, Department of Biotechnology, Ghent University, Ghent, Belgium
| | - Rafael Sanjuán
- Institute for Integrative Systems Biology (I2SysBio), Universitat de Valencia-CSIC, Paterna, Spain
| | - Pilar Domingo-Calap
- Institute for Integrative Systems Biology (I2SysBio), Universitat de Valencia-CSIC, Paterna, Spain
| | - Dimitri Boeckaerts
- KERMIT, Department of Data Analysis and Mathematical Modelling, Ghent University, Ghent, Belgium
- Laboratory of Applied Biotechnology, Department of Biotechnology, Ghent University, Ghent, Belgium
| |
Collapse
|
14
|
Tetz V, Kardava K, Vecherkovskaya M, Khodadadi-Jamayran A, Tsirigos A, Tetz G. Previously unknown regulatory role of extracellular RNA on bacterial directional migration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.11.603110. [PMID: 39026763 PMCID: PMC11257571 DOI: 10.1101/2024.07.11.603110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Bacterial directional migration plays a significant role in bacterial adaptation. However, the regulation of this process, particularly in young biofilms, remains unclear. Here, we demonstrated the critical role of extracellular RNA as part of the Universal Receptive System in bacterial directional migration using a multidisciplinary approach, including bacterial culture, biochemistry, and genetics. We found that the destruction or inactivation of extracellular RNA with RNase or RNA-specific antibodies in the presence of the chemoattractant triggered the formation of bacterial "runner cells» in what we call a "panic state" capable of directional migration. These cells quickly migrated even on the surface of 1.5% agar and formed evolved colonies that were transcriptionally and biochemically different from the ancestral cells. We have also shown that cell-free DNA from blood plasma can act as a potent bacterial chemoattractant. Our data revealed a previously unknown role of bacterial extracellular RNA in the regulation of bacterial migration and have shown that its destruction or inhibition triggered the directional migration of developing and mature biofilms towards the chemoattractant.
Collapse
|
15
|
Choi HY, Kim WG. Tyrosol blocks E. coli anaerobic biofilm formation via YbfA and FNR to increase antibiotic susceptibility. Nat Commun 2024; 15:5683. [PMID: 38971825 PMCID: PMC11227560 DOI: 10.1038/s41467-024-50116-3] [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: 09/04/2023] [Accepted: 06/28/2024] [Indexed: 07/08/2024] Open
Abstract
Bacteria within mature biofilms are highly resistant to antibiotics than planktonic cells. Oxygen limitation contributes to antibiotic resistance in mature biofilms. Nitric oxide (NO) induces biofilm dispersal; however, low NO levels stimulate biofilm formation, an underexplored process. Here, we introduce a mechanism of anaerobic biofilm formation by investigating the antibiofilm activity of tyrosol, a component in wine. Tyrosol inhibits E. coli and Pseudomonas aeruginosa biofilm formation by enhancing NO production. YbfA is identified as a target of tyrosol and its downstream targets are sequentially determined. YbfA activates YfeR, which then suppresses the anaerobic regulator FNR. This suppression leads to decreased NO production, elevated bis-(3'-5')-cyclic dimeric GMP levels, and finally stimulates anaerobic biofilm formation in the mature stage. Blocking YbfA with tyrosol treatment renders biofilm cells as susceptible to antibiotics as planktonic cells. Thus, this study presents YbfA as a promising antibiofilm target to address antibiotic resistance posed by biofilm-forming bacteria, with tyrosol acting as an inhibitor.
Collapse
Affiliation(s)
- Ha-Young Choi
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Yusong, Daejeon, 34141, Republic of Korea
| | - Won-Gon Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Yusong, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
16
|
Zuberi A, Ahmad N, Ahmad H, Saeed M, Ahmad I. Beyond antibiotics: CRISPR/Cas9 triumph over biofilm-associated antibiotic resistance infections. Front Cell Infect Microbiol 2024; 14:1408569. [PMID: 39035353 PMCID: PMC11257871 DOI: 10.3389/fcimb.2024.1408569] [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: 03/28/2024] [Accepted: 05/27/2024] [Indexed: 07/23/2024] Open
Abstract
A complex structure known as a biofilm is formed when a variety of bacterial colonies or a single type of cell in a group sticks to a surface. The extracellular polymeric compounds that encase these cells, often consisting of proteins, eDNA, and polysaccharides, exhibit strong antibiotic resistance. Concerns about biofilm in the pharmaceutical industry, public health, and medical fields have sparked a lot of interest, as antibiotic resistance is a unique capacity exhibited by these biofilm-producing bacteria, which increases morbidity and death. Biofilm formation is a complicated process that is controlled by several variables. Insights into the processes to target for the therapy have been gained from multiple attempts to dissect the biofilm formation process. Targeting pathogens within a biofilm is profitable because the bacterial pathogens become considerably more resistant to drugs in the biofilm state. Although biofilm-mediated infections can be lessened using the currently available medications, there has been a lot of focus on the development of new approaches, such as bioinformatics tools, for both treating and preventing the production of biofilms. Technologies such as transcriptomics, metabolomics, nanotherapeutics and proteomics are also used to develop novel anti-biofilm agents. These techniques help to identify small compounds that can be used to inhibit important biofilm regulators. The field of appropriate control strategies to avoid biofilm formation is expanding quickly because of this spurred study. As a result, the current article addresses our current knowledge of how biofilms form, the mechanisms by which bacteria in biofilms resist antibiotics, and cutting-edge treatment approaches for infections caused by biofilms. Furthermore, we have showcased current ongoing research utilizing the CRISPR/Cas9 gene editing system to combat bacterial biofilm infections, particularly those brought on by lethal drug-resistant pathogens, concluded the article with a novel hypothesis and aspirations, and acknowledged certain limitations.
Collapse
Affiliation(s)
- Azna Zuberi
- Department of Molecular, Cellular & Developmental Biology, University of Colorado Boulder, Boulder, CO, United States
- Department of Obs & Gynae, Northwestern University, Chicago, IL, United States
| | - Nayeem Ahmad
- Department of Biophysics, All India Institute of Medical Science, New Delhi, India
- Department of Microbiology, Immunology, and Infectious Diseases, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Hafiz Ahmad
- Department of Medical Microbiology & Immunology, Ras Al Khaimah (RAK) College of Medical Sciences, Ras Al Khaimah (RAK) Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Mohd Saeed
- Department of Biology, College of Science University of Hail, Hail, Saudi Arabia
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
17
|
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
|
18
|
Qiao K, Zhao T, Wang L, Zhang W, Meng W, Liu F, Gao X, Zhu J. Screening and identification of functional bacterial attachment genes in aerobic granular sludge. J Environ Sci (China) 2024; 141:205-214. [PMID: 38408821 DOI: 10.1016/j.jes.2023.07.011] [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: 01/17/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 02/28/2024]
Abstract
The screening and identification of attachment genes is important to exploring the formation mechanism of biofilms at the gene level. It is helpful to the development of key culture technologies for aerobic granular sludge (AGS). In this study, genome-wide sequencing and gene editing were employed for the first time to investigate the effects and functions of attachment genes in AGS. With the help of whole-genome analysis, ten attachment genes were screened from thirteen genes, and the efficiency of gene screening was greatly improved. Then, two attachment genes were selected as examples to further confirm the gene functions by constructing gene-knockout recombinant mutants of Stenotrophomonas maltophilia; when the two attachment genes were knocked out, the attachment potential was reduced by 50.67% and 43.93%, respectively. The results provide a new theoretical principle and efficient method for the development of AGS from the perspective of attachment genes.
Collapse
Affiliation(s)
- Kai Qiao
- School of Environment, Beijing Normal University, Beijing 100875, China; State Key Laboratory of Water Simulation, Beijing 100875, China
| | - Tingting Zhao
- School of Environment, Beijing Normal University, Beijing 100875, China; R & D Centre of Aerobic Granule Technology, Beijing 100875, China
| | - Lei Wang
- School of Environment, Beijing Normal University, Beijing 100875, China; R & D Centre of Aerobic Granule Technology, Beijing 100875, China
| | - Wei Zhang
- School of Environment, Beijing Normal University, Beijing 100875, China
| | - Wei Meng
- School of Environment, Beijing Normal University, Beijing 100875, China
| | - Fan Liu
- School of Environment, Beijing Normal University, Beijing 100875, China
| | - Xu Gao
- School of Environment, Beijing Normal University, Beijing 100875, China; State Key Laboratory of Water Simulation, Beijing 100875, China
| | - Jianrong Zhu
- School of Environment, Beijing Normal University, Beijing 100875, China; R & D Centre of Aerobic Granule Technology, Beijing 100875, China.
| |
Collapse
|
19
|
Hu XM, Peng L, Wang Y, Ma F, Tao Y, Liang X, Yang JL. Bacterial c-di-GMP triggers metamorphosis of mussel larvae through a STING receptor. NPJ Biofilms Microbiomes 2024; 10:51. [PMID: 38902226 PMCID: PMC11190208 DOI: 10.1038/s41522-024-00523-7] [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: 12/21/2023] [Accepted: 06/07/2024] [Indexed: 06/22/2024] Open
Abstract
Bacteria induced metamorphosis observed in nearly all marine invertebrates. However, the mechanism of bacteria regulating the larvae-juvenile metamorphosis remains unknown. Here, we test the hypothesis that c-di-GMP, a ubiquitous bacterial second-messenger molecule, directly triggers the mollusc Mytilus coruscus larval metamorphosis via the stimulator of interferon genes (STING) receptor. We determined that the deletion of c-di-GMP synthesis genes resulted in reduced c-di-GMP levels and biofilm-inducing activity on larval metamorphosis, accompanied by alterations in extracellular polymeric substances. Additionally, c-di-GMP extracted from tested varying marine bacteria all exhibited inducing activity on larval metamorphosis. Simultaneously, through pharmacological and molecular experiments, we demonstrated that M. coruscus STING (McSTING) participates in larval metamorphosis by binding with c-di-GMP. Our findings reveal that new role of bacterial c-di-GMP that triggers mussel larval metamorphosis transition, and extend knowledge in the interaction of bacteria and host development in marine ecosystems.
Collapse
Affiliation(s)
- Xiao-Meng Hu
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
- Shanghai Collaborative Innovation Center for Cultivating Elite Breeds and Green-Culture of Aquaculture Animals, Shanghai, 201306, China
| | - Lihua Peng
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
- Shanghai Collaborative Innovation Center for Cultivating Elite Breeds and Green-Culture of Aquaculture Animals, Shanghai, 201306, China
| | - Yuyi Wang
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
- Shanghai Collaborative Innovation Center for Cultivating Elite Breeds and Green-Culture of Aquaculture Animals, Shanghai, 201306, China
| | - Fan Ma
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
- Shanghai Collaborative Innovation Center for Cultivating Elite Breeds and Green-Culture of Aquaculture Animals, Shanghai, 201306, China
| | - Yu Tao
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
- Shanghai Collaborative Innovation Center for Cultivating Elite Breeds and Green-Culture of Aquaculture Animals, Shanghai, 201306, China
| | - Xiao Liang
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China.
- Shanghai Collaborative Innovation Center for Cultivating Elite Breeds and Green-Culture of Aquaculture Animals, Shanghai, 201306, China.
| | - Jin-Long Yang
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China.
- Shanghai Collaborative Innovation Center for Cultivating Elite Breeds and Green-Culture of Aquaculture Animals, Shanghai, 201306, China.
| |
Collapse
|
20
|
Catania S, Bottinelli M, Fincato A, Tondo A, Matucci A, Nai G, Righetti V, Abbate F, Ramírez AS, Gobbo F, Merenda M. Pathogenic avian mycoplasmas show phenotypic differences in their biofilm forming ability compared to non-pathogenic species in vitro. Biofilm 2024; 7:100190. [PMID: 38515541 PMCID: PMC10955283 DOI: 10.1016/j.bioflm.2024.100190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 03/23/2024] Open
Abstract
Mycoplasmas are known as the minimalist microorganisms in the microbes' world. Their minimalist nature makes them highly sensitive to the environmental conditions and limits their ability to survive for extended periods outside their animal host. Nevertheless, there are documented instances of mycoplasma transmission over significant distances and this phenomenon may be linked to relatively unexplored abilities of mycoplasmas, such as their capacity to synthesize biofilm-the predominant mode of bacterial growth in nature. The authors decided to establish a method aimed at inducing the clustering of mycoplasma planktonic cells within a biofilm in vitro and subsequently assess the capacity of certain avian mycoplasmas to synthesize a biofilm. A total of 299 avian mycoplasma isolates were included in the study, encompassing both pathogenic (Mycoplasma gallisepticum, M. synoviae, M. meleagridis, M. iowae) and non-pathogenic species (M. gallinaceum, M. gallinarum, M. iners and M. pullorum). The authors successfully demonstrated the feasibility of inducing avian mycoplasmas to synthetize in vitro a biofilm, which can be visually quantified. The only species that did not produce any biofilm was M. iowae. In general, the pathogenic mycoplasmas produced greater quantities of biofilm compared to the non-pathogenic ones. Furthermore, it was observed that the ability to produce biofilm appeared to vary, both qualitatively and quantitatively, not only among different species but also among isolates of a single species. Future studies will be necessary to determine whether biofilm production plays a pivotal epidemiological role for the pathogenic avian mycoplasmas.
Collapse
Affiliation(s)
- Salvatore Catania
- Unità Micoplasmi, WOAH Reference Laboratory for Avian Mycoplasmosis (M. Gallisepticum, M. Synoviae), Istituto Zooprofilattico Sperimentale delle Venezie, 37060, Buttapietra, (VR), Italy
| | - Marco Bottinelli
- Unità Micoplasmi, WOAH Reference Laboratory for Avian Mycoplasmosis (M. Gallisepticum, M. Synoviae), Istituto Zooprofilattico Sperimentale delle Venezie, 37060, Buttapietra, (VR), Italy
| | - Alice Fincato
- Unità Micoplasmi, WOAH Reference Laboratory for Avian Mycoplasmosis (M. Gallisepticum, M. Synoviae), Istituto Zooprofilattico Sperimentale delle Venezie, 37060, Buttapietra, (VR), Italy
| | - Annalucia Tondo
- Unità Micoplasmi, WOAH Reference Laboratory for Avian Mycoplasmosis (M. Gallisepticum, M. Synoviae), Istituto Zooprofilattico Sperimentale delle Venezie, 37060, Buttapietra, (VR), Italy
| | - Andrea Matucci
- Unità Micoplasmi, WOAH Reference Laboratory for Avian Mycoplasmosis (M. Gallisepticum, M. Synoviae), Istituto Zooprofilattico Sperimentale delle Venezie, 37060, Buttapietra, (VR), Italy
| | - Giorgia Nai
- Unità Micoplasmi, WOAH Reference Laboratory for Avian Mycoplasmosis (M. Gallisepticum, M. Synoviae), Istituto Zooprofilattico Sperimentale delle Venezie, 37060, Buttapietra, (VR), Italy
| | - Verdiana Righetti
- Unità Micoplasmi, WOAH Reference Laboratory for Avian Mycoplasmosis (M. Gallisepticum, M. Synoviae), Istituto Zooprofilattico Sperimentale delle Venezie, 37060, Buttapietra, (VR), Italy
| | - Francesco Abbate
- Dipartimento di Scienze Veterinarie, Università di Messina, 98168, Messina, ME, Italy
| | - Ana S. Ramírez
- Unidad de Epidemiología y Medicina Preventiva, Instituto Universitario de Sanidad Animal y Seguridad Alimentaria (IUSA), Universidad de Las Palmas de Gran Canaria, 35413, Arucas, Spain
| | - Federica Gobbo
- Unità Micoplasmi, WOAH Reference Laboratory for Avian Mycoplasmosis (M. Gallisepticum, M. Synoviae), Istituto Zooprofilattico Sperimentale delle Venezie, 37060, Buttapietra, (VR), Italy
| | - Marianna Merenda
- Unità Micoplasmi, WOAH Reference Laboratory for Avian Mycoplasmosis (M. Gallisepticum, M. Synoviae), Istituto Zooprofilattico Sperimentale delle Venezie, 37060, Buttapietra, (VR), Italy
| |
Collapse
|
21
|
Long J, Yang C, Liu J, Ma C, Jiao M, Hu H, Xiong J, Zhang Y, Wei W, Yang H, He Y, Zhu M, Yu Y, Fu L, Chen H. Tannic acid inhibits Escherichia coli biofilm formation and underlying molecular mechanisms: Biofilm regulator CsgD. Biomed Pharmacother 2024; 175:116716. [PMID: 38735084 DOI: 10.1016/j.biopha.2024.116716] [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: 02/29/2024] [Revised: 04/27/2024] [Accepted: 05/06/2024] [Indexed: 05/14/2024] Open
Abstract
Biofilms often engender persistent infections, heightened antibiotic resistance, and the recurrence of infections. Therefor, infections related to bacterial biofilms are often chronic and pose challenges in terms of treatment. The main transcription regulatory factor, CsgD, activates csgABC-encoded curli to participate in the composition of extracellular matrix, which is an important skeleton for biofilm development in enterobacteriaceae. In our previous study, a wide range of natural bioactive compounds that exhibit strong affinity to CsgD were screened and identified via molecular docking. Tannic acid (TA) was subsequently chosen, based on its potent biofilm inhibition effect as observed in crystal violet staining. Therefore, the aim of this study was to investigate the specific effects of TA on the biofilm formation of clinically isolated Escherichia coli (E. coli). Results demonstrated a significant inhibition of E. coli Ec032 biofilm formation by TA, while not substantially affecting the biofilm of the ΔcsgD strain. Moreover, deletion of the csgD gene led to a reduction in Ec032 biofilm formation, alongside diminished bacterial motility and curli synthesis inhibition. Transcriptomic analysis and RT-qPCR revealed that TA repressed genes associated with the csg operon and other biofilm-related genes. In conclusion, our results suggest that CsgD is one of the key targets for TA to inhibit E. coli biofilm formation. This work preliminarily elucidates the molecular mechanisms of TA inhibiting E. coli biofilm formation, which could provide a lead structure for the development of future antibiofilm drugs.
Collapse
Affiliation(s)
- Jinying Long
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; National Center of Technology Innovation for Pigs, Chongqing 402460, China; Immunology Research Center, Medical Research Institute, Southwest University, Chongqing 402460, China
| | - Can Yang
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; National Center of Technology Innovation for Pigs, Chongqing 402460, China; Immunology Research Center, Medical Research Institute, Southwest University, Chongqing 402460, China
| | - JingJing Liu
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; National Center of Technology Innovation for Pigs, Chongqing 402460, China
| | - Chengjun Ma
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; National Center of Technology Innovation for Pigs, Chongqing 402460, China
| | - Min Jiao
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; National Center of Technology Innovation for Pigs, Chongqing 402460, China
| | - Huiming Hu
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Jing Xiong
- National Center of Technology Innovation for Pigs, Chongqing 402460, China; Chongqing Academy of Animal Sciences, Chongqing 402460, China
| | - Yang Zhang
- National Center of Technology Innovation for Pigs, Chongqing 402460, China; Chongqing Academy of Animal Sciences, Chongqing 402460, China
| | - Wei Wei
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; National Center of Technology Innovation for Pigs, Chongqing 402460, China; Traditional Chinese Veterinary Research Institute, Southwest University, Chongqing 402460, China
| | - Hongzao Yang
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; National Center of Technology Innovation for Pigs, Chongqing 402460, China; Traditional Chinese Veterinary Research Institute, Southwest University, Chongqing 402460, China
| | - Yuzhang He
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; Immunology Research Center, Medical Research Institute, Southwest University, Chongqing 402460, China
| | - Maixun Zhu
- National Center of Technology Innovation for Pigs, Chongqing 402460, China; Chongqing Academy of Animal Sciences, Chongqing 402460, China
| | - Yuandi Yu
- National Center of Technology Innovation for Pigs, Chongqing 402460, China; Chongqing Academy of Animal Sciences, Chongqing 402460, China
| | - Lizhi Fu
- National Center of Technology Innovation for Pigs, Chongqing 402460, China; Chongqing Academy of Animal Sciences, Chongqing 402460, China.
| | - Hongwei Chen
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; National Center of Technology Innovation for Pigs, Chongqing 402460, China; Immunology Research Center, Medical Research Institute, Southwest University, Chongqing 402460, China; Traditional Chinese Veterinary Research Institute, Southwest University, Chongqing 402460, China.
| |
Collapse
|
22
|
Jeong GJ, Khan F, Tabassum N, Kim YM. Alteration of oral microbial biofilms by sweeteners. Biofilm 2024; 7:100171. [PMID: 38197082 PMCID: PMC10772577 DOI: 10.1016/j.bioflm.2023.100171] [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: 09/14/2023] [Revised: 11/14/2023] [Accepted: 12/11/2023] [Indexed: 01/11/2024] Open
Abstract
There is a growing interest in using sweeteners for taste improvement in the food and drink industry. Sweeteners were found to regulate the formation or dispersal of structural components of microbial biofilms. Dietary sugars may enhance biofilm formation and facilitate the development of antimicrobial resistance, which has become a major health issue worldwide. In contrast, bulk and non-nutritive sweeteners are also beneficial for managing microbial infections. This review discusses the clinical significance of oral biofilms formed upon the administration of nutritive and non-nutritive sweeteners. The underlying mechanism of action of sweeteners in the regulation of mono- or poly-microbial biofilm formation and destruction is comprehensively discussed. Bulk and non-nutritive sweeteners have also been used in conjunction with antimicrobial substances to reduce microbial biofilm formation. Formulations with bulk and non-nutritive sweeteners have been demonstrated to be particularly efficient in this regard. Finally, future perspectives with respect to advancing our understanding of mechanisms underlying biofilm regulation activities of sweeteners are presented as well. Several alternative strategies for the application of bulk sweeteners and non-nutritive sweeteners have been employed to control the biofilm-forming microbial pathogens. Gaining insight into the underlying mechanisms responsible for enhancing or inhibiting biofilm formation and virulence properties by both mono- and poly-microbial species in the presence of the sweetener is crucial for developing a therapeutic agent to manage microbial infections.
Collapse
Affiliation(s)
- Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Fazlurrahman Khan
- Institute of Fisheries Sciences, Pukyong National University, Busan, 48513, Republic of Korea
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Young-Mog Kim
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
| |
Collapse
|
23
|
Zhou W, Hao J, Guo Y, Zhao C, Zhang M, Zhang S, Han F. Revealing bioresponses of biofilm and flocs to salinity gradient in halophilic biofilm reactor. BIORESOURCE TECHNOLOGY 2024; 401:130727. [PMID: 38643952 DOI: 10.1016/j.biortech.2024.130727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/09/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
Understanding the different biological responses to salinity gradient between coexisting biofilm and flocs is crucial for regulating the ecological function of biofilm system. This study investigated performance, dynamics, and community assembly of biofilm system under 3 %-7% salinity gradient. The removal efficiency of NH4+-N remained stable and exceeded 93 % at 3 %-6% salinity, but decreased to below 80 % at 7 % salinity. The elevated salinity promoted the synthesis of extracellular polymer substrates, inhibited microbial respiration, and significantly regulated the microbial community structure. Compared to flocs, biofilm exhibited greater species diversity and richer Nitrosomonas. It was found diffusion limitations dominated the microbial community assembly under the salinity gradient. And microbial network revealed positive interactions predominated the microbial relationships, designating norank Spirochaetaceae, unclassified Micrococcales, Corynebacterium, and Pusillimonas as keystone species. Moreover, distinct salinity preferences in nitrogen transformation-related genes were observed. This study can improve the understanding to the regulation of biofilm systems to salt stresses.
Collapse
Affiliation(s)
- Weizhi Zhou
- School of Civil Engineering, Shandong University, Jinan, Shandong 250002, China; School of Environmental Science and Engineering, Shandong University, Qingdao, Shandong, China
| | - Jie Hao
- School of Environmental Science and Engineering, Shandong University, Qingdao, Shandong 266000, China; School of Environmental Science and Engineering, Shandong University, Qingdao, Shandong, China
| | - Yiting Guo
- School of Civil Engineering, Shandong University, Jinan, Shandong 250002, China; School of Environmental Science and Engineering, Shandong University, Qingdao, Shandong, China
| | - Chuanfu Zhao
- School of Civil Engineering, Shandong University, Jinan, Shandong 250002, China; School of Environmental Science and Engineering, Shandong University, Qingdao, Shandong, China
| | - Mengru Zhang
- School of Civil Engineering, Shandong University, Jinan, Shandong 250002, China; School of Environmental Science and Engineering, Shandong University, Qingdao, Shandong, China
| | - Shuhui Zhang
- School of Civil Engineering, Shandong University, Jinan, Shandong 250002, China; School of Environmental Science and Engineering, Shandong University, Qingdao, Shandong, China
| | - Fei Han
- School of Civil Engineering, Shandong University, Jinan, Shandong 250002, China; School of Environmental Science and Engineering, Shandong University, Qingdao, Shandong, China.
| |
Collapse
|
24
|
Sil M, Mukherjee D, Goswami A, Nag M, Lahiri D, Bhattacharya D. Antibiofilm activity of mesoporous silica nanoparticles against the biofilm associated infections. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3617-3633. [PMID: 38051365 DOI: 10.1007/s00210-023-02872-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/21/2023] [Indexed: 12/07/2023]
Abstract
In pharmaceutical industries, various chemical carriers are present which are used for drug delivery to the correct target sites. The most popular and upcoming drug delivery carriers are mesoporous silica nanoparticles (MSN). The main reason for its popularity is its ability to be specific and optimize the drug delivery process in a controlled manner. Nowadays, MSNs are widely used to eradicate various microbial infections, especially the ones related to biofilms. Biofilms are sessile groups of cells that live by forming a consortium and exhibit antibacterial resistance (AMR). They exhibit AMR by extracellular polymeric substances (EPS) and various quorum sensing (QS) signaling molecules. Usually, bacterial and fungal cells are capable of forming biofilms. These biofilms are pathogenic. In the majority of the cases, biofilms cause nosocomial diseases. This review will focus on the antibiofilm activities of MSN, its mechanism of target-specific drug delivery, and its ability to disrupt the bacterial biofilms inhibiting the infection. The review will also discuss various mechanisms for the delivery of pharmaceutical molecules by the MSNs to inhibit the bacterial biofilms, and lastly, we will talk about the different types of MSNs and their antibiofilm activities.
Collapse
Affiliation(s)
- Moumita Sil
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, India
| | - Dipro Mukherjee
- Agricultural and Ecological Research Unit, Biological Sciences Division, Indian Statistical Institute, Kolkata, India
| | - Arunava Goswami
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, India
| | - Moupriya Nag
- Department of Biotechnology, Institute of Engineering and Management, Kolkata, New Town, University of Engineering and Management, Kolkata, India
| | - Dibyajit Lahiri
- Department of Biotechnology, Institute of Engineering and Management, Kolkata, New Town, University of Engineering and Management, Kolkata, India.
| | - Debasmita Bhattacharya
- Department of Basic Science and Humanities, Institute of Engineering and Management, Kolkata, Salt Lake, University of Engineering and Management, Kolkata, India
| |
Collapse
|
25
|
Li S, Wang Y, Xu G, Xu Y, Fu C, Zhao Q, Xu L, Jia X, Zhang Y, Liu Y, Qiao J. The combination of allicin with domiphen is effective against microbial biofilm formation. Front Microbiol 2024; 15:1341316. [PMID: 38873153 PMCID: PMC11169630 DOI: 10.3389/fmicb.2024.1341316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 05/15/2024] [Indexed: 06/15/2024] Open
Abstract
Background Microorganisms in biofilms are particularly difficult to control because of their increased survival and antibiotic resistance. Allicin and domiphen were employed to inhibit the microbial growth and biofilm formation of Staphylococcus aureus, Escherichia coli, and Candida albicans strains. Methods Broth microdilution method and checkerboard assay were conducted to determine the efficacy of allicin combined with domiphen against S. aureus, E. coli, and C. albicans. Microbial biofilm formation was measured using the crystal violet staining method and fluorescence microscopy. And the total viable count of the biofilm cells on material surface after the treatment with antimicrobial reagents was calculated with the plate count technique. Results The two drugs showed synergistic effects against the pathogens with a fractional bactericidal concentration of less than 0.38. The combination of 64 μg/mL allicin with 1 μg/mL domiphen dispersed over 50% of the biofilm mass of S. aureus, E. coli, and C. albicans. In addition, the drug combination reduced the total viable counts of E. coli and C. albicans biofilm cells on stainless steel and polyethylene surfaces by more than 102 CFU/mL. Conclusion The combination of allicin and domiphen is an effective strategy for efficiently decreasing biofilms formation on various industrial materials surfaces.
Collapse
Affiliation(s)
- Shang Li
- Department of Biotechnology, School of Life Sciences, Xuzhou Medical University, Xuzhou, China
| | - Yutong Wang
- Department of Biotechnology, School of Life Sciences, Xuzhou Medical University, Xuzhou, China
| | - Geweirong Xu
- Department of Biotechnology, School of Life Sciences, Xuzhou Medical University, Xuzhou, China
| | - Yuqing Xu
- Department of Biotechnology, School of Life Sciences, Xuzhou Medical University, Xuzhou, China
| | - Cuiyan Fu
- Department of Biotechnology, School of Life Sciences, Xuzhou Medical University, Xuzhou, China
| | - Quanlin Zhao
- Department of Biotechnology, School of Life Sciences, Xuzhou Medical University, Xuzhou, China
| | - Linjie Xu
- Department of Biotechnology, School of Life Sciences, Xuzhou Medical University, Xuzhou, China
| | - Xinzhou Jia
- Department of Biotechnology, School of Life Sciences, Xuzhou Medical University, Xuzhou, China
| | - Yumeng Zhang
- Department of Biotechnology, School of Life Sciences, Xuzhou Medical University, Xuzhou, China
| | - Yi Liu
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
- School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiaju Qiao
- Department of Biotechnology, School of Life Sciences, Xuzhou Medical University, Xuzhou, China
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
26
|
Shao L, Shen Z, Li M, Guan C, Fan B, Chai Y, Zhao Y. ccdC Regulates Biofilm Dispersal in Bacillus velezensis FZB42. Int J Mol Sci 2024; 25:5201. [PMID: 38791239 PMCID: PMC11120784 DOI: 10.3390/ijms25105201] [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: 03/14/2024] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Bacillus velezensis FZB42 is a plant growth-promoting rhizobacterium (PGPR) and a model microorganism for biofilm studies. Biofilms are required for the colonization and promotion of plant growth in the rhizosphere. However, little is known about how the final stage of the biofilm life cycle is regulated, when cells regain their motility and escape the mature biofilm to spread and colonize new niches. In this study, the non-annotated gene ccdC was found to be involved in the process of biofilm dispersion. We found that the ccdC-deficient strain maintained a wrinkled state at the late stage of biofilm formation in the liquid-gas interface culture, and the bottom solution showed a clear state, indicating that no bacterial cells actively escaped, which was further evidenced by the formation of a cellular ring (biofilm pellicle) located on top of the preformed biofilm. It can be concluded that dispersal, a biofilm property that relies on motility proficiency, is also positively affected by the unannotated gene ccdC. Furthermore, we found that the level of cyclic diguanylate (c-di-GMP) in the ccdC-deficient strain was significantly greater than that in the wild-type strain, suggesting that B. velezensis exhibits a similar mechanism by regulating the level of c-di-GMP, the master regulator of biofilm formation, dispersal, and cell motility, which controls the fitness of biofilms in Pseudomonas aeruginosain. In this study, we investigated the mechanism regulating biofilm dispersion in PGPR.
Collapse
Affiliation(s)
- Lin Shao
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Forestry and Grass, Nanjing Forestry University, Nanjing 210037, China
- College of Life Science, Nanjing Forestry University, Nanjing 210037, China
| | - Zizhu Shen
- College of Life Science, Nanjing Forestry University, Nanjing 210037, China
| | - Meiju Li
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Forestry and Grass, Nanjing Forestry University, Nanjing 210037, China
| | - Chenyun Guan
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Forestry and Grass, Nanjing Forestry University, Nanjing 210037, China
| | - Ben Fan
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Forestry and Grass, Nanjing Forestry University, Nanjing 210037, China
- College of Life Science, Nanjing Forestry University, Nanjing 210037, China
| | - Yunrong Chai
- Department of Biology, Northeastern University, Boston, MA 02115, USA
| | - Yinjuan Zhao
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Forestry and Grass, Nanjing Forestry University, Nanjing 210037, China
| |
Collapse
|
27
|
Mishra AK, Thakare RP, Santani BG, Yabaji SM, Dixit SK, Srivastava KK. Unlocking the enigma of phenotypic drug tolerance: Mechanisms and emerging therapeutic strategies. Biochimie 2024; 220:67-83. [PMID: 38168626 DOI: 10.1016/j.biochi.2023.12.009] [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: 10/11/2023] [Revised: 12/09/2023] [Accepted: 12/27/2023] [Indexed: 01/05/2024]
Abstract
In the ongoing battle against antimicrobial resistance, phenotypic drug tolerance poses a formidable challenge. This adaptive ability of microorganisms to withstand drug pressure without genetic alterations further complicating global healthcare challenges. Microbial populations employ an array of persistence mechanisms, including dormancy, biofilm formation, adaptation to intracellular environments, and the adoption of L-forms, to develop drug tolerance. Moreover, molecular mechanisms like toxin-antitoxin modules, oxidative stress responses, energy metabolism, and (p)ppGpp signaling contribute to this phenomenon. Understanding these persistence mechanisms is crucial for predicting drug efficacy, developing strategies for chronic bacterial infections, and exploring innovative therapies for refractory infections. In this comprehensive review, we dissect the intricacies of drug tolerance and persister formation, explore their role in acquired drug resistance, and highlight emerging therapeutic approaches to combat phenotypic drug tolerance. Furthermore, we outline the future landscape of interventions for persistent bacterial infections.
Collapse
Affiliation(s)
- Alok K Mishra
- Division of Microbiology, CSIR-Central Drug Research Institute (CDRI), Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India; Department of Molecular Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA, 01605, USA.
| | - Ritesh P Thakare
- Division of Microbiology, CSIR-Central Drug Research Institute (CDRI), Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India; Department of Molecular Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Bela G Santani
- Department of Microbiology, Sant Gadge Baba Amravati University (SGBAU), Amravati, Maharashtra, India
| | - Shivraj M Yabaji
- Division of Microbiology, CSIR-Central Drug Research Institute (CDRI), Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India; National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, USA
| | - Shivendra K Dixit
- Division of Medicine ICAR-Indian Veterinary Research Institute (IVRI), Izatnagar Bareilly, Uttar Pradesh, 243122, India.
| | - Kishore K Srivastava
- Division of Microbiology, CSIR-Central Drug Research Institute (CDRI), Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India.
| |
Collapse
|
28
|
Suardi V, Baroni D, Shahein AHA, Morena V, Logoluso N, Mangiavini L, Pellegrini AV. Microbiology of Prosthetic Joint Infections: A Retrospective Study of an Italian Orthopaedic Hospital. Antibiotics (Basel) 2024; 13:399. [PMID: 38786128 PMCID: PMC11117340 DOI: 10.3390/antibiotics13050399] [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: 03/05/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
The most frequent cause of periprosthetic infections (PJIs) is intraoperative contamination; hence, antibiotic prophylaxis plays a crucial role in prevention. Modifications to standard prophylaxis can be considered if there is a high incidence of microorganisms resistant to current protocols. To date, very few studies regarding microbial etiology have been published in Italy. In this single-center, retrospective study conducted at IRCCS Ospedale Galeazzi-Sant'Ambrogio in Milan, we analyzed hip, knee, and shoulder PJIs in patients undergoing first implantation between 1 January 17 and 31 December 2021. The primary aim was to derive a local microbiological case history. The secondary aim was to evaluate the adequacy of preoperative antibiotic prophylaxis in relation to the identified bacteria. A total of 57 PJIs and 65 pathogens were identified: 16 S. aureus, 15 S. epidermidis, and 10 other coagulase-negative staphylococci (CoNS), which accounted for 63% of the isolations. A total of 86.7% of S. epidermidis were methicillin-resistant (MRSE). In line with other case reports, we found a predominance of staphylococcal infections, with a lower percentage of MRSA than the Italian average, while we found a high percentage of MRSE. We estimated that 44.6% of the bacteria isolated were resistant to cefazolin, our standard prophylaxis. These PJIs could be prevented by using glycopeptide alone or in combination with cefazolin, but the literature reports conflicting results regarding the adequacy of such prophylaxis. In conclusion, our study showed that in our local hospital, our standard antibiotic prophylaxis is ineffective for almost half of the cases, highlighting the importance of defining specific antibiotic guidelines based on the local bacterial prevalence of each institution.
Collapse
Affiliation(s)
- Virginia Suardi
- IRCCS Ospedale Galeazzi–Sant’Ambrogio, 20157 Milan, Italy; (V.S.); (N.L.); (A.V.P.)
| | - Daniele Baroni
- Department of Orthopedics and Traumatology, Alessandro Manzoni Hospital, 23900 Lecco, Italy;
| | | | - Valentina Morena
- Infectious Disease Unit, Alessandro Manzoni Hospital, 23900 Lecco, Italy;
| | - Nicola Logoluso
- IRCCS Ospedale Galeazzi–Sant’Ambrogio, 20157 Milan, Italy; (V.S.); (N.L.); (A.V.P.)
| | - Laura Mangiavini
- IRCCS Ospedale Galeazzi–Sant’Ambrogio, 20157 Milan, Italy; (V.S.); (N.L.); (A.V.P.)
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | | |
Collapse
|
29
|
Hu XM, Peng L, Wu J, Wu G, Liang X, Yang JL. Bacterial c-di-GMP signaling gene affects mussel larval metamorphosis through outer membrane vesicles and lipopolysaccharides. NPJ Biofilms Microbiomes 2024; 10:38. [PMID: 38575604 PMCID: PMC10994910 DOI: 10.1038/s41522-024-00508-6] [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: 10/16/2023] [Accepted: 03/20/2024] [Indexed: 04/06/2024] Open
Abstract
Biofilms serve as crucial cues for settlement and metamorphosis in marine invertebrates. Within bacterial systems, c-di-GMP functions as a pivotal signaling molecule regulating both biofilm formation and dispersion. However, the molecular mechanism of how c-di-GMP modulates biofilm-induced larval metamorphosis remains elusive. Our study reveals that the deletion of a c-di-GMP related gene in Pseudoalteromonas marina led to an increase in the level of bacterial c-di-GMP by knockout technique, and the mutant strain had an enhanced ability to produce more outer membrane vesicles (OMVs) and lipopolysaccharides (LPS). The mutant biofilms had higher induction activity for larval metamorphosis in mussels Mytilus coruscus, and OMVs play a major role in the induction activity. We further explored the function of LPS in OMVs. Extracted LPS induced high larval metamorphosis rate, and LPS content were subject to c-di-GMP and LPS-biosynthesis gene. Thus, we postulate that the impact of c-di-GMP on biofilm-induced metamorphosis is mediated through OMVs and LPS.
Collapse
Affiliation(s)
- Xiao-Meng Hu
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
- Shanghai Collaborative Innovation Center for Cultivating Elite Breeds and Green-Culture of Aquaculture Animals, Shanghai, 201306, China
| | - Lihua Peng
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
- Shanghai Collaborative Innovation Center for Cultivating Elite Breeds and Green-Culture of Aquaculture Animals, Shanghai, 201306, China
| | - Jingxian Wu
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
- Shanghai Collaborative Innovation Center for Cultivating Elite Breeds and Green-Culture of Aquaculture Animals, Shanghai, 201306, China
| | - Guanju Wu
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China
- Shanghai Collaborative Innovation Center for Cultivating Elite Breeds and Green-Culture of Aquaculture Animals, Shanghai, 201306, China
| | - Xiao Liang
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China.
- Shanghai Collaborative Innovation Center for Cultivating Elite Breeds and Green-Culture of Aquaculture Animals, Shanghai, 201306, China.
| | - Jin-Long Yang
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, China.
- Shanghai Collaborative Innovation Center for Cultivating Elite Breeds and Green-Culture of Aquaculture Animals, Shanghai, 201306, China.
| |
Collapse
|
30
|
Jin C, Sengupta A. Microbes in porous environments: from active interactions to emergent feedback. Biophys Rev 2024; 16:173-188. [PMID: 38737203 PMCID: PMC11078916 DOI: 10.1007/s12551-024-01185-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/27/2024] [Indexed: 05/14/2024] Open
Abstract
Microbes thrive in diverse porous environments-from soil and riverbeds to human lungs and cancer tissues-spanning multiple scales and conditions. Short- to long-term fluctuations in local factors induce spatio-temporal heterogeneities, often leading to physiologically stressful settings. How microbes respond and adapt to such biophysical constraints is an active field of research where considerable insight has been gained over the last decades. With a focus on bacteria, here we review recent advances in self-organization and dispersal in inorganic and organic porous settings, highlighting the role of active interactions and feedback that mediates microbial survival and fitness. We discuss open questions and opportunities for using integrative approaches to advance our understanding of the biophysical strategies which microbes employ at various scales to make porous settings habitable.
Collapse
Affiliation(s)
- Chenyu Jin
- Physics of Living Matter Group, Department of Physics and Materials Science, University of Luxembourg, 162 A, Avenue de la Faïencerie, Luxembourg City, L-1511 Luxembourg
| | - Anupam Sengupta
- Physics of Living Matter Group, Department of Physics and Materials Science, University of Luxembourg, 162 A, Avenue de la Faïencerie, Luxembourg City, L-1511 Luxembourg
- Institute for Advanced Studies, University of Luxembourg, 2 Avenue de l’Université, Esch-sur-Alzette, L-4365 Luxembourg
| |
Collapse
|
31
|
Ratsoma FM, Mokoena NZ, Santana QC, Wingfield BD, Steenkamp ET, Motaung TE. Characterization of the Fusarium circinatum biofilm environmental response role. J Basic Microbiol 2024; 64:e2300536. [PMID: 38314962 DOI: 10.1002/jobm.202300536] [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: 09/15/2023] [Revised: 01/10/2024] [Accepted: 01/13/2024] [Indexed: 02/07/2024]
Abstract
The capacity to form biofilms is a common trait among many microorganisms present on Earth. In this study, we demonstrate for the first time that the fatal pine pitch canker agent, Fusarium circinatum, can lead a biofilm-like lifestyle with aggregated hyphal bundles wrapped in extracellular matrix (ECM). Our research shows F. circinatum's ability to adapt to environmental changes by assuming a biofilm-like lifestyle. This was demonstrated by varying metabolic activities exhibited by the biofilms in response to factors like temperature and pH. Further analysis revealed that while planktonic cells produced small amounts of ECM per unit of the biomass, heat- and azole-exposed biofilms produced significantly more ECM than nonexposed biofilms, further demonstrating the adaptability of F. circinatum to changing environments. The increased synthesis of ECM triggered by these abiotic factors highlights the link between ECM production in biofilm and resistance to abiotic stress. This suggests that ECM-mediated response may be one of the key survival strategies of F. circinatum biofilms in response to changing environments. Interestingly, azole exposure also led to biofilms that were resistant to DNase, which typically uncouples biofilms by penetrating the biofilm and degrading its extracellular DNA; we propose that DNases were likely hindered from reaching target cells by the ECM barricade. The interplay between antifungal treatment and DNase enzyme suggests a complex relationship between eDNA, ECM, and antifungal agents in F. circinatum biofilms. Therefore, our results show how a phytopathogen's sessile (biofilm) lifestyle could influence its response to the surrounding environment.
Collapse
Affiliation(s)
- Francinah M Ratsoma
- Department of Biochemistry, Genetics, and Microbiology, Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Pretoria, South Africa
| | - Nthabiseng Z Mokoena
- Department of Biochemistry, Genetics, and Microbiology, Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Pretoria, South Africa
| | - Quentin C Santana
- Department of Biochemistry, Genetics, and Microbiology, Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Pretoria, South Africa
- Agricultural Research Council (ARC) Biotechnology Platform, Private Bag X5 Onderstepoort, Pretoria, South Africa
| | - Brenda D Wingfield
- Department of Biochemistry, Genetics, and Microbiology, Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Pretoria, South Africa
| | - Emma T Steenkamp
- Department of Biochemistry, Genetics, and Microbiology, Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Pretoria, South Africa
| | - Thabiso E Motaung
- Department of Biochemistry, Genetics, and Microbiology, Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Pretoria, South Africa
| |
Collapse
|
32
|
Saini P, Bandsode V, Singh A, Mendem SK, Semmler T, Alam M, Ahmed N. Genomic insights into virulence, antimicrobial resistance, and adaptation acumen of Escherichia coli isolated from an urban environment. mBio 2024; 15:e0354523. [PMID: 38376265 PMCID: PMC10936179 DOI: 10.1128/mbio.03545-23] [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: 01/11/2024] [Accepted: 01/23/2024] [Indexed: 02/21/2024] Open
Abstract
Populations of common commensal bacteria such as Escherichia coli undergo genetic changes by the acquisition of certain virulence and antimicrobial resistance (AMR) encoding genetic elements leading to the emergence of pathogenic strains capable of surviving in the previously uninhabited or protected niches. These bacteria are also reported to be prevalent in the environment where they survive by adopting various recombination strategies to counter microflora of the soil and water, under constant selection pressure(s). In this study, we performed molecular characterization, phenotypic AMR analysis, and whole genome sequencing (WGS) of E. coli (n = 37) isolated from soil and surface water representing the urban and peri-urban areas. The primary aim of this study was to understand the genetic architecture and pathogenic acumen exhibited by environmental E. coli. WGS-based analysis entailing resistome and virulome profiling indicated the presence of various virulence (adherence, iron uptake, and toxins) and AMR encoding genes, including blaNDM-5 in the environmental isolates. A majority of our isolates belonged to phylogroup B1 (73%). A few isolates in our collection were of sequence type(s) (ST) 58 and 224 that could have emerged recently as clonal lineages and might pose risk of infection/transmission. Mobile genetic elements (MGEs) such as plasmids (predominantly) of the IncF family, prophages, pipolins, and insertion elements such as IS1 and IS5 were also observed to exist, which may presumably aid in the propagation of genes encoding resistance against antimicrobial drugs. The observed high prevalence of MGEs associated with multidrug resistance in pathogenic E. coli isolates belonging to the phylogroup B1 underscores the need for extended surveillance to keep track of and prevent the transmission of the bacterium to certain vulnerable human and animal populations. IMPORTANCE Evolutionary patterns of E. coli bacteria convey that they evolve into highly pathogenic forms by acquiring fitness advantages, such as AMR, and various virulence factors through the horizontal gene transfer (HGT)-mediated acquisition of MGEs. However, limited research on the genetic profiles of environmental E. coli, particularly from India, hinders our understanding of their transition to pathogenic forms and impedes the adoption of a comprehensive approach to address the connection between environmentally dwelling E. coli populations and human and veterinary public health. This study focuses on high-resolution genomic analysis of the environmental E. coli isolates aiming to understand the genetic similarities and differences among isolates from different environmental niches and uncover the survival strategies employed by these bacteria to thrive in their surroundings. Our approach involved molecular characterization of environmental samples using PCR-based DNA fingerprinting and subsequent WGS analysis. This multidisciplinary approach is likely to provide valuable insights into the understanding of any potential spill-over to human and animal populations and locales. Investigating these environmental isolates has significant potential for developing epidemiological strategies against transmission and understanding niche-specific evolutionary patterns.
Collapse
Affiliation(s)
- Poorvi Saini
- Department of Biotechnology and Bioinformatics, Pathogen Biology Laboratory, University of Hyderabad, Hyderabad, Telangana State, India
| | - Viraj Bandsode
- Department of Biotechnology and Bioinformatics, Pathogen Biology Laboratory, University of Hyderabad, Hyderabad, Telangana State, India
| | - Anuradha Singh
- Department of Biotechnology and Bioinformatics, Pathogen Biology Laboratory, University of Hyderabad, Hyderabad, Telangana State, India
| | - Suresh Kumar Mendem
- Department of Biotechnology and Bioinformatics, Pathogen Biology Laboratory, University of Hyderabad, Hyderabad, Telangana State, India
| | | | - Munirul Alam
- International Centre for Diarrheal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Niyaz Ahmed
- Department of Biotechnology and Bioinformatics, Pathogen Biology Laboratory, University of Hyderabad, Hyderabad, Telangana State, India
| |
Collapse
|
33
|
Cooney C, Sommer B, Marzinelli EM, Figueira WF. The role of microbial biofilms in range shifts of marine habitat-forming organisms. Trends Microbiol 2024; 32:190-199. [PMID: 37633773 DOI: 10.1016/j.tim.2023.07.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/28/2023]
Abstract
Marine species, such as corals and kelp, are responding to climate change by altering their distributions. Microbial biofilms underpin key processes that affect the establishment, maintenance, and function of these dominant habitat-formers. Climate-mediated changes to microbial biofilms can therefore strongly influence species' range shifts. Here, we review emerging research on the interactions between benthic biofilms and habitat-formers and identify two key areas of interaction where climate change can impact this dynamic: (i) via direct effects on biofilm composition, and (ii) via impacts on the complex feedback loops which exist between the biofilm microbes and habitat-forming organisms. We propose that these key interactions will be fundamental in driving the speed and extent of tropicalisation of coastal ecosystems under climate change.
Collapse
Affiliation(s)
- Christopher Cooney
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Brigitte Sommer
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia; School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Ezequiel M Marzinelli
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia; Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore
| | - Will F Figueira
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
34
|
Ioannou P, Baliou S, Samonis G. Nanotechnology in the Diagnosis and Treatment of Antibiotic-Resistant Infections. Antibiotics (Basel) 2024; 13:121. [PMID: 38391507 PMCID: PMC10886108 DOI: 10.3390/antibiotics13020121] [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: 01/04/2024] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
The development of antimicrobial resistance (AMR), along with the relative reduction in the production of new antimicrobials, significantly limits the therapeutic options in infectious diseases. Thus, novel treatments, especially in the current era, where AMR is increasing, are urgently needed. There are several ongoing studies on non-classical therapies for infectious diseases, such as bacteriophages, antimicrobial peptides, and nanotechnology, among others. Nanomaterials involve materials on the nanoscale that could be used in the diagnosis, treatment, and prevention of infectious diseases. This review provides an overview of the applications of nanotechnology in the diagnosis and treatment of infectious diseases from a clinician's perspective, with a focus on pathogens with AMR. Applications of nanomaterials in diagnosis, by taking advantage of their electrochemical, optic, magnetic, and fluorescent properties, are described. Moreover, the potential of metallic or organic nanoparticles (NPs) in the treatment of infections is also addressed. Finally, the potential use of NPs in the development of safe and efficient vaccines is also reviewed. Further studies are needed to prove the safety and efficacy of NPs that would facilitate their approval by regulatory authorities for clinical use.
Collapse
Affiliation(s)
- Petros Ioannou
- School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Stella Baliou
- School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - George Samonis
- School of Medicine, University of Crete, 71003 Heraklion, Greece
- First Department of Medical Oncology, Metropolitan Hospital of Neon Faliron, 18547 Athens, Greece
| |
Collapse
|
35
|
Scotti R, Casciaro B, Stringaro A, Maggi F, Colone M, Gabbianelli R. Fighting Microbial Infections from Escherichia coli O157:H7: The Combined Use of Three Essential Oils of the Cymbopogon Genus and a Derivative of Esculentin-1a Peptide. Antibiotics (Basel) 2024; 13:86. [PMID: 38247645 PMCID: PMC10812396 DOI: 10.3390/antibiotics13010086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/23/2024] Open
Abstract
The absence of effective therapy against Escherichia coli O157:H7 infections has led to the need to develop new antimicrobial agents. As the use of synergistic combinations of natural antimicrobial compounds is growing as a new weapon in the fight against multidrug-resistant bacteria, here, we have tested new synergistic combinations of natural agents. Notably, we investigated a possible synergistic effect of combinations of essential oils and natural peptides to counteract the formation of biofilm. We chose three essential oils (i.e., Cymbopogon citratus, C. flexuosus and C. martinii) and one peptide already studied in our previous works. We determined the fractional inhibitory concentration (FIC) by analyzing the combination of the peptide derived from esculentin-1a, Esc(1-21), with the three essential oils. We also studied the effects of combinations by time-kill curves, scanning electron microscopy on biofilm and Sytox Green on cell membrane permeability. Finally, we analyzed the expression of different genes implicated in motility, biofilm formation and stress responses. The results showed a different pattern of gene expression in bacteria treated with the mixtures compared to those treated with the peptide or the single C. citratus essential oil. In conclusion, we demonstrated that the three essential oils used in combination with the peptide showed synergy against the E. coli O157:H7, proving attractive as an alternative strategy against E. coli pathogen infections.
Collapse
Affiliation(s)
- Raffaella Scotti
- Biological Service, Italian National Institute of Health, 00161 Rome, Italy;
| | - Bruno Casciaro
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy;
| | - Annarita Stringaro
- National Center for Drug Research and Evaluation, Italian National Institute of Health, 00161 Rome, Italy; (A.S.); (M.C.)
| | - Filippo Maggi
- Chemistry Interdisciplinary Project (ChIP) Research Center, School of Pharmacy, University of Camerino, 62032 Camerino, Italy;
| | - Marisa Colone
- National Center for Drug Research and Evaluation, Italian National Institute of Health, 00161 Rome, Italy; (A.S.); (M.C.)
| | - Roberta Gabbianelli
- Biological Service, Italian National Institute of Health, 00161 Rome, Italy;
| |
Collapse
|
36
|
Liu B, Lee CW, Bong CW, Wang AJ. Investigating Escherichia coli habitat transition from sediments to water in tropical urban lakes. PeerJ 2024; 12:e16556. [PMID: 38223759 PMCID: PMC10788090 DOI: 10.7717/peerj.16556] [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: 07/26/2023] [Accepted: 11/09/2023] [Indexed: 01/16/2024] Open
Abstract
Background Escherichia coli is a commonly used faecal indicator bacterium to assess the level of faecal contamination in aquatic habitats. However, extensive studies have reported that sediment acts as a natural reservoir of E. coli in the extraintestinal environment. E. coli can be released from the sediment, and this may lead to overestimating the level of faecal contamination during water quality surveillance. Thus, we aimed to investigate the effects of E. coli habitat transition from sediment to water on its abundance in the water column. Methods This study enumerated the abundance of E. coli in the water and sediment at five urban lakes in the Kuala Lumpur-Petaling Jaya area, state of Selangor, Malaysia. We developed a novel method for measuring habitat transition rate of sediment E. coli to the water column, and evaluated the effects of habitat transition on E. coli abundance in the water column after accounting for its decay in the water column. Results The abundance of E. coli in the sediment ranged from below detection to 12,000 cfu g-1, and was about one order higher than in the water column (1 to 2,300 cfu mL-1). The habitat transition rates ranged from 0.03 to 0.41 h-1. In contrast, the E. coli decay rates ranged from 0.02 to 0.16 h-1. In most cases (>80%), the habitat transition rates were higher than the decay rates in our study. Discussion Our study provided a possible explanation for the persistence of E. coli in tropical lakes. To the best of our knowledge, this is the first quantitative study on habitat transition of E. coli from sediments to water column.
Collapse
Affiliation(s)
- Boyu Liu
- Laboratory of Microbial Ecology, Institute of Biological Science, Faculty of Science, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Choon Weng Lee
- Laboratory of Microbial Ecology, Institute of Biological Science, Faculty of Science, Universiti Malaya, Kuala Lumpur, Malaysia
- Institute of Ocean and Earth Sciences, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Chui Wei Bong
- Laboratory of Microbial Ecology, Institute of Biological Science, Faculty of Science, Universiti Malaya, Kuala Lumpur, Malaysia
- Institute of Ocean and Earth Sciences, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Ai-Jun Wang
- Laboratory of Coastal and Marine Geology, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, Fujian, China
- Fujian Provincial Key Laboratory of Marine Physical and Geological Processes, Xiamen, Fujian, China
| |
Collapse
|
37
|
Khan MA, Shahid M, Celik I, Khan HM, Shahzad A, Husain FM, Adil M. Attenuation of quorum sensing regulated virulence functions and biofilm of pathogenic bacteria by medicinal plant Artemisia annua and its phytoconstituent 1, 8-cineole. Microsc Res Tech 2024; 87:133-148. [PMID: 37728140 DOI: 10.1002/jemt.24418] [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: 05/27/2023] [Revised: 07/26/2023] [Accepted: 09/03/2023] [Indexed: 09/21/2023]
Abstract
The emergence of multidrug resistance (MDR) in bacterial pathogens is a serious public health concern. A significant therapeutic target for MDR infections is the quorum sensing-regulated bacterial pathogenicity. Determining the anti-quorum sensing abilities of certain medicinal plants against bacterial pathogens as well as the in-silico interactions of particular bioactive phytocompounds with QS and biofilm-associated proteins were the objectives of the present study. In this study, 6 medicinal plants were selected based on their ethnopharmacological usage, screened for Anti-QS activity and Artemisia annua leaf extract (AALE) demonstrated pigment inhibitory activity against Chromobacterium violaceum CV12472. Further, the methanol active fraction significantly inhibited the virulence factors (pyocyanin, pyoverdine, rhamnolipid and swarming motility) of Pseudomonas aeruginosa PAO1 and Serratia marcescens MTCC 97 at respective sub-MICs. The inhibition of biofilm was determined using a microtiter plate test and scanning electron microscopy. Biofilm formation was impaired by 70%, 72% and 74% in P. aeruginosa, C. violaceum and S. marcescens, respectively at 0.5xMIC of the extract. The phytochemical content of the extract was studied using GC-MS and 1, 8-cineole was identified as major bioactive compound. Furthermore, 1, 8-cineole was docked with quorum sensing (QS) proteins (LasI, LasR, CviR, and rhlR) and biofilm proteins (PilY1 and PilT). In silico docking and dynamics simulations studies suggested interactions with QS-receptors CviR', LasI, LasR, and biofilm proteins PilY1, PilT for anti-QS activity. Further, 1, 8-cineole demonstrated 66% and 51% reduction in violacein production and biofilm formation, respectively to validate the findings of computational analysis. Findings of the present investigation suggests that 1, 8-cineole plays a crucial role in the QS and biofilm inhibitory activity demonstrated by Artemisia annua extract. RESEARCH HIGHLIGHTS: Artemisia annua leaf extract (AALE) methanol fraction demonstrated broad-spectrum QS and biofilm inhibition Scanning electron microscopy (SEM) confirmed biofilm inhibition Molecular docking and simulation studies suggested positive interactions of 1,8-cineol with QS-receptors and biofilm proteins.
Collapse
Affiliation(s)
- Mo Ahamad Khan
- Department of Microbiology, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Mohammad Shahid
- Department of Microbiology, Immunology and Infectious Diseases, College of Medicine and Medical Science, Arabian Gulf University, Manama, Bahrain
| | - Ismail Celik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey
| | - Haris M Khan
- Department of Microbiology, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Anwar Shahzad
- Department of Botany, Faculty of Life Science, Aligarh Muslim University, Aligarh, India
| | - Fohad Mabood Husain
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Mohd Adil
- Department of Environmental Sciences, Dalhousie University, Truro, Nova Scotia, Canada
| |
Collapse
|
38
|
Suresh G, Srivastava S. A concise review on genes involved in biofilm-related disease and differential gene expression in medical-related biofilms. MICROBIAL BIOFILMS 2024:215-235. [DOI: 10.1016/b978-0-443-19252-4.00012-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
39
|
Jung H. A pore-scale reactive transport modeling study for quorum sensing-driven biofilm dispersal in heterogeneous porous media. Math Biosci 2024; 367:109126. [PMID: 38070765 DOI: 10.1016/j.mbs.2023.109126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/26/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
Microorganisms regulate the expression of energetically expensive phenotypes via a collective decision-making mechanism known as quorum sensing (QS). This study investigates the intricate dynamics of biofilm growth and QS-controlled biofilm dispersal in heterogeneous porous media, employing a pore-scale reactive transport modeling approach. Model simulations carried out under various fluid flow conditions and biofilm growth scenarios reveal that QS processes are influenced not only by the biomass density of biofilm colonies but also by a complex interplay between pore architecture, flow velocity, and the rates of biofilm growth and dispersal. This study demonstrates that pore architecture controls the initiation of QS processes and advection gives rise to oscillatory growth of biofilms. Such oscillation is suppressed if biofilm dynamics are in favor of sustaining a sufficiently high signal concentration, such as fast growth or slow dispersal rates. By establishing a mathematical framework, this study contributes to the fundamental understanding of QS-controlled biofilm dynamics in complex environments.
Collapse
Affiliation(s)
- Heewon Jung
- Department of Geological Sciences, Chungnam National University, Daejeon 34134, South Korea.
| |
Collapse
|
40
|
Jiang S, Chen H, Shen P, Zhou Y, Li Q, Zhang J, Chen Y. Gasotransmitter Research Advances in Respiratory Diseases. Antioxid Redox Signal 2024; 40:168-185. [PMID: 37917094 DOI: 10.1089/ars.2023.0410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Significance: Gasotransmitters are small gas molecules that are endogenously generated and have well-defined physiological functions. The most well-defined gasotransmitters currently are nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S), while other potent gasotransmitters include ammonia, methane, cyanide, hydrogen gas, and sulfur dioxide. Gasotransmitters play a role in various respiratory diseases such as asthma, chronic obstructive pulmonary disease, obstructive sleep apnea, lung infection, bronchiectasis, cystic fibrosis, primary ciliary dyskinesia, and COVID-19. Recent Advances: Gasotransmitters can act as biomarkers that facilitate disease diagnosis, indicate disease severity, predict disease exacerbation, and evaluate disease outcomes. They also have cell-protective properties, and many studies have been conducted to explore their pharmacological applications. Innovative drug donors and drug delivery methods have been invented to amplify their therapeutic effects. Critical Issues: In this article, we briefly reviewed the physiological and pathophysiological functions of some gasotransmitters in the respiratory system, the progress in detecting exhaled gasotransmitters, as well as innovative drugs derived from these molecules. Future Directions: The current challenge for gasotransmitter research includes further exploring their physiological and pathological functions, clarifying their complicated interactions, exploring suitable drug donors and delivery devices, and characterizing new members of gasotransmitters. Antioxid. Redox Signal. 40, 168-185.
Collapse
Affiliation(s)
- Simin Jiang
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Haijie Chen
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Pu Shen
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Yumou Zhou
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Qiaoyu Li
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Jing Zhang
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Yahong Chen
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| |
Collapse
|
41
|
Wang Z, Peng D, Fu C, Luo X, Guo S, Li L, Yin H. Pan-metagenome reveals the abiotic stress resistome of cigar tobacco phyllosphere microbiome. FRONTIERS IN PLANT SCIENCE 2023; 14:1248476. [PMID: 38179476 PMCID: PMC10765411 DOI: 10.3389/fpls.2023.1248476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 11/03/2023] [Indexed: 01/06/2024]
Abstract
The important role of microbial associations in mediating plant protection and responses to abiotic stresses has been widely recognized. However, there have been limited studies on the functional profile of the phyllosphere microbiota from tobacco (Nicotiana tabacum), hindering our understanding of the mechanisms underlying stress resilience in this representative and easy-to-cultivate model species from the solanaceous family. To address this knowledge gap, our study employed shotgun metagenomic sequencing for the first time to analyze the genetic catalog and identify putative plant growth promoting bacteria (PGPB) candidates that confer abiotic stress resilience throughout the growth period of cigar tobacco in the phyllosphere. We identified abundant genes from specific bacterial lineages, particularly Pseudomonas, within the cigar tobacco phyllospheric microbiome. These genes were found to confer resilience against a wide range of stressors, including osmotic and drought stress, heavy metal toxicity, temperature perturbation, organic pollutants, oxidative stress, and UV light damage. In addition, we conducted a virome mining analysis on the metagenome to explore the potential roles of viruses in driving microbial adaptation to environmental stresses. Our results identified a total of 3,320 scaffolds predicted to be viral from the cigar tobacco phyllosphere metagenome, with various phages infecting Pseudomonas, Burkholderia, Enterobacteria, Ralstonia, and related viruses. Within the virome, we also annotated genes associated with abiotic stress resilience, such as alkaline phosphatase D (phoD) for nutrient solubilization and glutamate-5-semialdehyde dehydrogenase (proA) for osmolyte synthesis. These findings shed light on the unexplored roles of viruses in facilitating and transferring abiotic stress resilience in the phyllospheric microbiome through beneficial interactions with their hosts. The findings from this study have important implications for agricultural practices, as they offer potential strategies for harnessing the capabilities of the phyllosphere microbiome to enhance stress tolerance in crop plants.
Collapse
Affiliation(s)
- Zhenhua Wang
- Zhangjiajie Tobacco Company of Hunan Province, Zhangjiajie, China
| | - Deyuan Peng
- Zhangjiajie Tobacco Company of Hunan Province, Zhangjiajie, China
| | - Changwu Fu
- Zhangjiajie Tobacco Company of Hunan Province, Zhangjiajie, China
| | - Xianxue Luo
- Zhangjiajie Tobacco Company of Hunan Province, Zhangjiajie, China
| | - Shijie Guo
- Zhangjiajie Tobacco Company of Hunan Province, Zhangjiajie, China
| | - Liangzhi Li
- School of Minerals Processing and Bioengineering, Central South University, Changsha, China
- Key Laboratory of Biometallurgy of Ministry of Education, Central South University, Changsha, China
| | - Huaqun Yin
- School of Minerals Processing and Bioengineering, Central South University, Changsha, China
- Key Laboratory of Biometallurgy of Ministry of Education, Central South University, Changsha, China
| |
Collapse
|
42
|
Khan MA, Celik I, Khan HM, Shahid M, Shahzad A, Kumar S, Ahmed B. Antibiofilm and anti-quorum sensing activity of Psidium guajava L. leaf extract: In vitro and in silico approach. PLoS One 2023; 18:e0295524. [PMID: 38113217 PMCID: PMC10729950 DOI: 10.1371/journal.pone.0295524] [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: 07/25/2023] [Accepted: 11/21/2023] [Indexed: 12/21/2023] Open
Abstract
The quorum sensing mechanism relies on the detection and response to chemical signals, termed autoinducers, which regulate the synthesis of virulence factors including toxins, enzymes, and biofilms. Emerging therapeutic strategies for infection control encompass approaches that attenuate quorum-sensing systems. In this study, we evaluated the antibacterial, anti-quorum sensing, and anti-biofilm activities of Psidium guajava L. methanolic leaf extracts (PGME). Minimum Inhibitory Concentrations (MICs) of PGME were determined as 500 μg/ml for C. violaceum and 1000 μg/ml for P. aeruginosa PAO1. Significantly, even at sub-MIC concentrations, PGME exhibited noteworthy anti-quorum sensing properties, as evidenced by concentration-dependent inhibition of pigment production in C. violaceum 12742. Furthermore, PGME effectively suppressed quorum-sensing controlled virulence factors in P. aeruginosa PAO1, including biofilm formation, pyoverdin, pyocyanin, and rhamnolipid production, with concentration-dependent inhibitory effects. Phytochemical analysis utilizing GC-MS revealed the presence of compounds such as alpha-copaene, caryophyllene, and nerolidol. In-silico docking studies indicated a plausible mechanism for the observed anti-quorum sensing activity, involving favorable binding and interactions with QS-receptors, including RhlR, CviR', LasI, and LasR proteins. These interactions were found to potentially disrupt QS pathways through suppression of AHL production and receptor protein blockade. Collectively, our findings propose PGME as a promising candidate for the treatment of bacterial infections. Its attributes that mitigate biofilm development and impede quorum-sensing mechanisms highlight its potential therapeutic value.
Collapse
Affiliation(s)
- Mo Ahamad Khan
- Department of Microbiology, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Ismail Celik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey
| | - Haris M. Khan
- Department of Microbiology, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Mohammad Shahid
- Department of Microbiology, Immunology and Infectious Diseases, College of Medicine and Medical Science, Arabian Gulf University, Manama, Kingdom of Bahrain
| | - Anwar Shahzad
- Department of Botany, Faculty of Life Science, Aligarh Muslim University, Aligarh, India
| | - Sachin Kumar
- Department of Microbiology, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Bilal Ahmed
- Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, United States of America
| |
Collapse
|
43
|
Kim Y, Anburajan P, Kim H, Oh HS. Inhibiting Biofilm Formation via Simultaneous Application of Nitric Oxide and Quorum Quenching Bacteria. MEMBRANES 2023; 13:836. [PMID: 37888008 PMCID: PMC10608578 DOI: 10.3390/membranes13100836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023]
Abstract
Membrane biofouling is an inevitable challenge in membrane-based water treatment systems such as membrane bioreactors. Recent studies have shown that biological approaches based on bacterial signaling can effectively control biofilm formation. Quorum quenching (QQ) is known to inhibit biofilm growth by disrupting quorum sensing (QS) signaling, while nitric oxide (NO) signaling helps to disperse biofilms. In this study, batch biofilm experiments were conducted to investigate the impact of simultaneously applying NO signaling and QQ for biofilm control using Pseudomonas aeruginosa PAO1 as a model microorganism. The NO treatment involved the injection of NONOates (NO donor compounds) into mature biofilms, while QQ was implemented by immobilizing QQ bacteria (Escherichia coli TOP10-AiiO or Rhodococcus sp. BH4) in alginate or polyvinyl alcohol/alginate beads to preserve the QQ activity. When QQ beads were applied together with (Z)-1-[N-(3-aminopropyl)-N-(n-propyl) amino]diazen-1-ium-1,2-diolate (PAPA NONOate), they achieved a 39.0% to 71.3% reduction in biofilm formation, which was substantially higher compared to their individual applications (16.0% to 54.4%). These findings highlight the significant potential of combining QQ and NO technologies for effective biofilm control across a variety of processes that require enhanced biofilm inhibition.
Collapse
Affiliation(s)
- Youkyoung Kim
- Department of Environmental Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea; (Y.K.); (P.A.); (H.K.)
| | - Parthiban Anburajan
- Department of Environmental Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea; (Y.K.); (P.A.); (H.K.)
- Institute of Environmental Technology, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| | - Hyeok Kim
- Department of Environmental Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea; (Y.K.); (P.A.); (H.K.)
| | - Hyun-Suk Oh
- Department of Environmental Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea; (Y.K.); (P.A.); (H.K.)
- Institute of Environmental Technology, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| |
Collapse
|
44
|
Zanditenas E, Trebicz-Geffen M, Kolli D, Domínguez-García L, Farhi E, Linde L, Romero D, Chapman M, Kolodkin-Gal I, Ankri S. Digestive exophagy of biofilms by intestinal amoeba and its impact on stress tolerance and cytotoxicity. NPJ Biofilms Microbiomes 2023; 9:77. [PMID: 37813896 PMCID: PMC10562373 DOI: 10.1038/s41522-023-00444-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 09/26/2023] [Indexed: 10/11/2023] Open
Abstract
The human protozoan parasite Entamoeba histolytica is responsible for amebiasis, a disease endemic to developing countries. E. histolytica trophozoites colonize the large intestine, primarily feeding on bacteria. However, in the gastrointestinal tract, bacterial cells form aggregates or structured communities called biofilms too large for phagocytosis. Remarkably, trophozoites are still able to invade and degrade established biofilms, utilizing a mechanism that mimics digestive exophagy. Digestive exophagy refers to the secretion of digestive enzymes that promote the digestion of objects too large for direct phagocytosis by phagocytes. E. histolytica cysteine proteinases (CPs) play a crucial role in the degradation process of Bacillus subtilis biofilm. These proteinases target TasA, a major component of the B. subtilis biofilm matrix, also contributing to the adhesion of the parasite to the biofilm. In addition, they are also involved in the degradation of biofilms formed by Gram-negative and Gram-positive enteric pathogens. Furthermore, biofilms also play an important role in protecting trophozoites against oxidative stress. This specific mechanism suggests that the amoeba has adapted to prey on biofilms, potentially serving as an untapped reservoir for novel therapeutic approaches to treat biofilms. Consistently, products derived from the amoeba have been shown to restore antibiotic sensitivity to biofilm cells. In addition, our findings reveal that probiotic biofilms can act as a protective shield for mammalian cells, hindering the progression of the parasite towards them.
Collapse
Affiliation(s)
- Eva Zanditenas
- Department of Molecular Microbiology, Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Meirav Trebicz-Geffen
- Department of Molecular Microbiology, Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Divya Kolli
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, USA
| | - Laura Domínguez-García
- Departamento de Microbiología, Instituto de Hortofruticultura Subtropical y Mediterránea 'La Mayora', Universidad de Málaga-Consejo Superior de Investigaciones Científicas (IHSM-UMA-CSIC), Universidad de Málaga, Málaga, Spain
| | - Einan Farhi
- Technion Genomics Center, Technion - Israel Institute of Technology, Haifa, Israel
| | - Liat Linde
- Technion Genomics Center, Technion - Israel Institute of Technology, Haifa, Israel
| | - Diego Romero
- Departamento de Microbiología, Instituto de Hortofruticultura Subtropical y Mediterránea 'La Mayora', Universidad de Málaga-Consejo Superior de Investigaciones Científicas (IHSM-UMA-CSIC), Universidad de Málaga, Málaga, Spain
| | - Matthew Chapman
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, USA
| | - Ilana Kolodkin-Gal
- Department of Plant Pathology and Microbiology, the Robert H. Smith Faculty of Agriculture, Food & Environment, The Hebrew University of Jerusalem, Rehovot, Israel.
- Scojen Institute for Synthetic Biology, Reichman University, Herzliya, Israel.
| | - Serge Ankri
- Department of Molecular Microbiology, Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel.
| |
Collapse
|
45
|
Liu T, Zhai Y, Jeong KC. Advancing understanding of microbial biofilms through machine learning-powered studies. Food Sci Biotechnol 2023; 32:1653-1664. [PMID: 37780593 PMCID: PMC10533454 DOI: 10.1007/s10068-023-01415-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/26/2023] [Accepted: 08/07/2023] [Indexed: 10/03/2023] Open
Abstract
Microbial biofilms are prevalent in various environments and pose significant challenges to food safety and public health. The biofilms formed by pathogens can cause food spoilage, foodborne illness, and infectious diseases, which are difficult to treat due to their enhanced antimicrobial resistance. While the composition and development of biofilms have been widely studied, their profound impact on food, the food industry, and public health has not been sufficiently recapitulated. This review aims to provide a comprehensive overview of microbial biofilms in the food industry and their implication on public health. It highlights the existence of biofilms along the food-producing chains and the underlying mechanisms of biofilm-associated diseases. Furthermore, this review thoroughly summarizes the enhanced understanding of microbial biofilms achieved through machine learning approaches in biofilm research. By consolidating existing knowledge, this review intends to facilitate developing effective strategies to combat biofilm-associated infections in both the food industry and public health.
Collapse
Affiliation(s)
- Ting Liu
- Emerging Pathogens Institute, University of Florida, 2055 Mowry Rd, Gainesville, FL 32610 USA
- Department of Animal Sciences, University of Florida, 2250 Shealy Dr, Gainesville, FL 32608 USA
| | - Yuting Zhai
- Emerging Pathogens Institute, University of Florida, 2055 Mowry Rd, Gainesville, FL 32610 USA
- Department of Animal Sciences, University of Florida, 2250 Shealy Dr, Gainesville, FL 32608 USA
| | - Kwangcheol Casey Jeong
- Emerging Pathogens Institute, University of Florida, 2055 Mowry Rd, Gainesville, FL 32610 USA
- Department of Animal Sciences, University of Florida, 2250 Shealy Dr, Gainesville, FL 32608 USA
| |
Collapse
|
46
|
van Gestel J, Wagner A, Ackermann M. Pleiotropic hubs drive bacterial surface competition through parallel changes in colony composition and expansion. PLoS Biol 2023; 21:e3002338. [PMID: 37844064 PMCID: PMC10578586 DOI: 10.1371/journal.pbio.3002338] [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: 03/22/2023] [Accepted: 09/18/2023] [Indexed: 10/18/2023] Open
Abstract
Bacteria commonly adhere to surfaces where they compete for both space and resources. Despite the importance of surface growth, it remains largely elusive how bacteria evolve on surfaces. We previously performed an evolution experiment where we evolved distinct Bacilli populations under a selective regime that favored colony spreading. In just a few weeks, colonies of Bacillus subtilis showed strongly advanced expansion rates, increasing their radius 2.5-fold relative to that of the ancestor. Here, we investigate what drives their rapid evolution by performing a uniquely detailed analysis of the evolutionary changes in colony development. We find mutations in diverse global regulators, RicT, RNAse Y, and LexA, with strikingly similar pleiotropic effects: They lower the rate of sporulation and simultaneously facilitate colony expansion by either reducing extracellular polysaccharide production or by promoting filamentous growth. Combining both high-throughput flow cytometry and gene expression profiling, we show that regulatory mutations lead to highly reproducible and parallel changes in global gene expression, affecting approximately 45% of all genes. This parallelism results from the coordinated manner by which regulators change activity both during colony development-in the transition from vegetative growth to dormancy-and over evolutionary time. This coordinated activity can however also break down, leading to evolutionary divergence. Altogether, we show how global regulators function as major pleiotropic hubs that drive rapid surface adaptation by mediating parallel changes in both colony composition and expansion, thereby massively reshaping gene expression.
Collapse
Affiliation(s)
- Jordi van Gestel
- Department of Evolutionary Biology and Environmental Studies, University of Zürich, Zürich, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Environmental Systems Science, ETH Zürich, Zürich, Switzerland
- Department of Environmental Microbiology, Swiss Federal Institute of Aquatic Science and Technology (Eawag), Dübendorf, Switzerland
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California, United States of America
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Andreas Wagner
- Department of Evolutionary Biology and Environmental Studies, University of Zürich, Zürich, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- The Santa Fe Institute, Santa Fe, New Mexico, United States of America
- Stellenbosch Institute for Advanced Study (STIAS), Wallenberg Research Centre at Stellenbosch University, Stellenbosch, South Africa
| | - Martin Ackermann
- Department of Environmental Systems Science, ETH Zürich, Zürich, Switzerland
- Department of Environmental Microbiology, Swiss Federal Institute of Aquatic Science and Technology (Eawag), Dübendorf, Switzerland
| |
Collapse
|
47
|
Hawas S, Qin J, Wiedbrauk S, Fairfull-Smith K, Totsika M. Preclinical Evaluation of Nitroxide-Functionalised Ciprofloxacin as a Novel Antibiofilm Drug Hybrid for Urinary Tract Infections. Antibiotics (Basel) 2023; 12:1479. [PMID: 37887180 PMCID: PMC10604439 DOI: 10.3390/antibiotics12101479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 10/28/2023] Open
Abstract
Urinary tract infections (UTIs) are the second most common bacterial infection with high recurrence rates and can involve biofilm formation on patient catheters. Biofilms are inherently tolerant to antimicrobials, making them difficult to eradicate. Many antibiofilm agents alone do not have bactericidal activity; therefore, linking them to antibiotics is a promising antibiofilm strategy. However, many of these hybrid agents have not been tested in relevant preclinical settings, limiting their potential for clinical translation. Here, we evaluate a ciprofloxacin di-nitroxide hybrid (CDN11), previously reported to have antibiofilm activity against uropathogenic Escherichia coli (UPEC) strain UTI89 in vitro, as a potential UTI therapeutic using multiple preclinical models that reflect various aspects of UTI pathogenesis. We report improved in vitro activity over the parent drug ciprofloxacin against mature UTI89 biofilms formed inside polyethylene catheters. In bladder cell monolayers infected with UTI89, treatment with CDN11 afforded significant reduction in bacterial titers, including intracellular UPEC. Infected mouse bladders containing biofilm-like intracellular reservoirs of UPEC UTI89 showed decreased bacterial loads after ex vivo bladder treatment with CDN11. Activity for CDN11 was reported across different models of UTI, showcasing nitroxide-antibiotic hybridization as a promising antibiofilm approach. The pipeline we described here could be readily used in testing other new therapeutic compounds, fast-tracking the development of novel antibiofilm therapeutics.
Collapse
Affiliation(s)
- Sophia Hawas
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4006, Australia; (S.H.); (J.Q.)
- Max Planck Queensland Centre, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Jilong Qin
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4006, Australia; (S.H.); (J.Q.)
| | - Sandra Wiedbrauk
- School of Chemistry and Physics, Faculty of Science, Queensland University of Technology, Brisbane, QLD 4000, Australia; (S.W.); (K.F.-S.)
- Centre for Materials Science, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Kathryn Fairfull-Smith
- School of Chemistry and Physics, Faculty of Science, Queensland University of Technology, Brisbane, QLD 4000, Australia; (S.W.); (K.F.-S.)
- Centre for Materials Science, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Makrina Totsika
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4006, Australia; (S.H.); (J.Q.)
- Max Planck Queensland Centre, Queensland University of Technology, Brisbane, QLD 4059, Australia
| |
Collapse
|
48
|
El-Saadony MT, Saad AM, Yang T, Salem HM, Korma SA, Ahmed AE, Mosa WFA, Abd El-Mageed TA, Selim S, Al Jaouni SK, Zaghloul RA, Abd El-Hack ME, El-Tarabily KA, Ibrahim SA. Avian campylobacteriosis, prevalence, sources, hazards, antibiotic resistance, poultry meat contamination, and control measures: a comprehensive review. Poult Sci 2023; 102:102786. [PMID: 37454641 PMCID: PMC10371856 DOI: 10.1016/j.psj.2023.102786] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/05/2023] [Accepted: 05/13/2023] [Indexed: 07/18/2023] Open
Abstract
Avian campylobacteriosis is a vandal infection that poses human health hazards. Campylobacter is usually colonized in the avian gut revealing mild signs in the infected birds, but retail chicken carcasses have high contamination levels of Campylobacter spp. Consequently, the contaminated avian products constitute the main source of human infection with campylobacteriosis and result in severe clinical symptoms such as diarrhea, abdominal pain, spasm, and deaths in sensitive cases. Thus, the current review aims to shed light on the prevalence of Campylobacter in broiler chickens, Campylobacter colonization, bird immunity against Campylobacter, sources of poultry infection, antibiotic resistance, poultry meat contamination, human health hazard, and the use of standard antimicrobial technology during the chicken processing of possible control strategies to overcome such problems.
Collapse
Affiliation(s)
- Mohamed T El-Saadony
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | - Ahmed M Saad
- Department of Biochemistry, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | - Tao Yang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Pharmacy, Hainan Medical University, Haikou, 571199, China
| | - Heba M Salem
- Department of Poultry Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Sameh A Korma
- Department of Food Science, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | - Ahmed Ezzat Ahmed
- Biology Department, College of Science, King Khalid University, Abha, 61413, Saudi Arabia; Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha, 61413, Saudi Arabia
| | - Walid F A Mosa
- Plant Production Department (Horticulture-Pomology), Faculty of Agriculture, Saba Basha, Alexandria University, Alexandria, 21531, Egypt
| | - Taia A Abd El-Mageed
- Department of Soils and Water, Faculty of Agriculture, Fayoum University, Fayoum, 63514, Egypt
| | - Samy Selim
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, 72388, Saudi Arabia
| | - Soad K Al Jaouni
- Department of Hematology/Oncology, Yousef Abdulatif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Rashed A Zaghloul
- Department Agricultural Microbiology, Faculty of Agriculture, Benha University, Moshtohor, Qaluybia, 13736, Egypt
| | - Mohamed E Abd El-Hack
- Poultry Department, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | - Khaled A El-Tarabily
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, 15551, United Arab Emirates.
| | - Salam A Ibrahim
- Food Microbiology and Biotechnology Laboratory, Carver Hall, College of Agriculture and Environmental Sciences, North Carolina A & T State University, Greensboro, NC, 27411-1064
| |
Collapse
|
49
|
Saha P, Rafe MR. Cyclodextrin: A prospective nanocarrier for the delivery of antibacterial agents against bacteria that are resistant to antibiotics. Heliyon 2023; 9:e19287. [PMID: 37662769 PMCID: PMC10472013 DOI: 10.1016/j.heliyon.2023.e19287] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/02/2023] [Accepted: 08/17/2023] [Indexed: 09/05/2023] Open
Abstract
Supramolecular chemistry introduces us to the macrocyclic host cyclodextrin, which has a hydrophobic cavity. The hydrophobic cavity has a higher affinity for hydrophobic guest molecules and forms host-guest complexation with non-covalent interaction. Three significant cyclodextrin kinds are α-cyclodextrin, β-cyclodextrin, and γ-cyclodextrin. The most often utilized is β-cyclodextrin (β-CD). An effective weapon against bacteria that are resistant to antibiotics is cyclodextrin. Several different kinds of cyclodextrin nanocarriers (β-CD, HP-β-CD, Meth-β-CD, cationic CD, sugar-grafted CD) are utilized to enhance the solubility, stability, dissolution, absorption, bioavailability, and permeability of the antibiotics. Cyclodextrin also improves the effectiveness of antibiotics, antimicrobial peptides, metallic nanoparticles, and photodynamic therapy (PDT). Again, cyclodextrin nanocarriers offer slow-release properties for sustained-release formulations where steady-state plasma antibiotic concentration is needed for an extended time. A novel strategy to combat bacterial resistance is a stimulus (pH, ROS)-responsive antibiotics released from cyclodextrin carrier. Once again, cyclodextrin traps autoinducer (AI), a crucial part of bacterial quorum sensing, and reduces virulence factors, including biofilm formation. Cyclodextrin helps to minimize MIC in particular bacterial strains, keep antibiotic concentrations above MIC in the infection site and minimize the possibility of antibiotic and biofilm resistance. Sessile bacteria trapped in biofilms are more resistant to antibiotic therapy than bacteria in a planktonic form. Cyclodextrin also involves delivering antibiotics to biofilm and resistant bacteria to combat bacterial resistance.
Collapse
Affiliation(s)
- Pranoy Saha
- Department of Pharmacy, Jagannath University, Dhaka, 1100, Bangladesh
| | - Md Rajdoula Rafe
- Department of Pharmacy, Jagannath University, Dhaka, 1100, Bangladesh
| |
Collapse
|
50
|
Ma Y, Deng Y, Hua H, Khoo BL, Chua SL. Distinct bacterial population dynamics and disease dissemination after biofilm dispersal and disassembly. THE ISME JOURNAL 2023; 17:1290-1302. [PMID: 37270584 PMCID: PMC10356768 DOI: 10.1038/s41396-023-01446-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/05/2023]
Abstract
Microbial communities that form surface-attached biofilms must release and disperse their constituent cells into the environment to colonize fresh sites for continued survival of their species. For pathogens, biofilm dispersal is crucial for microbial transmission from environmental reservoirs to hosts, cross-host transmission, and dissemination of infections across tissues within the host. However, research on biofilm dispersal and its consequences in colonization of fresh sites remain poorly understood. Bacterial cells can depart from biofilms via stimuli-induced dispersal or disassembly due to direct degradation of the biofilm matrix, but the complex heterogeneity of bacterial populations released from biofilms rendered their study difficult. Using a novel 3D-bacterial "biofilm-dispersal-then-recolonization" (BDR) microfluidic model, we demonstrated that Pseudomonas aeruginosa biofilms undergo distinct spatiotemporal dynamics during chemical-induced dispersal (CID) and enzymatic disassembly (EDA), with contrasting consequences in recolonization and disease dissemination. Active CID required bacteria to employ bdlA dispersal gene and flagella to depart from biofilms as single cells at consistent velocities but could not recolonize fresh surfaces. This prevented the disseminated bacteria cells from infecting lung spheroids and Caenorhabditis elegans in on-chip coculture experiments. In contrast, EDA by degradation of a major biofilm exopolysaccharide (Psl) released immotile aggregates at high initial velocities, enabling the bacteria to recolonize fresh surfaces and cause infections in the hosts efficiently. Hence, biofilm dispersal is more complex than previously thought, where bacterial populations adopting distinct behavior after biofilm departure may be the key to survival of bacterial species and dissemination of diseases.
Collapse
Affiliation(s)
- Yeping Ma
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Yanlin Deng
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China
| | - Haojun Hua
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China
| | - Bee Luan Khoo
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China.
- Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE), Kowloon, Hong Kong SAR, 999077, China.
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen-Futian Research Institute, Shenzhen, 518057, China.
| | - Song Lin Chua
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China.
- State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China.
- Shenzhen Key Laboratory of Food Biological Safety Control, Shenzhen, China.
- Research Centre for Deep Space Explorations (RCDSE), The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China.
| |
Collapse
|