1
|
Mei Y, Cao Y, Wang W. Emerging Violet Phosphorus Nanomaterial for Biomedical Applications. Adv Healthc Mater 2025:e2403576. [PMID: 39791284 DOI: 10.1002/adhm.202403576] [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: 09/19/2024] [Revised: 12/07/2024] [Indexed: 01/12/2025]
Abstract
Violet phosphorus (VP) is a phosphorus allotrope first discovered by Hittorf in 1865, which has aroused more attention in the biomedical field in recent years attributed to its gradually discovered unique properties. VP can be further categorized into bulk VP, VP nanosheets (VPNs), and VP quantum dots (VPQDs), and chemical vapor transport (CVT), liquid-phase/mechanical/laser exfoliation, and solvothermal synthesis are the common preparation approaches of bulk VP, VPNs, and VPQDs, respectively. Compared with another phosphorus allotrope (black phosphorus, BP) that is once highly regarded in biomedical applications, VP nanomaterial (namely VPNs and VPQDs) not only exhibits tunable bandgap, moderate on/off current ratio, and good biodegradability, but shows enhanced stability and biosafety as well, allowing it to be a promising candidate for a variety of biomedical applications like antibacterial therapy, anticancer therapy, and biosensing and disease diagnosis. In this review, the classification and the relevant synthesis routes of VP are initially summarized, and the unique properties of VP nanomaterial momentous to its biomedical applications are subsequently expounded. The latest research advances of this emerging nanomaterial in the biomedical field are then introduced in detail, and both the existing challenges and future prospects are also discussed.
Collapse
Affiliation(s)
- Yijun Mei
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, No. 639 Longmian Avenue, Nanjing, 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, No. 639 Longmian Avenue, Nanjing, 211198, P. R. China
| | - Yuanyuan Cao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, No. 639 Longmian Avenue, Nanjing, 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, No. 639 Longmian Avenue, Nanjing, 211198, P. R. China
| | - Wei Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, No. 639 Longmian Avenue, Nanjing, 211198, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, No. 639 Longmian Avenue, Nanjing, 211198, P. R. China
| |
Collapse
|
2
|
Honegger AL, Schweizer TA, Achermann Y, Bosshard PP. Antimicrobial Efficacy of Five Wound Irrigation Solutions in the Periprosthetic Joint Infection Microenvironment In Vitro and Ex Vivo. Antibiotics (Basel) 2025; 14:25. [PMID: 39858311 PMCID: PMC11762658 DOI: 10.3390/antibiotics14010025] [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: 11/20/2024] [Revised: 12/16/2024] [Accepted: 01/01/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Periprosthetic joint infections (PJI) are difficult to treat due to biofilm formation on implant surfaces and the surrounding tissue, often requiring removal or exchange of prostheses along with long-lasting antibiotic treatment. Antiseptic irrigation during revision surgery might decrease bacterial biofilm load and thereby improve treatment success. This in vitro study investigated and compared the effect of five advanced wound irrigation solutions to reduce bacterial burden in the PJI microenvironment. Methods: We treated in vitro biofilms grown on titanium alloy implant discs with clinical bacterial strains isolated from patients with PJIs, as well as abscess communities in a plasma-supplemented collagen matrix. The biofilms were exposed for 1 min to the following wound irrigation solutions: Preventia®, Prontosan®, Granudacyn®, ActiMaris® forte ('Actimaris'), and Octenilin®. We measured the bacterial reduction of these irrigation solutions compared to Ringer-Lactate and to the strong bactericidal but not approved Betaseptic solution. Additionally, ex vivo free-floating bacteria isolated directly from clinical sonication fluids were treated in the same way, and regrowth or lack of regrowth was recorded as the outcome. Results: Irrigation solutions demonstrated variable efficacy. The mean CFU log10 reduction was as follows: Octenilin, 3.07, Preventia, 1.17, Actimaris, 1.11, Prontosan, 1.03, and Granudacyn, 0.61. For SACs, the reduction was: Actimaris, 8.27, Octenilin, 0.58, Prontosan, 0.56, Preventia, 0.35, and Granudacyn, 0.24. Conclusions: All solutions achieved complete bacterial eradication in all tested ex vivo sonication fluids, except Granudacyn, which was ineffective in 33% of the samples (2 out of 6). Advanced wound irrigation solutions have the potential to reduce bacterial burden in the PJI microenvironment during revision surgery. However, their efficacy varies depending on bacterial species, growth state, and the composition of the irrigation solution. This underscores the importance of considering these factors when developing future PJI-specific irrigation solutions.
Collapse
Affiliation(s)
- Anja L. Honegger
- Department of Dermatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland; (A.L.H.); (T.A.S.); (P.P.B.)
| | - Tiziano A. Schweizer
- Department of Dermatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland; (A.L.H.); (T.A.S.); (P.P.B.)
- Department of Cranio-Maxillo-Facial and Oral Surgery, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Yvonne Achermann
- Department of Dermatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland; (A.L.H.); (T.A.S.); (P.P.B.)
- Internal Medicine, Hospital Zollikerberg, 8125 Zollikerberg, Switzerland
| | - Philipp P. Bosshard
- Department of Dermatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland; (A.L.H.); (T.A.S.); (P.P.B.)
| |
Collapse
|
3
|
Li Y, Dong Y, Zhang Z, Lin ZT, Liang C, Wu MX. Efficient Photolysis of Multidrug-Resistant Polymicrobial Biofilms. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2407898. [PMID: 39708333 DOI: 10.1002/advs.202407898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 11/04/2024] [Indexed: 12/23/2024]
Abstract
Chronic wounds are prone to infections with multidrug-resistant bacteria, forming polymicrobial biofilms that limit treatment options and increase the risk of severe complications. Current cleansing options are insufficient to disrupt and remove tenacious biofilms; antibiotic treatments, on the other hand, often fall short against these biofilm-embedded bacteria. This study explores an non-antibiotic approach that extends beyond conventional porphyrin-based phototherapy by using blue light (BL) in conjunction with ferric ions (Fe(III)) to disrupt and eradicate biofilms. The dual not only degraded biofilm extracellular polymeric substances (EPS) in mono-species and polymicrobial biofilms by specifically targeting carboxyl-containing polysaccharides within the matrix but also exhibited broad-spectrum antimicrobial activity by affecting key components of the outer membrane and cell wall. Bacteria, such as K. pneumoniae, with compromised EPS after photolysis, demonstrated increased susceptibility to macrophage phagocytosis. Disruption of the polymicrobial biofilm structure also enhanced the bacterial susceptibility to bactericidal drugs. Treating wounds infected by mixed-species biofilm in diabetic mice demonstrated a substantial reduction in bacterial colonization and improved tissue repair. The BL-Fe(III) modality offers a safe, efficient alternative for managing chronic wound infections, making it ideal for repeated, non-invasive use at home, especially in resource-limited areas.
Collapse
Affiliation(s)
- Yongli Li
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, 50 Blossom Street, Boston, MA, 02114, USA
| | - Yan Dong
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, 50 Blossom Street, Boston, MA, 02114, USA
| | - ZhengKun Zhang
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, 50 Blossom Street, Boston, MA, 02114, USA
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Zuan-Tao Lin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, 50 Blossom Street, Boston, MA, 02114, USA
| | - Chen Liang
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, 50 Blossom Street, Boston, MA, 02114, USA
- Department of Stomatology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing, 100053, P. R. China
| | - Mei X Wu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, 50 Blossom Street, Boston, MA, 02114, USA
| |
Collapse
|
4
|
Yu S, Pan J, Xu M, Chen Y, Li P, Hu H. Antibacterial activity and mechanism of colistin-loaded polymeric nanoparticles for combating multidrug-resistant Pseudomonas aeruginosa biofilms: A synergistic approach. Int J Biol Macromol 2024; 282:136757. [PMID: 39437944 DOI: 10.1016/j.ijbiomac.2024.136757] [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: 03/17/2024] [Revised: 10/16/2024] [Accepted: 10/19/2024] [Indexed: 10/25/2024]
Abstract
Multidrug-resistant P. aeruginosa (MDR-P. aeruginosa), associated with elevated morbidity, mortality, and readmission rates, presents a formidable challenge to eradication due to its robust resistance to antimicrobial agents and biofilm formation. Herein, self-assembling nanoparticles (NO-PE/PLL NPs) comprised of NO donor-conjugated γ-polyglutamic acid (GSNO-PGA), epsilon-poly-l-lysine (PLL) and colistin were fabricated. The negatively charged NO-PE/PLL NPs exhibited effective penetration through airway mucus, reaching the infection site where GSNO-PGA released NO in response to glutathione within biofilm. PLL worked synergistically with colistin (fractional inhibitory concentration index: 0.281), reducing the minimum inhibitory concentration (MIC) of colistin from 2 to 0.5 μg/mL. Benefiting from this synergistic antibacterial action and NO-mediated biofilm disruption, NO-PE/PLL NPs achieved a 99.99 % eradication rate against MDR-P. aeruginosa biofilms. Additionally, NO-PE/PLL NPs efficiently inhibited endotoxins-stimulated inflammation response. In a chronic pulmonary infection model, NO-PE/PLL NPs displayed the highest eradication efficiency (99.78 %) to infected mice, while having no adverse effects on their major organs or pulmonary functions. These results highlight NO-PE/PLL NPs as a promising therapeutic strategy for treating recalcitrant infections caused by MDR-P. aeruginosa biofilms.
Collapse
Affiliation(s)
- Shihui Yu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jieyi Pan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Mao Xu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yujun Chen
- The First Affiliated Hospital of Guangxi Medical University, Guangxi 530000, China
| | - Pengyu Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; Department of Pharmacy, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha 410004, China.
| | - Haiyan Hu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
5
|
Kaushik A, Kest H, Sood M, Thieman C, Steussy BW, Padomek M, Gupta S. Infective Endocarditis by Biofilm-Producing Methicillin-Resistant Staphylococcus aureus-Pathogenesis, Diagnosis, and Management. Antibiotics (Basel) 2024; 13:1132. [PMID: 39766522 PMCID: PMC11672591 DOI: 10.3390/antibiotics13121132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/15/2024] [Accepted: 11/17/2024] [Indexed: 01/11/2025] Open
Abstract
Infective endocarditis (IE) is a life-threatening condition with increasing global incidence, primarily caused by Staphylococcus aureus, especially methicillin-resistant strains (MRSA). Biofilm formation by S. aureus is a critical factor in pathogenesis, contributing to antimicrobial resistance and complicating the treatment of infections involving prosthetic valves and cardiovascular devices. Biofilms provide a protective matrix for MRSA, shielding it from antibiotics and host immune defenses, leading to persistent infections and increased complications, particularly in cases involving prosthetic materials. Clinical manifestations range from acute to chronic presentations, with complications such as heart failure, embolic events, and neurological deficits. Diagnosis relies on the Modified Duke Criteria, which have been updated to incorporate modern cardiovascular interventions and advanced imaging techniques, such as PET/CT (positron emission tomography, computed tomography), to improve the detection of biofilm-associated infections. Management of MRSA-associated IE requires prolonged antimicrobial therapy, often with vancomycin or daptomycin, needing a combination of antimicrobials in the setting of prosthetic materials and frequently necessitates surgical intervention to remove infected prosthetic material or repair damaged heart valves. Anticoagulation remains controversial, with novel therapies like dabigatran showing potential benefits in reducing thrombus formation. Despite progress in treatment, biofilm-associated resistance poses ongoing challenges. Emerging therapeutic strategies, including combination antimicrobial regimens, bacteriophage therapy, antimicrobial peptides (AMPs), quorum sensing inhibitors (QSIs), hyperbaric oxygen therapy, and nanoparticle-based drug delivery systems, offer promising approaches to overcoming biofilm-related resistance and improving patient outcomes. This review provides an overview of the pathogenesis, current management guidelines, and future directions for treating biofilm-related MRSA IE.
Collapse
Affiliation(s)
- Ashlesha Kaushik
- Division of Pediatric Infectious Diseases, Unity Point Health at St. Luke’s Regional Medical Center and University of Iowa Carver College of Medicine, 2720 Stone Park Blvd, Sioux City, IA 51104, USA
| | - Helen Kest
- Division of Pediatric Infectious Diseases, St. Joseph’s Children’s Hospital, 703 Main Street, Paterson, NJ 07503, USA;
| | - Mangla Sood
- Department of Pediatrics, Indira Gandhi Medical College, Shimla 171006, HP, India;
| | - Corey Thieman
- Division of Pharmacology, Unity Point Health at St. Luke’s Regional Medical Center, 2720 Stone Park Blvd, Sioux City, IA 51104, USA; (C.T.); (M.P.)
| | - Bryan W. Steussy
- Division of Microbiology, Unity Point Health at St. Luke’s Regional Medical Center, 2720 Stone Park Blvd, Sioux City, IA 51104, USA;
| | - Michael Padomek
- Division of Pharmacology, Unity Point Health at St. Luke’s Regional Medical Center, 2720 Stone Park Blvd, Sioux City, IA 51104, USA; (C.T.); (M.P.)
| | - Sandeep Gupta
- Division of Pulmonary and Critical Care, Unity Point Health at St. Luke’s Regional Medical Center, 2720 Stone Park Blvd, Sioux City, IA 51104, USA;
| |
Collapse
|
6
|
Walke G, Santi C, Haydon C, Joshi P, Takebayashi Y, Rama S, Dorh J, Hotha S, Spencer J, Galan MC. C2-Linked Arabinose-Functionalized Polystyrene Microbeads Selectively Target Staphylococcus aureus. JACS AU 2024; 4:4537-4543. [PMID: 39610754 PMCID: PMC11600162 DOI: 10.1021/jacsau.4c00931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/30/2024]
Abstract
Carbohydrates play pivotal roles in the first stages of microbial infections and can be exploited as decoys to hijack the interactions between bacteria and the host cell. Multivalent glycan probes mimicking the natural presentation of glycans in living cells have been successfully employed to study fundamental carbohydrate/protein interactions in microbial systems; however, most pathogenic glycan receptors exhibit a shared specificity for commonly found sugars present in both healthy and pathogenic cells, posing a challenge for target selectivity. In this study, we report the synthesis of a small library of d-arabinose multivalent probes, a sugar absent in human physiology, and their evaluation in a bacteria agglutination assay using cluster analysis. Our findings reveal preferential binding to Staphylococcus aureus of C2-linked arabinose moieties over C1- or C5-linked probes, underscoring the importance of glycan presentation in targeting specificity. Furthermore, we demonstrate the selectivity of the C2-linked probe toward S. aureus across a panel of common bacterial pathogens. Additionally, these probes are able to disrupt biofilm formation in S. aureus SH1000, thereby further proving the cell surface interactions with S. aureus.
Collapse
Affiliation(s)
- Gulab Walke
- School of
Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, United Kingdom
| | - Cristina Santi
- School of
Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, United Kingdom
| | - Calum Haydon
- School of
Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, United Kingdom
| | - Pooja Joshi
- Department
of Chemistry, Indian Institute of Science
Education and Research Pune, Pune 411 008, India
| | - Yuiko Takebayashi
- School of
Cellular and Molecular Medicine, University
of Bristol Biomedical Sciences Building, Bristol BS8 1TD, United Kingdom
| | - Sylvain Rama
- FluoretiQ
Ltd., Futurespace, Filton Road, Stoke Gifford, Bristol BS34 8RB, United
Kingdom
| | - Josephine Dorh
- FluoretiQ
Ltd., Futurespace, Filton Road, Stoke Gifford, Bristol BS34 8RB, United
Kingdom
| | - Srinivas Hotha
- Department
of Chemistry, Indian Institute of Science
Education and Research Pune, Pune 411 008, India
| | - James Spencer
- School of
Cellular and Molecular Medicine, University
of Bristol Biomedical Sciences Building, Bristol BS8 1TD, United Kingdom
| | - M. Carmen Galan
- School of
Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, United Kingdom
| |
Collapse
|
7
|
Wang Z, Vanbever R, Lorent JH, Solis J, Knoop C, Van Bambeke F. Repurposing DNase I and alginate lyase to degrade the biofilm matrix of dual-species biofilms of Staphylococcus aureus and Pseudomonas aeruginosa grown in artificial sputum medium: In-vitro assessment of their activity in combination with broad-spectrum antibiotics. J Cyst Fibros 2024; 23:1146-1152. [PMID: 38402083 DOI: 10.1016/j.jcf.2024.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND Biofilm-associated pulmonary infections pose therapeutic challenges in cystic fibrosis patients, especially when involving multiple bacterial species. Enzymatic degradation of the biofilm matrix may offer a potential solution to enhance antibiotic efficacy. This study investigated the repurposing of DNase I, commonly used for its mucolytic activity in cystic fibrosis, to target extracellular DNA within biofilms, as well as potential synergies with alginate lyase and broad-spectrum antibiotics in dual-species biofilms of Pseudomonas aeruginosa and Staphylococcus aureus. METHODS Dual-species biofilms were grown in artificial sputum medium using S. aureus and P. aeruginosa isolated by pairs from the same patients and exposed to various combinations of enzymes, meropenem, or tobramycin. Activity was assessed by measuring biofilm biomass and viable counts. Matrix degradation and decrease in bacterial load were visualized using confocal microscopy. Biofilm viscoelasticity was estimated by rheology. RESULTS Nearly complete destruction of the biofilms was achieved only if combining the enzymatic cocktail with the two antibiotics, and if using supratherapeutic levels of DNase I and high concentrations of alginate lyase. Biofilms containing non-pigmented mucoid P. aeruginosa required higher antibiotic concentrations, despite low viscoelasticity. In contrast, for biofilms with pigmented mucoid P. aeruginosa, a correlation was observed between the efficacy of different treatments and the reduction they caused in elasticity and viscosity of the biofilm. CONCLUSIONS In this complex, highly drug-tolerant biofilm model, enzymes prove useful adjuvants to enhance antibiotic activity. However, the necessity for high enzyme concentrations emphasizes the need for thorough concentration-response evaluations and safety assessments before considering clinical applications.
Collapse
Affiliation(s)
- Zhifen Wang
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Rita Vanbever
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Joseph H Lorent
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Jessica Solis
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Christiane Knoop
- Erasme Hospital, Université libre de Bruxelles, Brussels, Belgium
| | - Françoise Van Bambeke
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
8
|
Liu JD, VanTreeck KE, Marston WA, Papadopoulou V, Rowe SE. Ultrasound-Mediated Antibiotic Delivery to In Vivo Biofilm Infections: A Review. Chembiochem 2024; 25:e202400181. [PMID: 38924307 PMCID: PMC11483220 DOI: 10.1002/cbic.202400181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 06/28/2024]
Abstract
Bacterial biofilms are a significant concern in various medical contexts due to their resilience to our immune system as well as antibiotic therapy. Biofilms often require surgical removal and frequently lead to recurrent or chronic infections. Therefore, there is an urgent need for improved strategies to treat biofilm infections. Ultrasound-mediated drug delivery is a technique that combines ultrasound application, often with the administration of acoustically-active agents, to enhance drug delivery to specific target tissues or cells within the body. This method involves using ultrasound waves to assist in the transportation or activation of medications, improving their penetration, distribution, and efficacy at the desired site. The advantages of ultrasound-mediated drug delivery include targeted and localized delivery, reduced systemic side effects, and improved efficacy of the drug at lower doses. This review scrutinizes recent advances in the application of ultrasound-mediated drug delivery for treating biofilm infections, focusing on in vivo studies. We examine the strengths and limitations of this technology in the context of wound infections, device-associated infections, lung infections and abscesses, and discuss current gaps in knowledge and clinical translation considerations.
Collapse
Affiliation(s)
- Jamie D. Liu
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Kelly E. VanTreeck
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, North Carolina 27599, USA
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - William A. Marston
- Department of Surgery, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Virginie Papadopoulou
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, North Carolina 27599, USA
- Department of Radiology, The University of North Carolina at Chapel Hill, NC, USA
| | - Sarah E. Rowe
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
9
|
Dias-Souza MV, Alves AL, Pagnin S, Veiga AA, Haq IU, Alonazi WB, Dos Santos VL. The activity of hydrolytic enzymes and antibiotics against biofilms of bacteria isolated from industrial-scale cooling towers. Microb Cell Fact 2024; 23:282. [PMID: 39415191 PMCID: PMC11484388 DOI: 10.1186/s12934-024-02502-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/07/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Cooling towers (CTs) are crucial to myriad industrial processes, supporting thermal exchange between fluids in heat exchangers using water from lakes and rivers as coolant. However, CT water can sometimes introduce microbial contaminants that adhere to and colonize various surfaces within the CT system. These microorganisms can form biofilms, significantly hindering the system's thermal exchange efficiency. Current treatment strategies employ oxidizing biocides to prevent microbial growth. However, despite their affordability, they do not eliminate biofilms effectively and can lead to corrosive damage within the system. Herein, we aim to devise an anti-biofilm strategy utilizing hydrolytic enzymes (such as α-amylase, glucoamylase, pectin-lyase, cellulase, protease, and DNase) alongside antibiotics (including meropenem, ciprofloxacin, gentamicin, erythromycin, chloramphenicol, and ceftriaxone) to combat microbial growth and biofilm formation in cooling systems. RESULTS All enzymes reduced the development of the biofilms significantly compared to controls (p < 0.05). The polysaccharidases exhibited biomass reduction of 90%, except for pectin-lyase (80%), followed by DNAse and protease at 43% and 49%, respectively. The antibiotics reduced the biofilms of 70% of isolates in concentration of > 2 mg/mL. The minimal biofilm eradication concentration (MBEC) lower than 1 mg/mL was detected for some 7-day-old sessile isolates. The enzymes and antibiotics were also used in combination against biofilms using the modified Chequerboard method. We found six synergistic combinations, with Fractional inhibitory concentrations (FIC) < 0.5, out of the ten tested. In the presence of the enzymatic mixture, MBECs presented a significant decrease (p < 0.05), at least 4-fold for antibiotics and 32-fold for enzymes. Moreover, we characterized high molecular weight (> 12 kDa) exopolysaccharides (EPS) from biofilms of ten isolates, and glycosyl composition analysis indicated a high frequency of glucose, mannose, erythrose, arabinose, and idose across isolates EPS contrasting with rhamnose, allose, and those carbohydrates, which were detected in only one isolate. CONCLUSION The synergistic approach of combining enzymes with antibiotics emerges as a highly effective and innovative strategy for anti-biofilm intervention, highlighting its potential to enhance biofilm management practices.
Collapse
Affiliation(s)
- Marcus Vinícius Dias-Souza
- Applied Microbiology Laboratory, Microbiology Department, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, C.P. 486, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Andrea Lima Alves
- Applied Microbiology Laboratory, Microbiology Department, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, C.P. 486, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Sérgio Pagnin
- Research and Development Center (CENPES), Petróleo Brasileiro S.A., Rio de Janeiro, Brazil
| | - Andrea Azevedo Veiga
- Research and Development Center (CENPES), Petróleo Brasileiro S.A., Rio de Janeiro, Brazil
| | - Ihtisham Ul Haq
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M. Strzody 9, Gliwice, 44-100, Poland
- Joint Doctoral School, Silesian University of Technology, Akademicka 2A, Gliwice, 44-100, Poland
- Programa de Pós-graduação em Inovação Tecnológica, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Wadi B Alonazi
- Health Administration Department, College of Business Administration, King Saud University, Riyadh, Saudi Arabia
| | - Vera Lúcia Dos Santos
- Applied Microbiology Laboratory, Microbiology Department, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, C.P. 486, Belo Horizonte, Minas Gerais, 31270-901, Brazil.
| |
Collapse
|
10
|
Dramé I, Rossez Y, Krzewinski F, Charbonnel N, Ollivier-Nakusi L, Briandet R, Dague E, Forestier C, Balestrino D. FabR, a regulator of membrane lipid homeostasis, is involved in Klebsiella pneumoniae biofilm robustness. mBio 2024; 15:e0131724. [PMID: 39240091 PMCID: PMC11481535 DOI: 10.1128/mbio.01317-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 08/06/2024] [Indexed: 09/07/2024] Open
Abstract
Biofilm is a dynamic structure from which individual bacteria and micro-aggregates are released to subsequently colonize new niches by either detachment or dispersal. Screening of a transposon mutant library identified genes associated with the alteration of Klebsiella pneumoniae biofilm including fabR, which encodes a transcriptional regulator involved in membrane lipid homeostasis. An isogenic ∆fabR mutant formed more biofilm than the wild-type (WT) strain and its trans-complemented strain. The thick and round aggregates observed with ∆fabR were resistant to extensive washes, unlike those of the WT strain. Confocal microscopy and BioFlux microfluidic observations showed that fabR deletion was associated with biofilm robustness and impaired erosion over time. The genes fabB and yqfA associated with fatty acid metabolism were significantly overexpressed in the ∆fabR strain, in both planktonic and biofilm conditions. Two monounsaturated fatty acids, palmitoleic acid (C16:1) and oleic acid (C18:1), were found in higher proportion in biofilm cells than in planktonic forms, whereas heptadecenoic acid (C17:1) and octadecanoic acid, 11-methoxy (C18:0-OCH3) were found in higher proportion in the planktonic lifestyle. The fabR mutation induced variations in the fatty acid composition, with no clear differences in the amounts of saturated fatty acids (SFA) and unsaturated fatty acids for the planktonic lifestyle but lower SFA in the biofilm form. Atomic force microscopy showed that deletion of fabR is associated with decreased K. pneumoniae cell rigidity in the biofilm lifestyle, as well as a softer, more elastic biofilm with increased cell cohesion compared to the wild-type strain.IMPORTANCEKlebsiella pneumoniae is an opportunistic pathogen responsible for a wide range of nosocomial infections. The success of this pathogen is due to its high resistance to antibiotics and its ability to form biofilms. The molecular mechanisms involved in biofilm formation have been largely described but the dispersal process that releases individual and aggregate cells from mature biofilm is less well documented while it is associated with the colonization of new environments and thus new threats. Using a multidisciplinary approach, we show that modifications of bacterial membrane fatty acid composition lead to variations in the biofilm robustness, and subsequent bacterial detachment and biofilm erosion over time. These results enhance our understanding of the genetic requirements for biofilm formation in K. pneumoniae that affect the time course of biofilm development and the embrittlement step preceding its dispersal that will make it possible to control K. pneumoniae infections.
Collapse
Affiliation(s)
- Ibrahima Dramé
- Université Clermont Auvergne, CNRS, LMGE, Clermont–Ferrand, France
| | - Yannick Rossez
- Université Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Frederic Krzewinski
- Université Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | | | | | - Romain Briandet
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Etienne Dague
- LAAS-CNRS, CNRS, Univeristé de Toulouse, Toulouse, France
| | | | | |
Collapse
|
11
|
Ragupathi H, Pushparaj MM, Gopi SM, Govindarajan DK, Kandaswamy K. Biofilm matrix: a multifaceted layer of biomolecules and a defensive barrier against antimicrobials. Arch Microbiol 2024; 206:432. [PMID: 39402397 DOI: 10.1007/s00203-024-04157-3] [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: 08/09/2024] [Revised: 09/24/2024] [Accepted: 10/03/2024] [Indexed: 11/10/2024]
Abstract
Bacterial cells often exist in the form of sessile aggregates known as biofilms, which are polymicrobial in nature and can produce slimy Extracellular Polymeric Substances (EPS). EPS is often referred to as a biofilm matrix and is a heterogeneous mixture of various biomolecules such as polysaccharides, proteins, and extracellular DNA/RNA (eDNA/RNA). In addition, bacteriophage (phage) was also found to be an integral component of the matrix and can serve as a protective barrier. In recent years, the roles of proteins, polysaccharides, and phages in the virulence of biofilms have been well studied. However, a mechanistic understanding of the release of such biomolecules and their interactions with antimicrobials requires a thorough review. Therefore, this article critically reviews the various mechanisms of release of matrix polymers. In addition, this article also provides a contemporary understanding of interactions between various biomolecules to protect biofilms against antimicrobials. In summary, this article will provide a thorough understanding of the functions of various biofilm matrix molecules.
Collapse
Affiliation(s)
- Harini Ragupathi
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India
| | - Mahamahima Muthuswamy Pushparaj
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India
| | - Sarves Mani Gopi
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India
| | - Deenadayalan Karaiyagowder Govindarajan
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Drive, 637371, Singapore, Singapore
| | - Kumaravel Kandaswamy
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India.
| |
Collapse
|
12
|
Klein EM, Heintz H, Wurst R, Schuldt S, Hähl H, Jacobs K, Gescher J. Comparative analysis of the influence of BpfA and BpfG on biofilm development and current density in Shewanella oneidensis under oxic, fumarate- and anode-respiring conditions. Sci Rep 2024; 14:23174. [PMID: 39369013 PMCID: PMC11455927 DOI: 10.1038/s41598-024-73474-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/17/2024] [Indexed: 10/07/2024] Open
Abstract
Biofilm formation by Shewanella oneidensis has been extensively studied under oxic conditions; however, relatively little is known about biofilm formation under anoxic conditions and how biofilm architecture and composition can positively influence current generation in bioelectrochemical systems. In this study, we utilized a recently developed microfluidic biofilm analysis setup with automated 3D imaging to investigate the effects of extracellular electron acceptors and synthetic modifications to the extracellular polymeric matrix on biofilm formation. Our results with the wild type strain demonstrate robust biofilm formation even under anoxic conditions when fumarate is used as the electron acceptor. However, this pattern shifts when a graphite electrode is employed as the electron acceptor, resulting in biofilm formation falling below the detection limit of the optical coherence tomography imaging system. To manipulate biofilm formation, we aimed to express BpfG with a single amino acid substitution in the catalytic center (C116S) and to overexpress bpfA. Our analyses indicate that, under oxic conditions, overarching mechanisms predominantly influence biofilm development, rather than the specific mutations we investigated. Under anoxic conditions, the bpfG mutation led to a quantitative increase in biofilm formation, but both strains exhibited significant qualitative changes in biofilm architecture compared to the controls. When an anode was used as the sole electron acceptor, both the bpfA and bpfG mutations positively impacted mean current density, yielding a 1.8-fold increase for each mutation.
Collapse
Affiliation(s)
- Edina Marlen Klein
- Institute of Technical Microbiology, University of Technology Hamburg, 21073, Hamburg, Germany
| | - Hannah Heintz
- Experimental Physics, Center for Biophysics, Saarland University, 66123, Saarbrücken, Germany
| | - René Wurst
- Institute of Technical Microbiology, University of Technology Hamburg, 21073, Hamburg, Germany
| | - Simon Schuldt
- Institute of Technical Microbiology, University of Technology Hamburg, 21073, Hamburg, Germany
| | - Hendrik Hähl
- Experimental Physics, Center for Biophysics, Saarland University, 66123, Saarbrücken, Germany
| | - Karin Jacobs
- Experimental Physics, Center for Biophysics, Saarland University, 66123, Saarbrücken, Germany
- Max Planck School Matter to Life, 69120, Heidelberg, Germany
| | - Johannes Gescher
- Institute of Technical Microbiology, University of Technology Hamburg, 21073, Hamburg, Germany.
| |
Collapse
|
13
|
Yun S, Min J, Han S, Sim HS, Kim SK, Lee JB, Yoon JW, Yeom J, Park W. Experimental evolution under different nutritional conditions changes the genomic architecture and virulence of Acinetobacter baumannii. Commun Biol 2024; 7:1274. [PMID: 39369115 PMCID: PMC11455985 DOI: 10.1038/s42003-024-06978-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 09/27/2024] [Indexed: 10/07/2024] Open
Abstract
This study uncovers the molecular processes governing the adaptive evolution of multidrug-resistant (MDR) pathogens without antibiotic pressure. Genomic analysis of MDR Acinetobacter baumannii cells cultured for 8000 generations under starvation conditions (EAB1) or nutrient-rich conditions (EAB2) revealed significant genomic changes, primarily by insertion sequence (IS)-mediated insertions and deletions. Only two Acinetobacter-specific prophage-related deletions and translocations were observed in the EAB1 strain. Both evolved strains exhibited higher virulence in mouse infection studies, each with different modes of action. The EAB1 strain displayed a heightened ability to cross the epithelial barrier of human lung tissue, evade the immune system, and spread to lung tissues, ultimately resulting in cellular mortality. In contrast, the EAB2 strain strongly attached to epithelial cells, leading to increased synthesis of proinflammatory cytokines and chemokines. The genomic alterations and increased virulence observed in evolved strains during short-term evolution underscore the need for caution when handling these pathogens, as these risks persist even without antibiotic exposure.
Collapse
Affiliation(s)
- Sohyeon Yun
- Laboratory of Molecular Environmental Microbiology, Department of Environmental Science and Ecological Engineering, Korea University, Seoul, Republic of Korea
| | - Jihyeon Min
- Laboratory of Molecular Environmental Microbiology, Department of Environmental Science and Ecological Engineering, Korea University, Seoul, Republic of Korea
| | - Sunyong Han
- Laboratory of Molecular Environmental Microbiology, Department of Environmental Science and Ecological Engineering, Korea University, Seoul, Republic of Korea
| | - Ho Seok Sim
- Department of Microbiology and Immunology, Department of Biomedical Science, and Cancer Research Institute, College of Medicine, Seoul National University, Jongno-gu, Seoul, Republic of Korea
| | - Se Kye Kim
- College of Veterinary Medicine & Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Jun Bong Lee
- College of Veterinary Medicine & Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Jang Won Yoon
- College of Veterinary Medicine & Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Jinki Yeom
- Department of Microbiology and Immunology, Department of Biomedical Science, and Cancer Research Institute, College of Medicine, Seoul National University, Jongno-gu, Seoul, Republic of Korea.
| | - Woojun Park
- Laboratory of Molecular Environmental Microbiology, Department of Environmental Science and Ecological Engineering, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
14
|
Carmona-Orozco ML, Echeverri F. Induction of biofilm in extended-spectrum beta-lactamase Staphylococcus aureus with drugs commonly used in pharmacotherapy. Microb Pathog 2024; 195:106863. [PMID: 39159772 DOI: 10.1016/j.micpath.2024.106863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 07/15/2024] [Accepted: 08/14/2024] [Indexed: 08/21/2024]
Abstract
Staphylococcus aureus is a bacterial pathogen that causes bloodstream infections, pneumonia, and skin abscesses and is the primary pathogen responsible for medical devices associated with biofilm infections, accounting for approximately 70 % of cases. Therefore, the World Health Organization (WHO) has designated this microorganism as a top priority due to its role in causing over 20,000 bacteremia-related deaths in the US each year. The issue of pathogen resistance to antibiotics, mainly by a biofilm, further complicates these infections since biofilms render the bacterial colony impervious to antibiotics. However, many natural and synthetic substances also induce bacterial biofilm formation. Therefore, we investigated whether the most common active pharmaceutical ingredients (APIs) could induce biofilm formation in two clinical isolates of extended-spectrum beta-lactamase Staphylococcus aureus, one of them also methicillin-resistant (A2M) and two medical devices. We detected biofilm inducers, inhibitors, and destabilizers. Microbial strain, medical devices, API structure, and concentration influenced the modulatory effects of biofilm. In all devices tested, including microplates, FR18 duodenal probe, and respiratory probe, the clinic isolate methicillin-resistant S. aureus A2M exhibited lower susceptibility to biofilm formation than S. aureus A1. The anti-inflammatory acetaminophen, the hypocholesterolemic lovastatin, and the diuretic hydrochlorothiazide all induced biofilm. However, verapamil, an antihypertensive, and cetirizine, an antihistamine, inhibited biofilm on S. aureus A2M, while propranolol, another antihypertensive, inhibited biofilm on S. aureus A1. Additionally, diclofenac, an analgesic, and cetirizine destabilized the biofilm, resulting in more free bacteria and possibly making them more susceptible to external agents such as antibiotics. Nonetheless, further epidemiologic analyses and in vivo assays are needed to confirm these findings and to establish a correlation between drug use, the onset of bacterial infections in patients, and the use of medical devices. This work provides information about the probable clinical implications of drugs in patients using medical devices or undergoing surgical procedures. Inhibitory APIs could also be used as drug repurposing or templates to design new, more potent biofilm inhibitors.
Collapse
Affiliation(s)
- Maria L Carmona-Orozco
- Química Orgánica de Productos Naturales, Instituto de Química, Universidad de Antioquia, Medellín, Colombia
| | - Fernando Echeverri
- Química Orgánica de Productos Naturales, Instituto de Química, Universidad de Antioquia, Medellín, Colombia.
| |
Collapse
|
15
|
Kungwani NA, Panda J, Mishra AK, Chavda N, Shukla S, Vikhe K, Sharma G, Mohanta YK, Sharifi-Rad M. Combating bacterial biofilms and related drug resistance: Role of phyto-derived adjuvant and nanomaterials. Microb Pathog 2024; 195:106874. [PMID: 39181190 DOI: 10.1016/j.micpath.2024.106874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024]
Abstract
The emergence of antimicrobial resistance (AMR) in clinical microbes has led to a search for novel antibiotics for combating bacterial infections. The treatment of bacterial infections becomes more challenging with the onset of biofilm formation. AMR is further accelerated by biofilm physiology and differential gene expression in bacteria with an inherent resistance to conventional antibiotics. In the search for innovative strategies to control the spread of AMR in clinical isolates, plant-derived therapeutic metabolites can be repurposed to control biofilm-associated drug resistance. Unlike antibiotics, designed to act on a single cellular process, phytochemicals can simultaneously target multiple cellular components. Furthermore, they can disrupt biofilm formation and inhibit quorum sensing, offering a comprehensive approach to combat bacterial infections. In bacterial biofilms, the first line of AMR is due to biofilms associated with the extracellular matrix, diffusion barriers, quorum sensing, and persister cells. These extracellular barriers can be overcome using phytochemical-based antibiotic adjuvants to increase the efficacy of antibiotic treatment and restrict the spread of AMR. Furthermore, phytochemicals can be used to target bacterial intracellular machinery such as DNA replication, protein synthesis, efflux pumps, and degrading enzymes. In parallel with pristine phytochemicals, phyto-derived nanomaterials have emerged as an effective means of fighting bacterial biofilms. These nanomaterials can be formulated to cross the biofilm barriers and function on cellular targets. This review focuses on the synergistic effects of phytochemicals and phyto-derived nanomaterials in controlling the progression of biofilm-related AMR. IT provides comprehensive insights into recent advancements and the underlying mechanisms of the use of phyto-derived adjuvants and nanomaterials.
Collapse
Affiliation(s)
- Neelam Amit Kungwani
- Department of Environmental Biotechnology, Gujarat Biotechnology University, Gandhinagar, Gujarat, 382355, India.
| | - Jibanjyoti Panda
- Nano-biotechnology and Translational Knowledge Laboratory, Department of Applied Biology, School of Biological Sciences, University of Science and Technology Meghalaya, Techno City, 9th Mile, Baridua, Ri-Bhoi, 793101, Meghalaya, India
| | - Awdhesh Kumar Mishra
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, Republic of Korea
| | | | - Sudhir Shukla
- Homi Bhabha National Institute, Biofouling and Biofilm Processes Section, WSCD, Chemistry Group, Bhabha Atomic Research Centre, Kalpakkam, Tamilnadu, 603102, India
| | - Kalyani Vikhe
- Department of Environmental Biotechnology, Gujarat Biotechnology University, Gandhinagar, Gujarat, 382355, India
| | - Gunjan Sharma
- Department of Plant Biotechnology, Gujarat Biotechnology University, Gandhinagar, Gujarat, 382355, India
| | - Yugal Kishore Mohanta
- Nano-biotechnology and Translational Knowledge Laboratory, Department of Applied Biology, School of Biological Sciences, University of Science and Technology Meghalaya, Techno City, 9th Mile, Baridua, Ri-Bhoi, 793101, Meghalaya, India; Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103, Tamil Nadu, India.
| | - Majid Sharifi-Rad
- Department of Range and Watershed Management, Faculty of Water and Soil, University of Zabol, Zabol, 98613-35856, Iran.
| |
Collapse
|
16
|
Biswas R, Jangra B, Ashok G, Ravichandiran V, Mohan U. Current Strategies for Combating Biofilm-Forming Pathogens in Clinical Healthcare-Associated Infections. Indian J Microbiol 2024; 64:781-796. [PMID: 39282194 PMCID: PMC11399387 DOI: 10.1007/s12088-024-01221-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 02/07/2024] [Indexed: 09/18/2024] Open
Abstract
The biofilm formation by various pathogens causes chronic infections and poses severe threats to industry, healthcare, and society. They can form biofilm on surfaces of medical implants, heart valves, pacemakers, contact lenses, vascular grafts, urinary catheters, dialysis catheters, etc. These biofilms play a central role in bacterial persistence and antibiotic tolerance. Biofilm formation occurs in a series of steps, and any interference in these steps can prevent its formation. Therefore, the hunt to explore and develop effective anti-biofilm strategies became necessary to decrease the rate of biofilm-related infections. In this review, we highlighted and discussed the current therapeutic approaches to eradicate biofilm formation and combat drug resistance by anti-biofilm drugs, phytocompounds, antimicrobial peptides (AMPs), antimicrobial lipids (AMLs), matrix-degrading enzymes, nanoparticles, phagebiotics, surface coatings, photodynamic therapy (PDT), riboswitches, vaccines, and antibodies. The clinical validation of these findings will provide novel preventive and therapeutic strategies for biofilm-associated infections to the medical world.
Collapse
Affiliation(s)
- Rashmita Biswas
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal India
| | - Bhawana Jangra
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab India
| | - Ganapathy Ashok
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal India
| | - Velayutham Ravichandiran
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal India
| | - Utpal Mohan
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal India
| |
Collapse
|
17
|
David A, Tahrioui A, Tareau AS, Forge A, Gonzalez M, Bouffartigues E, Lesouhaitier O, Chevalier S. Pseudomonas aeruginosa Biofilm Lifecycle: Involvement of Mechanical Constraints and Timeline of Matrix Production. Antibiotics (Basel) 2024; 13:688. [PMID: 39199987 PMCID: PMC11350761 DOI: 10.3390/antibiotics13080688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 09/01/2024] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen causing acute and chronic infections, especially in immunocompromised patients. Its remarkable adaptability and resistance to various antimicrobial treatments make it difficult to eradicate. Its persistence is enabled by its ability to form a biofilm. Biofilm is a community of sessile micro-organisms in a self-produced extracellular matrix, which forms a scaffold facilitating cohesion, cell attachment, and micro- and macro-colony formation. This lifestyle provides protection against environmental stresses, the immune system, and antimicrobial treatments, and confers the capacity for colonization and long-term persistence, often characterizing chronic infections. In this review, we retrace the events of the life cycle of P. aeruginosa biofilm, from surface perception/contact to cell spreading. We focus on the importance of extracellular appendages, mechanical constraints, and the kinetics of matrix component production in each step of the biofilm life cycle.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sylvie Chevalier
- Univ Rouen Normandie, Univ Caen Normandie, Normandie Univ, CBSA UR 4312, F-76000 Rouen, France
| |
Collapse
|
18
|
Sedighi O, Bednarke B, Sherriff H, Doiron AL. Nanoparticle-Based Strategies for Managing Biofilm Infections in Wounds: A Comprehensive Review. ACS OMEGA 2024; 9:27853-27871. [PMID: 38973924 PMCID: PMC11223148 DOI: 10.1021/acsomega.4c02343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 05/28/2024] [Accepted: 06/04/2024] [Indexed: 07/09/2024]
Abstract
Chronic wounds containing opportunistic bacterial pathogens are a growing problem, as they are the primary cause of morbidity and mortality in developing and developed nations. Bacteria can adhere to almost every surface, forming architecturally complex communities called biofilms that are tolerant to an individual's immune response and traditional treatments. Wound dressings are a primary source and potential treatment avenue for biofilm infections, and research has recently focused on using nanoparticles with antimicrobial activity for infection control. This Review categorizes nanoparticle-based approaches into four main types, each leveraging unique mechanisms against biofilms. Metallic nanoparticles, such as silver and copper, show promising data due to their ability to disrupt bacterial cell membranes and induce oxidative stress, although their effectiveness can vary based on particle size and composition. Phototherapy-based nanoparticles, utilizing either photodynamic or photothermal therapy, offer targeted microbial destruction by generating reactive oxygen species or localized heat, respectively. However, their efficacy depends on the presence of light and oxygen, potentially limiting their use in deeper or more shielded biofilms. Nanoparticles designed to disrupt extracellular polymeric substances directly target the biofilm structure, enhancing the penetration and efficacy of antimicrobial agents. Lastly, nanoparticles that induce biofilm dispersion represent a novel strategy, aiming to weaken the biofilm's defense and restore susceptibility to antimicrobials. While each method has its advantages, the selection of an appropriate nanoparticle-based treatment depends on the specific requirements of the wound environment and the type of biofilm involved. The integration of these nanoparticles into wound dressings not only promises enhanced treatment outcomes but also offers a reduction in the overall use of antibiotics, aligning with the urgent need for innovative solutions in the fight against antibiotic-tolerant infections. The overarching objective of employing these diverse nanoparticle strategies is to replace antibiotics or substantially reduce their required dosages, providing promising avenues for biofilm infection management.
Collapse
Affiliation(s)
- Omid Sedighi
- Department
of Electrical and Biomedical Engineering, University of Vermont, Burlington, Vermont 05405, United States
| | - Brooke Bednarke
- Department
of Electrical and Biomedical Engineering, University of Vermont, Burlington, Vermont 05405, United States
| | - Hannah Sherriff
- Department
of Electrical and Biomedical Engineering, University of Vermont, Burlington, Vermont 05405, United States
| | - Amber L. Doiron
- Department
of Electrical and Biomedical Engineering, University of Vermont, Burlington, Vermont 05405, United States
| |
Collapse
|
19
|
Tian S, Shi L, Ren Y, van der Mei HC, Busscher HJ. A normalized parameter for comparison of biofilm dispersants in vitro. Biofilm 2024; 7:100188. [PMID: 38495770 PMCID: PMC10943042 DOI: 10.1016/j.bioflm.2024.100188] [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: 12/01/2023] [Revised: 02/12/2024] [Accepted: 02/28/2024] [Indexed: 03/19/2024] Open
Abstract
Dispersal of infectious biofilms increases bacterial concentrations in blood. To prevent sepsis, the strength of a dispersant should be limited to allow the immune system to remove dispersed bacteria from blood, preferably without antibiotic administration. Biofilm bacteria are held together by extracellular polymeric substances that can be degraded by dispersants. Currently, comparison of the strength of dispersants is not possible by lack of a suitable comparison parameter. Here, a biofilm dispersal parameter is proposed that accounts for differences in initial biofilm properties, dispersant concentration and exposure time by using PBS as a control and normalizing outcomes with respect to concentration and time. The parameter yielded near-identical values based on dispersant-induced reductions in biomass or biofilm colony-forming-units and appeared strain-dependent across pathogens. The parameter as proposed is largely independent of experimental methods and conditions and suitable for comparing different dispersants with respect to different causative strains in particular types of infection.
Collapse
Affiliation(s)
- Shuang Tian
- University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Antonius Deusinglaan 1, 9713, AV, Groningen, the Netherlands
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Yijin Ren
- University of Groningen and University Medical Center Groningen, Department of Orthodontics, Hanzeplein 1, 9700, RB, Groningen, the Netherlands
| | - Henny C. van der Mei
- University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Antonius Deusinglaan 1, 9713, AV, Groningen, the Netherlands
| | - Henk J. Busscher
- University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Antonius Deusinglaan 1, 9713, AV, Groningen, the Netherlands
| |
Collapse
|
20
|
Barbosa A, Azevedo NF, Goeres DM, Cerqueira L. Ecology of Legionella pneumophila biofilms: The link between transcriptional activity and the biphasic cycle. Biofilm 2024; 7:100196. [PMID: 38601816 PMCID: PMC11004079 DOI: 10.1016/j.bioflm.2024.100196] [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: 12/29/2023] [Revised: 03/10/2024] [Accepted: 03/29/2024] [Indexed: 04/12/2024] Open
Abstract
There has been considerable discussion regarding the environmental life cycle of Legionella pneumophila and its virulence potential in natural and man-made water systems. On the other hand, the bacterium's morphogenetic mechanisms within host cells (amoeba and macrophages) have been well documented and are linked to its ability to transition from a non-virulent, replicative state to an infectious, transmissive state. Although the morphogenetic mechanisms associated with the formation and detachment of the L. pneumophila biofilm have also been described, the capacity of the bacteria to multiply extracellularly is not generally accepted. However, several studies have shown genetic pathways within the biofilm that resemble intracellular mechanisms. Understanding the functionality of L. pneumophila cells within a biofilm is fundamental for assessing the ecology and evaluating how the biofilm architecture influences L. pneumophila survival and persistence in water systems. This manuscript provides an overview of the biphasic cycle of L. pneumophila and its implications in associated intracellular mechanisms in amoeba. It also examines the molecular pathways and gene regulation involved in L. pneumophila biofilm formation and dissemination. A holistic analysis of the transcriptional activities in L. pneumophila biofilms is provided, combining the information of intracellular mechanisms in a comprehensive outline. Furthermore, this review discusses the techniques that can be used to study the morphogenetic states of the bacteria within biofilms, at the single cell and population levels.
Collapse
Affiliation(s)
- Ana Barbosa
- LEPABE – Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal
- ALiCE – Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal
| | - Nuno F. Azevedo
- LEPABE – Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal
- ALiCE – Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal
| | - Darla M. Goeres
- LEPABE – Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal
- ALiCE – Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal
- The Center for Biofilm Engineering, Montana State University, Bozeman, MT, USA
| | - Laura Cerqueira
- LEPABE – Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal
- ALiCE – Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal
| |
Collapse
|
21
|
Sousa AM, Ferreira D, Rodrigues LR, Pereira MO. Aptamer-based therapy for fighting biofilm-associated infections. J Control Release 2024; 367:522-539. [PMID: 38295992 DOI: 10.1016/j.jconrel.2024.01.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 01/06/2024] [Accepted: 01/27/2024] [Indexed: 02/06/2024]
Abstract
Biofilms are key players in the pathogenesis of most of chronic infections associated with host tissue or fluids and indwelling medical devices. These chronic infections are hard to be treated due to the increased biofilms tolerance towards antibiotics in comparison to planktonic (or free living) cells. Despite the advanced understanding of their formation and physiology, biofilms continue to be a challenge and there is no standardized therapeutic approach in clinical practice to eradicate them. Aptamers offer distinctive properties, including excellent affinity, selectivity, stability, making them valuable tools for therapeutic purposes. This review explores the flexibility and designability of aptamers as antibiofilm drugs but, importantly, as targeting tools for diverse drug and delivery systems. It highlights specific examples of application of aptamers in biofilms of diverse species according to different modes of action including inhibition of motility and adhesion, blocking of quorum sensing molecules, and dispersal of biofilm-cells to planktonic state. Moreover, it discusses the limitations and challenges that impaired an increased success of the use of aptamers on biofilm management, as well as the opportunities related to aptamers modifications that can significantly expand their applicability on the biofilm field.
Collapse
Affiliation(s)
- Ana Margarida Sousa
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal.
| | - Débora Ferreira
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal
| | - Lígia Raquel Rodrigues
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal
| | - Maria Olívia Pereira
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal.
| |
Collapse
|
22
|
Habimana O, Bridier A, Giaouris E. Editorial: The ever so elusive pathogen-harboring biofilms on abiotic surfaces in the food and clinical sectors: the good, the bad and the slimy. Front Cell Infect Microbiol 2024; 14:1374693. [PMID: 38404285 PMCID: PMC10884827 DOI: 10.3389/fcimb.2024.1374693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 01/30/2024] [Indexed: 02/27/2024] Open
Affiliation(s)
- Olivier Habimana
- Department of Biotechnology and Food Engineering, Guangdong Technion-Israel Institute of Technology, Shantou, China
| | - Arnaud Bridier
- Antibiotics, Biocides, Residues and Resistance Unit, Fougeres Laboratory, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), Fougeres, France
| | - Efstathios Giaouris
- Laboratory of Food Microbiology and Hygiene, Department of Food Science and Nutrition, School of the Environment, University of the Aegean, Myrina, Lemnos, Greece
| |
Collapse
|
23
|
Kaushik A, Kest H, Sood M, Steussy BW, Thieman C, Gupta S. Biofilm Producing Methicillin-Resistant Staphylococcus aureus (MRSA) Infections in Humans: Clinical Implications and Management. Pathogens 2024; 13:76. [PMID: 38251383 PMCID: PMC10819455 DOI: 10.3390/pathogens13010076] [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: 12/11/2023] [Revised: 01/10/2024] [Accepted: 01/13/2024] [Indexed: 01/23/2024] Open
Abstract
Since its initial description in the 1960s, methicillin-resistant Staphylococcus aureus (MRSA) has developed multiple mechanisms for antimicrobial resistance and evading the immune system, including biofilm production. MRSA is now a widespread pathogen, causing a spectrum of infections ranging from superficial skin issues to severe conditions like osteoarticular infections and endocarditis, leading to high morbidity and mortality. Biofilm production is a key aspect of MRSA's ability to invade, spread, and resist antimicrobial treatments. Environmental factors, such as suboptimal antibiotics, pH, temperature, and tissue oxygen levels, enhance biofilm formation. Biofilms are intricate bacterial structures with dense organisms embedded in polysaccharides, promoting their resilience. The process involves stages of attachment, expansion, maturation, and eventually disassembly or dispersion. MRSA's biofilm formation has a complex molecular foundation, involving genes like icaADBC, fnbA, fnbB, clfA, clfB, atl, agr, sarA, sarZ, sigB, sarX, psm, icaR, and srtA. Recognizing pivotal genes for biofilm formation has led to potential therapeutic strategies targeting elemental and enzymatic properties to combat MRSA biofilms. This review provides a practical approach for healthcare practitioners, addressing biofilm pathogenesis, disease spectrum, and management guidelines, including advances in treatment. Effective management involves appropriate antimicrobial therapy, surgical interventions, foreign body removal, and robust infection control practices to curtail spread within healthcare environments.
Collapse
Affiliation(s)
- Ashlesha Kaushik
- Division of Pediatric Infectious Diseases, St. Luke’s Regional Medical Center, Unity Point Health, 2720 Stone Park Blvd, Sioux City, IA 51104, USA
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Master of Science, Healthcare Quality and Safety, Harvard Medical School, Boston, MA 02115, USA
| | - Helen Kest
- Division of Pediatric Infectious Diseases, St. Joseph’s Children’s Hospital, 703 Main Street, Paterson, NJ 07503, USA;
| | - Mangla Sood
- Department of Pediatrics, Indira Gandhi Medical College, Shimla 171006, India;
| | - Bryan W. Steussy
- Division of Microbiology, St. Luke’s Regional Medical Center, Unity Point Health, 2720 Stone Park Blvd, Sioux City, IA 51104, USA;
| | - Corey Thieman
- Division of Pharmacology, St. Luke’s Regional Medical Center, Unity Point Health, 2720 Stone Park Blvd, Sioux City, IA 51104, USA;
| | - Sandeep Gupta
- Division of Pulmonary and Critical Care, St. Luke’s Regional Medical Center, Unity Point Health, 2720 Stone Park Blvd, Sioux City, IA 51104, USA;
| |
Collapse
|
24
|
Ghosh S, Sett U, Pal A, Nandy S, Nandi S, Chakrabarty S, Das A, Bandopadhyay P, Basu T. Antibiofilm potential of nanonized eugenol against Pseudomonas aeruginosa. J Appl Microbiol 2024; 135:lxad305. [PMID: 38093454 DOI: 10.1093/jambio/lxad305] [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: 06/19/2023] [Revised: 12/03/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024]
Abstract
AIMS The purpose of this study was to synthesize a nanoform of eugenol (an important phytochemical with various pharmacological potentials) and to investigate its antibiofilm efficacy on Pseudomonas aeruginosa biofilm. METHODS AND RESULTS Colloidal suspension of eugenol-nanoparticles (ENPs) was synthesized by the simple ultrasonic cavitation method through the emulsification of hydrophobic eugenol into hydrophilic gelatin. Thus, the nanonization process made water-insoluble eugenol into water-soluble nano-eugenol, making the nanoform bioavailable. The size of the ENPs was 20-30 nm, entrapment efficiency of eugenol within gelatin was 80%, and release of eugenol from the gelatin cap was slow and sustained over 5 days. Concerning the clinically relevant pathogen P. aeruginosa, ENPs had higher antibiofilm (for both formation and eradication) activities than free eugenol. Minimal biofilm inhibitory concentration and minimal biofilm eradication concentration of ENP on P. aeruginosa biofilm were 2.0 and 4.0 mM, respectively. In addition, the measurement of P. aeruginosa biofilm biomass, biofilm thickness, amount of biofilm extra-polymeric substance, cell surface hydrophobicity, cell swarming and twitching efficiencies, cellular morphology, and biofilm formation in catheter demonstrated that the antibiofilm efficacy of nano-eugenol was 30%-40% higher than that of bulk eugenol. CONCLUSION These results signify that future pharmacological and clinical studies are very much required to investigate whether ENPs can act as an effective drug against P. aeruginosa biofilm-mediated diseases. Thus, the problem of intrinsic antibiotic tolerance of biofilm-forming cells may be minimized by ENPs. Moreover, ENP may be used as a potential catheter-coating agent to inhibit pseudomonal colonization on catheter surfaces and, therefore, to reduce catheter-associated infections and complications.
Collapse
Affiliation(s)
- Sourav Ghosh
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741 235, India
| | - Upasana Sett
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741 235, India
| | - Anabadya Pal
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741 235, India
| | - Sanchita Nandy
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741 235, India
| | - Susmita Nandi
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741 235, India
| | - Soumajit Chakrabarty
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741 235, India
| | - Abhijit Das
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741 235, India
| | - Pathikrit Bandopadhyay
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741 235, India
| | - Tarakdas Basu
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741 235, India
| |
Collapse
|
25
|
Jiang H, Wang Z, Jia AQ. Methyl gallate from Camellia nitidissima Chi flowers reduces quorum sensing related virulence and biofilm formation against Aeromonas hydrophila. BIOFOULING 2024; 40:64-75. [PMID: 38373897 DOI: 10.1080/08927014.2024.2316611] [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: 09/07/2023] [Accepted: 02/02/2024] [Indexed: 02/21/2024]
Abstract
Aeromonas hydrophila, a Gram-negative zoonotic bacterium, causes high mortality in fish farming and immunocompromised patients. This study aimed to extract methyl gallate (MG) from the flowers of Camellia nitidissima Chi and evaluate its potential as a quorum sensing inhibitor (QSI) against Aeromonas hydrophila SHAe 115. MG reduced QS-associated virulence factors, including hemolysis, protease, and lipase, while impairing swimming motility and biofilm formation. Additionally, MG down-regulated positive regulatory genes (ahyR, fleQ) and up-regulated negative regulators (litR, fleN). This highlights MG's promise as a potent QSI for A. hydrophila SHAe 115, advancing strategies against infections in aquaculture and human health.
Collapse
Affiliation(s)
- Huan Jiang
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, China
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Zhennan Wang
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, China
| | - Ai-Qun Jia
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, China
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
26
|
Kumar V, Yasmeen N, Pandey A, Ahmad Chaudhary A, Alawam AS, Ahmad Rudayni H, Islam A, Lakhawat SS, Sharma PK, Shahid M. Antibiotic adjuvants: synergistic tool to combat multi-drug resistant pathogens. Front Cell Infect Microbiol 2023; 13:1293633. [PMID: 38179424 PMCID: PMC10765517 DOI: 10.3389/fcimb.2023.1293633] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/14/2023] [Indexed: 01/06/2024] Open
Abstract
The rise of multi-drug resistant (MDR) pathogens poses a significant challenge to the field of infectious disease treatment. To overcome this problem, novel strategies are being explored to enhance the effectiveness of antibiotics. Antibiotic adjuvants have emerged as a promising approach to combat MDR pathogens by acting synergistically with antibiotics. This review focuses on the role of antibiotic adjuvants as a synergistic tool in the fight against MDR pathogens. Adjuvants refer to compounds or agents that enhance the activity of antibiotics, either by potentiating their effects or by targeting the mechanisms of antibiotic resistance. The utilization of antibiotic adjuvants offers several advantages. Firstly, they can restore the effectiveness of existing antibiotics against resistant strains. Adjuvants can inhibit the mechanisms that confer resistance, making the pathogens susceptible to the action of antibiotics. Secondly, adjuvants can enhance the activity of antibiotics by improving their penetration into bacterial cells, increasing their stability, or inhibiting efflux pumps that expel antibiotics from bacterial cells. Various types of antibiotic adjuvants have been investigated, including efflux pump inhibitors, resistance-modifying agents, and compounds that disrupt bacterial biofilms. These adjuvants can act synergistically with antibiotics, resulting in increased antibacterial activity and overcoming resistance mechanisms. In conclusion, antibiotic adjuvants have the potential to revolutionize the treatment of MDR pathogens. By enhancing the efficacy of antibiotics, adjuvants offer a promising strategy to combat the growing threat of antibiotic resistance. Further research and development in this field are crucial to harness the full potential of antibiotic adjuvants and bring them closer to clinical application.
Collapse
Affiliation(s)
- Vikram Kumar
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
- Amity Institute of Pharmacy, Amity University Rajasthan, Jaipur, Rajasthan, India
| | - Nusrath Yasmeen
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
| | - Aishwarya Pandey
- INRS, Eau Terre Environnement Research Centre, Québec, QC, Canada
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Abdullah S. Alawam
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Hassan Ahmad Rudayni
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Asimul Islam
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Sudarshan S. Lakhawat
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
| | - Pushpender K. Sharma
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
| | - Mohammad Shahid
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| |
Collapse
|
27
|
Wang S, Ma C, Long J, Cheng P, Zhang Y, Peng L, Fu L, Yu Y, Xu D, Zhang S, Qiu J, He Y, Yang H, Chen H. Impact of CRAMP-34 on Pseudomonas aeruginosa biofilms and extracellular metabolites. Front Cell Infect Microbiol 2023; 13:1295311. [PMID: 38162583 PMCID: PMC10757720 DOI: 10.3389/fcimb.2023.1295311] [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: 09/16/2023] [Accepted: 11/30/2023] [Indexed: 01/03/2024] Open
Abstract
Biofilm is a structured community of bacteria encased within a self-produced extracellular matrix. When bacteria form biofilms, they undergo a phenotypic shift that enhances their resistance to antimicrobial agents. Consequently, inducing the transition of biofilm bacteria to the planktonic state may offer a viable approach for addressing infections associated with biofilms. Our previous study has shown that the mouse antimicrobial peptide CRAMP-34 can disperse Pseudomonas aeruginosa (P. aeruginosa) biofilm, and the potential mechanism of CRAMP-34 eradicate P. aeruginosa biofilms was also investigated by combined omics. However, changes in bacterial extracellular metabolism have not been identified. To further explore the mechanism by which CRAMP-34 disperses biofilm, this study analyzed its effects on the extracellular metabolites of biofilm cells via metabolomics. The results demonstrated that a total of 258 significantly different metabolites were detected in the untargeted metabolomics, of which 73 were downregulated and 185 were upregulated. Pathway enrichment analysis of differential metabolites revealed that metabolic pathways are mainly related to the biosynthesis and metabolism of amino acids, and it also suggested that CRAMP-34 may alter the sensitivity of biofilm bacteria to antibiotics. Subsequently, it was confirmed that the combination of CRAMP-34 with vancomycin and colistin had a synergistic effect on dispersed cells. These results, along with our previous findings, suggest that CRAMP-34 may promote the transition of PAO1 bacteria from the biofilm state to the planktonic state by upregulating the extracellular glutamate and succinate metabolism and eventually leading to the dispersal of biofilm. In addition, increased extracellular metabolites of myoinositol, palmitic acid and oleic acid may enhance the susceptibility of the dispersed bacteria to the antibiotics colistin and vancomycin. CRAMP-34 also delayed the development of bacterial resistance to colistin and ciprofloxacin. These results suggest the promising development of CRAMP-34 in combination with antibiotics as a potential candidate to provide a novel therapeutic approach for the prevention and treatment of biofilm-associated infections.
Collapse
Affiliation(s)
- Shiyuan Wang
- College of Veterinary Medicine, Southwest University, Chongqing, China
- Collaborative Innovation Institute National Center of Technology Innovation for Pigs, Chongqing, China
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Chengjun Ma
- College of Veterinary Medicine, Southwest University, Chongqing, China
- Collaborative Innovation Institute National Center of Technology Innovation for Pigs, Chongqing, China
- Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Jinying Long
- College of Veterinary Medicine, Southwest University, Chongqing, China
- Collaborative Innovation Institute National Center of Technology Innovation for Pigs, Chongqing, China
- Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Peng Cheng
- College of Veterinary Medicine, Southwest University, Chongqing, China
- Collaborative Innovation Institute National Center of Technology Innovation for Pigs, Chongqing, China
| | - Yang Zhang
- Collaborative Innovation Institute National Center of Technology Innovation for Pigs, Chongqing, China
- Institute of Veterinary Medicine Academy of Animal Sciences, Chongqing, China
| | - Lianci Peng
- College of Veterinary Medicine, Southwest University, Chongqing, China
- Collaborative Innovation Institute National Center of Technology Innovation for Pigs, Chongqing, China
- Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Lizhi Fu
- Collaborative Innovation Institute National Center of Technology Innovation for Pigs, Chongqing, China
- Institute of Veterinary Medicine Academy of Animal Sciences, Chongqing, China
| | - Yuandi Yu
- Collaborative Innovation Institute National Center of Technology Innovation for Pigs, Chongqing, China
- Institute of Veterinary Medicine Academy of Animal Sciences, Chongqing, China
| | - Dengfeng Xu
- Collaborative Innovation Institute National Center of Technology Innovation for Pigs, Chongqing, China
- Institute of Veterinary Medicine Academy of Animal Sciences, Chongqing, China
| | - Suhui Zhang
- Collaborative Innovation Institute National Center of Technology Innovation for Pigs, Chongqing, China
- Institute of Veterinary Medicine Academy of Animal Sciences, Chongqing, China
| | - Jinjie Qiu
- Collaborative Innovation Institute National Center of Technology Innovation for Pigs, Chongqing, China
- Institute of Veterinary Medicine Academy of Animal Sciences, Chongqing, China
| | - Yuzhang He
- College of Veterinary Medicine, Southwest University, Chongqing, China
- Collaborative Innovation Institute National Center of Technology Innovation for Pigs, Chongqing, China
- Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Hongzao Yang
- College of Veterinary Medicine, Southwest University, Chongqing, China
- Collaborative Innovation Institute National Center of Technology Innovation for Pigs, Chongqing, China
- Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Hongwei Chen
- College of Veterinary Medicine, Southwest University, Chongqing, China
- Collaborative Innovation Institute National Center of Technology Innovation for Pigs, Chongqing, China
- Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China
| |
Collapse
|
28
|
Louis M, Tahrioui A, Tremlett CJ, Clamens T, Leprince J, Lefranc B, Kipnis E, Grandjean T, Bouffartigues E, Barreau M, Defontaine F, Cornelis P, Feuilloley MG, Harmer NJ, Chevalier S, Lesouhaitier O. The natriuretic peptide receptor agonist osteocrin disperses Pseudomonas aeruginosa biofilm. Biofilm 2023; 5:100131. [PMID: 37252226 PMCID: PMC10220261 DOI: 10.1016/j.bioflm.2023.100131] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/02/2023] [Accepted: 05/18/2023] [Indexed: 05/31/2023] Open
Abstract
Biofilms are highly tolerant to antimicrobials and host immune defense, enabling pathogens to thrive in hostile environments. The diversity of microbial biofilm infections requires alternative and complex treatment strategies. In a previous work we demonstrated that the human Atrial Natriuretic Peptide (hANP) displays a strong anti-biofilm activity toward Pseudomonas aeruginosa and that the binding of hANP by the AmiC protein supports this effect. This AmiC sensor has been identified as an analog of the human natriuretic peptide receptor subtype C (h-NPRC). In the present study, we evaluated the anti-biofilm activity of the h-NPRC agonist, osteocrin (OSTN), a hormone that displays a strong affinity for the AmiC sensor at least in vitro. Using molecular docking, we identified a pocket in the AmiC sensor that OSTN reproducibly docks into, suggesting that OSTN might possess an anti-biofilm activity as well as hANP. This hypothesis was validated since we observed that OSTN dispersed established biofilm of P. aeruginosa PA14 strain at the same concentrations as hANP. However, the OSTN dispersal effect is less marked than that observed for the hANP (-61% versus -73%). We demonstrated that the co-exposure of P. aeruginosa preformed biofilm to hANP and OSTN induced a biofilm dispersion with a similar effect to that observed with hANP alone suggesting a similar mechanism of action of these two peptides. This was confirmed by the observation that OSTN anti-biofilm activity requires the activation of the complex composed by the sensor AmiC and the regulator AmiR of the ami pathway. Using a panel of both P. aeruginosa laboratory reference strains and clinical isolates, we observed that the OSTN capacity to disperse established biofilms is highly variable from one strain to another. Taken together, these results show that similarly to the hANP hormone, OSTN has a strong potential to be used as a tool to disperse P. aeruginosa biofilms.
Collapse
Affiliation(s)
- Melissande Louis
- Univ Rouen Normandie, Unité de Recherche Communication Bactérienne et Stratégies Anti-infectieuses, CBSA UR4312, 27000, Evreux, France
| | - Ali Tahrioui
- Univ Rouen Normandie, Unité de Recherche Communication Bactérienne et Stratégies Anti-infectieuses, CBSA UR4312, 27000, Evreux, France
| | - Courtney J. Tremlett
- Living Systems Institute, Stocker Road, University of Exeter, Exeter, EX4 4QD, UK
| | - Thomas Clamens
- Univ Rouen Normandie, Unité de Recherche Communication Bactérienne et Stratégies Anti-infectieuses, CBSA UR4312, 27000, Evreux, France
| | - Jérôme Leprince
- PRIMACEN, University of Rouen Normandy, 76821, Mont-Saint-Aignan, France
| | - Benjamin Lefranc
- PRIMACEN, University of Rouen Normandy, 76821, Mont-Saint-Aignan, France
| | - Eric Kipnis
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017-CIIL-Centre d'Infection et d'Immunité de Lille, University Lille, F-59000, Lille, France
| | - Teddy Grandjean
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017-CIIL-Centre d'Infection et d'Immunité de Lille, University Lille, F-59000, Lille, France
| | - Emeline Bouffartigues
- Univ Rouen Normandie, Unité de Recherche Communication Bactérienne et Stratégies Anti-infectieuses, CBSA UR4312, 27000, Evreux, France
| | - Magalie Barreau
- Univ Rouen Normandie, Unité de Recherche Communication Bactérienne et Stratégies Anti-infectieuses, CBSA UR4312, 27000, Evreux, France
| | - Florian Defontaine
- Univ Rouen Normandie, Unité de Recherche Communication Bactérienne et Stratégies Anti-infectieuses, CBSA UR4312, 27000, Evreux, France
| | - Pierre Cornelis
- Univ Rouen Normandie, Unité de Recherche Communication Bactérienne et Stratégies Anti-infectieuses, CBSA UR4312, 27000, Evreux, France
| | - Marc G.J. Feuilloley
- Univ Rouen Normandie, Unité de Recherche Communication Bactérienne et Stratégies Anti-infectieuses, CBSA UR4312, 27000, Evreux, France
| | - Nicholas J. Harmer
- Living Systems Institute, Stocker Road, University of Exeter, Exeter, EX4 4QD, UK
| | - Sylvie Chevalier
- Univ Rouen Normandie, Unité de Recherche Communication Bactérienne et Stratégies Anti-infectieuses, CBSA UR4312, 27000, Evreux, France
| | - Olivier Lesouhaitier
- Univ Rouen Normandie, Unité de Recherche Communication Bactérienne et Stratégies Anti-infectieuses, CBSA UR4312, 27000, Evreux, France
| |
Collapse
|
29
|
Grayton QE, Nguyen HK, Broberg CA, Ocampo J, Nagy SG, Schoenfisch MH. Biofilm Dispersal, Reduced Viscoelasticity, and Antibiotic Sensitization via Nitric Oxide-Releasing Biopolymers. ACS Infect Dis 2023; 9:1730-1741. [PMID: 37566512 DOI: 10.1021/acsinfecdis.3c00198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2023]
Abstract
Compared to planktonic bacteria, biofilms are notoriously difficult to eradicate due to their inherent protection against the immune response and antimicrobial agents. Inducing biofilm dispersal to improve susceptibility to antibiotics is an attractive therapeutic avenue for eradicating biofilms. Nitric oxide (NO), an endogenous antibacterial agent, has previously been shown to induce biofilm dispersal, but with limited understanding of the effects of NO-release properties. Herein, the antibiofilm effects of five promising NO-releasing biopolymer candidates were studied by assessing dispersal, changes in biofilm viscoelasticity, and increased sensitization to tobramycin after treatment with NO. A threshold level of NO was needed to achieve biofilm dispersal, with longer-releasing systems requiring lower concentrations. The most positively charged NO-release systems (from the presence of primary amines) led to the greatest reduction in viscoelasticity of Pseudomonas aeruginosa biofilms. Co-treatment of tobramycin with the NO-releasing biopolymer greatly decreased the dose of tobramycin required to eradicate tobramycin-susceptible and -resistant biofilms in both cellular and tissue models.
Collapse
|
30
|
Maliszewska I, Goldeman W. Increasing photoeradication's efficiency of Acinetobacter baumannii by polyphosphonic chelating agents. Photodiagnosis Photodyn Ther 2023; 43:103672. [PMID: 37364665 DOI: 10.1016/j.pdpdt.2023.103672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/17/2023] [Accepted: 06/22/2023] [Indexed: 06/28/2023]
Abstract
Ethylenediamine-N,N,N',N'-tetrakis(methylenephosphonic acid (EDTMP), nitrilotri(methylphosphonic acid (ATMP) and zoledronic acid were studied to enhance the methylene blue-mediated photodynamic inactivation of Acinetobacter baumannii. Laser light (wavelength 638 nm; standard light output 40 mW) was used in all experiment. Planktonic cultures were irradiated for 10, 20 and 30 min which corresponded to the light dose of 63 Jcm‒ 2, 126 Jcm‒2 and 189 Jcm‒2. Biocidal effect depended on the exposure time and it was shown that MB alone caused the highest reduction in the number of viable cells by 3.10 ± 0.2 log10 units after 30 min of irradiation. A significantly more effective killing effect was achieved when the bacteria were pre-treated with zoledronate, ATMP, or EDTMP (prior to photosensitisation) as the number of viable bacteria was reduced by 4.04±0.2 log10, 3.95±0.2 log10 and 4.01 ± 0.2 log10, respectively. The photo-killing effect caused by MB against biofilm pre-incubated with zoledronate, ATMP, or EDTMP and the number of viable bacteria was reduced by 0.80±0.1 log10, 1.25±0.05 log10 and 0.65±0.05 log10, respectively. Polyphosphonic chelating agents increased the efficiency of photo-destruction of A. baumannii by increasing the amount of bound photosensitizer by planktonic cells and biofilm, and increasing the detachment of live planktonic cells from the biofilm. The presence of glucose in the photosensitizing system significantly affected the bacterial photo-elimination. Pre-incubation of planktonic bacteria with the studied polyphosphonic chelating agents in the presence of glucose, and then exposure to light (with MB) for 30 min caused the lethal effect. This photo-eradication protocol (in the case of biofilms) reduced the number of viable bacteria by 2.05±0.2 log10, 3.2±0.2 log10 and 2.02±0.2 log10 for zoledronic acid, ATMP and EDTMP, respectively.
Collapse
Affiliation(s)
- Irena Maliszewska
- Department of Organic and Medicinal Chemistry, Faculty of Chemistry, Wrocaw University of Science and Technology, Wybrzeże Wyspiańskiego 27 Wrocław 50-370, Poland.
| | - Waldemar Goldeman
- Department of Organic and Medicinal Chemistry, Faculty of Chemistry, Wrocaw University of Science and Technology, Wybrzeże Wyspiańskiego 27 Wrocław 50-370, Poland
| |
Collapse
|
31
|
Li P, Yin R, Cheng J, Lin J. Bacterial Biofilm Formation on Biomaterials and Approaches to Its Treatment and Prevention. Int J Mol Sci 2023; 24:11680. [PMID: 37511440 PMCID: PMC10380251 DOI: 10.3390/ijms241411680] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Bacterial biofilms can cause widespread infection. In addition to causing urinary tract infections and pulmonary infections in patients with cystic fibrosis, biofilms can help microorganisms adhere to the surfaces of various medical devices, causing biofilm-associated infections on the surfaces of biomaterials such as venous ducts, joint prostheses, mechanical heart valves, and catheters. Biofilms provide a protective barrier for bacteria and provide resistance to antimicrobial agents, which increases the morbidity and mortality of patients. This review summarizes biofilm formation processes and resistance mechanisms, as well as the main features of clinically persistent infections caused by biofilms. Considering the various infections caused by clinical medical devices, we introduce two main methods to prevent and treat biomaterial-related biofilm infection: antibacterial coatings and the surface modification of biomaterials. Antibacterial coatings depend on the covalent immobilization of antimicrobial agents on the coating surface and drug release to prevent and combat infection, while the surface modification of biomaterials affects the adhesion behavior of cells on the surfaces of implants and the subsequent biofilm formation process by altering the physical and chemical properties of the implant material surface. The advantages of each strategy in terms of their antibacterial effect, biocompatibility, limitations, and application prospects are analyzed, providing ideas and research directions for the development of novel biofilm infection strategies related to therapeutic materials.
Collapse
Affiliation(s)
| | | | | | - Jinshui Lin
- Shaanxi Key Laboratory of Chinese Jujube, College of Life Sciences, Yan’an University, Yan’an 716000, China; (P.L.); (R.Y.); (J.C.)
| |
Collapse
|
32
|
Wu D, Zhao B, Zhang P, An Q. Insight into the effect of nitrate on AGS granulation: Granular characteristics, microbial community and metabolomics response. WATER RESEARCH 2023; 236:119949. [PMID: 37054606 DOI: 10.1016/j.watres.2023.119949] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 06/19/2023]
Abstract
As a promising wastewater treatment technology, aerobic granular sludge (AGS) process is still hindered by slow granule formation and easy disintegration in the application. While nitrate, one of the target pollutants in wastewater, showed a potential effect on AGS granulation process. Herein, this study attempted to reveal the role of nitrate in AGS granulation. By adding exogenous nitrate (10 mg L-1), the AGS formation was markedly improved and accomplished at 63 d, while the control group achieved AGS formation at 87 d. However, a disintegration was observed under a long-term nitrate feeding. A positive correlation was observed among granule size, extracellular polymeric substances (EPS) and intracellular c-di-GMP level in both formation and disintegration phases. The subsequent static biofilm assays indicated that nitrate might upregulate c-di-GMP via denitrification-derived NO, and c-di-GMP further upregulated EPS, thereby promoting AGS formation. However, excessive NO probably caused disintegration by downregulating c-di-GMP and EPS. Microbial community showed that nitrate favored the enrichment of denitrifiers and EPS producing microbes, which were responsible for the regulation of NO, c-di-GMP and EPS. Metabolomics analysis showed that amino acid metabolism was the most affected metabolism by nitrate. Some amino acids, such as Arg, His and Asp, were upregulated in the granule formation phase and downregulated in the disintegration phase, indicating the potential contribution to EPS biosynthesis. This study provides metabolic insight into how nitrate promotes/inhibits granulation, which may contribute to unwrapping the mystery of granulation and overcoming the limitations of AGS application.
Collapse
Affiliation(s)
- Danqing Wu
- Key Laboratory of the Three Gorges Reservoir Region's Eco-Environment, Ministry of Education, Chongqing University, 400045 Chongqing, China; College of Environment and Ecology, Chongqing University, 400045 Chongqing, China
| | - Bin Zhao
- Key Laboratory of the Three Gorges Reservoir Region's Eco-Environment, Ministry of Education, Chongqing University, 400045 Chongqing, China; College of Environment and Ecology, Chongqing University, 400045 Chongqing, China.
| | - Peng Zhang
- Key Laboratory of the Three Gorges Reservoir Region's Eco-Environment, Ministry of Education, Chongqing University, 400045 Chongqing, China; College of Environment and Ecology, Chongqing University, 400045 Chongqing, China
| | - Qiang An
- Key Laboratory of the Three Gorges Reservoir Region's Eco-Environment, Ministry of Education, Chongqing University, 400045 Chongqing, China; College of Environment and Ecology, Chongqing University, 400045 Chongqing, China
| |
Collapse
|
33
|
Van Alin A, Corbett MK, Fathollahzadeh H, Tjiam MC, Putnis A, Eksteen J, Kaksonen AH, Watkin E. Klebsiella aerogenes Adhesion Behaviour during Biofilm Formation on Monazite. Microorganisms 2023; 11:1331. [PMID: 37317305 PMCID: PMC10222597 DOI: 10.3390/microorganisms11051331] [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: 03/23/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 06/16/2023] Open
Abstract
The adsorption behaviour of micro-organisms during the initial attachment stage of biofilm formation affects subsequent stages. The available area for attachment and the chemophysical properties of a surface affect microbial attachment performance. This study focused on the initial attachment behaviour of Klebsiella aerogenes on monazite by measuring the ratio of planktonic against sessile subpopulations (P:S ratio), and the potential role of extracellular DNA (eDNA). eDNA production, effects of physicochemical properties of the surface, particle size, total available area for attachment, and the initial inoculation size on the attachment behaviour were tested. K. aerogenes attached to monazite immediately after exposure to the ore; however, the P:S ratio significantly (p = 0.05) changed in response to the particle size, available area, and inoculation size. Attachment occurred preferentially on larger-sized (~50 µm) particles, and either decreasing the inoculation size or increasing the available area further promoted attachment. Nevertheless, a portion of the inoculated cells always remained in a planktonic state. K. aerogenes produced lower eDNA in response to the changed surface chemical properties when monazite was replaced by xenotime. Using pure eDNA to cover the monazite surface significantly (p ≤ 0.05) hindered bacterial attachment due to the repulsive interaction between the eDNA layer and bacteria.
Collapse
Affiliation(s)
- Arya Van Alin
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; (A.V.A.); (H.F.); (M.C.T.)
- The Institute for Geoscience Research, School of Earth and Planetary Sciences, Curtin University, Bentley, WA 6102, Australia
| | - Melissa K. Corbett
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; (A.V.A.); (H.F.); (M.C.T.)
- The Institute for Geoscience Research, School of Earth and Planetary Sciences, Curtin University, Bentley, WA 6102, Australia
| | - Homayoun Fathollahzadeh
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; (A.V.A.); (H.F.); (M.C.T.)
- The Institute for Geoscience Research, School of Earth and Planetary Sciences, Curtin University, Bentley, WA 6102, Australia
| | - M. Christian Tjiam
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; (A.V.A.); (H.F.); (M.C.T.)
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Nedlands, WA 6009, Australia
- Centre for Child Health Research, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Andrew Putnis
- The Institute for Geoscience Research, School of Earth and Planetary Sciences, Curtin University, Bentley, WA 6102, Australia
- Institut für Mineralogie, University of Münster, 48149 Münster, Germany
| | - Jacques Eksteen
- WA School of Mines, Minerals, Energy and Chemical Engineering, Curtin University, Waterford, WA 6152, Australia; (J.E.); (A.H.K.)
| | - Anna H. Kaksonen
- WA School of Mines, Minerals, Energy and Chemical Engineering, Curtin University, Waterford, WA 6152, Australia; (J.E.); (A.H.K.)
- CSIRO Environment, Floreat, WA 6014, Australia
| | - Elizabeth Watkin
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; (A.V.A.); (H.F.); (M.C.T.)
- The Institute for Geoscience Research, School of Earth and Planetary Sciences, Curtin University, Bentley, WA 6102, Australia
- School of Science, Edith Cowan University, Joondalup, WA 6027, Australia
| |
Collapse
|
34
|
Smith WPJ, Wucher BR, Nadell CD, Foster KR. Bacterial defences: mechanisms, evolution and antimicrobial resistance. Nat Rev Microbiol 2023:10.1038/s41579-023-00877-3. [PMID: 37095190 DOI: 10.1038/s41579-023-00877-3] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 04/26/2023]
Abstract
Throughout their evolutionary history, bacteria have faced diverse threats from other microorganisms, including competing bacteria, bacteriophages and predators. In response to these threats, they have evolved sophisticated defence mechanisms that today also protect bacteria against antibiotics and other therapies. In this Review, we explore the protective strategies of bacteria, including the mechanisms, evolution and clinical implications of these ancient defences. We also review the countermeasures that attackers have evolved to overcome bacterial defences. We argue that understanding how bacteria defend themselves in nature is important for the development of new therapies and for minimizing resistance evolution.
Collapse
Affiliation(s)
- William P J Smith
- Division of Genomics, Infection and Evolution, University of Manchester, Manchester, UK.
- Department of Biology, University of Oxford, Oxford, UK.
- Department of Biochemistry, University of Oxford, Oxford, UK.
| | - Benjamin R Wucher
- Department of Biological sciences, Dartmouth College, Hanover, NH, USA
| | - Carey D Nadell
- Department of Biological sciences, Dartmouth College, Hanover, NH, USA
| | - Kevin R Foster
- Department of Biology, University of Oxford, Oxford, UK.
- Department of Biochemistry, University of Oxford, Oxford, UK.
| |
Collapse
|
35
|
Zhao A, Sun J, Liu Y. Understanding bacterial biofilms: From definition to treatment strategies. Front Cell Infect Microbiol 2023; 13:1137947. [PMID: 37091673 PMCID: PMC10117668 DOI: 10.3389/fcimb.2023.1137947] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/09/2023] [Indexed: 04/08/2023] Open
Abstract
Bacterial biofilms are complex microbial communities encased in extracellular polymeric substances. Their formation is a multi-step process. Biofilms are a significant problem in treating bacterial infections and are one of the main reasons for the persistence of infections. They can exhibit increased resistance to classical antibiotics and cause disease through device-related and non-device (tissue) -associated infections, posing a severe threat to global health issues. Therefore, early detection and search for new and alternative treatments are essential for treating and suppressing biofilm-associated infections. In this paper, we systematically reviewed the formation of bacterial biofilms, associated infections, detection methods, and potential treatment strategies, aiming to provide researchers with the latest progress in the detection and treatment of bacterial biofilms.
Collapse
Affiliation(s)
- Ailing Zhao
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Jiazheng Sun
- Department of Vasculocardiology, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yipin Liu
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
- *Correspondence: Yipin Liu,
| |
Collapse
|
36
|
Bravo P, Lung Ng S, MacGillivray KA, Hammer BK, Yunker PJ. Vertical growth dynamics of biofilms. Proc Natl Acad Sci U S A 2023; 120:e2214211120. [PMID: 36881625 PMCID: PMC10089195 DOI: 10.1073/pnas.2214211120] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 02/02/2023] [Indexed: 03/08/2023] Open
Abstract
During the biofilm life cycle, bacteria attach to a surface and then reproduce, forming crowded, growing communities. Many theoretical models of biofilm growth dynamics have been proposed; however, difficulties in accurately measuring biofilm height across relevant time and length scales have prevented testing these models, or their biophysical underpinnings, empirically. Using white light interferometry, we measure the heights of microbial colonies with nanometer precision from inoculation to their final equilibrium height, producing a detailed empirical characterization of vertical growth dynamics. We propose a heuristic model for vertical growth dynamics based on basic biophysical processes inside a biofilm: diffusion and consumption of nutrients and growth and decay of the colony. This model captures the vertical growth dynamics from short to long time scales (10 min to 14 d) of diverse microorganisms, including bacteria and fungi.
Collapse
Affiliation(s)
- Pablo Bravo
- School of Physics, Georgia Institute of Technology, Atlanta, GA30332
- Interdisciplinary Program in Quantitative Biosciences, Georgia Institute of Technology, Atlanta, GA30332
| | - Siu Lung Ng
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA30332
| | - Kathryn A. MacGillivray
- Interdisciplinary Program in Quantitative Biosciences, Georgia Institute of Technology, Atlanta, GA30332
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA30332
| | - Brian K. Hammer
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA30332
| | - Peter J. Yunker
- School of Physics, Georgia Institute of Technology, Atlanta, GA30332
| |
Collapse
|
37
|
Khanum R, Chung PY, Clarke SC, Chin BY. Lactoferrin modulates the biofilm formation and bap gene expression of methicillin-resistant Staphylococcus epidermidis. Can J Microbiol 2023; 69:117-122. [PMID: 36265186 DOI: 10.1139/cjm-2022-0135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Lactoferrin is an innate glycoprotein with broad antibacterial and antibiofilm properties. The autonomous antibiofilm activity of lactoferrin against Gram-positive bacteria is postulated to involve the cell wall and biofilm components. Thus, the prevention of biomass formation and eradication of preformed biofilms by lactoferrin was investigated using a methicillin-resistant Staphylococcus epidermidis (MRSE) strain. Additionally, the ability of lactoferrin to modulate the expression of the biofilm-associated protein gene (bap) was studied. The bap gene regulates the production of biofilm-associated proteins responsible for bacterial adhesion and aggregation. In the in vitro biofilm assays, lactoferrin prevented biofilm formation and eradicated established biofilms for up to 24 and 72 h, respectively. Extensive eradication of MRSE biofilm biomass was accompanied by the significant upregulation of bap gene expression. These data suggest the interaction of lactoferrin with the biofilm components and cell wall of MRSE, including the biofilm-associated protein.
Collapse
Affiliation(s)
- Ramona Khanum
- School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia
| | - Pooi Yin Chung
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Stuart C Clarke
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, UK.,Institute for Research, Development and Innovation, International Medical University, Kuala Lumpur, Malaysia
| | - Beek Yoke Chin
- School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
38
|
Kulišová M, Maťátková O, Brányik T, Zelenka J, Drábová L, Kolouchová IJ. Detection of microscopic filamentous fungal biofilms - Choosing the suitable methodology. J Microbiol Methods 2023; 205:106676. [PMID: 36693497 DOI: 10.1016/j.mimet.2023.106676] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/18/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
Microscopic filamentous fungi are ubiquitous microorganisms that adapt very easily to a variety of environmental conditions. Due to this adaptability, they can colonize a number of various surfaces where they are able to start forming biofilms. Life in the form of biofilms provides them with many benefits (increased resistance to desiccation, UV radiation, antimicrobial compounds, and host immune response). The aim of this study is to find a reliable and reproducible methodology to determine biofilm growth of selected microscopic filamentous fungi strains. Several methods (crystal violet staining, MTT assay, XTT assay, resazurin assay) for the determination of total biofilm biomass and its metabolic activity were tested on four fungi - Alternaria alternata, Aspergillus niger, Fusarium culmorum and Fusarium graminearum, and their biofilm was also imaged by spinning disc confocal microscopy using fluorescent dyes. A reproducible biofilm quantification method is essential for the subsequent testing of the biofilm growth suppression using antifungal agents or physical methods. Crystal violet staining was found to be a suitable method for the determination of total biofilm biomass of selected strains, and the MTT assay for the determination of metabolic activity of the biofilms. Calcofluor white and Nile red fluorescent stains successfully dyed the hyphae of microscopic fungi.
Collapse
Affiliation(s)
- Markéta Kulišová
- Department of Biotechnology, University of Chemistry and Technology, Prague, Technická 5, Prague 166 28, Czech Republic.
| | - Olga Maťátková
- Department of Biotechnology, University of Chemistry and Technology, Prague, Technická 5, Prague 166 28, Czech Republic.
| | - Tomáš Brányik
- Research Institute of Brewing and Malting, Lipová 511/15, Prague 120 44, Czech Republic.
| | - Jaroslav Zelenka
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Technická 5, Prague 166 28, Czech Republic.
| | - Lucie Drábová
- Department of Food Analysis and Nutrition, University of Chemistry and Technology, Prague, Technická 5, Prague 166 28, Czech Republic.
| | - Irena Jarošová Kolouchová
- Department of Biotechnology, University of Chemistry and Technology, Prague, Technická 5, Prague 166 28, Czech Republic.
| |
Collapse
|
39
|
Sharma A, Rishi P, Singh R. In vitro and in vivo evaluation of DNase I in reinstating antibiotic efficacy against Klebsiella pneumoniae biofilms. Pathog Dis 2023; 81:6986254. [PMID: 36633541 DOI: 10.1093/femspd/ftad001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/06/2023] [Accepted: 01/11/2023] [Indexed: 01/13/2023] Open
Abstract
Klebsiella pneumoniae is an opportunistic pathogen associated with biofilm-based infections, which are intrinsically antibiotic resistant. Extracellular DNA plays a crucial role in biofilm formation and self-defence, with nucleases being proposed as promising agents for biofilm disruption. This study evaluated the in vitro and in vivo efficacy of DNase I in improving the activity of cefotaxime, amikacin, and ciprofloxacin against K. pneumoniae biofilms. K. pneumoniae ATCC 700603 and a clinical isolate from catheter-related bloodstream infection were cultured for biofilm formation on microtiter plates, and the antibiofilm activity of the antibiotics (0.03-64 mg/L), with or without bovine pancreatic DNase I (1-32 mg/L) was determined by XTT dye reduction test and viable counting. The effect of ciprofloxacin (2 mg/L) and DNase I (16 mg/L) was further evaluated in vitro on 1-cm-long silicon catheter segments, and in a mouse model of subcutaneous catheter-associated infection. Combination with DNase I did not improve the biofilm-preventive capacity of the three antibiotics or the biofilm-eradicating capacity of cefotaxime and amikacin. The biofilm-eradicating capacity of ciprofloxacin was increased by 8-fold and 4-fold in K. pneumoniae ATCC 700603 and clinical isolate, respectively, with DNase I. The combination therapy caused 99% reduction in biofilm biomass in the mouse model.
Collapse
Affiliation(s)
- Anayata Sharma
- Department of Microbial Biotechnology, Panjab University, Chandigarh, 160014, India
| | - Praveen Rishi
- Department of Microbiology, Panjab University, Chandigarh, 160014, India
| | - Rachna Singh
- Department of Microbial Biotechnology, Panjab University, Chandigarh, 160014, India
| |
Collapse
|
40
|
Plant Growth-Promoting Bacteria (PGPB) with Biofilm-Forming Ability: A Multifaceted Agent for Sustainable Agriculture. DIVERSITY 2023. [DOI: 10.3390/d15010112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Plant growth-promoting bacteria (PGPB) enhance plant growth, as well as protect plants from several biotic and abiotic stresses through a variety of mechanisms. Therefore, the exploitation of PGPB in agriculture is feasible as it offers sustainable and eco-friendly approaches to maintaining soil health while increasing crop productivity. The vital key of PGPB application in agriculture is its effectiveness in colonizing plant roots and the phyllosphere, and in developing a protective umbrella through the formation of microcolonies and biofilms. Biofilms offer several benefits to PGPB, such as enhancing resistance to adverse environmental conditions, protecting against pathogens, improving the acquisition of nutrients released in the plant environment, and facilitating beneficial bacteria–plant interactions. Therefore, bacterial biofilms can successfully compete with other microorganisms found on plant surfaces. In addition, plant-associated PGPB biofilms are capable of protecting colonization sites, cycling nutrients, enhancing pathogen defenses, and increasing tolerance to abiotic stresses, thereby increasing agricultural productivity and crop yields. This review highlights the role of biofilms in bacterial colonization of plant surfaces and the strategies used by biofilm-forming PGPB. Moreover, the factors influencing PGPB biofilm formation at plant root and shoot interfaces are critically discussed. This will pave the role of PGPB biofilms in developing bacterial formulations and addressing the challenges related to their efficacy and competence in agriculture for sustainability.
Collapse
|
41
|
da Silva Filho PM, Paz IA, Nascimento NRFD, Abreu DS, Lopes LGDF, Sousa EHS, Longhinotti E. Nitroprusside─Expanding the Potential Use of an Old Drug Using Nanoparticles. Mol Pharm 2023; 20:6-22. [PMID: 36350781 DOI: 10.1021/acs.molpharmaceut.2c00661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
For more than 70 years, sodium nitroprusside (SNP) has been used to treat severe hypertension in hospital emergency settings. During this time, a few other clinical uses have also emerged such as in the treatment of acute heart failure as well as improving mitral incompetence and in the intra- and perioperative management during heart surgery. This drug functions by releasing nitric oxide (NO), which modulates several biological processes with many potential therapeutic applications. However, this small molecule has a short lifetime, and it has been administered through the use of NO donor molecules such as SNP. On the other hand, SNP also has some setbacks such as the release of cyanide ions, high water solubility, and very fast NO release kinetics. Currently, there are many drug delivery strategies that can be applied to overcome many of these limitations, providing novel opportunities for the use of old drugs, including SNP. This Perspective describes some nitroprusside properties and highlights new potential therapeutic uses arising from the use of drug delivery systems, mainly silica-based nanoparticles. There is a series of great opportunities to further explore SNP in many medical issues as reviewed, which deserves a closer look by the scientific community.
Collapse
Affiliation(s)
- Pedro Martins da Silva Filho
- Laboratório de Métodos de Análises e Modificação de Materiais (LABMA), Departamento de Química Analítica e Físico-Química, Universidade Federal do Ceará, 60440-900, Fortaleza, Ceará, Brazil.,Laboratório de Bioinorgânica, Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, PO Box 12200, Campus do Pici s/n, 60440-900, Fortaleza, Ceará, Brazil
| | - Iury Araújo Paz
- Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, 60714-110, Fortaleza, Ceará, Brazil
| | | | - Dieric S Abreu
- Laboratory of Materials & Devices (Lab MaDe), Departamento de Química Analítica e Físico-Química, Universidade Federal do Ceará, 60440-900, Fortaleza, Ceará, Brazil
| | - Luiz Gonzaga de França Lopes
- Laboratório de Bioinorgânica, Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, PO Box 12200, Campus do Pici s/n, 60440-900, Fortaleza, Ceará, Brazil
| | - Eduardo Henrique Silva Sousa
- Laboratório de Bioinorgânica, Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, PO Box 12200, Campus do Pici s/n, 60440-900, Fortaleza, Ceará, Brazil
| | - Elisane Longhinotti
- Laboratório de Métodos de Análises e Modificação de Materiais (LABMA), Departamento de Química Analítica e Físico-Química, Universidade Federal do Ceará, 60440-900, Fortaleza, Ceará, Brazil.,Laboratório de Bioinorgânica, Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, PO Box 12200, Campus do Pici s/n, 60440-900, Fortaleza, Ceará, Brazil
| |
Collapse
|
42
|
Xue Y, Ma H, Li YY. Anammox-based granulation cycle for sustainable granular sludge biotechnology from mechanisms to strategies: A critical review. WATER RESEARCH 2023; 228:119353. [PMID: 36423549 DOI: 10.1016/j.watres.2022.119353] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 11/06/2022] [Accepted: 11/11/2022] [Indexed: 06/16/2023]
Abstract
Anaerobic ammonium oxidation (anammox) granular sludge is a promising biotechnological process for treating low-carbon nitrogenous wastewater, and is featured with low energy consumption and footprint. Previous theoretical and experimental research on anammox granular sludge processes mainly focused on granulation (flocs → granules), but pay little attention to the granulation cycle including granulation and regeneration. This work reviewed the previous studies from the perspective of anammox granules lifecycle and proposed various sustainable formation mechanisms of anammox granules. By reviewing the anaerobic, aerobic, and anammox granulation mechanisms, we summarize the mechanisms of thermodynamic theory, heterogeneous growth, extracellular polymeric substance (EPS)-based adhesion, quorum sensing (QS)-based regulation, biomineralization-based growth, and stratification of microorganisms to understand anammox granulation. In the regeneration process, the formation of precursors for re-granulation is explained by the mechanisms of physical crushing, quorum quenching and dispersion cue sensing. Based on the granulation cycle mechanism, the rebuilding of the normal regeneration process is considered essential to avoid granule floatation and the wash-out of granules. This comprehensive review indicates that future research on anammox granulation cycle should focus on the effects of filamentous bacteria in denitrification-anammox granulation cycle, the role of QS/ quorum quenching (QQ)-based autoinducers, development of diversified mechanisms to understand the cycle and the cycle mechanisms of stored granules.
Collapse
Affiliation(s)
- Yi Xue
- Graduate School of Environmental Studies, Tohoku University, 6-6-06 Aza-Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8579, Japan
| | - Haiyuan Ma
- College of Environment and Ecology, Chongqing University, Chongqing 40045, China
| | - Yu-You Li
- Graduate School of Environmental Studies, Tohoku University, 6-6-06 Aza-Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8579, Japan; Department of Civil and Environmental Engineering, Graduate School of Engineering, Tohoku University, 6-6-06 Aza-Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8579, Japan.
| |
Collapse
|
43
|
Yum SJ, Jeong HG, Kim SM. Anti-biofilm effects of sinomenine against Staphylococcus aureus. Food Sci Biotechnol 2023; 32:83-90. [PMID: 36606087 PMCID: PMC9807730 DOI: 10.1007/s10068-022-01174-0] [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: 07/20/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 01/07/2023] Open
Abstract
Staphylococcus aureus is a gram-positive foodborne pathogen capable of forming strong biofilms. This study identified that anti-biofilm natural compound against S. aureus. Sinomenine, a natural compound, showed significantly reduced biofilm formation (31.97-39.86%), but no effect on bacterial growth was observed. The dispersion of preformed biofilms was observed by confocal laser scanning microscopy (CLSM). qRT-PCR revealed that sinomenine treatment significantly up-regulated agrA by 3.8-fold and down-regulated icaA gene by 3.1-fold. These indicate that sinomenine treatment induces biofilm dispersal due to cell-cell adhesion, polysaccharide intercellular adhesin (PIA), and phenol-soluble modulin (PSM) peptides production. Our results suggest that sinomenine can be used as a promising agent for effectively controlling biofilm formation and dispersion, thereby making S. aureus more susceptible to the action of antimicrobial agents.
Collapse
Affiliation(s)
- Su-Jin Yum
- Department of Food Science and Technology, College of Agriculture and Life Sciences, Chungnam National University, Daejeon, 34134 Korea
| | - Hee Gon Jeong
- Department of Food Science and Technology, College of Agriculture and Life Sciences, Chungnam National University, Daejeon, 34134 Korea
| | - Seung Min Kim
- Division of Human Ecology, Korea National Open University, Seoul, 03087 Korea
| |
Collapse
|
44
|
Zaуtsev EM, Britsina MV, Ozeretskovskaya MN, Bazhanova IG. Effect of trypsin, lidase and fluimucil on the growth of <i>Bordetella pertussis</i> biofilms on an abiotic substrate. JOURNAL OF MICROBIOLOGY, EPIDEMIOLOGY AND IMMUNOBIOLOGY 2022. [DOI: 10.36233/0372-9311-218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Aim. Study the effect of trypsin, lidase (hyaluronidase) and fluimucil (N-acetyl-L-cysteine) on the growth of biofilms of Bordetella pertussis strains on the abiotic substrate.
Materials and methods. In the experiments, the strains of the main B. pertussis serotypes isolated in the Russian Federation from whooping cough patients in 20012010 were used: No. 178 (serotype 1.2.0), No. 287 (serotype 1.0.3) and No. 317 (serotype 1.2.3), grown on a dense nutrient medium. The intensity of biofilm formation in a liquid nutrient medium in the presence of trypsin, lidase and fluimucil in round-bottomed polystyrene 96-well plates was estimated by staining with 0.1% gentian-violet solution.
Results. Trypsin suppressed the growth of biofilms and destroyed the formed biofilms. Lidase suppressed the growth of biofilms less actively, without affecting the formed biofilms. Fluimucil did not affect both the growth of biofilms and the formed biofilms. The growth of colonies typical for B. pertussis was noted when planting fluids from cultures in the presence of preparations, as well as from culture control wells on a dense nutrient medium.
Conclusion. The different effect of the drugs studied by us may be related to the different quantitative content of targets for trypsin (proteins), lidase (mucopolysaccharides, containing uronic acids), fluimucil (acid mucopolysaccharides) in the biofilm matrix. The growth of the typical morphological properties of the colony of B. pertussis during the sowing of culture seedlings on a dense nutrient medium testifies to the destruction of the biofilm matrix by trypsin and lidase in the absence of influence on plankton cells.
Collapse
|
45
|
Agarwalla SV, Ellepola K, Sorokin V, Ihsan M, Silikas N, Neto AHC, Seneviratne CJ, Rosa V. Antimicrobial-free graphene nanocoating decreases fungal yeast-to-hyphal switching and maturation of cross-kingdom biofilms containing clinical and antibiotic-resistant bacteria. BIOMATERIALS AND BIOSYSTEMS 2022; 8:100069. [PMID: 36824379 PMCID: PMC9934433 DOI: 10.1016/j.bbiosy.2022.100069] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 10/23/2022] [Accepted: 10/23/2022] [Indexed: 12/05/2022] Open
Abstract
Candida albicans and methicillin-resistant Staphylococcus aureus (MRSA) synergize in cross-kingdom biofilms to increase the risk of mortality and morbidity due to high resistance to immune and antimicrobial defenses. Biomedical devices and implants made with titanium are vulnerable to infections that may demand their surgical removal from the infected sites. Graphene nanocoating (GN) has promising anti-adhesive properties against C. albicans. Thus, we hypothesized that GN could prevent fungal yeast-to-hyphal switching and the development of cross-kingdom biofilms. Herein, titanium (Control) was coated with high-quality GN (coverage > 99%). Thereafter, mixed-species biofilms (C. albicans combined with S. aureus or MRSA) were allowed to develop on GN and Control. There were significant reductions in the number of viable cells, metabolic activity, and biofilm biomass on GN compared with the Control (CFU counting, XTT reduction, and crystal violet assays). Also, biofilms on GN were sparse and fragmented, whereas the Control presented several bacterial cells co-aggregating with intertwined hyphal elements (confocal and scanning electronic microscopy). Finally, GN did not induce hemolysis, an essential characteristic for blood-contacting biomaterials and devices. Thus, GN significantly inhibited the formation and maturation of deadly cross-kingdom biofilms, which can be advantageous to avoid infection and surgical removal of infected devices.
Collapse
Affiliation(s)
| | - Kassapa Ellepola
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, USA
| | - Vitaly Sorokin
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore,Department of Cardiac, Thoracic and Vascular Surgery, National University Hospital, National University Health System, Singapore
| | - Mario Ihsan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Nikolaos Silikas
- Dentistry, The University of Manchester, Manchester, United Kingdom
| | - AH Castro Neto
- Centre for Advanced 2D Materials, National University of Singapore, Singapore
| | - Chaminda Jayampath Seneviratne
- School of Dentistry, The University of Queensland, Australia,Co-corresponding author at: School of Dentistry, The University of Queensland, 288 Herston Road, Cnr Bramston Terrace & Herston Road Herston QLD 4006, Australia.
| | - Vinicius Rosa
- Faculty of Dentistry, National University of Singapore, Singapore,Centre for Advanced 2D Materials, National University of Singapore, Singapore,ORCHIDS: Oral Care Health Innovations and Designs Singapore, National University of Singapore, Singapore,Corresponding author at: Faculty of Dentistry, National University of Singapore, 9 Lower Kent Ridge Road, 119085, Singapore.
| |
Collapse
|
46
|
Bu F, Liu M, Xie Z, Chen X, Li G, Wang X. Targeted Anti-Biofilm Therapy: Dissecting Targets in the Biofilm Life Cycle. Pharmaceuticals (Basel) 2022; 15:1253. [PMID: 36297365 PMCID: PMC9611117 DOI: 10.3390/ph15101253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 06/13/2024] Open
Abstract
Biofilm is a crucial virulence factor for microorganisms that causes chronic infection. After biofilm formation, the bacteria present improve drug tolerance and multifactorial defense mechanisms, which impose significant challenges for the use of antimicrobials. This indicates the urgent need for new targeted technologies and emerging therapeutic strategies. In this review, we focus on the current biofilm-targeting strategies and those under development, including targeting persistent cells, quorum quenching, and phage therapy. We emphasize biofilm-targeting technologies that are supported by blocking the biofilm life cycle, providing a theoretical basis for design of targeting technology that disrupts the biofilm and promotes practical application of antibacterial materials.
Collapse
Affiliation(s)
| | | | | | | | | | - Xing Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
47
|
Lee H, Kharel S, Loo SCJ. Lipid-Coated Hybrid Nanoparticles for Enhanced Bacterial Biofilm Penetration and Antibiofilm Efficacy. ACS OMEGA 2022; 7:35814-35824. [PMID: 36249378 PMCID: PMC9558607 DOI: 10.1021/acsomega.2c04008] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/23/2022] [Indexed: 06/01/2023]
Abstract
Up to 80% of all infections are biofilm-mediated and they are often challenging to treat as the underlying bacterial cells can become 100- to 1000-fold more tolerant toward antibiotics. Antibiotic-loaded nanoparticles have gained traction as a potential drug delivery system to treat biofilm infections. In particular, lipid-coated hybrid nanoparticles (LCHNPs) were investigated on their capability to deliver antibiotics into biofilms. In this study, LCHNPs composed of a poly(lactic-co-glycolic acid) (PLGA) core and dioleoyl-3-trimethylammonium propane (DOTAP) lipid shell were developed and loaded with vancomycin (Van). In vitro antibacterial and antibiofilm tests were performed to evaluate the antimicrobial efficacy of the LCHNPs. LCHNPs were successfully fabricated with high vancomycin encapsulation and loading efficiencies, and exhibited enhanced antibacterial effects against planktonic Staphylococcus aureus USA300 when compared against Free-Van and Van-PLGANPs. When used to treat USA300 biofilms, Van-LCHNPs eradicated up to 99.99% of the underlying biofilm cells, an effect which was not observed for Free-Van and Van-PLGANPs. Finally, we showed that by possessing a robust DOTAP shell, LCHNPs were able to penetrate deeply into the biofilms.
Collapse
Affiliation(s)
- Hiang
Wee Lee
- School
of Materials Science & Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Sharad Kharel
- School
of Materials Science & Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Say Chye Joachim Loo
- School
of Materials Science & Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
- Singapore
Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551, Singapore
- Lee
Kong Chian School of Medicine, Nanyang Technological
University, Singapore 308232, Singapore
| |
Collapse
|
48
|
Ordek A, Gordesli-Duatepe FP. Impact of sodium nitroprusside concentration added to batch cultures of Escherichia coli biofilms on the c-di-GMP levels, morphologies and adhesion of biofilm-dispersed cells. BIOFOULING 2022; 38:796-813. [PMID: 36229918 DOI: 10.1080/08927014.2022.2131399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/19/2022] [Accepted: 09/28/2022] [Indexed: 06/16/2023]
Abstract
Biofilm dispersion can be triggered by the application of dispersing agents such as nitric oxide (NO)-donors, resulting in the release of biofilm-dispersed cells into the environment. In this work, biofilm-dispersed cells were obtained by adding different concentrations of NO-donor sodium nitroprusside (0.5, 5, 50 µM, and 2.5 mM of SNP) to batch cultures of pre-formed Escherichia coli biofilms. Except for those dispersed by 5 µM of SNP, biofilm-dispersed cells were found to be wider and longer than the planktonic cells and to have higher c-di-GMP levels and greater adhesion forces to silicon nitride surfaces in water as measured by atomic force microscope. Consequently, the optimum concentration of SNP to disperse E. coli biofilms was found to be 5 µM of SNP, whose addition to batch cultures resulted in a significant biofilm dispersion and the dispersed cells having c-di-GMP levels, morphologies and adhesion strengths similar to their planktonic counterparts.
Collapse
Affiliation(s)
- Ayse Ordek
- Bioengineering Graduate Program, Graduate School, Izmir University of Economics, Izmir, Turkey
| | - F Pinar Gordesli-Duatepe
- Department of Genetics and Bioengineering, Faculty of Engineering, Izmir University of Economics, Izmir, Turkey
| |
Collapse
|
49
|
Dey A, Yadav M, Kumar D, Dey AK, Samal S, Tanwar S, Sarkar D, Pramanik SK, Chaudhuri S, Das A. A combination therapy strategy for treating antibiotic resistant biofilm infection using a guanidinium derivative and nanoparticulate Ag(0) derived hybrid gel conjugate. Chem Sci 2022; 13:10103-10118. [PMID: 36128224 PMCID: PMC9430544 DOI: 10.1039/d2sc02980d] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 07/22/2022] [Indexed: 11/25/2022] Open
Abstract
Bacteria organized in biofilms show significant tolerance to conventional antibiotics compared to their planktonic counterparts and form the basis for chronic infections. Biofilms are composites of different types of extracellular polymeric substances that help in resisting several host-defense measures, including phagocytosis. These are increasingly being recognized as a passive virulence factor that enables many infectious diseases to proliferate and an essential contributing facet to anti-microbial resistance. Thus, inhibition and dispersion of biofilms are linked to addressing the issues associated with therapeutic challenges imposed by biofilms. This report is to address this complex issue using a self-assembled guanidinium-Ag(0) nanoparticle (AD-L@Ag(0)) hybrid gel composite for executing a combination therapy strategy for six difficult to treat biofilm-forming and multidrug-resistant bacteria. Improved efficacy was achieved primarily through effective biofilm inhibition and dispersion by the cationic guanidinium ion derivative, while Ag(0) contributes to the subsequent bactericidal activity on planktonic bacteria. Minimum Inhibitory Concentration (MIC) of the AD-L@Ag(0) formulation was tested against Acinetobacter baumannii (25 μg mL-1), Pseudomonas aeruginosa (0.78 μg mL-1), Staphylococcus aureus (0.19 μg mL-1), Klebsiella pneumoniae (0.78 μg mL-1), Escherichia coli (clinical isolate (6.25 μg mL-1)), Klebsiella pneumoniae (clinical isolate (50 μg mL-1)), Shigella flexneri (clinical isolate (0.39 μg mL-1)) and Streptococcus pneumoniae (6.25 μg mL-1). Minimum bactericidal concentration, and MBIC50 and MBIC90 (Minimum Biofilm Inhibitory Concentration at 50% and 90% reduction, respectively) were evaluated for these pathogens. All these results confirmed the efficacy of the formulation AD-L@Ag(0). Minimum Biofilm Eradication Concentration (MBEC) for the respective pathogens was examined by following the exopolysaccharide quantification method to establish its potency in inhibition of biofilm formation, as well as eradication of mature biofilms. These effects were attributed to the bactericidal effect of AD-L@Ag(0) on biofilm mass-associated bacteria. The observed efficacy of this non-cytotoxic therapeutic combination (AD-L@Ag(0)) was found to be better than that reported in the existing literature for treating extremely drug-resistant bacterial strains, as well as for reducing the bacterial infection load at a surgical site in a small animal BALB/c model. Thus, AD-L@Ag(0) could be a promising candidate for anti-microbial coatings on surgical instruments, wound dressing, tissue engineering, and medical implants.
Collapse
Affiliation(s)
- Ananta Dey
- CSIR - Central Salt and Marine Chemical Research Institute Bhavnagar Gujarat India
- Indian Institute of Science Education and Research Kolkata Mohanpur 741246 West Bengal India
| | - Manisha Yadav
- Translational Health Science and Technology Institute (THSTI) Faridabad 121001 Haryana India
| | - Deepak Kumar
- Translational Health Science and Technology Institute (THSTI) Faridabad 121001 Haryana India
| | - Anik Kumar Dey
- CSIR - Central Salt and Marine Chemical Research Institute Bhavnagar Gujarat India
| | - Sweety Samal
- Translational Health Science and Technology Institute (THSTI) Faridabad 121001 Haryana India
| | - Subhash Tanwar
- Translational Health Science and Technology Institute (THSTI) Faridabad 121001 Haryana India
| | - Debrupa Sarkar
- Translational Health Science and Technology Institute (THSTI) Faridabad 121001 Haryana India
| | - Sumit Kumar Pramanik
- CSIR - Central Salt and Marine Chemical Research Institute Bhavnagar Gujarat India
| | - Susmita Chaudhuri
- Translational Health Science and Technology Institute (THSTI) Faridabad 121001 Haryana India
| | - Amitava Das
- Indian Institute of Science Education and Research Kolkata Mohanpur 741246 West Bengal India
| |
Collapse
|
50
|
Control Measurements of Escherichia coli Biofilm: A Review. Foods 2022; 11:foods11162469. [PMID: 36010469 PMCID: PMC9407607 DOI: 10.3390/foods11162469] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/04/2022] [Accepted: 08/11/2022] [Indexed: 12/05/2022] Open
Abstract
Escherichia coli (E. coli) is a common pathogen that causes diarrhea in humans and animals. In particular, E. coli can easily form biofilm on the surface of living or non-living carriers, which can lead to the cross-contamination of food. This review mainly summarizes the formation process of E. coli biofilm, the prevalence of biofilm in the food industry, and inhibition methods of E. coli biofilm, including chemical and physical methods, and inhibition by bioactive extracts from plants and animals. This review aims to provide a basis for the prevention and control of E. coli biofilm in the food industry.
Collapse
|