1
|
Rao W, Wu J, Fang Z, Chen Z, Wu J, Fang X. Antibacterial mechanism of metabolites of Lactobacillus plantarum against Pseudomonas lundensis and their application in dry-aged beef preservation. Food Chem 2024; 460:140463. [PMID: 39047473 DOI: 10.1016/j.foodchem.2024.140463] [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: 04/24/2024] [Revised: 06/26/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024]
Abstract
In this study, the antibacterial mechanism of metabolites of Lactobacillus plantarum SCB2505 (MLp SCB2505) against Pseudomonas lundensis (P. lundensis) SCB2605 was investigated, along with evaluation of their preservative effects on dry-aged beef. The results demonstrated the effective inhibition of MLp SCB2505 on the growth and biofilm synthesis of P. lundensis. The treatment with MLp SCB2505 led to the compromised membrane integrity, as evidenced by reduced intracellular ATP content, increased extracellular AKPase, K+ and protein content, as well as disrupted cell morphology. Further metabolomics analysis revealed that MLp SCB2505 interfered amino acid metabolism, nucleotide metabolism, cofactor and vitamin metabolism, lipid metabolism and respiratory chain in P. lundensis, ultimately leading to the interrupted life activities and even death of the bacteria. Besides, MLp SCB2505 could effectively inhibit the growth of Pseudomonas in dry-aged beef and delay spoilage. These findings propose the potential application of MLp SCB2505 as an antibacterial agent in meat products.
Collapse
Affiliation(s)
- Wei Rao
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Jinchong Wu
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Ziying Fang
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Zhaomin Chen
- Weyran Food Biotechnology (Shenzhen) Co., LTD., Shenzhen 518048, China
| | - Jianfeng Wu
- College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | - Xiang Fang
- College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
2
|
Christensen MH, Jakobsen TH, Lichtenberg M, Hertz FB, Dahl B, Bjarnsholt T. Antimicrobial susceptibility testing of bone and joint pathogens using isothermal microcalorimetry. APMIS 2024; 132:814-823. [PMID: 39301971 DOI: 10.1111/apm.13470] [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: 08/21/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
The rise in osteomyelitis and periprosthetic joint infections, in combination with increasing life expectancy and the prevalence of diabetes, underscores the urgent need for rapid and accurate diagnostic tools. Conventional culture-based methods are often time-consuming and prone to false-negatives, leading to prolonged and inappropriate antibiotic treatments. This study aims to improve osteomyelitis diagnostics by decreasing the time to detection and the time to an antibiotic susceptibility result to enable a targeted treatment using isothermal microcalorimetry (IMC). IMC measures heat flow in real-time, providing insights into bacterial metabolism without the need for labeling. Using clinical isolates from bone infections, assessing their response to antibiotics through IMC, we demonstrated that IMC could detect bacteria within 4 h and determine antimicrobial susceptibility profiles within 2-22 h (median 4.85, range 1.28-21.78). This is significantly faster than traditional methods. A decision tree, based on antibiotic susceptibility, accurately categorized pathogens, achieving high accuracy (74-100%), sensitivity (100%), and specificity (65-100%). These findings suggest that IMC could redefine diagnostics of bone and joint infections and potentially infections in general, offering timely and precise treatment guidance, thereby improving patient outcomes and reducing health care burdens. Further optimization and clinical validation are needed to fully integrate IMC into routine diagnostics.
Collapse
Affiliation(s)
- Mads H Christensen
- Department of Immunology and Microbiology, Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
| | - Tim H Jakobsen
- Department of Immunology and Microbiology, Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
| | - Mads Lichtenberg
- Department of Immunology and Microbiology, Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
| | - Frederik B Hertz
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Benny Dahl
- Department of Orthopedics Surgery, Copenhagen University Hospital, Copenhagen, Denmark
| | - Thomas Bjarnsholt
- Department of Immunology and Microbiology, Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
3
|
Pradhan L, Hazra S, Singh SV, Bajrang, Upadhyay A, Pal BN, Mukherjee S. Surface modification of medical grade biomaterials by using a low-temperature-processed dual functional Ag-TiO 2 coating for preventing biofilm formation. J Mater Chem B 2024; 12:10093-10109. [PMID: 39264339 DOI: 10.1039/d4tb00701h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Biofilm development in medical devices is considered the major virulence component that leads to increased mortality and morbidity among patients. Removing a biofilm once formed is challenging and frequently results in persistent infections. Many current antibiofilm coating strategies involve harsh conditions causing damage to the surface of the medical devices. To address the issue of bacterial attachment in medical devices, we propose a novel antibacterial surface modification approach. In this paper, we developed a novel low-temperature based solution-processed approach to deposit silver nanoparticles (Ag NPs) inside a titanium oxide (TiO2) matrix to obtain a Ag-TiO2 nanoparticle coating. The low temperature (120 °C)-based UV annealed drop cast method is novel and ensures no surface damage to the medical devices. Various medical-grade biomaterials were then coated using Ag-TiO2 to modify the surface of the materials. Several studies were performed to observe the antibacterial and antibiofilm properties of Ag-TiO2-coated medical devices and biomaterials. Moreover, the Ag-TiO2 NPs did not show any skin irritation in rats and showed biocompatibility in the chicken egg model. This study indicates that Ag-TiO2 coating has promising potential for healthcare applications to combat microbial infection and biofilm formation.
Collapse
Affiliation(s)
- Lipi Pradhan
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, UP, India.
| | - Sobhan Hazra
- School of Materials Science and Technology, Indian Institute of Technology (BHU), Varanasi, UP, 221005, India.
| | - Satya Veer Singh
- School of Materials Science and Technology, Indian Institute of Technology (BHU), Varanasi, UP, 221005, India.
| | - Bajrang
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, UP, India.
| | - Anjali Upadhyay
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, UP, India.
| | - Bhola Nath Pal
- School of Materials Science and Technology, Indian Institute of Technology (BHU), Varanasi, UP, 221005, India.
| | - Sudip Mukherjee
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, UP, India.
| |
Collapse
|
4
|
Chelliah R, Park CR, Park SJ, Barathikannan K, Kim EJ, Wei S, Sultan G, Hirad AH, Vijayalakshmi S, Oh DH. Novel approach on the evaluation of enzyme-aided alkaline extraction of polysaccharide from Hordeum vulgare husk and molecular insight on the multifunctional scaffold. Int J Biol Macromol 2024; 278:134153. [DOI: https:/doi.10.1016/j.ijbiomac.2024.134153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
|
5
|
Chelliah R, Park CR, Park SJ, Barathikannan K, Kim EJ, Wei S, Sultan G, Hirad AH, Vijayalakshmi S, Oh DH. Novel approach on the evaluation of enzyme-aided alkaline extraction of polysaccharide from Hordeum vulgare husk and molecular insight on the multifunctional scaffold. Int J Biol Macromol 2024; 278:134153. [PMID: 39127270 DOI: 10.1016/j.ijbiomac.2024.134153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 06/03/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024]
Abstract
Hordeum vulgare husk, a cereal grain, is rich in dietary fiber and prebiotics beneficial for the gut microbiota and host organism. This study investigates the effects of barley husk-derived water-soluble xylan (BH-WSX) on gut homeostasis and the microbiome. We enzymatically extracted BH-WSX and evaluated its prebiotic and antioxidant properties. A 40.0 % (w/v) xylan yield was achieved, with the extracted xylan having a molecular mass of 212.0885 and a xylose to glucuronic acid molar ratio of 6:1. Specialized optical rotation research indicated that the isolated xylan is composed of monomeric sugars such as D-xylose, glucose, and arabinose. Fourier Transform Infrared (FTIR) spectroscopy revealed that the xylan comprises β (1 → 4) linked xylose units, randomly substituted with glucose residues, α-arabinofuranose, and acetyl groups. Nuclear Magnetic Resonance (NMR) analysis showed that the barley husk extract's backbone is substituted with 4-O-methyl glucuronic acid at the O2 position. Thermogravimetric analysis indicated that WSX exhibits a single sharp peak at 266 °C on the Differential Thermal Gravimetry (DTG) curve. Furthermore, a combination of in vitro, in vivo models, and molecular docking analysis elaborated on the anti-adhesion properties of BH-WSX. This study presents a novel approach to utilizing barley husk as an efficient source of functional polysaccharides for food-related industrial applications.
Collapse
Affiliation(s)
- Ramachandran Chelliah
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon 24341, South Korea; Future F Biotech Co., Ltd., Chuncheon 24341, South Korea; Saveetha School of Engineering, (SIMATS) University, Sriperumbudur, India; College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Centre of Seafood, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China
| | - Chae Rin Park
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon 24341, South Korea
| | - Se Jin Park
- School of Natural Resources and Environmental Sciences, Kangwon National University, Chuncheon 24341, South Korea
| | - Kaliyan Barathikannan
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon 24341, South Korea; Future F Biotech Co., Ltd., Chuncheon 24341, South Korea; Agriculture and Life Science Research Institute, Kangwon National University, Chuncheon 24341, South Korea
| | - Eun Ji Kim
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon 24341, South Korea
| | - Shuai Wei
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Centre of Seafood, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China; Collaborative Innovation Centre of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Ghazala Sultan
- Department of Computer Science, Aligarh Muslim University, Aligarh 202002, India
| | - Abdurahman Hajinur Hirad
- Department of Botany and Microbiology, College of Science, King Saud University, P. O. Box.2455, Riyadh, 11451, Saudi Arabia
| | - Selvakumar Vijayalakshmi
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon 24341, South Korea; Future F Biotech Co., Ltd., Chuncheon 24341, South Korea
| | - Deog Hwan Oh
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon 24341, South Korea; Future F Biotech Co., Ltd., Chuncheon 24341, South Korea.
| |
Collapse
|
6
|
Alexander PJ, Oyama LB, Olleik H, Godoy Santos F, O'Brien S, Cookson A, Cochrane SA, Gilmore BF, Maresca M, Huws SA. Microbiome-derived antimicrobial peptides show therapeutic activity against the critically important priority pathogen, Acinetobacter baumannii. NPJ Biofilms Microbiomes 2024; 10:92. [PMID: 39349945 PMCID: PMC11443000 DOI: 10.1038/s41522-024-00560-2] [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: 02/25/2024] [Accepted: 08/21/2024] [Indexed: 10/04/2024] Open
Abstract
Acinetobacter baumannii is designated by the World Health Organisation as a critical priority pathogen. Previously we discovered antimicrobial peptides (AMPs), namely Lynronne-1, -2 and -3, with efficacy against bacterial pathogens, such as Staphylococcus aureus and Pseudomonas aeruginosa. Here we assessed Lynronne-1, -2 and -3 structure by circular dichroism and efficacy against clinical strains of A. baumannii. All Lynronne AMPs demonstrated alpha-helical secondary structures and had antimicrobial activity towards all tested strains of A. baumannii (Minimum Inhibitory Concentrations 2-128 μg/ml), whilst also having anti-biofilm activity. Lynronne-2 and -3 demonstrated additive effects with amoxicillin and erythromycin, and synergy with gentamicin. The AMPs demonstrated little toxicity towards mammalian cell lines or Galleria mellonella. Fluorescence-based assay data demonstrated that Lynronne-1 and -3 had higher membrane-destabilising action against A. baumannii in comparison with Lynronne-2, which was corroborated by transcriptomic analysis. For the first time, we demonstrate the therapeutic activity of Lynronne AMPs against A. baumannii.
Collapse
Affiliation(s)
- P J Alexander
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, UK
| | - L B Oyama
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, UK
| | - H Olleik
- Aix Marseille Univ, CNRS, Centrale Marseille, iSm2 (UMR7313), Marseille, France
| | - F Godoy Santos
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, UK
| | - S O'Brien
- School of Pharmacy, QUB, Medical Biology Centre, Belfast, UK
| | - A Cookson
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, UK
| | - S A Cochrane
- School of Chemistry and Chemical Engineering, Queen's University Belfast, Belfast, UK
| | - B F Gilmore
- School of Pharmacy, QUB, Medical Biology Centre, Belfast, UK
| | - M Maresca
- Aix Marseille Univ, CNRS, Centrale Marseille, iSm2 (UMR7313), Marseille, France
| | - S A Huws
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, UK.
| |
Collapse
|
7
|
Chen C, Gu Q, Ge Y, Tian J, Zhang Y, Wang T, Wang C, Zhao Y, Xu H, Fan X. Antibiofilm Mechanisms of the Helical G3 Peptide against Staphylococcus epidermidis. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:11806-11816. [PMID: 38770910 DOI: 10.1021/acs.langmuir.4c01474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Antibacterial peptides (ABPs) have been recognized as promising alternatives to conventional antibiotics due to their broad antibacterial spectrum, high antibacterial activity, and low possibility of inducing bacterial resistance. However, their antibiofilm mechanisms have not yet reached a consensus. In this study, we investigated the antibiofilm activity of a short helical peptide G3 against Staphylococcus epidermidis, one of the most important strains of medical device contamination. Studies show that G3 inhibits S. epidermidis biofilm formation in a variety of ways. In the initial adhesion stage, G3 changes the properties of bacterial surfaces, such as charges, hydrophobicity, and permeability, by rapidly binding to them, thus interfering with their initial adhesion. In the mature stage, G3 prefers to target extracellular polysaccharides, leading to the death of outside bacteria and the disruption of the three-dimensional (3D) architecture of the bacterial biofilm. Such efficient antibiofilm activity of G3 endows it with great potential in the treatment of infections induced by the S. epidermidis biofilm.
Collapse
Affiliation(s)
- Cuixia Chen
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Qilong Gu
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Yangyang Ge
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Jingyun Tian
- Marine Science Research Institute of Shandong Province (National Oceanographic Center, Qingdao), 7 Youyun Road, Laoshan District, Qingdao 266104, China
| | - Yusen Zhang
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Tianling Wang
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Chen Wang
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Yurong Zhao
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Hai Xu
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Xinglong Fan
- Department of Thoracic Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China
| |
Collapse
|
8
|
Acosta RB, Durantini EN, Spesia MB. Evaluation of quantification methods to determine photodynamic action on mono- and dual-species bacterial biofilms. Photochem Photobiol Sci 2024; 23:1195-1208. [PMID: 38703274 DOI: 10.1007/s43630-024-00586-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/23/2024] [Indexed: 05/06/2024]
Abstract
The effect of photodynamic inactivation (PDI) sensitized by 5,10,15,20-tetra(4-N,N,N-trimethylammoniophenyl)porphyrin (TMAP4+) on different components of mono- and dual-species biofilms of Staphylococcus aureus and Escherichia coli was determined by different methods. First, the plate count technique showed that TMAP4+-PDI was more effective on S. aureus than E. coli biofilm. However, crystal violet staining revealed no significant differences between before and after PDI biofilms of both bacteria. On the other hand, the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide method indicated a reduction in viable cells as the light exposure time increases in both, mono- and dual-species biofilms. Furthermore, it was determined that as the irradiation time increases, the amount of extracellular polymeric substances present in the biofilms decreased. This effect was presented in both strains and in the mixed biofilm, being more evident in S. aureus mono-specie biofilm. Finally, scanning electron microscopy analysis showed a decrease in the number of cells forming the biofilm after photosensitization treatments. This information makes it possible to determine whether the photodynamic action is based on damage to metabolic activity, extracellular matrix and/or biomass, which may be useful in establishing a fully effective PDI protocol for the treatment of microorganisms growing as biofilms.
Collapse
Affiliation(s)
- Rocío B Acosta
- IDAS-CONICET, Departamento de Química, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Ruta Nacional 36 Km 601, X5804BYA, Río Cuarto, Córdoba, Argentina
| | - Edgardo N Durantini
- IDAS-CONICET, Departamento de Química, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Ruta Nacional 36 Km 601, X5804BYA, Río Cuarto, Córdoba, Argentina
| | - Mariana B Spesia
- IDAS-CONICET, Departamento de Química, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Ruta Nacional 36 Km 601, X5804BYA, Río Cuarto, Córdoba, Argentina.
| |
Collapse
|
9
|
Sousa AM, Pereira MO. Challenges with drug efficacy prediction of in vitro models of biofilms infecting cystic fibrosis airway. Expert Opin Drug Discov 2024; 19:635-638. [PMID: 38712907 DOI: 10.1080/17460441.2024.2350567] [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: 02/04/2024] [Accepted: 04/29/2024] [Indexed: 05/08/2024]
Affiliation(s)
- Ana Margarida Sousa
- CEB - Centre of Biological Engineering, University of Minho, Braga, Portugal
- LABBELS - Associate Laboratory, Braga, Portugal
| | - Maria Olívia Pereira
- CEB - Centre of Biological Engineering, University of Minho, Braga, Portugal
- LABBELS - Associate Laboratory, Braga, Portugal
| |
Collapse
|
10
|
Lutfi LL, Shaaban MI, Elshaer SL. Vitamin D and vitamin K1 as novel inhibitors of biofilm in Gram-negative bacteria. BMC Microbiol 2024; 24:173. [PMID: 38762474 PMCID: PMC11102130 DOI: 10.1186/s12866-024-03293-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/04/2024] [Indexed: 05/20/2024] Open
Abstract
BACKGROUND The persistent surge in antimicrobial resistance represents a global disaster. The initial attachment and maturation of microbial biofilms are intimately related to antimicrobial resistance, which in turn exacerbates the challenge of eradicating bacterial infections. Consequently, there is a pressing need for novel therapies to be employed either independently or as adjuvants to diminish bacterial virulence and pathogenicity. In this context, we propose a novel approach focusing on vitamin D and vitamin K1 as potential antibiofilm agents that target Gram-negative bacteria which are hazardous to human health. RESULTS Out of 130 Gram-negative bacterial isolates, 117 were confirmed to be A. baumannii (21 isolates, 17.9%), K. pneumoniae (40 isolates, 34.2%) and P. aeruginosa (56 isolates, 47.9%). The majority of the isolates were obtained from blood and wound specimens (27.4% each). Most of the isolates exhibited high resistance rates to β-lactams (60.7-100%), ciprofloxacin (62.5-100%), amikacin (53.6-76.2%) and gentamicin (65-71.4%). Approximately 93.2% of the isolates were biofilm producers, with 6.8% categorized as weak, 42.7% as moderate, and 50.4% as strong biofilm producers. The minimum inhibitory concentrations (MICs) of vitamin D and vitamin K1 were 625-1250 µg mL-1 and 2500-5000 µg mL-1, respectively, against A. baumannii (A5, A20 and A21), K. pneumoniae (K25, K27 and K28), and P. aeruginosa (P8, P16, P24 and P27) clinical isolates and standard strains A. baumannii (ATCC 19606 and ATCC 17978), K. pneumoniae (ATCC 51503) and P. aeruginosa PAO1 and PAO14. Both vitamins significantly decreased bacterial attachment and significantly eradicated mature biofilms developed by the selected standard and clinical Gram-negative isolates. The anti-biofilm effects of both supplements were confirmed by a notable decrease in the relative expression of the biofilm-encoding genes cusD, bssS and pelA in A. baumannii A5, K. pneumoniae K28 and P. aeruginosa P16, respectively. CONCLUSION This study highlights the anti-biofilm activity of vitamins D and K1 against the tested Gram-negative strains, which emphasizes the potential of these vitamins for use as adjuvant therapies to increase the efficacy of treatment for infections caused by multidrug-resistant (MDR) strains and biofilm-forming phenotypes. However, further validation through in vivo studies is needed to confirm these promising results.
Collapse
Affiliation(s)
- Lekaa L Lutfi
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Mona I Shaaban
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Soha Lotfy Elshaer
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
11
|
Nafee N, Gaber DM, Abouelfetouh A, Alseqely M, Empting M, Schneider M. Enzyme-Linked Lipid Nanocarriers for Coping Pseudomonal Pulmonary Infection. Would Nanocarriers Complement Biofilm Disruption or Pave Its Road? Int J Nanomedicine 2024; 19:3861-3890. [PMID: 38708178 PMCID: PMC11068056 DOI: 10.2147/ijn.s445955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/28/2024] [Indexed: 05/07/2024] Open
Abstract
Introduction Cystic fibrosis (CF) is associated with pulmonary Pseudomonas aeruginosa infections persistent to antibiotics. Methods To eradicate pseudomonal biofilms, solid lipid nanoparticles (SLNs) loaded with quorum-sensing-inhibitor (QSI, disrupting bacterial crosstalk), coated with chitosan (CS, improving internalization) and immobilized with alginate lyase (AL, destroying alginate biofilms) were developed. Results SLNs (140-205 nm) showed prolonged release of QSI with no sign of acute toxicity to A549 and Calu-3 cells. The CS coating improved uptake, whereas immobilized-AL ensured >1.5-fold higher uptake and doubled SLN diffusion across the artificial biofilm sputum model. Respirable microparticles comprising SLNs in carbohydrate matrix elicited aerodynamic diameters MMAD (3.54, 2.48 µm) and fine-particle-fraction FPF (65, 48%) for anionic and cationic SLNs, respectively. The antimicrobial and/or antibiofilm activity of SLNs was explored in Pseudomonas aeruginosa reference mucoid/nonmucoid strains as well as clinical isolates. The full growth inhibition of planktonic bacteria was dependent on SLN type, concentration, growth medium, and strain. OD measurements and live/dead staining proved that anionic SLNs efficiently ceased biofilm formation and eradicated established biofilms, whereas cationic SLNs unexpectedly promoted biofilm progression. AL immobilization increased biofilm vulnerability; instead, CS coating increased biofilm formation confirmed by 3D-time lapse confocal imaging. Incubation of SLNs with mature biofilms of P. aeruginosa isolates increased biofilm density by an average of 1.5-fold. CLSM further confirmed the binding and uptake of the labeled SLNs in P. aeruginosa biofilms. Considerable uptake of CS-coated SLNs in non-mucoid strains could be observed presumably due to interaction of chitosan with LPS glycolipids in the outer cell membrane of P. aeruginosa. Conclusion The biofilm-destructive potential of QSI/SLNs/AL inhalation is promising for site-specific biofilm-targeted interventional CF therapy. Nevertheless, the intrinsic/extrinsic fundamentals of nanocarrier-biofilm interactions require further investigation.
Collapse
Affiliation(s)
- Noha Nafee
- Department of Pharmaceutics, College of Pharmacy, Kuwait University, Safat, 13110, Kuwait
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Dina M Gaber
- Department of Pharmaceutics, Division of Pharmaceutical Sciences, College of Pharmacy, Arab Academy for Science, Technology and Maritime Transport, Alexandria, 1029, Egypt
| | - Alaa Abouelfetouh
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alamein International University, Alamein, 5060335, Egypt
| | - Mustafa Alseqely
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Martin Empting
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Department of Antiviral and Antivirulence Drugs (AVID), Saarland University, Saarbrücken, 66123, Germany
| | - Marc Schneider
- Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, Saarland University, Saarbrücken, 66123, Germany
| |
Collapse
|
12
|
Pradhan L, Sah P, Nayak M, Upadhyay A, Pragya P, Tripathi S, Singh G, Mounika B, Paik P, Mukherjee S. Biosynthesized silver nanoparticles prevent bacterial infection in chicken egg model and mitigate biofilm formation on medical catheters. J Biol Inorg Chem 2024; 29:353-373. [PMID: 38744691 DOI: 10.1007/s00775-024-02050-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/20/2024] [Indexed: 05/16/2024]
Abstract
Investigating the application of innovative antimicrobial surface coatings on medical devices is an important field of research. Many of these coatings have significant drawbacks, including biocompatibility, coating stability and the inability to effectively combat multiple drug-resistant bacteria. In this research, we developed an antibiofilm surface coating for medical catheters using biosynthesized silver nanoparticles (b-Cs-AgNPs) developed using leaves extract of Calliandra surinamensis. Various characterization techniques were employed to thoroughly characterize the synthesized b-Cs-AgNPs and c-AgNPs. b-Cs-AgNPs were compatible with human normal kidney cells and chicken embryos. It did not trigger any skin inflammatory response in in vivo rat model. b-Cs-AgNPs demonstrated potent zone of inhibition of 19.09 mm when subjected to the disc diffusion method in E. coli confirming strong antibacterial property. Different anti-bacterial assays including liquid growth curve, colony counting assay, biofilm formation assay supported the potent antimicrobial efficacy of b-Cs-AgNPs alone and when coated to medical grade catheters. Mechanistic studies reveal the presence of ferulic acid, that was important for the synthesis of b-AgNPs along with enhanced antibacterial effects of b-Cs-AgNPs compared to c-AgNPs, supported by molecular docking analysis. These results together demonstrated the effective role b-Cs-AgNPs in combating infections and mitigating biofilm formations, highlighting their need for further study in the field of biomedical applications.
Collapse
Affiliation(s)
- Lipi Pradhan
- School of Biomedical Engineering, IIT (BHU), Varanasi, India
| | - Prince Sah
- School of Biomedical Engineering, IIT (BHU), Varanasi, India
| | - Malay Nayak
- School of Biomedical Engineering, IIT (BHU), Varanasi, India
| | - Anjali Upadhyay
- School of Biomedical Engineering, IIT (BHU), Varanasi, India
| | - Pragya Pragya
- School of Biomedical Engineering, IIT (BHU), Varanasi, India
| | - Shikha Tripathi
- Department of Physics, IIT (BHU), Uttar Pradesh, Varanasi, India
| | - Gurmeet Singh
- School of Biomedical Engineering, IIT (BHU), Varanasi, India
| | - B Mounika
- School of Biomedical Engineering, IIT (BHU), Varanasi, India
| | - Pradip Paik
- School of Biomedical Engineering, IIT (BHU), Varanasi, India
| | - Sudip Mukherjee
- School of Biomedical Engineering, IIT (BHU), Varanasi, India.
| |
Collapse
|
13
|
Pandey P, Pradhan S, Meher K, Lopus M, Vavilala SL. Exploring the efficacy of tryptone-stabilized silver nanoparticles against respiratory tract infection-causing bacteria: a study on planktonic and biofilm forms. Biomed Mater 2024; 19:025047. [PMID: 38364289 DOI: 10.1088/1748-605x/ad2a40] [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] [Accepted: 02/16/2024] [Indexed: 02/18/2024]
Abstract
Respiratory tract infections (RTIs) are a common cause of mortality and morbidity in the human population. The overuse of antibiotics to overcome such infections has led to antibiotic resistance. The emergence of multidrug resistant bacteria is necessitating the development of novel therapeutic techniques in order to avoid a major global clinical threat. Our study aims to investigate the potential of tryptone stabilised silver nanoparticles (Ts-AgNPs) on planktonic and biofilms produced byKlebsiella pneumoniae(K. pneumoniae)and Pseudomonas aeruginosa(P. aeruginosa). The MIC50of Ts-AgNPs was found to be as low as 1.7 μg ml-1and 2.7 μg ml-1forK. pneumoniae and P.aeruginosarespectively. Ts-AgNPs ability to alter redox environment by producing intracellular ROS, time-kill curves showing substantial decrease in the bacterial growth and significantly reduced colony forming units further validate its antimicrobial effect. The biofilm inhibition and eradication ability of Ts-AgNPs was found to be as high as 93% and 97% in both the tested organisms. A significant decrease in the eDNA and EPS quantity in Ts-AgNPs treated cells proved its ability to successfully distort the matrix and matured biofilms. Interestingly Ts-AgNPs also attenuated QS-induced virulence factors production. This study paves way to develop Ts-AgNPs as novel antibiotics against RTIs causing bacterial biofilms.
Collapse
Affiliation(s)
- Pooja Pandey
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidyanagari, Kalina Campus, Mumbai 400098, India
| | - Sristi Pradhan
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidyanagari, Kalina Campus, Mumbai 400098, India
| | - Kimaya Meher
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidyanagari, Kalina Campus, Mumbai 400098, India
| | - Manu Lopus
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidyanagari, Kalina Campus, Mumbai 400098, India
| | - Sirisha L Vavilala
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidyanagari, Kalina Campus, Mumbai 400098, India
| |
Collapse
|
14
|
Elsayed EM, Farghali AA, Zanaty MI, Abdel-Fattah M, Alkhalifah DHM, Hozzein WN, Mahmoud AM. Poly-Gamma-Glutamic Acid Nanopolymer Effect against Bacterial Biofilms: In Vitro and In Vivo Study. Biomedicines 2024; 12:251. [PMID: 38397853 PMCID: PMC10887140 DOI: 10.3390/biomedicines12020251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/01/2024] [Accepted: 01/09/2024] [Indexed: 02/25/2024] Open
Abstract
In this study, a biodegradable poly-gamma-glutamic-acid nanopolymer (Ɣ-PGA NP) was investigated for its activity against clinical strains of Gram-positive (Staphylococcus aureus and Streptococcus pyogenes) and Gram-negative (Klebsiella pneumoniae and Escherichia coli), and reference strains of S. aureus ATCC 6538, S. pyogenes ATCC 19615 (Gram-positive), and Gram-negative E. coli ATCC 25922, and K. pneumoniae ATCC 13884 bacterial biofilms. The minimum inhibitory concentration (MIC) effect of Ɣ-PGA NP showed inhibitory effects of 0.2, 0.4, 1.6, and 3.2 μg/mL for S. pyogenes, S. aureus, E. coli, and K. pneumoniae, respectively. Also, MIC values were 1.6, 0.8, 0.2, and 0.2 μg/mL for K. pneumoniae ATCC 13884, E. coli ATCC 25922, S. aureus ATCC 6538, and S. pyogenes ATCC 19615, respectively. Afterwards, MBEC (minimum biofilm eradication concentration) and MBIC (minimum biofilm inhibitory concentration) were investigated to detect Ɣ-PGA NPs efficiency against the biofilms. MBEC and MBIC increased with increasing Ɣ-PGA NPs concentration or time of exposure. Interestingly, MBIC values were at lower concentrations of Ɣ-PGA NPs than those of MBEC. Moreover, MBEC values showed that K. pneumoniae was more resistant to Ɣ-PGA NPs than E. coli, S. aureus, and S. pyogenes, and the same pattern was observed in the reference strains. The most effective results for MBEC were after 48 h, which were 1.6, 0.8, 0.4, and 0.2 µg/mL for K. pneumoniae, E. coli, S. aureus, and S. pyogenes, respectively. Moreover, MBIC results were the most impactful after 24 h but some were the same after 48 h. MBIC values after 48 h were 0.2, 0.2, 0.2, and 0.1 μg/mL for K. pneumoniae, E. coli, S. aureus, and S. pyogenes, respectively. The most effective results for MBEC were after 24 h, which were 1.6, 0.8, 0.4, and 0.4 µg/mL for K. pneumoniae ATCC 13884, E. coli ATCC 25922, S. aureus ATCC 6538, and S. pyogenes ATCC 19615, respectively. Also, MBIC results were the most impactful after an exposure time of 12 h. MBIC values after exposure time of 12 h were 0.4, 0.4, 0.2, and 0.2 μg/mL for K. pneumoniae ATCC 13884, E. coli ATCC 25922, S. aureus ATCC 6538, and S. pyogenes ATCC 19615, respectively. Besides that, results were confirmed using confocal laser scanning microscopy (CLSM), which showed a decrease in the number of living cells to 80% and 60% for MBEC and MBIC, respectively, for all the clinical bacterial strains. Moreover, living bacterial cells decreased to 70% at MBEC while decreasing up to 50% at MBIC with all bacterial refence strains. These data justify the CFU quantification. After that, ImageJ software was used to count the attached cells after incubating with the NPs, which proved the variation in live cell count between the manual counting and image analysis methods. Also, a scanning electron microscope (SEM) was used to detect the biofilm architecture after incubation with the Ɣ-PGA NP. In in vivo wound healing experiments, treated wounds of mice showed faster healing (p < 0.00001) than both the untreated mice and those that were only wounded, as the bacterial count was eradicated. Briefly, the infected mice were treated faster (p < 0.0001) when infected with S. pyogenes > S. aureus > E. coli > K. pneumoniae. The same pattern was observed for mice infected with the reference strains. Wound lengths after 2 h showed slightly healing (p < 0.001) for the clinical strains, while treatment became more obvious after 72 h > 48 h > 24 h (p < 0.0001) as wounds began to heal after 24 h up to 72 h. For reference strains, wound lengths after 2 h started to heal up to 72 h.
Collapse
Affiliation(s)
- Eman M. Elsayed
- Department of Botany and Microbiology, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt; (M.A.-F.); (W.N.H.); (A.M.M.)
| | - Ahmed A. Farghali
- Department of Materials Science and Nanotechnology, Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef 62521, Egypt;
| | - Mohamed I. Zanaty
- Department of Biotechnology and Life Sciences, Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef 62521, Egypt;
| | - Medhat Abdel-Fattah
- Department of Botany and Microbiology, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt; (M.A.-F.); (W.N.H.); (A.M.M.)
| | - Dalal Hussien M. Alkhalifah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia;
| | - Wael N. Hozzein
- Department of Botany and Microbiology, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt; (M.A.-F.); (W.N.H.); (A.M.M.)
| | - Ahmed M. Mahmoud
- Department of Botany and Microbiology, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt; (M.A.-F.); (W.N.H.); (A.M.M.)
| |
Collapse
|
15
|
Huang Y, Afolabi MA, Gan L, Liu S, Chen Y. MXene-Coated Ion-Selective Electrode Sensors for Highly Stable and Selective Lithium Dynamics Monitoring. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:1359-1368. [PMID: 38079615 PMCID: PMC10795166 DOI: 10.1021/acs.est.3c06235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 01/17/2024]
Abstract
Lithium holds immense significance in propelling sustainable energy and environmental systems forward. However, existing sensors used for lithium monitoring encounter issues concerning their selectivity and long-term durability. Addressing these challenges is crucial to ensure accurate and reliable lithium measurements during the lithium recovery processes. In response to these concerns, this study proposes a novel approach involving the use of an MXene composite membrane with incorporated poly(sodium 4-styrenesulfonate) (PSS) as an antibiofouling layer on the Li+ ion selective electrode (ISE) sensors. The resulting MXene-PSS Li+ ISE sensor demonstrates exceptional electrochemical performance, showcasing a superior slope (59.42 mV/dec), lower detection limit (10-7.2 M), quicker response time (∼10 s), higher selectivity to Na+ (-2.37) and K+ (-2.54), and reduced impedance (106.9 kΩ) when compared to conventional Li+ ISE sensors. These improvements are attributed to the unique electronic conductivity and layered structure of the MXene-PSS nanosheet coating layer. In addition, the study exhibits the long-term accuracy and durability of the MXene-PSS Li+ ISE sensor by subjecting it to real wastewater testing for 14 days, resulting in sensor reading errors of less than 10% when compared to laboratory validation results. This research highlights the great potential of MXene nanosheet coatings in advancing sensor technology, particularly in challenging applications, such as detecting emerging contaminants and developing implantable biosensors. The findings offer promising prospects for future advancements in sensor technology, particularly in the context of sustainable energy and environmental monitoring.
Collapse
Affiliation(s)
| | | | - Lan Gan
- School of Civil and Environmental
Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Su Liu
- School of Civil and Environmental
Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Yongsheng Chen
- School of Civil and Environmental
Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
16
|
Kvich L, Fritz BG, Zschach H, Terkelsen T, Raskov H, Høst-Rasmussen K, Jakobsen MR, Gheorghe AG, Gögenur I, Bjarnsholt T. Biofilms and core pathogens shape the tumor microenvironment and immune phenotype in colorectal cancer. Gut Microbes 2024; 16:2350156. [PMID: 38726597 PMCID: PMC11093030 DOI: 10.1080/19490976.2024.2350156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 04/26/2024] [Indexed: 05/16/2024] Open
Abstract
Extensive research has explored the role of gut microbiota in colorectal cancer (CRC). Nonetheless, metatranscriptomic studies investigating the in situ functional implications of host-microbe interactions in CRC are scarce. Therefore, we characterized the influence of CRC core pathogens and biofilms on the tumor microenvironment (TME) in 40 CRC, paired normal, and healthy tissue biopsies using fluorescence in situ hybridization (FISH) and dual-RNA sequencing. FISH revealed that Fusobacterium spp. was associated with increased bacterial biomass and inflammatory response in CRC samples. Dual-RNA sequencing demonstrated increased expression of pro-inflammatory cytokines, defensins, matrix-metalloproteases, and immunomodulatory factors in CRC samples with high bacterial activity. In addition, bacterial activity correlated with the infiltration of several immune cell subtypes, including M2 macrophages and regulatory T-cells in CRC samples. Specifically, Bacteroides fragilis and Fusobacterium nucleatum correlated with the infiltration of neutrophils and CD4+ T-cells, respectively. The collective bacterial activity/biomass appeared to exert a more significant influence on the TME than core pathogens, underscoring the intricate interplay between gut microbiota and CRC. These results emphasize how biofilms and core pathogens shape the immune phenotype and TME in CRC while highlighting the need to extend the bacterial scope beyond CRC pathogens to advance our understanding and identify treatment targets.
Collapse
Affiliation(s)
- Lasse Kvich
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Region Zealand, Denmark
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Blaine Gabriel Fritz
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Henrike Zschach
- Center for Health Data Science, University of Copenhagen, Copenhagen, Denmark
| | - Thilde Terkelsen
- Center for Health Data Science, University of Copenhagen, Copenhagen, Denmark
| | - Hans Raskov
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Region Zealand, Denmark
| | - Kathrine Høst-Rasmussen
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Region Zealand, Denmark
| | - Morten Ragn Jakobsen
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Region Zealand, Denmark
| | - Alexandra Gabriella Gheorghe
- Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Region Zealand, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Bjarnsholt
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
17
|
Coenye T. Biofilm antimicrobial susceptibility testing: where are we and where could we be going? Clin Microbiol Rev 2023; 36:e0002423. [PMID: 37812003 PMCID: PMC10732061 DOI: 10.1128/cmr.00024-23] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/27/2023] [Indexed: 10/10/2023] Open
Abstract
Our knowledge about the fundamental aspects of biofilm biology, including the mechanisms behind the reduced antimicrobial susceptibility of biofilms, has increased drastically over the last decades. However, this knowledge has so far not been translated into major changes in clinical practice. While the biofilm concept is increasingly on the radar of clinical microbiologists, physicians, and healthcare professionals in general, the standardized tools to study biofilms in the clinical microbiology laboratory are still lacking; one area in which this is particularly obvious is that of antimicrobial susceptibility testing (AST). It is generally accepted that the biofilm lifestyle has a tremendous impact on antibiotic susceptibility, yet AST is typically still carried out with planktonic cells. On top of that, the microenvironment at the site of infection is an important driver for microbial physiology and hence susceptibility; but this is poorly reflected in current AST methods. The goal of this review is to provide an overview of the state of the art concerning biofilm AST and highlight the knowledge gaps in this area. Subsequently, potential ways to improve biofilm-based AST will be discussed. Finally, bottlenecks currently preventing the use of biofilm AST in clinical practice, as well as the steps needed to get past these bottlenecks, will be discussed.
Collapse
Affiliation(s)
- Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
18
|
Wächter J, Vestweber PK, Planz V, Windbergs M. Unravelling host-pathogen interactions by biofilm infected human wound models. Biofilm 2023; 6:100164. [PMID: 38025836 PMCID: PMC10656240 DOI: 10.1016/j.bioflm.2023.100164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Approximately 80 % of persistent wound infections are affected by the presence of bacterial biofilms, resulting in a severe clinical challenge associated with prolonged healing periods, increased morbidity, and high healthcare costs. Unfortunately, in vitro models for wound infection research almost exclusively focus on early infection stages with planktonic bacteria. In this study, we present a new approach to emulate biofilm-infected human wounds by three-dimensional human in vitro systems. For this purpose, a matured biofilm consisting of the clinical key wound pathogen Pseudomonas aeruginosa was pre-cultivated on electrospun scaffolds allowing for non-destructive transfer of the matured biofilm to human in vitro wound models. We infected tissue-engineered human in vitro skin models as well as ex vivo human skin explants with the biofilm and analyzed structural tissue characteristics, biofilm growth behavior, and biofilm-tissue interactions. The structural development of biofilms in close proximity to the tissue, resulting in high bacterial burden and in vivo-like morphology, confirmed a manifest wound infection on all tested wound models, validating their applicability for general investigations of biofilm growth and structure. The extent of bacterial colonization of the wound bed, as well as the subsequent changes in molecular composition of skin tissue, were inherently linked to the characteristics of the underlying wound models including their viability and origin. Notably, the immune response observed in viable ex vivo and in vitro models was consistent with previous in vivo reports. While ex vivo models offered greater complexity and closer similarity to the in vivo conditions, in vitro models consistently demonstrated higher reproducibility. As a consequence, when focusing on direct biofilm-skin interactions, the viability of the wound models as well as their advantages and limitations should be aligned to the particular research question of future studies. Altogether, the novel model allows for a systematic investigation of host-pathogen interactions of bacterial biofilms and human wound tissue, also paving the way for development and predictive testing of novel therapeutics to combat biofilm-infected wounds.
Collapse
Affiliation(s)
| | | | - Viktoria Planz
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Maike Windbergs
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
19
|
Childs SK, Jones AAD. A microtiter peg lid with ziggurat geometry for medium-throughput antibiotic testing and in situ imaging of biofilms. Biofilm 2023; 6:100167. [PMID: 38078058 PMCID: PMC10700155 DOI: 10.1016/j.bioflm.2023.100167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/09/2023] [Accepted: 11/09/2023] [Indexed: 01/12/2024] Open
Abstract
Bacteria biofilm responses to disinfectants and antibiotics are quantified and observed using multiple methods, though microscopy, particularly confocal laser scanning microscopy (CLSM) is preferred due to speed, a reduction in user error, and in situ analysis. CLSM can resolve biological and spatial heterogeneity of biofilms in 3D with limited throughput. The microplate peg-lid-based assay, described in ASTM E2799-22, is a medium-throughput method for testing biofilms but does not permit in situ imaging. Breaking off the peg, as recommended by the manufacturer, risks sample damage, and is limited to easily accessible pegs. Here we report modifications to the peg optimized for in situ visualization and visualization of all pegs. We report similar antibiotic challenge recovery via colony formation following the ASTM E2799-22 protocol and in situ imaging. We report novel quantifiable effects of antibiotics on biofilm morphologies, specifically biofilm streamers. The new design bridges the MBEC® assays design that selects for biofilm phenotypes with in situ imaging needs.
Collapse
Affiliation(s)
| | - A-Andrew D. Jones
- Department of Civil & Environmental Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| |
Collapse
|
20
|
Wu M, Xu X, Hu R, Chen Q, Chen L, Yuan Y, Li J, Zhou L, Feng S, Wang L, Chen S, Gu M. A Membrane-Targeted Photosensitizer Prevents Drug Resistance and Induces Immune Response in Treating Candidiasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207736. [PMID: 37875397 PMCID: PMC10724446 DOI: 10.1002/advs.202207736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 09/20/2023] [Indexed: 10/26/2023]
Abstract
Candida albicans (C. albicans), a ubiquitous polymorphic fungus in humans, causes different types of candidiasis, including oral candidiasis (OC) and vulvovaginal candidiasis (VVC), which are physically and mentally concerning and financially costly. Thus, developing alternative antifungals that prevent drug resistance and induce immunity to eliminate Candida biofilms is crucial. Herein, a novel membrane-targeted aggregation-induced emission (AIE) photosensitizer (PS), TBTCP-QY, is developed for highly efficient photodynamic therapy (PDT) of candidiasis. TBTCP-QY has a high molar absorption coefficient and an excellent ability to generate 1 O2 and •OH, entering the interior of biofilms due to its high permeability. Furthermore, TBTCP-QY can efficiently inhibit biofilm formation by suppressing the expression of genes related to the adhesion (ALS3, EAP1, and HWP1), invasion (SAP1 and SAP2), and drug resistance (MDR1) of C. albicans, which is also advantageous for eliminating potential fungal resistance to treat clinical infectious diseases. TBTCP-QY-mediated PDT efficiently targets OC and VVC in vivo in a mouse model, induces immune response, relieves inflammation, and accelerates the healing of mucosal defects to combat infections caused by clinically isolated fluconazole-resistant strains. Moreover, TBTCP-QY demonstrates excellent biocompatibility, suggesting its potential applications in the clinical treatment of OC and VVC.
Collapse
Affiliation(s)
- Ming‐Yu Wu
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural DrugsSchool of Life Science and EngineeringSouthwest Jiaotong UniversityChengduSichuan610031China
| | - Xiaoyu Xu
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Rui Hu
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Department of Respiratory DiseasesThe Research and Application Center of Precision MedicineThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhou450014China
| | - Qingrong Chen
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Luojia Chen
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Yuncong Yuan
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Jie Li
- Department of Medical Intensive Care UnitMaternal and Child Health Hospital of Hubei ProvinceTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430070China
| | - Li Zhou
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Shun Feng
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural DrugsSchool of Life Science and EngineeringSouthwest Jiaotong UniversityChengduSichuan610031China
| | - Lianrong Wang
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Department of Respiratory DiseasesThe Research and Application Center of Precision MedicineThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhou450014China
| | - Shi Chen
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
| | - Meijia Gu
- Department of GastroenterologyMinistry of Education Key Laboratory of Combinatorial Biosynthesis and Drug DiscoveryTaiKang Center for Life and Medical SciencesZhongnan Hospital of Wuhan UniversitySchool of Pharmaceutical SciencesWuhan UniversityWuhan430071China
- Department of Respiratory DiseasesThe Research and Application Center of Precision MedicineThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhou450014China
| |
Collapse
|
21
|
Artini M, Papa R, Vrenna G, Trecca M, Paris I, D’Angelo C, Tutino ML, Parrilli E, Selan L. Antarctic Marine Bacteria as a Source of Anti-Biofilm Molecules to Combat ESKAPE Pathogens. Antibiotics (Basel) 2023; 12:1556. [PMID: 37887257 PMCID: PMC10604463 DOI: 10.3390/antibiotics12101556] [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: 09/21/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023] Open
Abstract
The ESKAPE pathogens, including bacteria such as Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species, pose a global health threat due to their ability to resist antimicrobial drugs and evade the immune system. These pathogens are responsible for hospital-acquired infections, especially in intensive care units, and contribute to the growing problem of multi-drug resistance. In this study, researchers focused on exploring the potential of Antarctic marine bacteria as a source of anti-biofilm molecules to combat ESKAPE pathogens. Four Antarctic bacterial strains were selected, and their cell-free supernatants were tested against 60 clinical ESKAPE isolates. The results showed that the supernatants did not exhibit antimicrobial activity but effectively prevented biofilm formation and dispersed mature biofilms. This research highlights the promising potential of Antarctic bacteria in producing compounds that can counteract biofilms formed by clinically significant bacterial species. These findings contribute to the development of new strategies for preventing and controlling infections caused by ESKAPE pathogens.
Collapse
Affiliation(s)
- Marco Artini
- Department of Public Health and Infectious Diseases, Sapienza University, p.le Aldo Moro 5, 00185 Rome, Italy; (M.A.); (G.V.); (M.T.); (I.P.); (L.S.)
| | - Rosanna Papa
- Department of Public Health and Infectious Diseases, Sapienza University, p.le Aldo Moro 5, 00185 Rome, Italy; (M.A.); (G.V.); (M.T.); (I.P.); (L.S.)
| | - Gianluca Vrenna
- Department of Public Health and Infectious Diseases, Sapienza University, p.le Aldo Moro 5, 00185 Rome, Italy; (M.A.); (G.V.); (M.T.); (I.P.); (L.S.)
- Research Unit of Diagnostical and Management Innovations, Children’s Hospital and Institute Research Bambino Gesù, 00165 Rome, Italy
| | - Marika Trecca
- Department of Public Health and Infectious Diseases, Sapienza University, p.le Aldo Moro 5, 00185 Rome, Italy; (M.A.); (G.V.); (M.T.); (I.P.); (L.S.)
| | - Irene Paris
- Department of Public Health and Infectious Diseases, Sapienza University, p.le Aldo Moro 5, 00185 Rome, Italy; (M.A.); (G.V.); (M.T.); (I.P.); (L.S.)
| | - Caterina D’Angelo
- Department of Chemical Sciences, Federico II University, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (M.L.T.); (E.P.)
| | - Maria Luisa Tutino
- Department of Chemical Sciences, Federico II University, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (M.L.T.); (E.P.)
| | - Ermenegilda Parrilli
- Department of Chemical Sciences, Federico II University, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (M.L.T.); (E.P.)
| | - Laura Selan
- Department of Public Health and Infectious Diseases, Sapienza University, p.le Aldo Moro 5, 00185 Rome, Italy; (M.A.); (G.V.); (M.T.); (I.P.); (L.S.)
| |
Collapse
|
22
|
Shehabeldine AM, Al-Askar AA, AbdElgawad H, Hagras FA, Ramadan AA, Kamel MR, Ahmed MA, Atia KH, Hashem AH. Wound Dressing Scaffold with High Anti-biofilm Performance Based on Ciprofloxacin-Loaded Chitosan-Hydrolyzed Starch Nanocomposite: In Vitro and In Vivo Study. Appl Biochem Biotechnol 2023; 195:6421-6439. [PMID: 37450215 DOI: 10.1007/s12010-023-04665-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] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
Today, the search for solutions to reduce wound infection and restore wound receptivity also reduces its side effects which are a difficult problem in medical science research. The greatest options for this purpose are hydrogel dressings since they are compatible with tissue and have an antibacterial effect on wound healing. Chronic wounds represent a significant burden on people and healthcare systems worldwide. Bacteria often enter such skin wounds, causing irritation and complicating the healing process. In addition, bacteria cause infection, which inhibits rejuvenation and the production of collagen. This study is aimed at developing novel chitosan (CS)-hydrolyzed starch nanocomposite (HS/Ch-NC) loaded with ciprofloxacin to enhance its skin retention and wound healing efficacy and anti-biofilm efficacy. Drug-loading on the (HS/Ch-NC) and encapsulation efficiency was 55.2% and 97.2%, respectively. The activity of HS-NC loaded with ciprofloxacin as anti-biofilm activity by 72% and 63% against Enterobacter aerogenes and Pseudomonas aeruginosa, respectively. The obtained (HS/Ch-NC) loaded with ciprofloxacin is a promising candidate for the development of improved bandage materials, as cell viability and proliferation was assessed using an SRB assay with half-maximal inhibitory concentrations (IC50) at 119.1 µg/ml. In vitro scratch wound healing assay revealed significant (p ≤ 0.05) acceleration in wound closure at 24 h enhanced by 56.04% 24-h and 100% 72-h post-exposure to (HS/Ch-NC) loaded ciprofloxacin, compared to the negative control. In vivo skin retention study revealed that (HS/Ch-NC)-loaded ciprofloxacin showed 3.65-fold higher retention, respectively, than ciprofloxacin. Thus, our study assumes that ciprofloxacin-loaded HS-NC is a potential delivery system for enhancing ciprofloxacin skin retention and wound healing activity.
Collapse
Affiliation(s)
- Amr M Shehabeldine
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, 11884, Egypt.
| | - Abdulaziz A Al-Askar
- Department of Botany and Microbiology, Faculty of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Hamada AbdElgawad
- Integrated Molecular Plant Physiology Research (IMPRES), Department of Biology, University of Antwerp, Antwerp, Belgium
| | - Fatouh A Hagras
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, 11884, Egypt
| | - Amr A Ramadan
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, 11884, Egypt
| | - Mohamed R Kamel
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, 11884, Egypt
| | - Mohamed A Ahmed
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, 11884, Egypt
| | - Kareem H Atia
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, 11884, Egypt
| | - Amr H Hashem
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, 11884, Egypt.
| |
Collapse
|
23
|
Rana S, Upadhyay LSB. Methylene Blue Assisted Electrochemical Detection of Bacterial Biofilm. Indian J Microbiol 2023; 63:299-306. [PMID: 37781013 PMCID: PMC10533774 DOI: 10.1007/s12088-023-01084-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 06/27/2023] [Indexed: 10/03/2023] Open
Abstract
This paper presents a novel electrochemical transduction method for the rapid and straightforward detection of bacterial biofilm. Briefly, fifteen isolates from various sources were collected and evaluated for their ability to generate biofilm. The Congo red-based agar method and the tube test were used for preliminary screening. A microtiter experiment was also performed to quantitatively examine the screening results and validate the outcomes of the proposed methylene blue-based electrochemical detection method. Electrochemical sensing was performed on the two selected isolates using methylene blue as a redox indicator. For optimization goals, several methylene blue concentrations were studied. Methylene blue at a concentration of 0.4 mM was used for the analysis conclusion. The developed electrochemical method displayed a linear R2 value of 0.9747. The new electrochemical approach demonstrated great sensitivity and rapid response compared to conventional microtiter test methods.
Collapse
Affiliation(s)
- Sonali Rana
- Department of Biotechnology, National Institute of Technology Raipur, Raipur, Chhattisgarh 492010 India
| | - Lata Sheo Bachan Upadhyay
- Department of Biotechnology, National Institute of Technology Raipur, Raipur, Chhattisgarh 492010 India
| |
Collapse
|
24
|
Kragh KN, Tolker-Nielsen T, Lichtenberg M. The non-attached biofilm aggregate. Commun Biol 2023; 6:898. [PMID: 37658117 PMCID: PMC10474055 DOI: 10.1038/s42003-023-05281-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 08/24/2023] [Indexed: 09/03/2023] Open
Abstract
Biofilms have conventionally been perceived as dense bacterial masses on surfaces, following the five-step model of development. Initial biofilm research focused on surface-attached formations, but detached aggregates have received increasing attention in the past decade due to their pivotal role in chronic infections. Understanding their nature sparked fervent discussions in biofilm conferences and scientific literature. This review consolidates current insights on non-attached aggregates, offering examples of their occurrence in nature and diseases. We discuss their formation and dispersion mechanisms, resilience to antibiotics and immune-responses, drawing parallels to surface-attached biofilms. Moreover, we outline available in vitro models for studying non-attached aggregates.
Collapse
Affiliation(s)
- Kasper N Kragh
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Tim Tolker-Nielsen
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Mads Lichtenberg
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
25
|
Ladewig L, Gloy L, Langfeldt D, Pinnow N, Weiland-Bräuer N, Schmitz RA. Antimicrobial Peptides Originating from Expression Libraries of Aurelia aurita and Mnemiopsis leidyi Prevent Biofilm Formation of Opportunistic Pathogens. Microorganisms 2023; 11:2184. [PMID: 37764028 PMCID: PMC10537229 DOI: 10.3390/microorganisms11092184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/17/2023] [Accepted: 08/26/2023] [Indexed: 09/29/2023] Open
Abstract
The demand for novel antimicrobial compounds is rapidly growing due to the rising appearance of antibiotic resistance in bacteria; accordingly, alternative approaches are urgently needed. Antimicrobial peptides (AMPs) are promising, since they are a naturally occurring part of the innate immune system and display remarkable broad-spectrum activity and high selectivity against various microbes. Marine invertebrates are a primary resource of natural AMPs. Consequently, cDNA expression (EST) libraries from the Cnidarian moon jellyfish Aurelia aurita and the Ctenophore comb jelly Mnemiopsis leidyi were constructed in Escherichia coli. Cell-free size-fractionated cell extracts (<3 kDa) of the two libraries (each with 29,952 clones) were consecutively screened for peptides preventing the biofilm formation of opportunistic pathogens using the crystal violet assay. The 3 kDa fraction of ten individual clones demonstrated promising biofilm-preventing activities against Klebsiella oxytoca and Staphylococcus epidermidis. Sequencing the respective activity-conferring inserts allowed for the identification of small ORFs encoding peptides (10-22 aa), which were subsequently chemically synthesized to validate their inhibitory potential. Although the peptides are likely artificial products from a random translation of EST inserts, the biofilm-preventing effects against K. oxytoca, Pseudomonas aeruginosa, S. epidermidis, and S. aureus were verified for five synthetic peptides in a concentration-dependent manner, with peptide BiP_Aa_5 showing the strongest effects. The impact of BiP_Aa_2, BiP_Aa_5, and BiP_Aa_6 on the dynamic biofilm formation of K. oxytoca was further validated in microfluidic flow cells, demonstrating a significant reduction in biofilm thickness and volume by BiP_Aa_2 and BiP_Aa_5. Overall, the structural characteristics of the marine invertebrate-derived AMPs, their physicochemical properties, and their promising antibiofilm effects highlight them as attractive candidates for discovering new antimicrobials.
Collapse
Affiliation(s)
- Lisa Ladewig
- General Microbiology, Kiel University, Am Botanischen Garten 1-9, 24118 Kiel, Germany
| | - Leon Gloy
- General Microbiology, Kiel University, Am Botanischen Garten 1-9, 24118 Kiel, Germany
| | - Daniela Langfeldt
- General Microbiology, Kiel University, Am Botanischen Garten 1-9, 24118 Kiel, Germany
- Institute of Clinical Molecular Biology (IKMB), Kiel University, Am Botanischen Garten 11, 24118 Kiel, Germany
| | - Nicole Pinnow
- General Microbiology, Kiel University, Am Botanischen Garten 1-9, 24118 Kiel, Germany
| | - Nancy Weiland-Bräuer
- General Microbiology, Kiel University, Am Botanischen Garten 1-9, 24118 Kiel, Germany
| | - Ruth A. Schmitz
- General Microbiology, Kiel University, Am Botanischen Garten 1-9, 24118 Kiel, Germany
| |
Collapse
|
26
|
Kurbatfinski N, Kramer CN, Goodman SD, Bakaletz LO. ESKAPEE pathogens newly released from biofilm residence by a targeted monoclonal are sensitized to killing by traditional antibiotics. Front Microbiol 2023; 14:1202215. [PMID: 37564292 PMCID: PMC10410267 DOI: 10.3389/fmicb.2023.1202215] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/30/2023] [Indexed: 08/12/2023] Open
Abstract
Introduction The "silent" antimicrobial resistance (AMR) pandemic is responsible for nearly five million deaths annually, with a group of seven biofilm-forming pathogens, known as the ESKAPEE pathogens, responsible for 70% of these fatalities. Biofilm-resident bacteria, as they exist within the disease site, are canonically highly resistant to antibiotics. One strategy to counter AMR and improve disease resolution involves developing methods to disrupt biofilms. These methods aim to release bacteria from the protective biofilm matrix to facilitate their killing by antibiotics or immune effectors. Several laboratories working on such strategies have demonstrated that bacteria newly released from a biofilm display a transient phenotype of significantly increased susceptibility to antibiotics. Similarly, we developed an antibody-based approach for biofilm disruption directed against the two-membered DNABII family of bacterial DNA-binding proteins, which serve as linchpins to stabilize the biofilm matrix. The incubation of biofilms with α-DNABII antibodies rapidly collapses them to induce a population of newly released bacteria (NRel). Methods In this study, we used a humanized monoclonal antibody (HuTipMab) directed against protective epitopes of a DNABII protein to determine if we could disrupt biofilms formed by the high-priority ESKAPEE pathogens as visualized by confocal laser scanning microscopy (CLSM) and COMSTAT2 analysis. Then, we demonstrated the potentiated killing of the induced NRel by seven diverse classes of traditional antibiotics by comparative plate count. Results To this end, ESKAPEE biofilms were disrupted by 50%-79% using a single tested dose and treatment period with HuTipMab. The NRel of each biofilm were significantly more sensitive to killing than their planktonically grown counterparts (heretofore, considered to be the most sensitive to antibiotic-mediated killing), even when tested at a fraction of the MIC (1/250-1/2 MIC). Moreover, the bacteria that remained within the biofilms of two representative ESKAPEE pathogens after HuTipMab disruption were also significantly more susceptible to killing by antibiotics. Discussion New data presented in this study support our continued development of a combinatorial therapy wherein HuTipMab is delivered to a patient with recalcitrant disease due to an ESKAPEE pathogen to disrupt a pathogenic biofilm, along with a co-delivered dose of an antibiotic whose ability to rapidly kill the induced NRel has been demonstrated. This novel regimen could provide a more successful clinical outcome to those with chronic, recurrent, or recalcitrant diseases, while limiting further contribution to AMR.
Collapse
Affiliation(s)
- Nikola Kurbatfinski
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Cameron N. Kramer
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Steven D. Goodman
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Lauren O. Bakaletz
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, United States
| |
Collapse
|
27
|
Chen T, Zhao MX, Tang XY, Wei WX, Wen X, Zhou SZ, Ma BH, Zou YD, Zhang N, Mi JD, Wang Y, Liao XD, Wu YB. The tigecycline resistance gene tetX has an expensive fitness cost based on increased outer membrane permeability and metabolic burden in Escherichia coli. JOURNAL OF HAZARDOUS MATERIALS 2023; 458:131889. [PMID: 37348375 DOI: 10.1016/j.jhazmat.2023.131889] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 05/23/2023] [Accepted: 06/16/2023] [Indexed: 06/24/2023]
Abstract
Livestock-derived tetX-positive Escherichia coli with tigecycline resistance poses a serious risk to public health. Fitness costs, antibiotic residues, and other tetracycline resistance genes (TRGs) are fundamental in determining the spread of tetX in the environment, but there is a lack of relevant studies. The results of this study showed that both tetO and tetX resulted in reduction in growth and an increased in the metabolic burden of E. coli, but the presence of doxycycline reversed this phenomenon. Moreover, the protection of E. coli growth and metabolism by tetO was superior to that of tetX in the presence of doxycycline, resulting in a much lower competitiveness of tetX-carrying E. coli than tetO-carrying E. coli. The results of RNA-seq showed that the increase in outer membrane proteins (ompC, ompF and ompT) of tetX-carrying E. coli resulted in increased membrane permeability and biofilm formation, which is an important reason for fitness costs. Overall, the increased membrane permeability and metabolic burden of E. coli is the mechanistic basis for the high fitness cost of tetX, and the spread of tetO may limit the spread of tetX. This study provides new insights into the rational use of tetracycline antibiotics to control the spread of tetX.
Collapse
Affiliation(s)
- Tao Chen
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Min-Xing Zhao
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Xiao-Yue Tang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Wen-Xiao Wei
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Xin Wen
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Shi-Zheng Zhou
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Bao-Hua Ma
- Foshan Customs Comprehensive Technology Center, Foshan 528200, China
| | - Yong-De Zou
- Foshan Customs Comprehensive Technology Center, Foshan 528200, China
| | - Na Zhang
- Foshan Customs Comprehensive Technology Center, Foshan 528200, China
| | - Jian-Dui Mi
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou 730000, China
| | - Yan Wang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Xin-Di Liao
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yin-Bao Wu
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
28
|
Mhade S, Kaushik KS. Tools of the Trade: Image Analysis Programs for Confocal Laser-Scanning Microscopy Studies of Biofilms and Considerations for Their Use by Experimental Researchers. ACS OMEGA 2023; 8:20163-20177. [PMID: 37332792 PMCID: PMC10268615 DOI: 10.1021/acsomega.2c07255] [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: 11/11/2022] [Accepted: 05/11/2023] [Indexed: 06/20/2023]
Abstract
Confocal laser-scanning microscopy (CLSM) is the bedrock of the microscopic visualization of biofilms. Previous applications of CLSM in biofilm studies have largely focused on observations of bacterial or fungal elements of biofilms, often seen as aggregates or mats of cells. However, the field of biofilm research is moving beyond qualitative observations alone, toward the quantitative analysis of the structural and functional features of biofilms, across clinical, environmental, and laboratory conditions. In recent times, several image analysis programs have been developed to extract and quantify biofilm properties from confocal micrographs. These tools not only vary in their scope and relevance to the specific biofilm features under study but also with respect to the user interface, compatibility with operating systems, and raw image requirements. Understanding these considerations is important when selecting tools for quantitative biofilm analysis, including at the initial experimental stages of image acquisition. In this review, we provide an overview of image analysis programs for confocal micrographs of biofilms, with a focus on tool selection and image acquisition parameters that are relevant for experimental researchers to ensure reliability and compatibility with downstream image processing.
Collapse
Affiliation(s)
- Shreeya Mhade
- Department
of Biotechnology, Savitribai Phule Pune
University, Pune 411007, India
| | - Karishma S Kaushik
- Department
of Biotechnology, Savitribai Phule Pune
University, Pune 411007, India
| |
Collapse
|
29
|
Thames HT, Pokhrel D, Willis E, Rivers O, Dinh TTN, Zhang L, Schilling MW, Ramachandran R, White S, Sukumaran AT. Salmonella Biofilm Formation under Fluidic Shear Stress on Different Surface Materials. Foods 2023; 12:foods12091918. [PMID: 37174455 PMCID: PMC10178852 DOI: 10.3390/foods12091918] [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: 03/31/2023] [Revised: 04/23/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
This study characterized biofilm formation of various Salmonella strains on common processing plant surface materials (stainless steel, concrete, rubber, polyethylene) under static and fluidic shear stress conditions. Surface-coupons were immersed in well-plates containing 1 mL of Salmonella (6 log CFU/mL) and incubated aerobically for 48 h at 37 °C in static or shear stress conditions. Biofilm density was determined using crystal violet assay, and biofilm cells were enumerated by plating on tryptic soy agar plates. Biofilms were visualized using scanning electron microscopy. Data were analyzed by SAS 9.4 at a significance level of 0.05. A surface-incubation condition interaction was observed for biofilm density (p < 0.001). On stainless steel, the OD600 was higher under shear stress than static incubation; whereas, on polyethylene, the OD600 was higher under static condition. Enumeration revealed surface-incubation condition (p = 0.024) and surface-strain (p < 0.001) interactions. Among all surface-incubation condition combinations, the biofilm cells were highest on polyethylene under fluidic shear stress (6.4 log/coupon; p < 0.001). Biofilms of S. Kentucky on polyethylene had the highest number of cells (7.80 log/coupon) compared to all other strain-surface combinations (p < 0.001). Electron microscopy revealed morphological and extracellular matrix differences between surfaces. Results indicate that Salmonella biofilm formation is influenced by serotype, surface, and fluidic shear stress.
Collapse
Affiliation(s)
- Hudson T Thames
- Department of Poultry Science, Mississippi State University, Starkville, MS 39762, USA
| | - Diksha Pokhrel
- Department of Poultry Science, Mississippi State University, Starkville, MS 39762, USA
| | - Emma Willis
- Department of Poultry Science, Mississippi State University, Starkville, MS 39762, USA
| | - Orion Rivers
- Institute for Imaging & Analytical Technologies, Mississippi State University, Starkville, MS 39762, USA
| | - Thu T N Dinh
- Tyson Foods, 2200 W. Don Tyson Parkway, Springdale, AR 72762, USA
| | - Li Zhang
- Department of Poultry Science, Mississippi State University, Starkville, MS 39762, USA
| | - Mark W Schilling
- Department of Food Science, Nutrition, and Health Promotion, Mississippi State University, Starkville, MS 39762, USA
| | - Reshma Ramachandran
- Department of Poultry Science, Mississippi State University, Starkville, MS 39762, USA
| | - Shecoya White
- Department of Food Science, Nutrition, and Health Promotion, Mississippi State University, Starkville, MS 39762, USA
| | - Anuraj T Sukumaran
- Department of Poultry Science, Mississippi State University, Starkville, MS 39762, USA
| |
Collapse
|
30
|
Paleczny J, Junka AF, Krzyżek P, Czajkowska J, Kramer A, Benkhai H, Żyfka-Zagrodzińska E, Bartoszewicz M. Comparison of antibiofilm activity of low-concentrated hypochlorites vs polyhexanide-containing antiseptic. Front Cell Infect Microbiol 2023; 13:1119188. [PMID: 37009512 PMCID: PMC10050698 DOI: 10.3389/fcimb.2023.1119188] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/27/2023] [Indexed: 03/17/2023] Open
Abstract
Chronic wound infection is highly associated with morbidity and endangers the patient's life. Therefore, wound care products must have a potent antimicrobial and biofilm-eradicating effect. In this work, the antimicrobial/antibiofilm activity of two low-concentrated chlorine-based and releasing solutions was investigated on a total of 78 strains of methicillin-resistant Staphylococcus aureus, Pseudomonas aeruginosa, and Candida albicans, using the cohesive spectrum of in vitro settings, including microtiter plate models, biofilm-oriented antiseptic test, cellulose-based biofilm model, biofilm bioreactors and Bioflux model. The antiseptic containing polyhexamethylene biguanide was used in the character of usability control of performed tests. The results obtained by static biofilm models indicate that low-concentrated chlorine-based and releasing solutions display none to moderate antibiofilm activity, while data obtained by means of the Bioflux model, providing flow conditions, indicate the moderate antibiofilm activity of substances compared with the polyhexanide antiseptic. Considering in vitro data presented in this manuscript, the earlier reported favorable clinical results of low-concentrated hypochlorites should be considered rather an effect of their rinsing activity combined with low cytotoxicity but not the antimicrobial effect per se. For the treatment of heavily biofilm-infected wounds, polyhexanide should be considered the agent of choice because of its higher efficacy against pathogenic biofilms.
Collapse
Affiliation(s)
- Justyna Paleczny
- Department of Pharmaceutical Microbiology and Parasitology, Unique Application Models Laboratory, Wroclaw Medical University, Wroclaw, Poland
- *Correspondence: Adam Junka, ; Justyna Paleczny,
| | - Adam Felix Junka
- Department of Pharmaceutical Microbiology and Parasitology, Unique Application Models Laboratory, Wroclaw Medical University, Wroclaw, Poland
- *Correspondence: Adam Junka, ; Justyna Paleczny,
| | - Paweł Krzyżek
- Department of Microbiology, Wroclaw Medical University, Wroclaw, Poland
| | - Joanna Czajkowska
- Department of Biochemistry and Molecular Biology, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| | - Axel Kramer
- Institute of Hygiene and Environmental Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Hicham Benkhai
- Institute of Hygiene and Environmental Medicine, University Medicine Greifswald, Greifswald, Germany
| | | | - Marzenna Bartoszewicz
- Department of Pharmaceutical Microbiology and Parasitology, Unique Application Models Laboratory, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
31
|
Ovcharova MA, Schelkunov MI, Geras’kina OV, Makarova NE, Sukhacheva MV, Martyanov SV, Nevolina ED, Zhurina MV, Feofanov AV, Botchkova EA, Plakunov VK, Gannesen AV. C-Type Natriuretic Peptide Acts as a Microorganism-Activated Regulator of the Skin Commensals Staphylococcus epidermidis and Cutibacterium acnes in Dual-Species Biofilms. BIOLOGY 2023; 12:436. [PMID: 36979128 PMCID: PMC10045295 DOI: 10.3390/biology12030436] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/06/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023]
Abstract
The effect of C-type natriuretic peptide in a concentration closer to the normal level in human blood plasma was studied on the mono-species and dual-species biofilms of the skin commensal bacteria Cutibacterium acnes HL043PA2 and Staphylococcus epidermidis ATCC14990. Despite the marginal effect of the hormone on cutibacteria in mono-species biofilms, the presence of staphylococci in the community resulted in a global shift of the CNP effect, which appeared to increase the competitive properties of C. acnes, its proliferation and the metabolic activity of the community. S. epidermidis was mostly inhibited in the presence of CNP. Both bacteria had a significant impact on the gene expression levels revealed by RNA-seq. CNP did not affect the gene expression levels in mono-species cutibacterial biofilms; however, in the presence of staphylococci, five genes were differentially expressed in the presence of the hormone, including two ribosomal proteins and metal ABC transporter permease. In staphylococci, the Na-translocating system protein MpsB NADH-quinone oxidoreductase subunit L was downregulated in the dual-species biofilms in the presence of CNP, while in mono-species biofilms, two proteins of unknown function were downregulated. Hypothetically, at least one of the CNP mechanisms of action is via the competition for zinc, at least on cutibacteria.
Collapse
Affiliation(s)
- Maria A. Ovcharova
- Federal Research Centre “Fundamentals of Biotechnology” of Russian Academy of Sciences, Moscow 119071, Russia
| | - Mikhail I. Schelkunov
- Skolkovo Institute of Science and Technology, Moscow 121205, Russia
- Institute for Information Transmission Problems of Russian Academy of Sciences, Moscow 127051, Russia
| | - Olga V. Geras’kina
- Biological Faculty, Lomonosov Moscow State University, Moscow 119192, Russia
| | | | - Marina V. Sukhacheva
- Federal Research Centre “Fundamentals of Biotechnology” of Russian Academy of Sciences, Moscow 119071, Russia
| | - Sergey V. Martyanov
- Federal Research Centre “Fundamentals of Biotechnology” of Russian Academy of Sciences, Moscow 119071, Russia
| | - Ekaterina D. Nevolina
- Federal Research Centre “Fundamentals of Biotechnology” of Russian Academy of Sciences, Moscow 119071, Russia
| | - Marina V. Zhurina
- Federal Research Centre “Fundamentals of Biotechnology” of Russian Academy of Sciences, Moscow 119071, Russia
| | - Alexey V. Feofanov
- Biological Faculty, Lomonosov Moscow State University, Moscow 119192, Russia
| | - Ekaterina A. Botchkova
- Federal Research Centre “Fundamentals of Biotechnology” of Russian Academy of Sciences, Moscow 119071, Russia
| | - Vladimir K. Plakunov
- Federal Research Centre “Fundamentals of Biotechnology” of Russian Academy of Sciences, Moscow 119071, Russia
| | - Andrei V. Gannesen
- Federal Research Centre “Fundamentals of Biotechnology” of Russian Academy of Sciences, Moscow 119071, Russia
| |
Collapse
|
32
|
Lichtenberg M, Kvich L, Larsen SLB, Jakobsen TH, Bjarnsholt T. Inoculum Concentration Influences Pseudomonas aeruginosa Phenotype and Biofilm Architecture. Microbiol Spectr 2022; 10:e0313122. [PMID: 36354337 PMCID: PMC9769529 DOI: 10.1128/spectrum.03131-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 10/21/2022] [Indexed: 11/12/2022] Open
Abstract
In infections, bacterial cells are often found as relatively small multicellular aggregates characterized by a heterogeneous distribution of phenotype, genotype, and growth rates depending on their surrounding microenvironment. Many laboratory models fail to mimic these characteristics, and experiments are often initiated from planktonic bacteria given optimal conditions for rapid growth without concerns about the microenvironmental characteristics during biofilm maturation. Therefore, we investigated how the initial bacterial concentration (henceforth termed the inoculum) influences the microenvironment during initial growth and how this affects the sizes and distribution of developed aggregates in an embedded biofilm model-the alginate bead biofilm model. Following 24 h of incubation, the viable biomass was independent of starting inoculum but with a radically different microenvironment which led to differences in metabolic activity depending on the inoculum. The inoculum also affected the number of cells surviving treatment with the antibiotic tobramycin, where the highest inoculum showed higher survival rates than the lowest inoculum. The change in antibiotic tolerance was correlated with cell-specific RNA content and O2 consumption rates, suggesting a direct role of metabolic activity. Thus, the starting number of bacteria results in different phenotypic trajectories governed by different microenvironmental characteristics, and we demonstrate some of the possible implications of such physiological gradients on the outcome of in vitro experiments. IMPORTANCE Biofilm aggregates grown in the alginate bead biofilm model bear resemblance to features of in vivo biofilms. Here, we show that changing the initial concentration of bacteria in the biofilm model leads to widely different behavior of the bacteria following an incubation period. This difference is influenced by the local conditions experienced by the bacteria during growth, which impact their response to antibiotic treatment. Our study provides a framework for manipulating aggregate sizes in in vitro biofilm models. It underlines the importance of how experiments are initiated, which can profoundly impact the outcomes and interpretation of microbiological experiments.
Collapse
Affiliation(s)
- Mads Lichtenberg
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Lasse Kvich
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Sara Louise Borregaard Larsen
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Tim Holm Jakobsen
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Thomas Bjarnsholt
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- Department of Clinical Microbiology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
33
|
Chen T, Zhao M, Tang X, Wang W, Zhang M, Tang J, Wang W, Wei W, Ma B, Zou Y, Zhang N, Mi J, Wang Y, Liao X, Wu Y. Serious Risk of Tigecycline Resistance in Escherichia coli Isolated from Swine Manure. MICROBIAL ECOLOGY 2022:10.1007/s00248-022-02133-2. [PMID: 36326874 DOI: 10.1007/s00248-022-02133-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/24/2022] [Indexed: 06/16/2023]
Abstract
The emergence of the plasmid-mediated tigecycline resistance gene tetX family in pig farms has attracted worldwide attention. The use of tetracycline antibiotics in pig farms has a facilitating effect on the prevalence of the tetX family, but the relationship among its presence, expression, and resistance phenotype in resistant bacteria is unknown. In this study, the presence and expression characteristics of tetracycline resistance genes (TRGs) in 89 strains of doxycycline-resistant E. coli (DRE) isolated from pig manure samples from 20 pig farms under low concentrations of doxycycline stress (2 μg/mL) were analyzed. The detection rate of tetO was 96.63%, which is higher than those of other TRGs, such as tetA (94.38%), tetX (76.40%), tetB (73.03%), and tet(X4) (69.66%). At least three TRG types were present in DRE strains, which thus showed extensive resistance to tetracycline antibiotics, and 37% of these strains were resistant to tigecycline. In the presence of a low concentration of doxycycline, tetA played an important role, and the expression and existence ratio of TRGs indicated low expression of TRGs. Furthermore, the doxycycline resistance of DRE was jointly determined by the total absolute abundance of TRGs, and the absolute abundance of tetX and tet(X4) was significantly positively associated with tigecycline resistance in DRE (P < 0.05). Overall, DRE isolated from swine manure is an important reservoir of the tetX family, which suggests that DRE in swine manure has a high risk of tigecycline resistance, poses a potential threat to human health, and should be of public concern.
Collapse
Affiliation(s)
- Tao Chen
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Minxing Zhao
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xiaoyue Tang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Wenqiang Wang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Miao Zhang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jing Tang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Wei Wang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Wenxiao Wei
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Baohua Ma
- Foshan Customs Comprehensive Technology Center, 528200, Foshan, China
| | - Yongde Zou
- Foshan Customs Comprehensive Technology Center, 528200, Foshan, China
| | - Na Zhang
- Foshan Customs Comprehensive Technology Center, 528200, Foshan, China
| | - Jiandui Mi
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, 525000, China
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Engineering Technology Research Center of Harmless Treatment and Resource Utilization of Livestock Waste, Yunfu, Xinxing, China
- Ministry of Agriculture Key Laboratory of Tropical Agricultural Environment, South China Agricultural University, Guangzhou, 510642, China
| | - Yan Wang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, 525000, China
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Engineering Technology Research Center of Harmless Treatment and Resource Utilization of Livestock Waste, Yunfu, Xinxing, China
- Ministry of Agriculture Key Laboratory of Tropical Agricultural Environment, South China Agricultural University, Guangzhou, 510642, China
| | - Xindi Liao
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, 525000, China
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Engineering Technology Research Center of Harmless Treatment and Resource Utilization of Livestock Waste, Yunfu, Xinxing, China
- Ministry of Agriculture Key Laboratory of Tropical Agricultural Environment, South China Agricultural University, Guangzhou, 510642, China
| | - Yinbao Wu
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, 525000, China.
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Engineering Technology Research Center of Harmless Treatment and Resource Utilization of Livestock Waste, Yunfu, Xinxing, China.
- Ministry of Agriculture Key Laboratory of Tropical Agricultural Environment, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
34
|
Grace A, Sahu R, Owen DR, Dennis VA. Pseudomonas aeruginosa reference strains PAO1 and PA14: A genomic, phenotypic, and therapeutic review. Front Microbiol 2022; 13:1023523. [PMID: 36312971 PMCID: PMC9607943 DOI: 10.3389/fmicb.2022.1023523] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/28/2022] [Indexed: 11/25/2022] Open
Abstract
Pseudomonas aeruginosa is a ubiquitous, motile, gram-negative bacterium that has been recently identified as a multi-drug resistant pathogen in critical need of novel therapeutics. Of the approximately 5,000 strains, PAO1 and PA14 are common laboratory reference strains, modeling moderately and hyper-virulent phenotypes, respectively. PAO1 and PA14 have been instrumental in facilitating the discovery of novel drug targets, testing novel therapeutics, and supplying critical genomic information on the bacterium. While the two strains have contributed to a wide breadth of knowledge on the natural behaviors and therapeutic susceptibilities of P. aeruginosa, they have demonstrated significant deviations from observations in human infections. Many of these deviations are related to experimental inconsistencies in laboratory strain environment that complicate and, at times, terminate translation from laboratory results to clinical applications. This review aims to provide a comparative analysis of the two strains and potential methods to improve their clinical relevance.
Collapse
Affiliation(s)
- Amber Grace
- Department of Biological Sciences, Alabama State University, Montgomery, AL, United States
| | - Rajnish Sahu
- Department of Biological Sciences, Alabama State University, Montgomery, AL, United States
| | | | - Vida A. Dennis
- Department of Biological Sciences, Alabama State University, Montgomery, AL, United States
- *Correspondence: Vida A. Dennis,
| |
Collapse
|
35
|
Lichtenberg M, Kragh KN, Fritz B, Kirkegaard JB, Tolker-Nielsen T, Bjarnsholt T. Cyclic-di-GMP signaling controls metabolic activity in Pseudomonas aeruginosa. Cell Rep 2022; 41:111515. [DOI: 10.1016/j.celrep.2022.111515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 07/13/2022] [Accepted: 09/26/2022] [Indexed: 11/03/2022] Open
|
36
|
Zhang Y, Cai Y, Zeng L, Liu P, Ma LZ, Liu J. A Microfluidic Approach for Quantitative Study of Spatial Heterogeneity in Bacterial Biofilms. SMALL SCIENCE 2022. [DOI: 10.1002/smsc.202200047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Yuzhen Zhang
- Center for Infectious Disease Research School of Medicine Tsinghua University Beijing 100084 China
- Tsinghua-Peking Center for Life Sciences Beijing 100084 China
| | - Yumin Cai
- Center for Infectious Disease Research School of Medicine Tsinghua University Beijing 100084 China
| | - Lingbin Zeng
- Center for Infectious Disease Research School of Medicine Tsinghua University Beijing 100084 China
| | - Peng Liu
- Department of Biomedical Engineering School of Medicine Tsinghua University Beijing 100084 China
| | - Luyan Z. Ma
- State Key Laboratory of Microbial Resources Institute of Microbiology Chinese Academy of Sciences Beijing 100101 China
| | - Jintao Liu
- Center for Infectious Disease Research School of Medicine Tsinghua University Beijing 100084 China
- Tsinghua-Peking Center for Life Sciences Beijing 100084 China
| |
Collapse
|
37
|
Moreno X, Ventura M, Panizo MM, Garcés MF. Assessment of biofilms formation of bacterial and fungal isolates using qualitative Congo red agar and semiquantitative crystal violet microtiter methods. BIOMEDICA : REVISTA DEL INSTITUTO NACIONAL DE SALUD 2022; 43:77-88. [PMID: 37721922 PMCID: PMC10574781 DOI: 10.7705/biomedica.6732] [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: 09/15/2022] [Accepted: 11/15/2022] [Indexed: 09/20/2023]
Abstract
Introduction. Sixty-five percent of human infections are caused by bacteria or yeasts able to form biofilms. This feature makes them more resistant to antimicrobials and antifungals. Objective. To determine biofilm formation capacity of bacterial and fungal isolates by quantitative crystal violet microtiter and qualitative Congo red agar methods. Materials and methods. Brain-heart infusion, trypticase soy broth and Müeller‑Hinton culture media were used in bacterial isolates for the quantitative method; brain-heart infusion broth and Sabouraud dextrose were used for yeasts. The same culture media plus 3% Congo red and 10% dextrose were used to apply the qualitative method in agar. The proposal by Stepanovic, et al. was used as a reference method. Results. We evaluated 103 bacterial isolates and 108 yeasts isolates. We did not recommend substitute brain-heart infusion broth for trypticase soy and Müeller-Hinton broths for biofilm formation assessment in bacterial isolates using the quantitative method. Sabouraud dextrose medium, both broth and agar, can replace brain-heart infusion to assess biofilm formation in yeasts, quantitatively and qualitatively. Conclusion. The study of biofilms in the microbiology laboratory, using Congo red agar qualitative method, is a simple, fast, and inexpensive procedure that provides precise information for the diagnosis and treatment of persistent infections caused by bacteria and yeasts.
Collapse
Affiliation(s)
- Xiomara Moreno
- Departamento de Microbiología, Instituto Médico La Floresta, Caracas, Venezuela; Cátedra de Bacteriología, Escuela de Bioanálisis, Facultad de Medicina, Universidad Central de Venezuela, Caracas, Venezuela.
| | - Melanie Ventura
- Escuela de Bioanálisis, Facultad de Medicina, Universidad Central de Venezuela, Caracas.
| | | | - María Fátima Garcés
- Laboratorio de Investigaciones Básicas y Aplicadas, Escuela de Bioanálisis, Facultad de Medicina, Universidad Central de Venezuela, Caracas, Venezuela.
| |
Collapse
|
38
|
Vidal JM, Ruiz P, Carrasco C, Barros J, Sepúlveda D, Ruiz-Tagle N, Romero A, Urrutia H, Oliver C. Piscirickettsia salmonis forms a biofilm on nylon surface using a CDC Biofilm Reactor. JOURNAL OF FISH DISEASES 2022; 45:1099-1107. [PMID: 35543448 DOI: 10.1111/jfd.13632] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/18/2022] [Accepted: 04/21/2022] [Indexed: 06/14/2023]
Abstract
Research into Piscirickettsia salmonis biofilms on materials commonly used in salmon farming is crucial for understanding its persistence and virulence. We used the CDC Biofilm Reactor to investigate P. salmonis (LF-89 and EM-90) biofilm formation on Nylon, Stainless steel (316L), Polycarbonate and High-Density Polyethylene (HDPE) surfaces. After 144 h of biofilm visualization by scanning confocal laser microscopy under batch growth conditions, Nylon coupons generated the greatest biofilm formation and coverage compared to Stainless steel (316L), Polycarbonate and HDPE. Additionally, P. salmonis biofilm formation on Nylon was significantly greater (p ≤ .01) than Stainless steel (316L), Polycarbonate and HDPE at 288 h. We used Nylon coupons to determine the kinetic parameters of the planktonic and biofilm phases of P. salmonis. The two strains had similar latencies in the planktonic phase; however, LF-89 maximum growth was 2.5 orders of magnitude higher (Log cell ml-1 ). Additionally, LF-89 had a specified growth rate (µmax) of 0.0177 ± 0.006 h-1 and a generation time of 39.2 h. This study contributes to a deeper understanding of the biofilm formation by P. salmonis and elucidates the impact of the biofilm on aquaculture systems.
Collapse
Affiliation(s)
- José Miguel Vidal
- Laboratorio de Biopelículas y Microbiología Ambiental, Centro de Biotecnología, Universidad de Concepción, Concepción, Chile
- Departamento de Investigación y Desarrollo, Ecombio limitada, Concepción, Chile
| | - Pamela Ruiz
- Laboratorio de Biopelículas y Microbiología Ambiental, Centro de Biotecnología, Universidad de Concepción, Concepción, Chile
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Talcahuano, Chile
| | - Carlos Carrasco
- Laboratorio de Biopelículas y Microbiología Ambiental, Centro de Biotecnología, Universidad de Concepción, Concepción, Chile
| | - Javier Barros
- Departamento de Investigación y Desarrollo, Micbiotech spa, Concepción, Chile
| | - Daniela Sepúlveda
- Laboratorio de Biopelículas y Microbiología Ambiental, Centro de Biotecnología, Universidad de Concepción, Concepción, Chile
| | - Nathaly Ruiz-Tagle
- Laboratorio de Biopelículas y Microbiología Ambiental, Centro de Biotecnología, Universidad de Concepción, Concepción, Chile
| | - Alex Romero
- Laboratorio de Inmunología y Estrés de Organismos Acuáticos, Instituto de Patología Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
- Interdisciplinary Center for Aquaculture Research, INCAR, Concepción, Chile
| | - Homero Urrutia
- Laboratorio de Biopelículas y Microbiología Ambiental, Centro de Biotecnología, Universidad de Concepción, Concepción, Chile
- Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Cristian Oliver
- Laboratorio de Inmunología y Estrés de Organismos Acuáticos, Instituto de Patología Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
39
|
Biofilm Detection by a Fiber-Tip Ball Resonator Optical Fiber Sensor. BIOSENSORS 2022; 12:bios12070481. [PMID: 35884284 PMCID: PMC9313161 DOI: 10.3390/bios12070481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 06/29/2022] [Accepted: 06/29/2022] [Indexed: 11/16/2022]
Abstract
Bacterial biofilms are one of the most important challenges that modern medicine faces due to the difficulties of diagnosis, antibiotic resistance, and protective mechanisms against aggressive environments. For these reasons, methods that ensure the inexpensive and rapid or real-time detection of biofilm formation on medical devices are needed. This study examines the possibilities of using optical- and fiber-based biosensors to detect and analyze early bacterial biofilms. In this study, the biofilm-forming model organism Pseudomonas aeruginosa was inoculated on the surface of the optical sensor and allowed to attach for 2 h. The biosensors were made by a fiber-tip ball resonator, fabricated through a CO2 laser splicer on a single-mode fiber, forming a weak reflective spectrum. An optical backscatter reflectometer was used to measure the refractive index detected by the sensors during different growth periods. The early biofilm concentration was determined by crystal violet (CV) binding assay; however, such a concentration was lower than the detection limit of this assay. This work presents a new approach of biofilm sensing in the early attachment stage with a low limit of detection up to 10−4 RIU (refractive index units) or 35 ± 20 × 103 CFU/mL (colony formed units).
Collapse
|
40
|
Yang Y, Zan J, Shuai Y, Yang L, Zhang L, Zhang H, Wang D, Peng S, Shuai C. In Situ Growth of a Metal-Organic Framework on Graphene Oxide for the Chemo-Photothermal Therapy of Bacterial Infection in Bone Repair. ACS APPLIED MATERIALS & INTERFACES 2022; 14:21996-22005. [PMID: 35512272 DOI: 10.1021/acsami.2c04841] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Bacterial infection with high morbidity (>30%) seriously affects the defect's healing after bone transplantation. To this end, chemotherapy and photothermal therapy have been utilized for antibacterial treatment owing to their high selectivity and minimal toxicity. However, they also face several dilemmas. For example, bacterial biofilms prevented the penetration of antibacterial agents and local temperatures (over 70 °C) caused by the photothermal therapy damaged normal tissue. Herein, a co-dispersion nanosystem with chemo-photothermal function was constructed via the in situ growth of zeolitic imidazolate framework-8 (ZIF-8) on graphene oxide (GO) nanosheets. In this nanosystem, GO generates a local temperature (∼50 °C) to increase the permeability of a bacterial biofilm under near-infrared laser irradiation. Then, Zn ions released by ZIF-8 seized this chance to react with the bacterial membrane and inactivate it, thus realizing efficient sterilization in a low-temperature environment. This antibacterial system was incorporated into a poly-l-lactic acid scaffold for bone repair. Results showed that the scaffold showed a high antibacterial rate of 85% against both Escherichia coli and Staphylococcus aureus. In vitro cell tests showed that the scaffold promoted cell proliferation.
Collapse
Affiliation(s)
- Youwen Yang
- Institute of Additive Manufacturing, Jiangxi University of Science and Technology, Nanchang 330013, China
| | - Jun Zan
- Institute of Additive Manufacturing, Jiangxi University of Science and Technology, Nanchang 330013, China
| | - Yang Shuai
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Liuyimei Yang
- Ganjiang Innovation Academy, Chinese Academy of Sciences, Ganzhou 341119, China
| | - Lemin Zhang
- Institute of Additive Manufacturing, Jiangxi University of Science and Technology, Nanchang 330013, China
| | - Hanqing Zhang
- Institute of Additive Manufacturing, Jiangxi University of Science and Technology, Nanchang 330013, China
| | - Dongsheng Wang
- Key Laboratory of Construction Hydraulic Robots of Anhui Higher Education Institutes, Tongling University, Tongling 244000, China
| | - Shuping Peng
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China
- NHC Key Laboratory of Carcinogenesis of Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410013, China
- School of Energy and Machinery Engineering, Jiangxi University of Science and Technology, Nanchang 330013, China
| | - Cijun Shuai
- Institute of Additive Manufacturing, Jiangxi University of Science and Technology, Nanchang 330013, China
- State Key Laboratory of High Performance Complex Manufacturing, Central South University, Changsha 410083, China
| |
Collapse
|
41
|
Tang PC, Eriksson O, Sjögren J, Fatsis-Kavalopoulos N, Kreuger J, Andersson DI. A Microfluidic Chip for Studies of the Dynamics of Antibiotic Resistance Selection in Bacterial Biofilms. Front Cell Infect Microbiol 2022; 12:896149. [PMID: 35619647 PMCID: PMC9128571 DOI: 10.3389/fcimb.2022.896149] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/05/2022] [Indexed: 01/01/2023] Open
Abstract
Biofilms are arguably the most important mode of growth of bacteria, but how antibiotic resistance emerges and is selected in biofilms remains poorly understood. Several models to study evolution of antibiotic resistance have been developed, however, their usability varies depending on the nature of the biological question. Here, we developed and validated a microfluidic chip (Brimor) for studying the dynamics of enrichment of antibiotic-resistant bacteria in biofilms using real-time monitoring with confocal microscopy. In situ extracellular cellulose staining and physical disruption of the biomass confirmed Escherichia coli growth as biofilms in the chip. We showed that seven generations of growth occur in 16 h when biofilms were established in the growth chambers of Brimor, and that bacterial death and growth rates could be estimated under these conditions using a plasmid with a conditional replication origin. Additionally, competition experiments between antibiotic-susceptible and -resistant bacteria at sub-inhibitory concentrations demonstrated that the antibiotic ciprofloxacin selected for antibiotic resistance in bacterial biofilms at concentrations 17-fold below the minimal inhibitory concentration of susceptible planktonic bacteria. Overall, the microfluidic chip is easy to use and a relevant model for studying the dynamics of selection of antibiotic resistance in bacterial biofilms and we anticipate that the Brimor chip will facilitate basic research in this area.
Collapse
Affiliation(s)
- Po-Cheng Tang
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Olle Eriksson
- U-Print, Uppsala University 3D-Printing Facility, Uppsala University, Uppsala, Sweden
| | | | | | - Johan Kreuger
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- *Correspondence: Dan I. Andersson, ; Johan Kreuger,
| | - Dan I. Andersson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- *Correspondence: Dan I. Andersson, ; Johan Kreuger,
| |
Collapse
|
42
|
Carriot N, Barry-Martinet R, Briand JF, Ortalo-Magné A, Culioli G. Impact of phosphate concentration on the metabolome of biofilms of the marine bacterium Pseudoalteromonas lipolytica. Metabolomics 2022; 18:18. [PMID: 35290545 DOI: 10.1007/s11306-022-01875-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 02/22/2022] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Marine biofilms are the most widely distributed mode of life on Earth and drive biogeochemical cycling processes of most elements. Phosphorus (P) is essential for many biological processes such as energy transfer mechanisms, biological information storage and membrane integrity. OBJECTIVES Our aim was to analyze the effect of a gradient of ecologically relevant phosphate concentrations on the biofilm-forming capacity and the metabolome of the marine bacterium Pseudoalteromonas lipolytica TC8. METHODS In addition to the evaluation of the effect of different phosphate concentration on the biomass, structure and gross biochemical composition of biofilms of P. lipolytica TC8, untargeted metabolomics based on liquid chromatography-mass spectrometry (LC-MS) analysis was used to determine the main metabolites impacted by P-limiting conditions. Annotation of the most discriminating and statistically robust metabolites was performed through the concomitant use of molecular networking and MS/MS fragmentation pattern interpretation. RESULTS At the lowest phosphate concentration, biomass, carbohydrate content and three-dimensional structures of biofilms tended to decrease. Furthermore, untargeted metabolomics allowed for the discrimination of the biofilm samples obtained at the five phosphate concentrations and the highlighting of a panel of metabolites mainly implied in such a discrimination. A large part of the metabolites of the resulting dataset were then putatively annotated. Ornithine lipids were found in increasing quantity when the phosphate concentration decreased, while the opposite trend was observed for oxidized phosphatidylethanolamines (PEs). CONCLUSION This study demonstrated the suitability of LC-MS-based untargeted metabolomics for evaluating the effect of culture conditions on marine bacterial biofilms. More precisely, these results supported the high plasticity of the membrane of P. lipolytica TC8, while the role of the oxidized PEs remains to be clarified.
Collapse
Affiliation(s)
- Nathan Carriot
- Laboratoire MAPIEM, Université de Toulon, EA 4323, La Garde, France
| | | | | | | | - Gérald Culioli
- Laboratoire MAPIEM, Université de Toulon, EA 4323, La Garde, France.
- Institut Méditerranéen de Biodiversité et d'Ecologie Marine et Continentale (IMBE), UMR CNRS-IRD-Avignon, Université-Aix-Marseille Université, Avignon, France.
| |
Collapse
|
43
|
Patel H, Gajjar D. Cell adhesion and twitching motility influence strong biofilm formation in Pseudomonas aeruginosa. BIOFOULING 2022; 38:235-249. [PMID: 35345952 DOI: 10.1080/08927014.2022.2054703] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 06/14/2023]
Abstract
In the present study, biofilm formation was quantified in UTI isolates of Pseudomonas aeruginosa (n = 22) using the crystal violet assay and was categorized into; strong (n = 16), weak (n = 4), and moderate (n = 2) biofilm producers. Further experiments were done using strong (n = 4) and weak (n = 4) biofilm producers. Biofilm formation was greater in Luria broth followed by natural urine and artificial urine on silicone and silicone-coated latex. Cell adhesion and twitching motility were greater in strong biofilm producers. The presence of thick biofilm with an increased number of dead and total number of cells of strong biofilm producers was observed using CLSM. The concentrations of exopolymeric substances (eDNA, protein, and pel polysaccharide) were high in strong biofilm producers. FEG-SEM visualization of biofilm produced by strong biofilm producers showed more cells encased in thick biofilm matrix than weak ones. Overall results provide evidence for increased cell adhesion and twitching motility in strong biofilm producers.
Collapse
Affiliation(s)
- Hiral Patel
- Department of Microbiology and Biotechnology Centre, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Devarshi Gajjar
- Department of Microbiology and Biotechnology Centre, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| |
Collapse
|
44
|
Medina Lopez AI, Fregoso DR, Gallegos A, Yoon DJ, Fuentes JJ, Crawford R, Kaba H, Yang H, Isseroff RR. Beta adrenergic receptor antagonist can modify
Pseudomonas aeruginosa
biofilm formation in vitro: Implications for chronic wounds. FASEB J 2022; 36:e22057. [DOI: 10.1096/fj.202100717rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 10/20/2021] [Accepted: 11/08/2021] [Indexed: 11/11/2022]
Affiliation(s)
| | - Daniel R. Fregoso
- Department of Dermatology University of California, Davis Davis California USA
| | - Anthony Gallegos
- Department of Dermatology University of California, Davis Davis California USA
| | - Daniel J. Yoon
- Department of Dermatology University of California, Davis Davis California USA
| | - Jaime J. Fuentes
- Department of Biological Sciences California State University Sacramento Sacramento California USA
| | - Robert Crawford
- Department of Biological Sciences California State University Sacramento Sacramento California USA
| | - Hawa Kaba
- Department of Dermatology University of California, Davis Davis California USA
| | - Hsin‐ya Yang
- Department of Dermatology University of California, Davis Davis California USA
| | - R. Rivkah Isseroff
- Department of Dermatology University of California, Davis Davis California USA
- Dermatology Section VA Northern California Health Care System Mather USA
| |
Collapse
|
45
|
A Humanized Monoclonal Antibody Potentiates Killing by Antibiotics of Diverse Biofilm-Forming Respiratory Tract Pathogens. Antimicrob Agents Chemother 2022; 66:e0187721. [DOI: 10.1128/aac.01877-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
New strategies to treat diseases wherein biofilms contribute significantly to pathogenesis are needed as biofilm-resident bacteria are highly recalcitrant to antibiotics due to physical biofilm architecture and a canonically quiescent metabolism, among many additional attributes. We, and others, have shown that when biofilms are dispersed or disrupted, bacteria released from biofilm residence are in a distinct physiologic state that, in part, renders these bacteria highly sensitive to killing by specific antibiotics. We sought to demonstrate the breadth of ability of a recently humanized monoclonal antibody against an essential biofilm structural element (DNABII protein) to disrupt biofilms formed by respiratory tract pathogens and potentiate antibiotic-mediated killing of bacteria released from biofilm residence.
Biofilms formed by six respiratory tract pathogens were significantly disrupted by the humanized monoclonal antibody in a dose- and time-dependent manner, as corroborated by CLSM imaging. Bacteria newly released from the biofilms of 3 of 6 species were significantly more sensitive than their planktonic counterparts to killing by 2 of 3 antibiotics currently used clinically and were now also equally as sensitive to killing by the 3
rd
antibiotic. The remaining 3 pathogens were significantly more susceptible to killing by all 3 antibiotics.
A humanized monoclonal antibody directed against protective epitopes of a DNABII protein effectively released six diverse respiratory tract pathogens from biofilm residence in a phenotypic state that was now as, or significantly more, sensitive to killing by three antibiotics currently indicated for use clinically. These data support this targeted, combinatorial, species-agnostic therapy to mitigate chronic bacterial diseases.
Collapse
|
46
|
Tam AKY, Harding B, Green JEF, Balasuriya S, Binder BJ. Thin-film lubrication model for biofilm expansion under strong adhesion. Phys Rev E 2022; 105:014408. [PMID: 35193209 DOI: 10.1103/physreve.105.014408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 11/26/2021] [Indexed: 06/14/2023]
Abstract
Understanding microbial biofilm growth is important to public health because biofilms are a leading cause of persistent clinical infections. In this paper, we develop a thin-film model for microbial biofilm growth on a solid substratum to which it adheres strongly. We model biofilms as two-phase viscous fluid mixtures of living cells and extracellular fluid. The model explicitly tracks the movement, depletion, and uptake of nutrients and incorporates cell proliferation via a nutrient-dependent source term. Notably, our thin-film reduction is two dimensional and includes the vertical dependence of cell volume fraction. Numerical solutions show that this vertical dependence is weak for biologically feasible parameters, reinforcing results from previous models in which this dependence was neglected. We exploit this weak dependence by writing and solving a simplified one-dimensional model that is computationally more efficient than the full model. We use both the one- and two-dimensional models to predict how model parameters affect expansion speed and biofilm thickness. This analysis reveals that expansion speed depends on cell proliferation, nutrient availability, cell-cell adhesion on the upper surface, and slip on the biofilm-substratum interface. Our numerical solutions provide a means to qualitatively distinguish between the extensional flow and lubrication regimes, and quantitative predictions that can be tested in future experiments.
Collapse
Affiliation(s)
- Alexander K Y Tam
- School of Mathematical Sciences, Queensland University of Technology, Brisbane Queensland 4000, Australia
- School of Mathematics and Physics, The University of Queensland, St. Lucia Queensland 4072, Australia
- School of Mathematical Sciences, The University of Adelaide, Adelaide SA 5005, Australia
| | - Brendan Harding
- School of Mathematical Sciences, The University of Adelaide, Adelaide SA 5005, Australia
- School of Mathematics and Statistics, Victoria University of Wellington, Wellington 6140, New Zealand
| | - J Edward F Green
- School of Mathematical Sciences, The University of Adelaide, Adelaide SA 5005, Australia
| | - Sanjeeva Balasuriya
- School of Mathematical Sciences, The University of Adelaide, Adelaide SA 5005, Australia
| | - Benjamin J Binder
- School of Mathematical Sciences, The University of Adelaide, Adelaide SA 5005, Australia
| |
Collapse
|
47
|
Kadam S, Madhusoodhanan V, Dhekane R, Bhide D, Ugale R, Tikhole U, Kaushik KS. Milieu matters: An in vitro wound milieu to recapitulate key features of, and probe new insights into, mixed-species bacterial biofilms. Biofilm 2021; 3:100047. [PMID: 33912828 PMCID: PMC8065265 DOI: 10.1016/j.bioflm.2021.100047] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/29/2021] [Accepted: 03/29/2021] [Indexed: 12/20/2022] Open
Abstract
Bacterial biofilms are a major cause of delayed wound healing. Consequently, the study of wound biofilms, particularly in host-relevant conditions, has gained importance. Most in vitro studies employ refined laboratory media to study biofilms, representing conditions that are not relevant to the infection state. To mimic the wound milieu, in vitro biofilm studies often incorporate serum or plasma in growth conditions, or employ clot or matrix-based biofilm models. While incorporating serum or plasma alone is a minimalistic approach, the more complex in vitro wound models are technically demanding, and poorly compatible with standard biofilm assays. Based on previous reports of clinical wound fluid composition, we have developed an in vitro wound milieu (IVWM) that includes, in addition to serum (to recapitulate wound fluid), matrix elements and biochemical factors. With Luria-Bertani broth and Fetal Bovine Serum (FBS) for comparison, the IVWM was used to study planktonic growth, biofilm features, and interspecies interactions, of common wound pathogens, Staphylococcus aureus and Pseudomonas aeruginosa. We demonstrate that the IVWM recapitulates widely reported in vivo biofilm features such as biomass formation, metabolic activity, increased antibiotic tolerance, 3D structure, and interspecies interactions for monospecies and mixed-species biofilms. Further, the IVWM is simple to formulate, uses laboratory-grade components, and is compatible with standard biofilm assays. Given this, it holds potential as a tractable approach to study wound biofilms under host-relevant conditions.
Collapse
Affiliation(s)
- Snehal Kadam
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Vandana Madhusoodhanan
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Radhika Dhekane
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Devyani Bhide
- MES Abasaheb Garware College of Arts and Science, Pune, India
| | - Rutuja Ugale
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Utkarsha Tikhole
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Karishma S. Kaushik
- Institute of Bioinformatics and Biotechnology, Savitribai Phule Pune University, Pune, India
| |
Collapse
|
48
|
Žiemytė M, Carda-Diéguez M, Rodríguez-Díaz JC, Ventero MP, Mira A, Ferrer MD. Real-time monitoring of Pseudomonas aeruginosa biofilm growth dynamics and persister cells' eradication. Emerg Microbes Infect 2021; 10:2062-2075. [PMID: 34663186 PMCID: PMC8583918 DOI: 10.1080/22221751.2021.1994355] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/16/2021] [Accepted: 10/12/2021] [Indexed: 01/11/2023]
Abstract
Biofilm formation and the appearance of persister cells with low metabolic rates are key factors affecting conventional treatment failure and antibiotic resistance. Using impedance-based measurements, crystal violet staining and traditional culture we have studied the biofilm growth dynamics of 13 Pseudomonas aeruginosa strains under the effect of seven conventional antibiotics. Real-time growth quantifications revealed that the exposure of established P. aeruginosa biofilms to certain concentrations of ciprofloxacin, ceftazidime and tobramycin induced the emergence of persister cells, that showed different morphology and pigmentation, as well increased antibiotic resistance. Whole-genome sequencing of wildtype and persister cells identified several SNPs, a genomic inversion and a genomic duplication in one of the strains. However, these mutations were not uniquely associated with persisters, suggesting that the persistent phenotype may be related to metabolic and transcriptional changes. Given that mannitol has been proposed to activate bacterial metabolism, the synergistic combination of mannitol and ciprofloxacin was evaluated on clinical 48 h P. aeruginosa biofilms. When administered at doses ≥320 mg/L, mannitol was capable of preventing persister cell formation by efficiently activating dormant bacteria and making them susceptible to the antibiotic. These results were confirmed using viable colony counting. As the tested ciprofloxacin-mannitol combination appeared to fully eradicate mature biofilms, we conclude that impedance-based biofilm diagnostics, which permits antibiotic susceptibility testing and the identification of persister cells, is of great potential for the clinical practice and could aid in establishing treatment breakpoints for emerging biofilm-related infections.
Collapse
Affiliation(s)
- Miglė Žiemytė
- Genomics & Health Department, FISABIO Foundation, Valencia, Spain
| | | | - Juan C. Rodríguez-Díaz
- Servicio de Microbiología, Hospital General Universitario de Alicante, ISABIAL, Alicante, Spain
| | - Maria P. Ventero
- Servicio de Microbiología, Hospital General Universitario de Alicante, ISABIAL, Alicante, Spain
| | - Alex Mira
- Genomics & Health Department, FISABIO Foundation, Valencia, Spain
- CIBER of Epidemiology and Public Health, Madrid, Spain
| | - María D. Ferrer
- Genomics & Health Department, FISABIO Foundation, Valencia, Spain
- CIBER of Epidemiology and Public Health, Madrid, Spain
| |
Collapse
|
49
|
Evaluation of the Antimicrobial Efficacy of N-Acetyl-l-Cysteine, Rhamnolipids, and Usnic Acid-Novel Approaches to Fight Food-Borne Pathogens. Int J Mol Sci 2021; 22:ijms222111307. [PMID: 34768739 PMCID: PMC8583417 DOI: 10.3390/ijms222111307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/13/2021] [Accepted: 10/15/2021] [Indexed: 11/21/2022] Open
Abstract
In the food industry, the increasing antimicrobial resistance of food-borne pathogens to conventional sanitizers poses the risk of food contamination and a decrease in product quality and safety. Therefore, we explored alternative antimicrobials N-Acetyl-l-cysteine (NAC), rhamnolipids (RLs), and usnic acid (UA) as a novel approach to prevent biofilm formation and reduce existing biofilms formed by important food-borne pathogens (three strains of Salmonella enterica and two strains of Escherichia coli, Listeria monocytogenes, Staphylococcus aureus). Their effectiveness was evaluated by determining minimum inhibitory concentrations needed for inhibition of bacterial growth, biofilm formation, metabolic activity, and biofilm reduction. Transmission electron microscopy and confocal scanning laser microscopy followed by image analysis were used to visualize and quantify the impact of tested substances on both planktonic and biofilm-associated cells. The in vitro cytotoxicity of the substances was determined as a half-maximal inhibitory concentration in five different cell lines. The results indicate relatively low cytotoxic effects of NAC in comparison to RLs and UA. In addition, NAC inhibited bacterial growth for all strains, while RLs showed overall lower inhibition and UA inhibited only the growth of Gram-positive bacteria. Even though tested substances did not remove the biofilms, NAC represents a promising tool in biofilm prevention.
Collapse
|
50
|
High-throughput screening alternative to crystal violet biofilm assay combining fluorescence quantification and imaging. J Microbiol Methods 2021; 190:106343. [PMID: 34619138 DOI: 10.1016/j.mimet.2021.106343] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 11/23/2022]
Abstract
The crystal violet assay is widely used for biofilm quantitation despite its toxicity and variability. Here, we instead combine fluorescence labelling with the Cytation 5 multi-mode plate reader, to enable simultaneous acquisition of both quantitative and imaging biofilm data. This high-throughput method produces more robust data and provides information about morphology and spatial species organization within the biofilm.
Collapse
|