1
|
Roque‐Borda CA, Primo LMDG, Medina‐Alarcón KP, Campos IC, Nascimento CDF, Saraiva MMS, Berchieri Junior A, Fusco‐Almeida AM, Mendes‐Giannini MJS, Perdigão J, Pavan FR, Albericio F. Antimicrobial Peptides: A Promising Alternative to Conventional Antimicrobials for Combating Polymicrobial Biofilms. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410893. [PMID: 39530703 PMCID: PMC11714181 DOI: 10.1002/advs.202410893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Polymicrobial biofilms adhere to surfaces and enhance pathogen resistance to conventional treatments, significantly contributing to chronic infections in the respiratory tract, oral cavity, chronic wounds, and on medical devices. This review examines antimicrobial peptides (AMPs) as a promising alternative to traditional antibiotics for treating biofilm-associated infections. AMPs, which can be produced as part of the innate immune response or synthesized therapeutically, have broad-spectrum antimicrobial activity, often disrupting microbial cell membranes and causing cell death. Many specifically target negatively charged bacterial membranes, unlike host cell membranes. Research shows AMPs effectively inhibit and disrupt polymicrobial biofilms and can enhance conventional antibiotics' efficacy. Preclinical and clinical research is advancing, with animal studies and clinical trials showing promise against multidrug-resistant bacteria and fungi. Numerous patents indicate increasing interest in AMPs. However, challenges such as peptide stability, potential cytotoxicity, and high production costs must be addressed. Ongoing research focuses on optimizing AMP structures, enhancing stability, and developing cost-effective production methods. In summary, AMPs offer a novel approach to combating biofilm-associated infections, with their unique mechanisms and synergistic potential with existing antibiotics positioning them as promising candidates for future treatments.
Collapse
Affiliation(s)
- Cesar Augusto Roque‐Borda
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
- iMed.ULisboa–Institute for Medicines ResearchFaculty of PharmacyUniversity of LisbonLisbon1649004Portugal
- Vicerrectorado de InvestigaciónUniversidad Católica de Santa MaríaArequipa04000Peru
| | - Laura Maria Duran Gleriani Primo
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Kaila Petronila Medina‐Alarcón
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Isabella C. Campos
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Camila de Fátima Nascimento
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Mauro M. S. Saraiva
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Angelo Berchieri Junior
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Ana Marisa Fusco‐Almeida
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Maria José Soares Mendes‐Giannini
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - João Perdigão
- iMed.ULisboa–Institute for Medicines ResearchFaculty of PharmacyUniversity of LisbonLisbon1649004Portugal
| | - Fernando Rogério Pavan
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Fernando Albericio
- Peptide Science LaboratorySchool of Chemistry and PhysicsUniversity of KwaZulu‐NatalDurban4001South Africa
- CIBER‐BBNNetworking Centre on BioengineeringBiomaterials and Nanomedicineand Department of Organic ChemistryUniversity of BarcelonaBarcelona08028Spain
| |
Collapse
|
2
|
Kashi M, Noei M, Chegini Z, Shariati A. Natural compounds in the fight against Staphylococcus aureus biofilms: a review of antibiofilm strategies. Front Pharmacol 2024; 15:1491363. [PMID: 39635434 PMCID: PMC11615405 DOI: 10.3389/fphar.2024.1491363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024] Open
Abstract
Staphylococcus aureus is an important pathogen due to its ability to form strong biofilms and antibiotic resistance. Biofilms play an important role in bacterial survival against the host immune system and antibiotics. Natural compounds (NCs) have diverse bioactive properties with a low probability of resistance, making them promising candidates for biofilm control. NC such as curcumin, cinnamaldehyde, carvacrol, eugenol, thymol, citral, linalool, 1,8-cineole, pinene, cymene, terpineol, quercetin, and limonene have been widely utilized for the inhibition and destruction of S. aureus biofilms. NCs influence biofilm formation through several procedures. Some of the antibiofilm mechanisms of NCs are direct bactericidal effect, disrupting the quorum sensing system, preventing bacteria from aggregation and attachment to surfaces, reducing the microbial surface components recognizing adhesive matrix molecules (MSCRAMMs), interfering with sortase A enzyme, and altering the expression of biofilm-associated genes such as icaADBC, agr, and sarA. Furthermore, these compounds affect extracellular polymeric substances (EPS) and their components, such as polysaccharide intercellular adhesin (PIA) and eDNA. However, some disadvantages, such as low water solubility and bioavailability, limit their clinical usage. Therefore, scientists have considered using nanotechnology and drug platforms to improve NC's efficacy. Some NC, such as thymol and curcumin, can also enhance photodynamic therapy against S. aurous biofilm community. This article evaluates the anti-biofilm potential of NC, their mechanisms of action against S. aureus biofilms, and various aspects of their application.
Collapse
Affiliation(s)
- Milad Kashi
- Student Research Committee, Arak University of Medical Sciences, Arak, Iran
| | - Milad Noei
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zahra Chegini
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Aref Shariati
- Infectious Diseases Research Center (IDRC), Arak University of Medical Sciences, Arak, Iran
| |
Collapse
|
3
|
Faghani-Eskandarkolaei P, Heli H, Akbari N, Koohi-Hosseinabadi O, Sari Aslani F, Sattarahmady N. Antibacterial and anti-biofilm activities of gold-curcumin nanohybrids and its polydopamine form upon photo-sonotherapy of Staphylococcus aureus infected implants: In vitro and animal model studies. Int J Biol Macromol 2024; 282:137430. [PMID: 39528199 DOI: 10.1016/j.ijbiomac.2024.137430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/27/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Implant-related infections are among the major post-surgery problems, and treatment of these infections is challenging due to the formation of biofilms by microorganisms such as Staphylococcus aureus. Herein, a novel gold-curcumin nanohybrid (GCNH) was synthesized for the first time and characterized. GCNH had a band gap energy of 2.41 eV, a zeta potential of -15 mV, and comprised uniform spherical particles with a mean diameter of 8 ± 2 nm. The biological macromolecule of polydopamine was then coated on GCNH to prepare a gold-curcumin-polydopamine nanohybrid (GCDNH). The nanohybrids were employed as novel dual photo-sonosensitizers for bacterial eradication by near-infrared (NIR) light and ultrasound (US) irradiations. GCNH and GCDNH represented photothermal conversion efficiencies of 26 and 32 %, respectively, and GCDNH represented a hemolysis rate of 2.3 % under both near-infrared (NIR) light and ultrasound (US) irradiations. NIR light and US irradiations (photo-sonotherapy) of Staphylococcus aureus using GCDNH depicted anti-bacterial and anti-biofilm efficiencies of 98 and 99 %, respectively, in synergistic manners, which are higher or as high as other sensitizers reported previously. The mechanism of photo-sonotherapy was related to generation of high levels of reactive oxygen species (ROS), and protein and nucleic acid leakages. In an in vivo infection model, NIR light and US irradiations annihilated Staphylococcus aureus on GCDNH-covered implants with high efficiency, without causing damage to normal tissues.
Collapse
Affiliation(s)
- P Faghani-Eskandarkolaei
- Department of Medical Physics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - H Heli
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - N Akbari
- Department of Microbiology, Faculty of Science, Arak Branch, Islamic Azad University, Arak, Iran
| | - O Koohi-Hosseinabadi
- Central Research Laboratory, Shiraz University of Medical Sciences, Shiraz, Iran
| | - F Sari Aslani
- Molecular Dermatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - N Sattarahmady
- Department of Medical Physics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
4
|
Beshiru A, Igbinosa IH, Salami JO, Uwhuba KE, Ogofure AG, Azazi GM, Igere BE, Anegbe B, Evuen UF, Igbinosa EO. Curcuma longa rhizome extract: a potential antibiofilm agent against antibiotic-resistant foodborne pathogens. BIOFOULING 2024; 40:932-947. [PMID: 39624852 DOI: 10.1080/08927014.2024.2432963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 10/28/2024] [Accepted: 11/15/2024] [Indexed: 12/13/2024]
Abstract
The traditional medicinal value of Curcuma longa (turmeric) and its potential relevance in modern healthcare suggests that traditional remedies and natural products can provide valuable solutions to contemporary challenges, such as combating biofilms and antibiotic-resistant pathogens, potentially offering new strategies for addressing health and safety issues in the fields of food and medicine. This study assessed the antibiofilm and antibacterial characterization of Curcuma longa rhizome extract against antibiotic-resistant foodborne pathogens. Gas Chromatography-Mass Spectrometry (GC-MS) and Fourier-transform infrared (FTIR) analysis were determined to check for the compounds, functional groups, and constituents of the plant extract. In-vitro antibiofilm and antibacterial bioassay of the extract were determined using standard bacteriological procedures. Potential mechanisms of the plant extract were also studied using standard biological methods. The important chemical constituents from the GC-MS extract of C. longa are arturmerone, cinnamyl angelate, tumerone, γ-atlantone, atlantone, α-atlantone, γ-atlantone and curlone. The FTIR analysis of the extract comprises alkyl halides, bromoalkanes, alkanes, ethylene molecules, arenes, amines, alcohols, sulfones, carboxylic acids and their derivatives, aromatic compounds, and phenols. The MIC of C. longa crude extract ranges from ethanol extract (0.03125 - 0.5 mg/mL) and acetone extract (0.0625 - 0.5 mg/mL). The MBC range is as follows: ethanol extract (0.125 - 1 mg/mL), acetone extract (0.125 - 1 mg/mL). The time-kill kinetics showed significant cell reduction with time. The bacterial isolates' nucleic acids and protein leakage were consistent with increased extract concentration and time. There was a reduction in the biofilm cell on the shrimp surface and EPS with increased concentration and time. C. longa exerted significant anti-biofilm activity by removing existing biofilms, disrupting cell connections, and decreasing cells in biofilms. These findings can aid food protection from microbial contamination and prevent biofilms-related infections.
Collapse
Affiliation(s)
- Abeni Beshiru
- Applied Microbial Processes & Environmental Health Research Group, University of Benin, Benin City, Nigeria
- Department of Microbiology, College of Natural and Applied Sciences, Western Delta University, Oghara, Nigeria
- Stellenbosch Institute for Advanced Study (STIAS), Wallenberg Research Centre at Stellenbosch University, Stellenbosch, South Africa
| | - Isoken H Igbinosa
- Applied Microbial Processes & Environmental Health Research Group, University of Benin, Benin City, Nigeria
- Department of Environmental Management and Toxicology, University of Benin, Benin City, Nigeria
| | - Joshua O Salami
- Applied Microbial Processes & Environmental Health Research Group, University of Benin, Benin City, Nigeria
| | - Kate E Uwhuba
- Department of Microbiology, College of Natural and Applied Sciences, Western Delta University, Oghara, Nigeria
| | - Abraham G Ogofure
- Applied Microbial Processes & Environmental Health Research Group, University of Benin, Benin City, Nigeria
| | - Gift M Azazi
- Department of Microbiology, College of Natural and Applied Sciences, Western Delta University, Oghara, Nigeria
| | - Bright E Igere
- Department of Microbiology, Biotechnology Unit, Delta State University, Abraka, Nigeria
| | - Bala Anegbe
- Department of Basic and Industrial Chemistry, College of Natural and Applied Science, Western Delta University, Oghara, Nigeria
| | - Uduenevwo F Evuen
- Department of Biochemistry, Delta State University of Science and Technology, Ozoro, Nigeria
| | - Etinosa O Igbinosa
- Applied Microbial Processes & Environmental Health Research Group, University of Benin, Benin City, Nigeria
- Stellenbosch Institute for Advanced Study (STIAS), Wallenberg Research Centre at Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
5
|
Jeong GJ, Khan F, Tabassum N, Cho KJ, Kim YM. Marine-derived bioactive materials as antibiofilm and antivirulence agents. Trends Biotechnol 2024; 42:1288-1304. [PMID: 38637243 DOI: 10.1016/j.tibtech.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/20/2024]
Abstract
Microbial infections are major human health issues, and, recently, the mortality rate owing to bacterial and fungal infections has been increasing. In addition to intrinsic and extrinsic antimicrobial resistance mechanisms, biofilm formation is a key adaptive resistance mechanism. Several bioactive compounds from marine organisms have been identified for use in biofilm therapy owing to their structural complexity, biocompatibility, and economic viability. In this review, we discuss recent trends in the application of marine natural compounds, marine-bioinspired nanomaterials, and marine polymer conjugates as possible therapeutic agents for controlling biofilms and virulence factors. We also comprehensively discuss the mechanisms underlying biofilm formation and inhibition of virulence factors by marine-derived materials and propose possible applications of novel and effective antibiofilm and antivirulence agents.
Collapse
Affiliation(s)
- Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea; Institute of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea.
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Kyung-Jin Cho
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Young-Mog Kim
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
6
|
Pasman R, Zhang J, Zaat SAJ, Brul S, Krom BP. A customizable and defined medium supporting culturing of Candida albicans, Staphylococcus aureus, and human oral epithelial cells. Appl Environ Microbiol 2024; 90:e0036024. [PMID: 39072650 PMCID: PMC11337806 DOI: 10.1128/aem.00360-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024] Open
Abstract
Candida albicans, an opportunistic oral pathogen, synergizes with Staphylococcus aureus, allowing bacteria to co-invade and systemically disseminate within the host. Studying human-microbe interactions creates the need for a universal culture medium that supports fungal, bacterial, and human cell culturing, while allowing sensitive analytical approaches such as OMICs and chromatography techniques. In this study, we established a fully defined, customizable adaptation of Dulbecco's modified Eagle medium (DMEM), allowing multi-kingdom culturing of S. aureus, C. albicans, and human oral cell lines, whereas minimal version of DMEM (mDMEM) did not support growth of S. aureus, and neither did supplementation with dextrose, MEM non-essential amino acids, pyruvate, and Glutamax. This new medium composition, designated as "mDMEM-DMP," promoted growth of all tested S. aureus strains. Addition of 25 mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) further improved growth, while higher concentrations did not improve growth any further. Higher concentrations of HEPES did result in prolonged stabilization of medium pH. mDMEM-DMP promoted (hyphal) C. albicans monoculturing and co-culturing on both solid and semi-solid surfaces. In contrast to S. aureus, addition of HEPES reduced C. albicans maximum culture optical density (OD). Finally, only buffered mDMEM-DMP (100 mM HEPES) was successful in maintaining the metabolic activity of human oral Ca9-22 and HO1N1 cell lines for 24 hours. Altogether, our findings show that mDMEM-DMP is a versatile and potent culture medium for both microbial and human cell culturing, providing a customizable platform to study human as well as microbial molecular physiology and putative interactions. IMPORTANCE Interaction between microbes and the host are in the center of interest both in disease and in health. In order to study the interactions between microbes of different kingdoms and the host, alternative media are required. Synthetic media are useful as they allow addition of specific components. In addition, well-defined media are required if high-resolution analyses such as metabolomics and proteomics are desired. We describe the development of a synthetic medium to study the interactions between C. albicans, S. aureus, and human oral epithelial cells. Our findings show that mDMEM-DMP is a versatile and potent culture medium for both microbial and human cell culturing, providing a customizable platform to study human as well as microbial molecular physiology and putative interactions.
Collapse
Affiliation(s)
- Raymond Pasman
- Department of Molecular Biology and Microbial Food Safety, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Jianbo Zhang
- Department of Molecular Biology and Microbial Food Safety, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Sebastian A. J. Zaat
- Department of Medical Microbiology and Infection Prevention, Amsterdam institute for Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Stanley Brul
- Department of Molecular Biology and Microbial Food Safety, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Bastiaan P. Krom
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Free University Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
7
|
Jeong GJ, Khan F, Tabassum N, Cho KJ, Kim YM. Strategies for controlling polymicrobial biofilms: A focus on antibiofilm agents. Int J Antimicrob Agents 2024; 64:107243. [PMID: 38908533 DOI: 10.1016/j.ijantimicag.2024.107243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/29/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024]
Abstract
Polymicrobial biofilms are among the leading causes of antimicrobial treatment failure. In these biofilms, bacterial and fungal pathogens interact synergistically at the interspecies, intraspecies, and interkingdom levels. Consequently, combating polymicrobial biofilms is substantially more difficult compared to single-species biofilms due to their distinct properties and the resulting potential variation in antimicrobial drug efficiency. In recent years, there has been an increased focus on developing alternative strategies for controlling polymicrobial biofilms formed by bacterial and fungal pathogens. Current approaches for controlling polymicrobial biofilms include monotherapy (using either natural or synthetic compounds), combination treatments, and nanomaterials. Here, a comprehensive review of different types of polymicrobial interactions between pathogenic bacterial species or bacteria and fungi is provided along with a discussion of their relevance. The mechanisms of action of individual compounds, combination treatments, and nanomaterials against polymicrobial biofilms are thoroughly explored. This review provides various future perspectives that can advance the strategies used to control polymicrobial biofilms and their likely modes of action. Since the majority of research on combating polymicrobial biofilms has been conducted in vitro, it would be an essential step in performing in vivo tests to determine the clinical effectiveness of different treatments against polymicrobial biofilms.
Collapse
Affiliation(s)
- Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea; Institute of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea; International Graduate Program of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea.
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Kyung-Jin Cho
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Young-Mog Kim
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
8
|
Song W, Kim C, Lee J, Han J, Jiang Z, Kim J, An S, Park Y, Kweon J. Low-biofouling membrane bioreactor: Effects of cis-2-Decenoic acid addition on EPS and biofouling mitigation. CHEMOSPHERE 2024; 358:142110. [PMID: 38657688 DOI: 10.1016/j.chemosphere.2024.142110] [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: 02/02/2024] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 04/26/2024]
Abstract
Biofouling is inevitable in the membrane process, particularly in membrane bioreactors (MBR) combined with activated sludge processes. Regulating microbial signaling systems with diffusible signal factors such as cis-2-Decenoic acid (CDA) can control biofilm formation without microbial death or growth inhibition. This study assessed the effectiveness of CDA in controlling biofouling in membrane bioreactors (MBRs), essential for wastewater treatment. By modulating microbial signaling, CDA mitigated biofilm formation without hindering microbial growth. Analysis using Confocal Laser Scanning Microscopy (CLSM) revealed structural alterations in the biofilm, reducing biomass and thickness upon CDA application. Moreover, examination of extracellular polymeric substances (EPS) highlighted a decrease in total EPS, particularly effective polysaccharides. In addition, the possibility of shifting from high molecular weight EPS to low molecular weight EPS was revealed through the change in dispersion activity. The 56% extension of MBR operational lifespan resulting from the reduction in EPS is anticipated to offer potential cost savings and improved performance. Despite these results, further investigation is crucial to validate any potential environmental risks associated with CDA and to comprehend its long-term effects at various conditions.
Collapse
Affiliation(s)
- Wonjung Song
- The Academy of Applied Science and Technology, Konkuk University, Seoul, 05029, Republic of Korea
| | - Chehyeun Kim
- Department of Environmental Engineering Konkuk University, Seoul, 05029, Republic of Korea
| | - Jihoon Lee
- Department of Environmental Engineering Konkuk University, Seoul, 05029, Republic of Korea
| | - Jiwon Han
- Department of Environmental Engineering Konkuk University, Seoul, 05029, Republic of Korea
| | - Zikang Jiang
- Department of Environmental Engineering Konkuk University, Seoul, 05029, Republic of Korea
| | - Jaehyeok Kim
- Environmetal & Bio Department, FITI Testing & Research Institute Cheongju-si, Chungcheongbuk-do, 28115, Republic of Korea
| | - Sunkyung An
- Department of Environmental Engineering Konkuk University, Seoul, 05029, Republic of Korea
| | - Yongmin Park
- Operation Business Division, EPS Solution Co.,Ltd, Anyang-si, Gyeonggi-do, 14059, Republic of Korea
| | - Jihyang Kweon
- Department of Environmental Engineering Konkuk University, Seoul, 05029, Republic of Korea.
| |
Collapse
|
9
|
Naz F, Ahmad A, Sarwar Y, Khan MM, Schierack P, Rauf W, Ali A. Characterization of Salmonella enterica Biofilms and Antibiofilm Effect of Carvacrol and 2-Aminobenzimidazole. Foodborne Pathog Dis 2024; 21:52-60. [PMID: 37819687 DOI: 10.1089/fpd.2023.0044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023] Open
Abstract
Biofilm-associated foodborne Salmonella infections in poultry have become increasingly challenging for veterinarians, particularly in developing countries, and warrant thorough investigation. We assessed the biofilm-forming tendency of poultry isolates of Salmonella enterica, namely Salmonella Typhimurium (n = 23), Salmonella Infantis (n = 28), and Salmonella Heidelberg (n = 18), in nutrient-rich Rappaport-Vassiliadis Soya (RVS) peptone broth and nutrient-deficient diluted Tryptone Soya Broth (TSB). Seven of the tested isolates exhibited moderate biofilm formation in diluted TSB, whereas two showed such formation in RVS. In addition, the Congo red agar assay revealed curli and cellulose production in seven isolates. Fourteen specific biofilm-associated genes were analyzed identifying sdiA and seqA to be the most prevalent (100%), and glyA the least prevalent (69.5%). The prevalence of the genes bcsA and csgA was significantly lower in moderate and weak biofilm formers, respectively, as compared with nonbiofilm formers in RVS peptone broth. Furthermore, the compounds carvacrol and 2-aminobenzimidazole (2-ABI) effectively inhibited biofilm formation by Salmonella serovars in RVS peptone and TSB media, respectively. Whereas the antibiofilm activity of 2-ABI against Salmonella has not been reported previously, we determined its most effective concentration at 1.5 mM among tested antibiofilm treatments. These findings indicate that Salmonella strains prevalent in poultry farms have the potential to form biofilms, and the tested compounds should be further explored as supportive or alternative antimicrobials.
Collapse
Affiliation(s)
- Fizza Naz
- Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Abrar Ahmad
- Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Yasra Sarwar
- Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Muhammad Moman Khan
- Faculty Environment and Natural Sciences, Institute of Biotechnology, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Peter Schierack
- Faculty Environment and Natural Sciences, Institute of Biotechnology, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus-Senftenberg, the Brandenburg Medical School Theodor Fontane and the University of Potsdam, Potsdam, Germany
| | - Waqar Rauf
- Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Aamir Ali
- Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| |
Collapse
|
10
|
Chen L, Li X, Wang Y, Guo Z, Wang G, Zhang Y. The performance of plant essential oils against lactic acid bacteria and adverse microorganisms in silage production. FRONTIERS IN PLANT SCIENCE 2023; 14:1285722. [PMID: 38023889 PMCID: PMC10667483 DOI: 10.3389/fpls.2023.1285722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023]
Abstract
Plant essential oils have played an important role in the field of antibiotic alternatives because of their efficient bacteriostatic and fungistatic activity. As plant essential oils are widely used, their activity to improve the quality of plant silage has also been explored. This review expounds on the active ingredients of essential oils, their bacteriostatic and fungistatic activity, and mechanisms, as well as discusses the application of plant essential oils in plant silage fermentation, to provide a reference for the development and application of plant essential oils as silage additives in plant silage fermentation feed.
Collapse
Affiliation(s)
- Lijuan Chen
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Xi Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Yili Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Zelin Guo
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Guoming Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Yunhua Zhang
- College of Resources and Environment, Anhui Agricultural University, Hefei, China
| |
Collapse
|
11
|
Khan F, Jeong GJ, Javaid A, Thuy Nguyen Pham D, Tabassum N, Kim YM. Surface adherence and vacuolar internalization of bacterial pathogens to the Candida spp. cells: Mechanism of persistence and propagation. J Adv Res 2023; 53:115-136. [PMID: 36572338 PMCID: PMC10658324 DOI: 10.1016/j.jare.2022.12.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/17/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The co-existence of Candida albicans with the bacteria in the host tissues and organs displays interactions at competitive, antagonistic, and synergistic levels. Several pathogenic bacteria take advantage of such types of interaction for their survival and proliferation. The chemical interaction involves the signaling molecules produced by the bacteria or Candida spp., whereas the physical attachment occurs by involving the surface proteins of the bacteria and Candida. In addition, bacterial pathogens have emerged to internalize inside the C. albicans vacuole, which is one of the inherent properties of the endosymbiotic relationship between the bacteria and the eukaryotic host. AIM OF REVIEW The interaction occurring by the involvement of surface protein from diverse bacterial species with Candida species has been discussed in detail in this paper. An in silico molecular docking study was performed between the surface proteins of different bacterial species and Als3P of C. albicans to explain the molecular mechanism involved in the Als3P-dependent interaction. Furthermore, in order to understand the specificity of C. albicans interaction with Als3P, the evolutionary relatedness of several bacterial surface proteins has been investigated. Furthermore, the environmental factors that influence bacterial pathogen internalization into the Candida vacuole have been addressed. Moreover, the review presented future perspectives for disrupting the cross-kingdom interaction and eradicating the endosymbiotic bacterial pathogens. KEY SCIENTIFIC CONCEPTS OF REVIEW With the involvement of cross-kingdom interactions and endosymbiotic relationships, the bacterial pathogens escape from the environmental stresses and the antimicrobial activity of the host immune system. Thus, the study of interactions between Candida and bacterial pathogens is of high clinical significance.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea.
| | - Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Aqib Javaid
- Department of Biotechnology and Bioinformatics, University of Hyderabad, India
| | - Dung Thuy Nguyen Pham
- Institute of Applied Technology and Sustainable Development, Nguyen Tat Thanh University, Ho Chi Minh City 70000, Vietnam
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Young-Mog Kim
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea; Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
12
|
Bonincontro G, Scuderi SA, Marino A, Simonetti G. Synergistic Effect of Plant Compounds in Combination with Conventional Antimicrobials against Biofilm of Staphylococcus aureus, Pseudomonas aeruginosa, and Candida spp. Pharmaceuticals (Basel) 2023; 16:1531. [PMID: 38004397 PMCID: PMC10675371 DOI: 10.3390/ph16111531] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/20/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
Bacterial and fungal biofilm has increased antibiotic resistance and plays an essential role in many persistent diseases. Biofilm-associated chronic infections are difficult to treat and reduce the efficacy of medical devices. This global problem has prompted extensive research to find alternative strategies to fight microbial chronic infections. Plant bioactive metabolites with antibiofilm activity are known to be potential resources to alleviate this problem. The phytochemical screening of some medicinal plants showed different active groups, such as stilbenes, tannins, alkaloids, terpenes, polyphenolics, flavonoids, lignans, quinones, and coumarins. Synergistic effects can be observed in the interaction between plant compounds and conventional drugs. This review analyses and summarises the current knowledge on the synergistic effects of plant metabolites in combination with conventional antimicrobials against biofilms of Staphylococcus aureus, Pseudomonas aeruginosa, and Candida albicans. The synergism of conventional antimicrobials with plant compounds can modify and inhibit the mechanisms of acquired resistance, reduce undesirable effects, and obtain an appropriate therapeutic effect at lower doses. A deeper knowledge of these combinations and of their possible antibiofilm targets is needed to develop next-generation novel antimicrobials and/or improve current antimicrobials to fight drug-resistant infections attributed to biofilm.
Collapse
Affiliation(s)
- Graziana Bonincontro
- Department of Environmental Biology, Sapienza University of Rome, P.le Aldo Moro, 5, 00185 Roma, Italy;
| | - Sarah Adriana Scuderi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 98100 Messina, Italy;
| | - Andreana Marino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 98100 Messina, Italy;
| | - Giovanna Simonetti
- Department of Environmental Biology, Sapienza University of Rome, P.le Aldo Moro, 5, 00185 Roma, Italy;
| |
Collapse
|
13
|
Gonçalves ASC, Leitão MM, Simões M, Borges A. The action of phytochemicals in biofilm control. Nat Prod Rep 2023; 40:595-627. [PMID: 36537821 DOI: 10.1039/d2np00053a] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Covering: 2009 to 2021Antimicrobial resistance is now rising to dangerously high levels in all parts of the world, threatening the treatment of an ever-increasing range of infectious diseases. This has becoming a serious public health problem, especially due to the emergence of multidrug-resistance among clinically important bacterial species and their ability to form biofilms. In addition, current anti-infective therapies have low efficacy in the treatment of biofilm-related infections, leading to recurrence, chronicity, and increased morbidity and mortality. Therefore, it is necessary to search for innovative strategies/antibacterial agents capable of overcoming the limitations of conventional antibiotics. Natural compounds, in particular those obtained from plants, have been exhibiting promising properties in this field. Plant secondary metabolites (phytochemicals) can act as antibiofilm agents through different mechanisms of action from the available antibiotics (inhibition of quorum-sensing, motility, adhesion, and reactive oxygen species production, among others). The combination of different phytochemicals and antibiotics have revealed synergistic or additive effects in biofilm control. This review aims to bring together the most relevant reports on the antibiofilm properties of phytochemicals, as well as insights into their structure and mechanistic action against bacterial pathogens, spanning December 2008 to December 2021.
Collapse
Affiliation(s)
- Ariana S C Gonçalves
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| | - Miguel M Leitão
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| | - Manuel Simões
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| | - Anabela Borges
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| |
Collapse
|
14
|
Fernandes S, Gomes IB, Simões M. Antimicrobial and antibiofilm potentiation by a triple combination of dual biocides and a phytochemical with complementary activity. Food Res Int 2023; 167:112680. [PMID: 37087211 DOI: 10.1016/j.foodres.2023.112680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/26/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023]
Abstract
The failure of current sanitation practices requires the development of effective solutions for microbial control. Although combinations using antibiotics have been extensively studied to look for additive/synergistic effects, biocide combinations are still underexplored. This study aims to evaluate the antimicrobial effectiveness of dual biocide and triple biocide/phytochemical combinations, where phytochemicals are used as quorum sensing (QS) inhibitors. The biocides selected were benzalkonium chloride (BAC) and peracetic acid (PAA) - as commonly used biocides, and glycolic acid (GA) and glyoxal (GO) - as alternative and sustainable biocides. Curcumin (CUR) and 10-undecenoic acid (UA) were the phytochemicals selected, based on their QS inhibition properties. A checkerboard assay was used for the screening of chemical interactions based on the cell growth inhibitory effects against Bacilluscereus and Pseudomonasfluorescens. It was observed that dual biocide combinations resulted in indifference, except the PAA + GA combination, which had a potential additive effect. PAA + GA + CUR and PAA + GA + UA combinations also triggered additive effects. The antimicrobial effects of the combinations were further evaluated on the inactivation of planktonic and biofilm cells after 30 min of exposure. These experiments corroborated the checkerboard results, in which PAA + GA was the most effective combination against planktonic cells (additive/synergistic effects). The antimicrobial effects of triple combinations were species- and biocide-specific. While CUR only potentiate the antimicrobial activity of GA against B.cereus, GA + UA and PAA + GA + UA combinations promoted additional antimicrobial effects against both bacteria. Biofilms were found to be highly tolerant, with modest antimicrobial effects being observed for all the combinations tested. However, this study demonstrated that low doses of biocides can be effective in bacterial control when combining biocides with a QS inhibitor, in particular, the combination of the phytochemical UA (as a QS inhibitor) with GA and PAA.
Collapse
Affiliation(s)
- Susana Fernandes
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Inês B Gomes
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Manuel Simões
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal.
| |
Collapse
|
15
|
Akbar M, Haque A, Liaquat S, Schierack P, Ali A. Biofilm Formation by Staphylococcus epidermidis and Its Inhibition Using Carvacrol, 2-Aminobenzemidazole, and 3-Indole Acetonitrile. ACS OMEGA 2023; 8:682-687. [PMID: 36643518 PMCID: PMC9835774 DOI: 10.1021/acsomega.2c05893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/16/2022] [Indexed: 06/17/2023]
Abstract
Biofilm-associated bacterial infections are problematic for physicians due to high antimicrobial resistance in biofilm-forming bacteria. Staphylococcus species, particularly Staphylococcus epidermidis, cause severe infections particularly associated with clinical implants. In this study, we have detected the biofilm formation potential of clinical S. epidermidis isolates using phenotypic and genotypic approaches in nutrient-rich and nutrient-deficient growth conditions. The Congo red agar method determined the biofilm formation potential with limited efficacy. However, the tissue culture plate method adroitly classified the isolates as strong, moderate, weak, and non-biofilm producers with five (10%) of the isolates as strong biofilm producers. Ten biofilm-associated genes were targeted, and the fruA gene was found to be the most prevalent (20%). Three antibiofilm compounds, carvacrol, 2-aminobenzemidazole, and 3-indole acetonitrile, were assessed against strong biofilm-producing S. epidermidis isolates. To the best of our knowledge, this is the first report of genotypic and phenotypic detection of biofilms formed by clinical S. epidermidis isolates from this region. The use of 3-indole acetonitrile against these biofilms and toluene as a solvent is novel. The study highlights the significance of biofilm and antibiofilm potential of the studied compounds for effective treatment and control of S. epidermidis infections.
Collapse
Affiliation(s)
- Muhammad
Umair Akbar
- Department
of Bioinformatics and Biotechnology, Government
College University, Faisalabad, Faisalabad 38000, Pakistan
- National
Institute for Biotechnology and Genetic Engineering College, Pakistan
Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Jhang Road, Faisalabad 38000, Pakistan
| | - Asma Haque
- Department
of Bioinformatics and Biotechnology, Government
College University, Faisalabad, Faisalabad 38000, Pakistan
| | - Sadia Liaquat
- Department
of Bioinformatics and Biotechnology, Government
College University, Faisalabad, Faisalabad 38000, Pakistan
| | - Peter Schierack
- Institute
of Biotechnology, Brandenburg University
of Technology, Cottbus−Senftenberg, Universitätsplatz 1, Senftenberg D-01968, Germany
| | - Aamir Ali
- National
Institute for Biotechnology and Genetic Engineering College, Pakistan
Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Jhang Road, Faisalabad 38000, Pakistan
| |
Collapse
|
16
|
In silico and in vitro insights into the prediction and analysis of natural photosensitive compounds targeting Acinetobacter baumannii biofilm-associated protein. Photodiagnosis Photodyn Ther 2022; 40:103134. [PMID: 36240659 DOI: 10.1016/j.pdpdt.2022.103134] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/03/2022] [Accepted: 09/21/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND The spread of Acinetobacter baumannii strains has become a global concern due to its extensive antibiotic resistance and biofilm formation. To overcome it, new antimicrobial strategies have been needed. Among them, antimicrobial photodynamic therapy (aPDT) is an efficient approach against various microorganisms. This study was focused on the use of curcumin (Cur) and quercetin (Qct) as natural photosensitive compounds to improve the activity of aPDT against A. baumannii biofilm-associated protein (Bap). MATERIALS AND METHODS In this in silico and in vitro study, after determining drug-likeness property, ADME/Toxicity profile, and pharmacological activity of Cur and Qct, virtual screening and molecular docking were assessed to determine the potential binding modes of Cur and Qct to Bap. Then, the anti-biofilm potential of natural photosensitizers-mediated aPDT against A. baumannii was evaluated after the determination of minimum inhibitory concentration (MIC). Subsequently, reverse transcription-quantitative real-time PCR (RT-qPCR) was used to exhibit the anti-virulent effect of aPDT against the gene involved in the biofilm formation of A. baumannii RESULTS: Cur and Qct showed almost similar pharmacokinetic and pharmacodynamics properties. These natural photosensitizers obeyed all the criteria of Lipinski's rule of five principles. According to the molecular docking analysis of protein-ligand complexes, Qct and Cur with a high affinity for Bap showed binding affinity of -6.34 and -6.98 kcal/mol, respectively. According to the findings, aPDT using 4 ×, and 8 × MIC of Cur and Qct could significantly reduce A. baumannii growth in biofilm structures in comparison with the control group (P < 0.05). Also, a significant downregulation by 3.7-, and 5.2-fold in gene expression of bap was observed after treatment with sub-MIC doses of Cur- and Qct-mediated aPDT, respectively (P < 0.05). CONCLUSIONS In summary, the in silico analysis showed that Cur and Qct had strong binding affinity with Bap as a stable protein of A. baumannii. Furthermore, in vitro results displayed that targeted aPDT based on these natural photosensitizers can be considered a treatment against A. baumannii infections by reducing the growth of microbial biofilm and reducing the expression of bap as a gene involved in A. baumannii biofilm formation.
Collapse
|
17
|
Shariati A, Didehdar M, Razavi S, Heidary M, Soroush F, Chegini Z. Natural Compounds: A Hopeful Promise as an Antibiofilm Agent Against Candida Species. Front Pharmacol 2022; 13:917787. [PMID: 35899117 PMCID: PMC9309813 DOI: 10.3389/fphar.2022.917787] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
The biofilm communities of Candida are resistant to various antifungal treatments. The ability of Candida to form biofilms on abiotic and biotic surfaces is considered one of the most important virulence factors of these fungi. Extracellular DNA and exopolysaccharides can lower the antifungal penetration to the deeper layers of the biofilms, which is a serious concern supported by the emergence of azole-resistant isolates and Candida strains with decreased antifungal susceptibility. Since the biofilms' resistance to common antifungal drugs has become more widespread in recent years, more investigations should be performed to develop novel, inexpensive, non-toxic, and effective treatment approaches for controlling biofilm-associated infections. Scientists have used various natural compounds for inhibiting and degrading Candida biofilms. Curcumin, cinnamaldehyde, eugenol, carvacrol, thymol, terpinen-4-ol, linalool, geraniol, cineole, saponin, camphor, borneol, camphene, carnosol, citronellol, coumarin, epigallocatechin gallate, eucalyptol, limonene, menthol, piperine, saponin, α-terpineol, β-pinene, and citral are the major natural compounds that have been used widely for the inhibition and destruction of Candida biofilms. These compounds suppress not only fungal adhesion and biofilm formation but also destroy mature biofilm communities of Candida. Additionally, these natural compounds interact with various cellular processes of Candida, such as ABC-transported mediated drug transport, cell cycle progression, mitochondrial activity, and ergosterol, chitin, and glucan biosynthesis. The use of various drug delivery platforms can enhance the antibiofilm efficacy of natural compounds. Therefore, these drug delivery platforms should be considered as potential candidates for coating catheters and other medical material surfaces. A future goal will be to develop natural compounds as antibiofilm agents that can be used to treat infections by multi-drug-resistant Candida biofilms. Since exact interactions of natural compounds and biofilm structures have not been elucidated, further in vitro toxicology and animal experiments are required. In this article, we have discussed various aspects of natural compound usage for inhibition and destruction of Candida biofilms, along with the methods and procedures that have been used for improving the efficacy of these compounds.
Collapse
Affiliation(s)
- Aref Shariati
- Molecular and Medicine Research Center, Khomein University of Medical Sciences, Khomein, Iran
| | - Mojtaba Didehdar
- Department of Medical Parasitology and Mycology, Arak University of Medical Sciences, Arak, Iran
| | - Shabnam Razavi
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohsen Heidary
- Department of Laboratory Sciences, School of Paramedical Sciences, Sabzevar University of Medical Sciences, Sabzevar, Iran
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Fatemeh Soroush
- Molecular and Medicine Research Center, Khomein University of Medical Sciences, Khomein, Iran
- Student Research Committee, Khomein University of Medical Sciences, Khomein, Iran
| | - Zahra Chegini
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
18
|
Khadke SK, Lee JH, Kim YG, Raj V, Lee J. Appraisal of Cinnamaldehyde Analogs as Dual-Acting Antibiofilm and Anthelmintic Agents. Front Microbiol 2022; 13:818165. [PMID: 35369516 PMCID: PMC8966877 DOI: 10.3389/fmicb.2022.818165] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/02/2022] [Indexed: 12/24/2022] Open
Abstract
Cinnamaldehyde has a broad range of biological activities, which include antibiofilm and anthelmintic activities. The ever-growing problem of drug resistance and limited treatment options have created an urgent demand for natural molecules with antibiofilm and anthelmintic properties. Hence, we hypothesized that molecules with a scaffold structurally similar to that of cinnamaldehyde might act as dual inhibitors against fungal biofilms and helminths. In this regard, eleven cinnamaldehyde analogs were tested to determine their effects on fungal Candida albicans biofilm and nematode Caenorhabditis elegans. α-Methyl and trans-4-methyl cinnamaldehydes efficiently inhibited C. albicans biofilm formation (>90% inhibition at 50 μg/mL) with minimum inhibitory concentrations (MICs) of ≥ 200 μg/mL and 4-bromo and 4-chloro cinnamaldehydes exhibited anthelmintic property at 20 μg/mL against C. elegans. α-Methyl and trans-4-methyl cinnamaldehydes inhibited hyphal growth and cell aggregation. Scanning electron microscopy was employed to determine the surface architecture of C. albicans biofilm and cuticle of C. elegans, and confocal laser scanning microscopy was used to determine biofilm characteristics. The perturbation in gene expression of C. albicans was investigated using qRT-PCR analysis and α-methyl and trans-4-methyl cinnamaldehydes exhibited down-regulation of ECE1, IFD6, RBT5, UCF1, and UME6 and up-regulation of CHT4 and YWP1. Additionally, molecular interaction of these two molecules with UCF1 and YWP1 were revealed by molecular docking simulation. Our observations collectively suggest α-methyl and trans-4-methyl cinnamaldehydes are potent biofilm inhibitors and that 4-bromo and 4-chloro cinnamaldehydes are anthelmintic agents. Efforts are required to determine the range of potential therapeutic applications of cinnamaldehyde analogs.
Collapse
Affiliation(s)
- Sagar Kiran Khadke
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| | - Jin-Hyung Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| | - Yong-Guy Kim
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| | - Vinit Raj
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| |
Collapse
|
19
|
Hwang G. In it together: Candida-bacterial oral biofilms and therapeutic strategies. ENVIRONMENTAL MICROBIOLOGY REPORTS 2022; 14:183-196. [PMID: 35218311 PMCID: PMC8957517 DOI: 10.1111/1758-2229.13053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 05/16/2023]
Abstract
Under natural environmental settings or in the human body, the majority of microorganisms exist in complex polymicrobial biofilms adhered to abiotic and biotic surfaces. These microorganisms exhibit symbiotic, mutualistic, synergistic, or antagonistic relationships with other species during biofilm colonization and development. These polymicrobial interactions are heterogeneous, complex and hard to control, thereby often yielding worse outcomes than monospecies infections. Concerning fungi, Candida spp., in particular, Candida albicans is often detected with various bacterial species in oral biofilms. These Candida-bacterial interactions may induce the transition of C. albicans from commensal to pathobiont or dysbiotic organism. Consequently, Candida-bacterial interactions are largely associated with various oral diseases, including dental caries, denture stomatitis, periodontitis, peri-implantitis, and oral cancer. Given the severity of oral diseases caused by cross-kingdom consortia that develop hard-to-remove and highly drug-resistant biofilms, fundamental research is warranted to strategically develop cost-effective and safe therapies to prevent and treat cross-kingdom interactions and subsequent biofilm development. While studies have shed some light, targeting fungal-involved polymicrobial biofilms has been limited. This mini-review outlines the key features of Candida-bacterial interactions and their impact on various oral diseases. In addition, current knowledge on therapeutic strategies to target Candida-bacterial polymicrobial biofilms is discussed.
Collapse
Affiliation(s)
- Geelsu Hwang
- Department of Preventive and Restorative Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Innovation & Precision Dentistry, School of Dental Medicine, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
- Corresponding Author: Geelsu Hwang,
| |
Collapse
|
20
|
Pohl CH. Recent Advances and Opportunities in the Study of Candida albicans Polymicrobial Biofilms. Front Cell Infect Microbiol 2022; 12:836379. [PMID: 35252039 PMCID: PMC8894716 DOI: 10.3389/fcimb.2022.836379] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/26/2022] [Indexed: 01/11/2023] Open
Abstract
It is well known that the opportunistic pathogenic yeast, Candida albicans, can form polymicrobial biofilms with a variety of bacteria, both in vitro and in vivo, and that these polymicrobial biofilms can impact the course and management of disease. Although specific interactions are often described as either synergistic or antagonistic, this may be an oversimplification. Polymicrobial biofilms are complex two-way interacting communities, regulated by inter-domain (inter-kingdom) signaling and various molecular mechanisms. This review article will highlight advances over the last six years (2016-2021) regarding the unique biology of polymicrobial biofilms formed by C. albicans and bacteria, including regulation of their formation. In addition, some of the consequences of these interactions, such as the influence of co-existence on antimicrobial susceptibility and virulence, will be discussed. Since the aim of this knowledge is to inform possible alternative treatment options, recent studies on the discovery of novel anti-biofilm compounds will also be included. Throughout, an attempt will be made to identify ongoing challenges in this area.
Collapse
|
21
|
Sohn SI, Priya A, Balasubramaniam B, Muthuramalingam P, Sivasankar C, Selvaraj A, Valliammai A, Jothi R, Pandian S. Biomedical Applications and Bioavailability of Curcumin-An Updated Overview. Pharmaceutics 2021; 13:2102. [PMID: 34959384 PMCID: PMC8703330 DOI: 10.3390/pharmaceutics13122102] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/25/2021] [Accepted: 12/03/2021] [Indexed: 02/07/2023] Open
Abstract
Curcumin, a yellow-colored molecule derived from the rhizome of Curcuma longa, has been identified as the bioactive compound responsible for numerous pharmacological activities of turmeric, including anticancer, antimicrobial, anti-inflammatory, antioxidant, antidiabetic, etc. Nevertheless, the clinical application of curcumin is inadequate due to its low solubility, poor absorption, rapid metabolism and elimination. Advancements in recent research have shown several components and techniques to increase the bioavailability of curcumin. Combining with adjuvants, encapsulating in carriers and formulating in nanoforms, in combination with other bioactive agents, synthetic derivatives and structural analogs of curcumin, have shown increased efficiency and bioavailability, thereby augmenting the range of applications of curcumin. The scope for incorporating biotechnology and nanotechnology in amending the current drawbacks would help in expanding the biomedical applications and clinical efficacy of curcumin. Therefore, in this review, we provide a comprehensive overview of the plethora of therapeutic potentials of curcumin, their drawbacks in efficient clinical applications and the recent advancements in improving curcumin's bioavailability for effective use in various biomedical applications.
Collapse
Affiliation(s)
- Soo-In Sohn
- Department of Agricultural Biotechnology, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju 54874, Korea
| | - Arumugam Priya
- Department of Biotechnology, Alagappa University, Karaikudi 630003, India; (A.P.); (P.M.); (R.J.)
| | | | - Pandiyan Muthuramalingam
- Department of Biotechnology, Alagappa University, Karaikudi 630003, India; (A.P.); (P.M.); (R.J.)
- Department of Biotechnology, Sri Shakthi Institute of Engineering and Technology, Coimbatore 641062, India
| | - Chandran Sivasankar
- Department of Food Science and Technology, Pondicherry University, Pondicherry 605014, India;
| | - Anthonymuthu Selvaraj
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA;
| | - Alaguvel Valliammai
- Department of Environmental Hydrology and Microbiology, Ben-Gurion University of the Negev, Beersheba 84990, Israel;
| | - Ravi Jothi
- Department of Biotechnology, Alagappa University, Karaikudi 630003, India; (A.P.); (P.M.); (R.J.)
| | - Subramani Pandian
- Department of Agricultural Biotechnology, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju 54874, Korea
| |
Collapse
|
22
|
Martinengo P, Arunachalam K, Shi C. Polyphenolic Antibacterials for Food Preservation: Review, Challenges, and Current Applications. Foods 2021; 10:foods10102469. [PMID: 34681518 PMCID: PMC8536111 DOI: 10.3390/foods10102469] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 11/16/2022] Open
Abstract
Natural alternatives replacing artificial additives have gained much attention in the consumer’s view because of the growing search for clean label products that are devoid of carcinogenic and toxic effects. Plant polyphenols are considered as suitable alternative natural preservatives with antioxidant and antimicrobial properties. However, their uses in the food industry are undermined by a series of limitations such as low solubility and stability during food processing and storage, lack of standardization, and undesirable organoleptic properties. Different approaches in the use of polyphenols have been proposed in order to overcome the current hurdles related to food preservation. This review article specifically focuses on the antibacterial activity of plant-derived polyphenols as well as their applications as food preservatives, main challenges, and other trends in the food industry.
Collapse
|
23
|
Trigo-Gutierrez JK, Vega-Chacón Y, Soares AB, Mima EGDO. Antimicrobial Activity of Curcumin in Nanoformulations: A Comprehensive Review. Int J Mol Sci 2021; 22:7130. [PMID: 34281181 PMCID: PMC8267827 DOI: 10.3390/ijms22137130] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/21/2021] [Accepted: 06/28/2021] [Indexed: 01/10/2023] Open
Abstract
Curcumin (CUR) is a natural substance extracted from turmeric that has antimicrobial properties. Due to its ability to absorb light in the blue spectrum, CUR is also used as a photosensitizer (PS) in antimicrobial Photodynamic Therapy (aPDT). However, CUR is hydrophobic, unstable in solutions, and has low bioavailability, which hinders its clinical use. To circumvent these drawbacks, drug delivery systems (DDSs) have been used. In this review, we summarize the DDSs used to carry CUR and their antimicrobial effect against viruses, bacteria, and fungi, including drug-resistant strains and emergent pathogens such as SARS-CoV-2. The reviewed DDSs include colloidal (micelles, liposomes, nanoemulsions, cyclodextrins, chitosan, and other polymeric nanoparticles), metallic, and mesoporous particles, as well as graphene, quantum dots, and hybrid nanosystems such as films and hydrogels. Free (non-encapsulated) CUR and CUR loaded in DDSs have a broad-spectrum antimicrobial action when used alone or as a PS in aPDT. They also show low cytotoxicity, in vivo biocompatibility, and improved wound healing. Although there are several in vitro and some in vivo investigations describing the nanotechnological aspects and the potential antimicrobial application of CUR-loaded DDSs, clinical trials are not reported and further studies should translate this evidence to the clinical scenarios of infections.
Collapse
Affiliation(s)
| | | | | | - Ewerton Garcia de Oliveira Mima
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (Unesp), Araraquara 14800-000, Brazil; (J.K.T.-G.); (Y.V.-C.); (A.B.S.)
| |
Collapse
|
24
|
Devi TS, Vijay K, Vidhyavathi RM, Kumar P, Govarthanan M, Kavitha T. Antifungal activity and molecular docking of phenol, 2,4-bis(1,1-dimethylethyl) produced by plant growth-promoting actinobacterium Kutzneria sp. strain TSII from mangrove sediments. Arch Microbiol 2021; 203:4051-4064. [PMID: 34046705 DOI: 10.1007/s00203-021-02397-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/14/2021] [Accepted: 05/20/2021] [Indexed: 10/21/2022]
Abstract
The present study reveals the plant growth-promoting (PGP) potentials and characterizes the antifungal metabolites of Kutzneria sp. strain TSII isolated from mangrove sediment soil through in vitro and in silico studies. In this study, Kutzneria sp. strain TSII was screened for PGP activities and the antifungal activities against Pithomyces atro-olivaceous, a leaf spot-associated pathogen in groundnut plants. The ethyl acetate extract of Kutzneria sp. strain TSII was purified using column chromatography, and the presence of various antimicrobial compounds was studied by gas chromatography-mass spectrometry (GC-MS) analysis. In silico modeling and docking were carried out to evaluate the antifungal potent of bioactive compound. Kutzneria sp. strain TSII produced proteases, phosphatases, ammonia, siderophores, cellulases, indole acetic acid (IAA), lipases, and amylases, indicating its ability to enhance the growth of plants. The ethyl acetate extract of Kutzneria sp strain TSII was found to be a potent inhibitor of fungal mycelial growth in the potato dextrose agar (PDA) plates. The GC-MS spectral study showed 24 antimicrobial compounds belonging to five chemical groups: phenolics, phthalates, fatty acid methyl esters (FAME), spiro, and fatty alcohols. In silico docking studies showed that phenol, 2,4-bis(1,1-dimethylethyl)-effectively attaches with the active site of mitochondrial F1F0 Adenosine triphosphate synthase enzymes of Pithomyces atro-olivaceous. Hence, it is clear that these antifungal compounds shall be formulated shortly to treat many plant fungal diseases in an eco-friendly manner.
Collapse
Affiliation(s)
- Thangarasu Suganya Devi
- Department of Microbiology, Science Campus, Alagappa University, Karaikudi, Tamilnadu, India
| | - Karuppiah Vijay
- Department of Microbiology, Science Campus, Alagappa University, Karaikudi, Tamilnadu, India
| | - R M Vidhyavathi
- Department of Bioinformatics Science Campus, Alagappa University, Karaikudi, Tamilnadu, India
| | - Ponnuchamy Kumar
- Department of Animal Health and Management, Science Campus, Alagappa University, Karaikudi, Tamilnadu, India
| | - Muthusamy Govarthanan
- Department of Environmental Engineering, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Thangavel Kavitha
- Department of Microbiology, Science Campus, Alagappa University, Karaikudi, Tamilnadu, India.
| |
Collapse
|
25
|
Khan F, Bamunuarachchi NI, Pham DTN, Tabassum N, Khan MSA, Kim YM. Mixed biofilms of pathogenic Candida-bacteria: regulation mechanisms and treatment strategies. Crit Rev Microbiol 2021; 47:699-727. [PMID: 34003065 DOI: 10.1080/1040841x.2021.1921696] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mixed-species biofilm is one of the most frequently recorded clinical problems. Mixed biofilms develop as a result of interactions between microorganisms of a single or multiple species (e.g. bacteria and fungi). Candida spp., particularly Candida albicans, are known to associate with various bacterial species to form a multi-species biofilm. Mixed biofilms of Candida spp. have been previously detected in vivo and on the surfaces of many biomedical instruments. Treating infectious diseases caused by mixed biofilms of Candida and bacterial species has been challenging due to their increased resistance to antimicrobial drugs. Here, we review and discuss the clinical significance of mixed Candida-bacteria biofilms as well as the signalling mechanisms involved in Candida-bacteria interactions. We also describe possible approaches for combating infections associated with mixed biofilms, such as the use of natural or synthetic drugs and combination therapy. The review presented here is expected to contribute to the advances in the biomedical field on the understanding of underlying interaction mechanisms of pathogens in mixed biofilm, and alternative approaches to treating the related infections.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, South Korea
| | - Nilushi Indika Bamunuarachchi
- Department of Food Science and Technology, Pukyong National University, Busan, South Korea.,Department of Fisheries and Marine Sciences, Ocean University of Sri Lanka, Tangalle, Sri Lanka
| | - Dung Thuy Nguyen Pham
- Center of Excellence for Biochemistry and Natural Products, Nguyen Tat Thanh University, Ho Chi Minh City, Vietnam.,NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City, Vietnam
| | - Nazia Tabassum
- Industrial Convergence Bionix Engineering, Pukyong National University, Busan, South Korea
| | - Mohd Sajjad Ahmad Khan
- Department of Basic Sciences, Deanship of Preparatory Year and Supporting Studies, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Young-Mog Kim
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, South Korea.,Department of Food Science and Technology, Pukyong National University, Busan, South Korea
| |
Collapse
|
26
|
Huang X, Ge Y, Yang B, Han Q, Zhou W, Liang J, Li M, Peng X, Ren B, Yang B, Weir MD, Guo Q, Wang H, Zhou X, Lu X, Oates TW, Xu HHK, Deng D, Zhou X, Cheng L. Novel dental implant modifications with two-staged double benefits for preventing infection and promoting osseointegration in vivo and in vitro. Bioact Mater 2021; 6:4568-4579. [PMID: 34095616 PMCID: PMC8141509 DOI: 10.1016/j.bioactmat.2021.04.041] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/24/2021] [Accepted: 04/24/2021] [Indexed: 02/05/2023] Open
Abstract
Peri-implantitis are a major problem causing implant failure these days. Accordingly, anti-infection during the early stage and subsequent promotion of osseointegration are two main key factors to solve this issue. Micro-arc oxidation (MAO) treatment is a way to form an oxidation film on the surface of metallic materials. The method shows good osteogenic properties but weak antibacterial effect. Therefore, we developed combined strategies to combat severe peri-implantitis, which included the use of a novel compound, PD, comprising dendrimers poly(amidoamine) (PAMAM) loading dimethylaminododecyl methacrylate (DMADDM) as well as MAO treatment. Here, we explored the chemical properties of the novel compound PD, and proved that this compound was successfully synthesized, with the loading efficiency and encapsulation efficiency of 23.91% and 31.42%, respectively. We further report the two-stage double benefits capability of PD + MAO: (1) in the first stage, PD + MAO could decrease the adherence and development of biofilms by releasing DMADDM in the highly infected first stage after implant surgery both in vitro and in vivo; (2) in the second stage, PD + MAO indicated mighty anti-infection and osteoconductive characteristics in a rat model of peri-implantitis in vivo. This study first reports the two-staged, double benefits of PD + MAO, and demonstrates its potential in clinical applications for inhibiting peri-implantitis, especially in patients with severe infection risk.
Collapse
Affiliation(s)
- Xiaoyu Huang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, China.,Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yang Ge
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, China.,Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, China.,Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China.,Department of Preventive Dentistry, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, the Netherlands
| | - Bina Yang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, China.,Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Qi Han
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, China.,Department of Pathology, West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Wen Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, China.,Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jingou Liang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, China.,Department of Pediatrics, West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Mingyun Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, China.,Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xian Peng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, China
| | - Biao Ren
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, China
| | - Bangcheng Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, China
| | - Michael D Weir
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, 21201, USA
| | - Qiang Guo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, China
| | - Haohao Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, China.,Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xinxuan Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, China
| | - Xugang Lu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, China
| | - Thomas W Oates
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, 21201, USA
| | - Hockin H K Xu
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, 21201, USA
| | - Dongmei Deng
- Department of Preventive Dentistry, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam, the Netherlands
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, China.,Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, China.,Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
27
|
Kart D, Reçber T, Nemutlu E, Sagiroglu M. Sub-Inhibitory Concentrations of Ciprofloxacin Alone and Combinations with Plant-Derived Compounds against P. aeruginosa Biofilms and Their Effects on the Metabolomic Profile of P. aeruginosa Biofilms. Antibiotics (Basel) 2021; 10:antibiotics10040414. [PMID: 33918895 PMCID: PMC8070142 DOI: 10.3390/antibiotics10040414] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION Alternative anti-biofilm agents are needed to combat Pseudomonas aeruginosa infections. The mechanisms behind these new agents also need to be revealed at a molecular level. MATERIALS AND METHODS The anti-biofilm effects of 10 plant-derived compounds on P. aeruginosa biofilms were investigated using minimum biofilm eradication concentration (MBEC) and virulence assays. The effects of ciprofloxacin and compound combinations on P. aeruginosa in mono and triple biofilms were compared. A metabolomic approach and qRT-PCR were applied to the biofilms treated with ciprofloxacin in combination with baicalein, esculin hydrate, curcumin, and cinnamaldehyde at sub-minimal biofilm inhibitory concentration (MBIC) concentrations to highlight the specific metabolic shifts between the biofilms and to determine the quorum sensing gene expressions, respectively. RESULTS The combinations of ciprofloxacin with curcumin, baicalein, esculetin, and cinnamaldehyde showed more reduced MBICs than ciprofloxacin alone. The quorum sensing genes were downregulated in the presence of curcumin and cinnamaldehyde, while upregulated in the presence of baicalein and esculin hydrate rather than for ciprofloxacin alone. The combinations exhibited different killing effects on P. aeruginosa in mono and triple biofilms without affecting its virulence. The findings of the decreased metabolite levels related to pyrimidine and lipopolysaccharide synthesis and to down-regulated alginate and lasI expressions strongly indicate the role of multifactorial mechanisms for curcumin-mediated P. aeruginosa growth inhibition. CONCLUSIONS The use of curcumin, baicalein, esculetin, and cinnamaldehyde with ciprofloxacin will help fight against P. aeruginosa biofilms. To the best of our knowledge, this is the first study of its kind to define the effect of plant-based compounds as possible anti-biofilm agents with low MBICs for the treatment of P. aeruginosa biofilms through metabolomic pathways.
Collapse
Affiliation(s)
- Didem Kart
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Hacettepe University, Ankara 06100, Turkey;
- Correspondence: ; Tel.: +90-533-690-7637
| | - Tuba Reçber
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara 06100, Turkey; (T.R.); (E.N.)
| | - Emirhan Nemutlu
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara 06100, Turkey; (T.R.); (E.N.)
- Bioanalytic and Omics Laboratory, Faculty of Pharmacy, Hacettepe University, Ankara 06100, Turkey
| | - Meral Sagiroglu
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Hacettepe University, Ankara 06100, Turkey;
| |
Collapse
|
28
|
Van Dyck K, Pinto RM, Pully D, Van Dijck P. Microbial Interkingdom Biofilms and the Quest for Novel Therapeutic Strategies. Microorganisms 2021; 9:412. [PMID: 33671126 PMCID: PMC7921918 DOI: 10.3390/microorganisms9020412] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/10/2021] [Accepted: 02/15/2021] [Indexed: 02/06/2023] Open
Abstract
Fungal and bacterial species interact with each other within polymicrobial biofilm communities in various niches of the human body. Interactions between these species can greatly affect human health and disease. Diseases caused by polymicrobial biofilms pose a major challenge in clinical settings because of their enhanced virulence and increased drug tolerance. Therefore, different approaches are being explored to treat fungal-bacterial biofilm infections. This review focuses on the main mechanisms involved in polymicrobial drug tolerance and the implications of the polymicrobial nature for the therapeutic treatment by highlighting clinically relevant fungal-bacterial interactions. Furthermore, innovative treatment strategies which specifically target polymicrobial biofilms are discussed.
Collapse
Affiliation(s)
- Katrien Van Dyck
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, Department of Biology, KU Leuven, 3001 Leuven, Belgium; (K.V.D.); (R.M.P.); (D.P.)
- VIB—KU Leuven Center for Microbiology, 3001 Leuven, Belgium
| | - Rita M. Pinto
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, Department of Biology, KU Leuven, 3001 Leuven, Belgium; (K.V.D.); (R.M.P.); (D.P.)
- VIB—KU Leuven Center for Microbiology, 3001 Leuven, Belgium
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade Do Porto, 4050-313 Porto, Portugal
| | - Durgasruthi Pully
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, Department of Biology, KU Leuven, 3001 Leuven, Belgium; (K.V.D.); (R.M.P.); (D.P.)
| | - Patrick Van Dijck
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, Department of Biology, KU Leuven, 3001 Leuven, Belgium; (K.V.D.); (R.M.P.); (D.P.)
- VIB—KU Leuven Center for Microbiology, 3001 Leuven, Belgium
| |
Collapse
|
29
|
Ceresa C, Rinaldi M, Tessarolo F, Maniglio D, Fedeli E, Tambone E, Caciagli P, Banat IM, Diaz De Rienzo MA, Fracchia L. Inhibitory Effects of Lipopeptides and Glycolipids on C. albicans-Staphylococcus spp. Dual-Species Biofilms. Front Microbiol 2021; 11:545654. [PMID: 33519721 PMCID: PMC7838448 DOI: 10.3389/fmicb.2020.545654] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 12/17/2020] [Indexed: 12/19/2022] Open
Abstract
Microbial biofilms strongly resist host immune responses and antimicrobial treatments and are frequently responsible for chronic infections in peri-implant tissues. Biosurfactants (BSs) have recently gained prominence as a new generation of anti-adhesive and antimicrobial agents with great biocompatibility and were recently suggested for coating implantable materials in order to improve their anti-biofilm properties. In this study, the anti-biofilm activity of lipopeptide AC7BS, rhamnolipid R89BS, and sophorolipid SL18 was evaluated against clinically relevant fungal/bacterial dual-species biofilms (Candida albicans, Staphylococcus aureus, Staphylococcus epidermidis) through quantitative and qualitative in vitro tests. C. albicans-S. aureus and C. albicans-S. epidermidis cultures were able to produce a dense biofilm on the surface of the polystyrene plates and on medical-grade silicone discs. All tested BSs demonstrated an effective inhibitory activity against dual-species biofilms formation in terms of total biomass, cell metabolic activity, microstructural architecture, and cell viability, up to 72 h on both these surfaces. In co-incubation conditions, in which BSs were tested in soluble form, rhamnolipid R89BS (0.05 mg/ml) was the most effective among the tested BSs against the formation of both dual-species biofilms, reducing on average 94 and 95% of biofilm biomass and metabolic activity at 72 h of incubation, respectively. Similarly, rhamnolipid R89BS silicone surface coating proved to be the most effective in inhibiting the formation of both dual-species biofilms, with average reductions of 93 and 90%, respectively. Scanning electron microscopy observations showed areas of treated surfaces that were free of microbial cells or in which thinner and less structured biofilms were present, compared to controls. The obtained results endorse the idea that coating of implant surfaces with BSs may be a promising strategy for the prevention of C. albicans-Staphylococcus spp. colonization on medical devices, and can potentially contribute to the reduction of the high economic efforts undertaken by healthcare systems for the treatment of these complex fungal-bacterial infections.
Collapse
Affiliation(s)
- Chiara Ceresa
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Maurizio Rinaldi
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Francesco Tessarolo
- BIOtech Center for Biomedical Technologies, Department of Industrial Engineering, Università di Trento, Trento, Italy.,Healthcare Research and Innovation Program (IRCS-FBK-PAT), Bruno Kessler Foundation, Trento, Italy
| | - Devid Maniglio
- BIOtech Center for Biomedical Technologies, Department of Industrial Engineering, Università di Trento, Trento, Italy
| | - Emanuele Fedeli
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Erica Tambone
- BIOtech Center for Biomedical Technologies, Department of Industrial Engineering, Università di Trento, Trento, Italy
| | - Patrizio Caciagli
- Section of Electron Microscopy, Department of Medicine Laboratory, Azienda Provinciale per i Servizi Sanitari di Trento, Trento, Italy
| | - Ibrahim M Banat
- School of Biomedical Sciences, Faculty of Life and Health Sciences, Ulster University, Coleraine, United Kingdom
| | - Mayri Alessandra Diaz De Rienzo
- School of Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Liverpool, United Kingdom
| | - Letizia Fracchia
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| |
Collapse
|
30
|
Cheraghipour K, Ezatpour B, Masoori L, Marzban A, Sepahvand A, Rouzbahani AK, Moridnia A, Khanizadeh S, Mahmoudvand H. Anti-Candida Activity of Curcumin: A Systematic Review. Curr Drug Discov Technol 2021; 18:379-390. [PMID: 32418527 DOI: 10.2174/1570163817666200518074629] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/25/2020] [Accepted: 03/25/2020] [Indexed: 06/11/2023]
Abstract
Curcumin is one of the important natural compounds that is extracted from turmeric. This compound and its derivatives have numerous biological properties, including antioxidant, anticancer, anti-inflammatory, antimicrobial, and healing effects. Extensive research in various fields has been conducted on turmeric as it is widely used as a food additive. The significant antifungal activity is one of the major effects of curcumin. In this paper, recent studies on the effects of different forms of curcumin drug on the candidiasis were systematically examined and discussed. The data in this study were extracted from the articles and reports published in the Web of Science, Google Scholar, PubMed, and Scopus databases. After the preliminary investigation, relevant reports were selected and classified based on the incorporated formulation and purpose of the study. After a systematic discussion of the data, it was found that the use of medicinal forms based on nanoparticles can increase the absorption and target the controlled release of curcumin with a more effective role compared to other formulations. Consequently, it can be concluded that new methods of modern medicine can be employed to increase the efficacy of natural pharmaceutical compounds used in the past. In this regard, the present study analyzed the effect of curcumin against various Candida infections, using the recent data. It was found that applying a combination of drug formulation or the formulation of curcumin and its derivatives can be an effective strategy to overcome the medicine resistance in fungal infections, especially candidiasis.
Collapse
Affiliation(s)
- Kourosh Cheraghipour
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Behrouz Ezatpour
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Leila Masoori
- Department of Laboratory Sciences, School of Allied Medical Sciences, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Abdolrazagh Marzban
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Asghar Sepahvand
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | | | - Abbas Moridnia
- Department of Genetics and Molecular Biology, School of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Sayyad Khanizadeh
- Hepatitis Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Hossein Mahmoudvand
- Nutritional Health Research Center, Lorestan University of Medical Sciencs, Khorramabad, Iran
| |
Collapse
|
31
|
Gao S, Liu G, Li J, Chen J, Li L, Li Z, Zhang X, Zhang S, Thorne RF, Zhang S. Antimicrobial Activity of Lemongrass Essential Oil ( Cymbopogon flexuosus) and Its Active Component Citral Against Dual-Species Biofilms of Staphylococcus aureus and Candida Species. Front Cell Infect Microbiol 2020; 10:603858. [PMID: 33415085 PMCID: PMC7783362 DOI: 10.3389/fcimb.2020.603858] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/20/2020] [Indexed: 12/22/2022] Open
Abstract
Compared to mono-species biofilm, biofilms formed by cross-kingdom pathogens are more refractory to conventional antibiotics, thus complicating clinical treatment and causing significant morbidity. Lemongrass essential oil and its bioactive component citral were previously demonstrated to possess strong antimicrobial efficacy against pathogenic bacteria and fungi. However, their effects on polymicrobial biofilms remain to be determined. In this study, the efficacy of lemongrass (Cymbopogon flexuosus) essential oil and its bioactive part citral against dual-species biofilms formed by Staphylococcus aureus and Candida species was evaluated in vitro. Biofilm staining and viability test showed both lemongrass essential oil and citral were able to reduce biofilm biomass and cell viability of each species in the biofilm. Microscopic examinations showed these agents interfered with adhesive characteristics of each species and disrupted biofilm matrix through counteracting nucleic acids, proteins and carbohydrates in the biofilm. Moreover, transcriptional analyses indicated citral downregulated hyphal adhesins and virulent factors of Candida albicans, while also reducing expression of genes involved in quorum sensing, peptidoglycan and fatty acids biosynthesis of S. aureus. Taken together, our results demonstrate the potential of lemongrass essential oil and citral as promising agents against polymicrobial biofilms as well as the underlying mechanisms of their activity in this setting.
Collapse
Affiliation(s)
- Shanjun Gao
- Microbiome Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Guangzhi Liu
- Microbiome Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianguo Li
- Department of Dermatology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Chen
- Microbiome Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Lina Li
- Department of Dermatology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhen Li
- Microbiome Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiulei Zhang
- Microbiome Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Shoumin Zhang
- Department of Dermatology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Rick Francis Thorne
- Translational Research Institute of Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China.,School of Environmental & Life Sciences, University of Newcastle, Newcastle, NSW, Australia
| | - Shuzhen Zhang
- Microbiome Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China.,Department of Dermatology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
32
|
Gao S, Zhang S, Zhang S. Enhanced in vitro antimicrobial activity of amphotericin B with berberine against dual-species biofilms of Candida albicans and Staphylococcus aureus. J Appl Microbiol 2020; 130:1154-1172. [PMID: 32996236 DOI: 10.1111/jam.14872] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022]
Abstract
AIMS Multi-species biofilms formed by fungi and bacteria are clinically common and confer the commensal micro-organisms with protection against antimicrobial therapies. Previously, the plant alkaloid berberine was reported to show antimicrobial efficacy to eliminate bacterial and fungal biofilms. In this study, the combination of berberine and amphotericin B, an antifungal agent, was evaluated against dual-species Candida albicans/Staphylococcus aureus biofilms. METHODS AND RESULTS Combinatorial treatment by berberine and amphotericin B significantly reduced the biomass and viability of residing species in biofilms. Moreover, morphological examination revealed hyphal filamentation of C. albicans and coadhesion between C. albicans/S. aureus were considerably impaired by the treatment. These effects coincided with the reduced expression of cell surface components and quorum-sensing-related genes in both C. albicans and S. aureus. Additionally, in C. albicans, the core transcription factors for controlling biofilm formation together with a crucial component of dual-species biofilms were also downregulated. CONCLUSIONS These results demonstrated synergistic effects of berberine and amphotericin B against C. albicans/S. aureus dual-species biofilms. SIGNIFICANCE AND IMPACT OF THE STUDY This study confirms the potential of berberine and amphotericin B for treating the C. albicans/S. aureus biofilms related infections and reveals molecular basis for the efficacy of combinatorial treatment.
Collapse
Affiliation(s)
- S Gao
- Microbiome Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - S Zhang
- Department of Dermatology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - S Zhang
- Microbiome Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.,Department of Dermatology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
33
|
Wu J, Wu D, Zhao Y, Si Y, Mei L, Shao J, Wang T, Yan G, Wang C. Sodium New Houttuyfonate Inhibits Candida albicans Biofilm Formation by Inhibiting the Ras1-cAMP-Efg1 Pathway Revealed by RNA-seq. Front Microbiol 2020; 11:2075. [PMID: 32983053 PMCID: PMC7477049 DOI: 10.3389/fmicb.2020.02075] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 08/06/2020] [Indexed: 12/23/2022] Open
Abstract
Here, we aim to investigate the antifungal effect and mechanism of action of sodium new houttuyfonate (SNH) against Candida albicans. Microdilution analysis results showed that SNH possesses potent inhibitory activity against C. albicans SC5314, with a MIC80 of 256 μg/mL. Furthermore, we found that SNH can effectively inhibit the initial adhesion of C. albicans. Inverted microscopy, crystal violet staining, scanning electron microscopy and confocal laser scanning microscopy results showed that morphological changes during the transition from yeast to hypha and the biofilm formation of C. albicans are repressed by SNH treatment. We also found that SNH can effectively inhibit the biofilm formation of clinical C. albicans strains (Z103, Z3044, Z1402, and Z1407) and SNH in combination with fluconazole, berberine chloride, caspofungin and itraconazole antifungal agents can synergistically inhibit the biofilm formation of C. albicans. Eukaryotic transcriptome sequencing and qRT-PCR results showed that SNH treatment resulted in significantly down-regulated expression in several biofilm formation related genes in the Ras1-cAMP-Efg1 pathway (ALS1, ALA1, ALS3, EAP1, RAS1, EFG1, HWP1, and TEC1) and significantly up-regulated expression in yeast form-associated genes (YWP1 and RHD1). We also found that SNH can effectively reduce the production of key messenger cAMP in the Ras1-cAMP-Efg1 pathway. Furthermore, using Galleria mellonella as an in vivo model we found that SNH can effectively treat C. albicans infection in vivo. Our presented results suggest that SNH exhibits potential antibiofilm effects related to inhibiting the Ras1-cAMP-Efg1 pathway in the biofilm formation of C. albicans.
Collapse
Affiliation(s)
- Jiadi Wu
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Daqiang Wu
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Key Laboratory of Chinese Herbal Compound Formula in Anhui Province, Hefei, China
| | - Yeye Zhao
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Yuanqing Si
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Longfei Mei
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Jing Shao
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Key Laboratory of Chinese Herbal Compound Formula in Anhui Province, Hefei, China
| | - Tianming Wang
- Key Laboratory of Chinese Herbal Compound Formula in Anhui Province, Hefei, China
| | - Guiming Yan
- Key Laboratory of Chinese Herbal Compound Formula in Anhui Province, Hefei, China
| | - Changzhong Wang
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Key Laboratory of Chinese Herbal Compound Formula in Anhui Province, Hefei, China
| |
Collapse
|
34
|
Qian W, Yang M, Li X, Sun Z, Li Y, Wang X, Wang T. Anti-microbial and anti-biofilm activities of combined chelerythrine-sanguinarine and mode of action against Candida albicans and Cryptococcus neoformans in vitro. Colloids Surf B Biointerfaces 2020; 191:111003. [DOI: 10.1016/j.colsurfb.2020.111003] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/10/2020] [Accepted: 03/26/2020] [Indexed: 12/17/2022]
|
35
|
Co-immobilization of cellobiose dehydrogenase and deoxyribonuclease I on chitosan nanoparticles against fungal/bacterial polymicrobial biofilms targeting both biofilm matrix and microorganisms. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 108:110499. [DOI: 10.1016/j.msec.2019.110499] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/16/2019] [Accepted: 11/26/2019] [Indexed: 11/21/2022]
|
36
|
He H, Qiao K, Wang C, Yang W, Xu Z, Zhang Z, Jia Y, Zhang C, Peng L. Hydrazinocurcumin Induces Apoptosis of Hepatocellular Carcinoma Cells Through the p38 MAPK Pathway. Clin Transl Sci 2020; 14:2075-2084. [PMID: 32100959 PMCID: PMC8504816 DOI: 10.1111/cts.12765] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 12/16/2019] [Indexed: 12/21/2022] Open
Abstract
Hydrazinocurcumin (HZC), a synthetic derivative of curcumin (CUR), has been documented to show anticancer potential in impeding tumor growth in several cancers, including hepatocellular carcinoma (HCC). However, the underlying molecular mechanisms remain unclear. This study aimed to explore the function and underlying mechanisms of HZC on HCC cells, which may involve the p38 mitogen activated protein kinase (MAPK) pathway. HZC was first purified and identified. HepG2 cells were then subjected to treatment with HZC or CUR of different concentrations and p38 MAPK signaling inhibitor (SB203580) to verify their effects on HCC cell apoptosis and proliferation. Furthermore, the functional relevance between HZC and the p38 MAPK pathway in HCC was examined. It was observed that 40 μM HZC exhibited the best pro-apoptosis effect in HCC cells. HZC was found to inhibit HCC cell proliferation and promote apoptosis, the effect of which was stronger than 5-fluorouracil (5-FU). More importantly, the anti-oncogenic effect of HZC and 5-FU was implicated with activation of the p38 MAPK pathway. In vivo experimental results showed that HZC inhibited tumor growth more effectively than 5-FU through the p38 MAPK pathway. These results provide evidence that HZC exerted anti-oncogenic and pro-apoptosis effects in HCC cells through activation of the p38 MAPK pathway.
Collapse
Affiliation(s)
- Hongtao He
- Department of Hepatobiliary Surgery, Fourth Hospital of Hebei Medical University Tumor Hospital of Hebei Province, Shijiazhuang, China
| | - Kuangyuan Qiao
- Basic Medical College of Hebei Medical University, Shijiazhuang, China
| | - Chao Wang
- Department of Hepatobiliary Surgery, Fourth Hospital of Hebei Medical University Tumor Hospital of Hebei Province, Shijiazhuang, China
| | - Wuhan Yang
- Department of Hepatobiliary Surgery, Fourth Hospital of Hebei Medical University Tumor Hospital of Hebei Province, Shijiazhuang, China
| | - Zhuo Xu
- Department of Hepatobiliary Surgery, Fourth Hospital of Hebei Medical University Tumor Hospital of Hebei Province, Shijiazhuang, China
| | - Zhilei Zhang
- Department of Hepatobiliary Surgery, Fourth Hospital of Hebei Medical University Tumor Hospital of Hebei Province, Shijiazhuang, China
| | - Yuming Jia
- Department of Hepatobiliary Surgery, Fourth Hospital of Hebei Medical University Tumor Hospital of Hebei Province, Shijiazhuang, China
| | - Chong Zhang
- Department of Hepatobiliary Surgery, Fourth Hospital of Hebei Medical University Tumor Hospital of Hebei Province, Shijiazhuang, China
| | - Li Peng
- Department of Hepatobiliary Surgery, Fourth Hospital of Hebei Medical University Tumor Hospital of Hebei Province, Shijiazhuang, China
| |
Collapse
|
37
|
Rodrigues ME, Gomes F, Rodrigues CF. Candida spp./Bacteria Mixed Biofilms. J Fungi (Basel) 2019; 6:jof6010005. [PMID: 31861858 PMCID: PMC7151131 DOI: 10.3390/jof6010005] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/13/2019] [Accepted: 12/14/2019] [Indexed: 12/21/2022] Open
Abstract
The ability to form biofilms is a common feature of microorganisms, such as bacteria or fungi. These consortiums can colonize a variety of surfaces, such as host tissues, dentures, and catheters, resulting in infections highly resistant to drugs, when compared with their planktonic counterparts. This refractory effect is particularly critical in polymicrobial biofilms involving both fungi and bacteria. This review emphasizes Candida spp.-bacteria biofilms, the epidemiology of this community, the challenges in the eradication of such biofilms, and the most relevant treatments.
Collapse
Affiliation(s)
- Maria Elisa Rodrigues
- CEB, Centre of Biological Engineering, LIBRO–Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, 4710-057 Braga, Portugal; (M.E.R.); (F.G.)
| | - Fernanda Gomes
- CEB, Centre of Biological Engineering, LIBRO–Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, 4710-057 Braga, Portugal; (M.E.R.); (F.G.)
| | - Célia F. Rodrigues
- LEPABE–Dep. of Chemical Engineering, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
- Correspondence:
| |
Collapse
|
38
|
Shin DS, Eom YB. Efficacy of zerumbone against dual-species biofilms of Candida albicans and Staphylococcus aureus. Microb Pathog 2019; 137:103768. [DOI: 10.1016/j.micpath.2019.103768] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 09/16/2019] [Accepted: 09/30/2019] [Indexed: 02/06/2023]
|
39
|
Sonthalia S, Goldust M. Innovative Physical Approaches for Onychomycosis: Peeling, Lasers and Beyond. Skin Appendage Disord 2019; 5:197-200. [PMID: 31367597 DOI: 10.1159/000497044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 01/16/2019] [Indexed: 01/19/2023] Open
Affiliation(s)
- Sidharth Sonthalia
- Department of Dermatology and Dermatosurgery, Skinnocence: The Skin Clinic and Research Center, Gurugram, India
| | | |
Collapse
|
40
|
Raorane CJ, Lee JH, Kim YG, Rajasekharan SK, García-Contreras R, Lee J. Antibiofilm and Antivirulence Efficacies of Flavonoids and Curcumin Against Acinetobacter baumannii. Front Microbiol 2019; 10:990. [PMID: 31134028 PMCID: PMC6517519 DOI: 10.3389/fmicb.2019.00990] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 04/18/2019] [Indexed: 01/08/2023] Open
Abstract
Acinetobacter baumannii is well adapted to hospital environments, and the persistence of its chronic infections is mainly due to its ability to form biofilms resistant to conventional antibiotics and host immune systems. Hence, the inhibitions of biofilm formation and virulence characteristics provide other means of addressing infections. In this study, the antibiofilm activities of twelve flavonoids were initially investigated. Three most active flavonoids, namely, fisetin, phloretin, and curcumin, dose-dependently inhibited biofilm formation by a reference A. baumannii strain and by several clinical isolates, including four multidrug-resistant isolates. Furthermore, the antibiofilm activity of curcumin (the most active flavonoid) was greater than that of the well-known biofilm inhibitor gallium nitrate. Curcumin inhibited pellicle formation and the surface motility of A. baumannii. Interestingly, curcumin also showed antibiofilm activity against Candida albicans and mixed cultures of C. albicans and A. baumannii. In silico molecular docking of the biofilm response regulator BfmR showed that the binding efficacy of flavonoids with BfmR was correlated with antibiofilm efficacy. In addition, curcumin treatment diminished A. baumannii virulence in an in vivo Caenorhabditis elegans model without cytotoxicity. The study shows curcumin and other flavonoids have potential for controlling biofilm formation by and the virulence of A. baumannii.
Collapse
Affiliation(s)
| | - Jin-Hyung Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| | - Yong-Guy Kim
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| | | | - Rodolfo García-Contreras
- Department of Microbiology and Parasitology, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| |
Collapse
|
41
|
Negrini TDC, Koo H, Arthur RA. Candida–Bacterial Biofilms and Host–Microbe Interactions in Oral Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1197:119-141. [DOI: 10.1007/978-3-030-28524-1_10] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|