1
|
Barman S, Kurnaz LB, Leighton R, Hossain MW, Decho AW, Tang C. Intrinsic antimicrobial resistance: Molecular biomaterials to combat microbial biofilms and bacterial persisters. Biomaterials 2024; 311:122690. [PMID: 38976935 PMCID: PMC11298303 DOI: 10.1016/j.biomaterials.2024.122690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 05/13/2024] [Accepted: 06/26/2024] [Indexed: 07/10/2024]
Abstract
The escalating rise in antimicrobial resistance (AMR) coupled with a declining arsenal of new antibiotics is imposing serious threats to global public health. A pervasive aspect of many acquired AMR infections is that the pathogenic microorganisms exist as biofilms, which are equipped with superior survival strategies. In addition, persistent and recalcitrant infections are seeded with bacterial persister cells at infection sites. Together, conventional antibiotic therapeutics often fail in the complete treatment of infections associated with bacterial persisters and biofilms. Novel therapeutics have been attempted to tackle AMR, biofilms, and persister-associated complex infections. This review focuses on the progress in designing molecular biomaterials and therapeutics to address acquired and intrinsic AMR, and the fundamental microbiology behind biofilms and persisters. Starting with a brief introduction of AMR basics and approaches to tackling acquired AMR, the emphasis is placed on various biomaterial approaches to combating intrinsic AMR, including (1) semi-synthetic antibiotics; (2) macromolecular or polymeric biomaterials mimicking antimicrobial peptides; (3) adjuvant effects in synergy; (4) nano-therapeutics; (5) nitric oxide-releasing antimicrobials; (6) antimicrobial hydrogels; (7) antimicrobial coatings. Particularly, the structure-activity relationship is elucidated in each category of these biomaterials. Finally, illuminating perspectives are provided for the future design of molecular biomaterials to bypass AMR and cure chronic multi-drug resistant (MDR) infections.
Collapse
Affiliation(s)
- Swagatam Barman
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, United States; Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, 29208, United States
| | - Leman Buzoglu Kurnaz
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, United States
| | - Ryan Leighton
- Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, 29208, United States
| | - Md Waliullah Hossain
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, United States
| | - Alan W Decho
- Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, 29208, United States.
| | - Chuanbing Tang
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, United States.
| |
Collapse
|
2
|
Ratrey P, Bhattacharya S, Coffey L, Thompson D, Hudson SP. Solid lipid nanoparticle formulation maximizes membrane-damaging efficiency of antimicrobial nisin Z peptide. Colloids Surf B Biointerfaces 2024; 245:114255. [PMID: 39303385 DOI: 10.1016/j.colsurfb.2024.114255] [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: 06/13/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Solid lipid nanoparticles (SLNs) can protect and deliver naturally derived or synthetic biologically active products to target sites in vivo. Here, an SLN formulation produces a measured four-fold reduction in inhibitory concentration of an antimicrobial peptide nisin Z against S. aureus as compared to the free peptide, indicating the successful delivery and enhanced effectiveness of the SLN-encapsulated bacteriocin. Spherical SLNs of size 79.47 ± 2.01 nm and zeta potential of -9.8 ± 0.3 mV were synthesised. The lipid formulation maximizes the membrane-damaging mode of action of the free peptide with more and larger-sized pores formed on bacterial membranes treated with nisin Z SLNs as measured from scanning electron microscopy and transmission electron microscopy. Flow cytometry measurements precisely quantified an enhanced dye leakage from pre-labeled bacterial cells when treated with nisin Z-loaded SLNs compared to free peptide. The lipid formulation accelerated cell death by killing all the cells within half an hour compared to the equivalent concentration of free peptide which was not bactericidal. Molecular dynamics simulations revealed a mechanism of SLN facilitated binding to the lipid II bacterial cell wall precursor via enhanced adsorption of nisin Z at the inner bacterial cell membrane bilayer. These findings confirmed the potential of SLN formulations for the effective delivery of therapeutic peptides for next-generation antibiotics that are active at low concentrations with the potential to mitigate antimicrobial resistance.
Collapse
Affiliation(s)
- Poonam Ratrey
- Department of Chemical Sciences, SSPC the SFI Research Centre for Pharmaceuticals, Bernal Institute, University of Limerick, Limerick V94 T9PX, Ireland.
| | - Shayon Bhattacharya
- Department of Physics, SSPC the SFI Research Centre for Pharmaceuticals, Bernal Institute, University of Limerick, Limerick V94 T9PX, Ireland.
| | - Laura Coffey
- Department of Chemical Sciences, SSPC the SFI Research Centre for Pharmaceuticals, Bernal Institute, University of Limerick, Limerick V94 T9PX, Ireland.
| | - Damien Thompson
- Department of Physics, SSPC the SFI Research Centre for Pharmaceuticals, Bernal Institute, University of Limerick, Limerick V94 T9PX, Ireland.
| | - Sarah P Hudson
- Department of Chemical Sciences, SSPC the SFI Research Centre for Pharmaceuticals, Bernal Institute, University of Limerick, Limerick V94 T9PX, Ireland.
| |
Collapse
|
3
|
Patri G, Chatterjee I, Lath H, Sinha Y, Agrawal P, Majee N, Bansal S. A Comparative Analysis of the Antimicrobial Efficacy of Nisin in Different Vehicles Against Enterococcus faecalis: An In Vitro Study. Cureus 2024; 16:e66204. [PMID: 39233962 PMCID: PMC11373878 DOI: 10.7759/cureus.66204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
Aim To evaluate and compare the antimicrobial efficacy of nisin in different carriers against Enterococcus faecalis. Materials and methods Test materials were divided into four groups of five samples each as follows: group 1 = nisin + 17% ethylenediaminetetraacetic acid (EDTA); group 2 = nisin + 2% chitosan; group 3 = nisin + 2% chlorhexidine; group 4 = nisin + distilled water (control). The antimicrobial effectiveness was assessed using the direct contact method, where a standardized E. faecalis suspension was applied to the test materials. Optical density (OD) was assessed using enzyme-linked immunosorbent assay (ELISA) at the end of days one and seven. Data were analyzed using ANOVA and Tukey's post hoc analysis. The level of significance was set at p < 0.05. Results On day one, there was a significant difference in the mean OD values (p < 0.001) with group 3 showing the highest, followed by groups 1, 2, and 4. On day seven, all groups demonstrated antibacterial activity (group 1 > group 3 > group 4 > group 2) but the differences were not statistically significant (p = 0.393). Intragroup analysis showed a decrease in the OD values from day one to day seven, the difference of which was not significant in all groups except group 1, which showed a significant difference (p = 0.035). Conclusion The antibacterial efficacy of nisin was synergistically enhanced with the addition of 17% EDTA and 2% chlorhexidine over seven days against E. faecalis.
Collapse
Affiliation(s)
- Gaurav Patri
- Department of Conservative Dentistry and Endodontics, Kalinga Institute of Dental Sciences, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, IND
| | - Ishika Chatterjee
- Department of Conservative Dentistry and Endodontics, Kalinga Institute of Dental Sciences, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, IND
| | - Harshita Lath
- Department of Conservative Dentistry and Endodontics, Kalinga Institute of Dental Sciences, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, IND
| | - Yash Sinha
- Department of Conservative Dentistry and Endodontics, Kalinga Institute of Dental Sciences, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, IND
| | - Pratik Agrawal
- Department of Conservative Dentistry and Endodontics, Kalinga Institute of Dental Sciences, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, IND
| | - Neelanjana Majee
- Department of Conservative Dentistry and Endodontics, Kalinga Institute of Dental Sciences, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, IND
| | - Sonali Bansal
- Department of Conservative Dentistry and Endodontics, Kalinga Institute of Dental Sciences, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, IND
| |
Collapse
|
4
|
Kong X, Vishwanath V, Neelakantan P, Ye Z. Harnessing antimicrobial peptides in endodontics. Int Endod J 2024; 57:815-840. [PMID: 38441321 DOI: 10.1111/iej.14043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 06/13/2024]
Abstract
Endodontic therapy includes various procedures such as vital pulp therapy, root canal treatment and retreatment, surgical endodontic treatment and regenerative endodontic procedures. Disinfection and tissue repair are crucial for the success of these therapies, necessitating the development of therapeutics that can effectively target microbiota, eliminate biofilms, modulate inflammation and promote tissue repair. However, no current endodontic agents can achieve these goals. Antimicrobial peptides (AMPs), which are sequences of amino acids, have gained attention due to their unique advantages, including reduced susceptibility to drug resistance, broad-spectrum antibacterial properties and the ability to modulate the immune response of the organism effectively. This review systematically discusses the structure, mechanisms of action, novel designs and limitations of AMPs. Additionally, it highlights the efforts made by researchers to overcome peptide shortcomings and emphasizes the potential applications of AMPs in endodontic treatments.
Collapse
Affiliation(s)
- Xinzi Kong
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong S.A.R., China
| | - Vijetha Vishwanath
- Division of Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong S.A.R., China
| | - Prasanna Neelakantan
- Department of Endodontics, University of the Pacific Arthur A. Dugoni School of Dentistry, San Francisco, California, USA
| | - Zhou Ye
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong S.A.R., China
| |
Collapse
|
5
|
Pereira ACC, Aguiar APS, Barbosa VL, Régis JR, Miyazima EM, Araujo LMP, Dantas LO, Mayer MPA, Andrade FB, Karygianni L, Pinheiro ET. Enhancing Antibiotic Efficacy in Regenerative Endodontics by Improving Biofilm Susceptibility. J Endod 2024; 50:962-965. [PMID: 38615826 DOI: 10.1016/j.joen.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 04/16/2024]
Abstract
INTRODUCTION Various strategies have been researched to enhance the susceptibility of biofilms, given their tolerance to antibiotics. This study evaluated the effect of the anti-microbial peptide nisin in association with antibiotics used in regenerative endodontics, exploring different treatment times and biofilm growth conditions. METHODS A mixture of 10 bacterial species was cultivated on dentin specimens anaerobically for 21 days. Biofilms were treated with 1 mL of high-purity nisin Z (nisin ZP, 200 μg/mL) and a triple antibiotic mixture (TAP: ciprofloxacin + metronidazole + minocycline, 5 mg/mL), alone or in combination. The effectiveness of antimicrobial agents was assessed after 1 and 7 days. During the 7-day period, biofilms were treated under 2 conditions: a single dose in a nutrient-depleted setting (ie, no replenishment of growth medium) and multiple doses in a nutrient-rich environment (ie, renewal of medium and antimicrobial agents every 48 h). After treatments, biofilm cells were dispersed, and total colony-forming units were counted. RESULTS After 1 d-treatment, nisin ZP + TAP resulted in 2-log cell reduction compared to TAP alone (P < .05). After 7 d-treatment with a single dose, nisin ZP + TAP and TAP reduced bacteria to nonculturable levels (P < .05), whereas repeated antimicrobial doses did not eliminate bacteria in a nutrient-rich environment. No bacterial reduction was observed with nisin ZP alone in any treatment time. CONCLUSIONS The additional use of nisin improved the TAP activity only after a short exposure time. Longer exposure to TAP or nisin + TAP in a nutrient-deprived environment effectively eliminated biofilms.
Collapse
Affiliation(s)
- Ana C C Pereira
- Department of Dentistry, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Alana P S Aguiar
- Department of Dentistry, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Vinícius L Barbosa
- Department of Dentistry, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Júlia R Régis
- Department of Dentistry, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Erica Mina Miyazima
- Department of Dentistry, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Leticia M P Araujo
- Department of Biomaterials and Oral Biology, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Larissa O Dantas
- Department of Dentistry, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Marcia P A Mayer
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Flaviana B Andrade
- Department of Operative Dentistry, Endodontics and Dental Materials, Bauru School of Dentistry, University of São Paulo, Bauru, São Paulo, Brazil
| | - Lamprini Karygianni
- Clinic of Conservative and Preventive Dentistry, Center of Dental Medicine, University of Zurich, Zürich, Switzerland
| | - Ericka T Pinheiro
- Department of Dentistry, School of Dentistry, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
6
|
Charest AM, Reed E, Bozorgzadeh S, Hernandez L, Getsey NV, Smith L, Galperina A, Beauregard HE, Charest HA, Mitchell M, Riley MA. Nisin Inhibition of Gram-Negative Bacteria. Microorganisms 2024; 12:1230. [PMID: 38930612 PMCID: PMC11205666 DOI: 10.3390/microorganisms12061230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Aims: This study investigates the activity of the broad-spectrum bacteriocin nisin against a large panel of Gram-negative bacterial isolates, including relevant plant, animal, and human pathogens. The aim is to generate supportive evidence towards the use/inclusion of bacteriocin-based therapeutics and open avenues for their continued development. Methods and Results: Nisin inhibitory activity was screened against a panel of 575 strains of Gram-negative bacteria, encompassing 17 genera. Nisin inhibition was observed in 309 out of 575 strains, challenging the prevailing belief that nisin lacks effectiveness against Gram-negative bacteria. The genera Acinetobacter, Helicobacter, Erwinia, and Xanthomonas exhibited particularly high nisin sensitivity. Conclusions: The findings of this study highlight the promising potential of nisin as a therapeutic agent for several key Gram-negative plant, animal, and human pathogens. These results challenge the prevailing notion that nisin is less effective or ineffective against Gram-negative pathogens when compared to Gram-positive pathogens and support future pursuits of nisin as a complementary therapy to existing antibiotics. Significance and Impact of Study: This research supports further exploration of nisin as a promising therapeutic agent for numerous human, animal, and plant health applications, offering a complementary tool for infection control in the face of multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Adam M. Charest
- Department of Biology, University of Massachusetts, Amherst, MA 01002, USA; (A.M.C.); (E.R.); (S.B.); (L.H.); (N.V.G.); (L.S.); (A.G.); (H.E.B.); (H.A.C.)
| | - Ethan Reed
- Department of Biology, University of Massachusetts, Amherst, MA 01002, USA; (A.M.C.); (E.R.); (S.B.); (L.H.); (N.V.G.); (L.S.); (A.G.); (H.E.B.); (H.A.C.)
| | - Samantha Bozorgzadeh
- Department of Biology, University of Massachusetts, Amherst, MA 01002, USA; (A.M.C.); (E.R.); (S.B.); (L.H.); (N.V.G.); (L.S.); (A.G.); (H.E.B.); (H.A.C.)
| | - Lorenzo Hernandez
- Department of Biology, University of Massachusetts, Amherst, MA 01002, USA; (A.M.C.); (E.R.); (S.B.); (L.H.); (N.V.G.); (L.S.); (A.G.); (H.E.B.); (H.A.C.)
| | - Natalie V. Getsey
- Department of Biology, University of Massachusetts, Amherst, MA 01002, USA; (A.M.C.); (E.R.); (S.B.); (L.H.); (N.V.G.); (L.S.); (A.G.); (H.E.B.); (H.A.C.)
| | - Liam Smith
- Department of Biology, University of Massachusetts, Amherst, MA 01002, USA; (A.M.C.); (E.R.); (S.B.); (L.H.); (N.V.G.); (L.S.); (A.G.); (H.E.B.); (H.A.C.)
| | - Anastasia Galperina
- Department of Biology, University of Massachusetts, Amherst, MA 01002, USA; (A.M.C.); (E.R.); (S.B.); (L.H.); (N.V.G.); (L.S.); (A.G.); (H.E.B.); (H.A.C.)
| | - Hadley E. Beauregard
- Department of Biology, University of Massachusetts, Amherst, MA 01002, USA; (A.M.C.); (E.R.); (S.B.); (L.H.); (N.V.G.); (L.S.); (A.G.); (H.E.B.); (H.A.C.)
| | - Hailey A. Charest
- Department of Biology, University of Massachusetts, Amherst, MA 01002, USA; (A.M.C.); (E.R.); (S.B.); (L.H.); (N.V.G.); (L.S.); (A.G.); (H.E.B.); (H.A.C.)
| | - Mathew Mitchell
- Organicin Scientific, 240 Thatcher Road, Amherst, MA 01003, USA;
| | - Margaret A. Riley
- Department of Biology, University of Massachusetts, Amherst, MA 01002, USA; (A.M.C.); (E.R.); (S.B.); (L.H.); (N.V.G.); (L.S.); (A.G.); (H.E.B.); (H.A.C.)
- Organicin Scientific, 240 Thatcher Road, Amherst, MA 01003, USA;
| |
Collapse
|
7
|
Kharga K, Jha S, Vishwakarma T, Kumar L. Current developments and prospects of the antibiotic delivery systems. Crit Rev Microbiol 2024:1-40. [PMID: 38425122 DOI: 10.1080/1040841x.2024.2321480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 02/16/2024] [Indexed: 03/02/2024]
Abstract
Antibiotics have remained the cornerstone for the treatment of bacterial infections ever since their discovery in the twentieth century. The uproar over antibiotic resistance among bacteria arising from genome plasticity and biofilm development has rendered current antibiotic therapies ineffective, urging the development of innovative therapeutic approaches. The development of antibiotic resistance among bacteria has further heightened the clinical failure of antibiotic therapy, which is often linked to its low bioavailability, side effects, and poor penetration and accumulation at the site of infection. In this review, we highlight the potential use of siderophores, antibodies, cell-penetrating peptides, antimicrobial peptides, bacteriophages, and nanoparticles to smuggle antibiotics across impermeable biological membranes to achieve therapeutically relevant concentrations of antibiotics and combat antimicrobial resistance (AMR). We will discuss the general mechanisms via which each delivery system functions and how it can be tailored to deliver antibiotics against the paradigm of mechanisms underlying antibiotic resistance.
Collapse
Affiliation(s)
- Kusum Kharga
- School of Biotechnology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Himachal Pradesh, India
| | - Shubhang Jha
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Himachal Pradesh, India
| | - Tanvi Vishwakarma
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Himachal Pradesh, India
| | - Lokender Kumar
- School of Biotechnology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Himachal Pradesh, India
| |
Collapse
|
8
|
Mariconda A, Iacopetta D, Sirignano M, Ceramella J, D'Amato A, Marra M, Pellegrino M, Sinicropi MS, Aquaro S, Longo P. Silver and Gold Complexes with NHC-Ligands Derived from Caffeine: Catalytic and Pharmacological Activity. Int J Mol Sci 2024; 25:2599. [PMID: 38473851 DOI: 10.3390/ijms25052599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
N-heterocyclic carbene (NHC) silver(I) and gold(I) complexes have found different applications in various research fields, as in medicinal chemistry for their antiproliferative, anticancer, and antibacterial activity, and in chemistry as innovative and effective catalysts. The possibility of modulating the physicochemical properties, by acting on their ligands and substituents, makes them versatile tools for the development of novel metal-based compounds, mostly as anticancer compounds. As it is known, chemotherapy is commonly adopted for the clinical treatment of different cancers, even though its efficacy is hampered by several factors. Thus, the development of more effective and less toxic drugs is still an urgent need. Herein, we reported the synthesis and characterization of new silver(I) and gold(I) complexes stabilized by caffeine-derived NHC ligands, together with their biological and catalytic activities. Our data highlight the interesting properties of this series as effective catalysts in A3-coupling and hydroamination reactions and as promising anticancer, anti-inflammatory, and antioxidant agents. The ability of these complexes in regulating different pathological aspects, and often co-promoting causes, of cancer makes them ideal leads to be further structurally functionalized and investigated.
Collapse
Affiliation(s)
- Annaluisa Mariconda
- Department of Science, University of Basilicata, Via dell'Ateneo Lucano 10, 85100 Potenza, Italy
| | - Domenico Iacopetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, 87036 Arcavacata di Rende, Italy
| | - Marco Sirignano
- Department of Chemistry and Biology "A. Zambelli", University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
| | - Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, 87036 Arcavacata di Rende, Italy
| | - Assunta D'Amato
- Department of Chemistry and Biology "A. Zambelli", University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
| | - Maria Marra
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, 87036 Arcavacata di Rende, Italy
| | - Michele Pellegrino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, 87036 Arcavacata di Rende, Italy
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, 87036 Arcavacata di Rende, Italy
| | - Stefano Aquaro
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, 87036 Arcavacata di Rende, Italy
| | - Pasquale Longo
- Department of Chemistry and Biology "A. Zambelli", University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
| |
Collapse
|
9
|
Sharafi T, Ghaemi EA, Rafiee M, Ardebili A. Combination antimicrobial therapy: in vitro synergistic effect of anti-staphylococcal drug oxacillin with antimicrobial peptide nisin against Staphylococcus epidermidis clinical isolates and Staphylococcus aureus biofilms. Ann Clin Microbiol Antimicrob 2024; 23:7. [PMID: 38245727 PMCID: PMC10800071 DOI: 10.1186/s12941-024-00667-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/04/2024] [Indexed: 01/22/2024] Open
Abstract
The ability of Staphylococcus epidermidis and S. aureus to form strong biofilm on plastic devices makes them the major pathogens associated with device-related infections (DRIs). Biofilm-embedded bacteria are more resistant to antibiotics, making biofilm infections very difficult to effectively treat. Here, we evaluate the in vitro activities of anti-staphylococcal drug oxacillin and antimicrobial peptide nisin, alone and in combination, against methicillin-resistant S. epidermidis (MRSE) clinical isolates and the methicillin-resistant S. aureus ATCC 43,300. The minimum inhibitory concentrations (MIC) and minimum biofilm eradication concentrations (MBEC) of oxacillin and nisin were determined using the microbroth dilution method. The anti-biofilm activities of oxacillin and nisin, alone or in combination, were evaluated. In addition, the effects of antimicrobial agents on the expression of icaA gene were examined by quantitative real-time PCR. MIC values for oxacillin and nisin ranged 4-8 µg/mL and 64-128 µg/mL, respectively. Oxacillin and nisin reduced biofilm biomass in all bacteria in a dose-dependent manner and this inhibitory effect was enhanced with combinatorial treatment. MBEC ranges for oxacillin and nisin were 2048-8192 µg/mL and 2048-4096 µg/mL, respectively. The addition of nisin significantly decreased the oxacillin MBECs from 8- to 32-fold in all bacteria. At the 1× MIC and 1/2× MIC, both oxacillin and nisin decreased significantly the expression of icaA gene in comparison with untreated control. When two antimicrobial agents were combined at 1/2× MIC concentration, the expression of icaA were significantly lower than when were used alone. Nisin/conventional oxacillin combination showed considerable anti-biofilm effects, including inhibition of biofilm formation, eradication of mature biofilm, and down-regulation of biofilm-related genes, proposing its applications for treating or preventing staphylococcal biofilm-associated infections, including device-related infections.
Collapse
Affiliation(s)
- Toktam Sharafi
- Infectious Disease Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ezzat Allah Ghaemi
- Infectious Disease Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Maryam Rafiee
- Infectious Disease Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Abdollah Ardebili
- Infectious Disease Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran.
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
10
|
Saraswat J, Firoz A, Kamli MR, Patel R. Improved Antibacterial Activity of Peptide Nisin with Pyrrole-Based Ionic Liquids Having Bis(trifluoromethylsulfonyl)imide as a Counterion: A Synergistic Approach to Combat Bacterial Infections. ACS OMEGA 2024; 9:2758-2769. [PMID: 38250392 PMCID: PMC10795159 DOI: 10.1021/acsomega.3c07824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/02/2023] [Accepted: 12/12/2023] [Indexed: 01/23/2024]
Abstract
Bacterial resistance against antimicrobial drugs is a forthcoming threat to the prevention and treatment of developing bacterial infections. Hence, the development of new antimicrobial therapy or therapeutic drugs is desperately needed. A combination of antibiotics exhibits synergistic antibacterial effects. As the combination approach of antibiotics has always shown better results against pathogens compared to monotherapy with an antibiotic, we focused on creating a new combination that may reduce the chances of strains attaining resistance, consequently lowering the toxicity factor associated with the consumption of high amounts of antibiotics. Nisin, a food preservative and potential antibiotic, shows antibacterial activity against Gram-positive strains. Since the past decade, ionic liquids (ILs) have proven to be an important class of potential antibacterial agents. In our study, we studied the effect of pyrrolidinium-based ILs and arrived at a noncovalent conjugate formed by combining nisin with ILs. The conjugates were tested against a couple of clinically relevant microorganisms, namely, Escherichia coli and Staphylococcus aureus. We reached a novel discovery that the combination of sodium/iodide symporter (NIS) and IL exhibited inhibitory effects against Gram-negative bacteria, which was not observed with NIS alone. The results showed remarkable improvement in the minimum inhibitory concentration (MIC) value of NIS in the presence of ILs targeted against both microorganisms. Further, flow cytometry and confocal microscopy results revealed the membrane disruption efficiency of the best combination obtained, leading to cell death. Additionally, the complexation of nisin and ILs was studied using various techniques, such as surface tension, dynamic light scattering, absorption spectroscopy, and molecular docking.
Collapse
Affiliation(s)
- Juhi Saraswat
- Biophysical
Chemistry Laboratory, Centre for Interdisciplinary Research in Basic
Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Ahmad Firoz
- Department
of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Princess
Dr. Najla Bint Saud Al-Saud Centre for Excellence Research in Biotechnology, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Majid Rasool Kamli
- Department
of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Rajan Patel
- Biophysical
Chemistry Laboratory, Centre for Interdisciplinary Research in Basic
Sciences, Jamia Millia Islamia, New Delhi 110025, India
| |
Collapse
|
11
|
Sefali S, Y N. Efficacy of oritavancin and nisin alone and their combination against vancomycin resistant enterococci strains in hospitalized patients in Turkiye. Indian J Med Microbiol 2024; 47:100489. [PMID: 37890415 DOI: 10.1016/j.ijmmb.2023.100489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 09/27/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023]
Abstract
PURPOSE Vancomycin-Resistant Enterococci (VREs) have emerged and become a problem that threatens the health of hospitalized patients. VRE can cause different serious infections of the urinary tract, the bloodstream, wound and other body sites. VREs are resistant to multiple antibiotics and treatment options are very limited. We aimed to investigate the efficacy of oritavancin and nisin alone and their combination against VRE strains. METHODS VRE isolates from rectal swabs of hospitalized patients were identified by conventional and commercial methods. The minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of oritavancin and nisin against VRE strains were determined. The synergistic effect of both agent combinations was examined by the Checkerboard test. RESULTS All VRE strains were identifined as Enterococcus faecium. The MIC value of oritavancin was found in the range of 0.015-0.24 μg/mL; in which 48 strains were susceptible (≤0.12 μg/mL) and two strains were resistant (>0.12 μg/mL). The MBC of oritavancin was determined in the range of 0.06-3.84 μg/mL. The MIC of nisin was found in the range of 12.5-100 μg/mL; in which 32 strains were susceptible (≤50 μg/mL) and 18 strains were resistant (>50 μg/mL). MBC of nisin was determined in the range of 25-800 μg/mL. Two oritavancin resistant strains were displayed indifference effect, whereas from 18 nisin resistant strains, 11 showed indifference, and seven displayed synergistic effect. Thirty-eight out of 48 strains which were sensitive to oritavancin showed indifference and 10 revealed synergistic effect, whereas 29 of 32 strains which were sensitive to nisin showed indifference and three had synergistic effect. CONCLUSIONS A synergistic combination of oritavansin and nisin was detected in 20 strains (40%), Our study is the first study in Turkiye.
Collapse
Affiliation(s)
- Sena Sefali
- Istanbul Faculty of Medicine, Department of Medical Microbiology, Istanbul University, 34390 Capa, Istanbul, Turkiye.
| | - Nakipoglu Y
- Istanbul Faculty of Medicine, Department of Medical Microbiology, Istanbul University, 34390 Capa, Istanbul, Turkiye.
| |
Collapse
|
12
|
Baindara P, Mandal SM. Gut-Antimicrobial Peptides: Synergistic Co-Evolution with Antibiotics to Combat Multi-Antibiotic Resistance. Antibiotics (Basel) 2023; 12:1732. [PMID: 38136766 PMCID: PMC10740742 DOI: 10.3390/antibiotics12121732] [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: 11/29/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Due to huge diversity and dynamic competition, the human gut microbiome produces a diverse array of antimicrobial peptides (AMPs) that play an important role in human health. The gut microbiome has an important role in maintaining gut homeostasis by the AMPs and by interacting with other human organs via established connections such as the gut-lung, and gut-brain axis. Additionally, gut AMPs play a synergistic role with other gut microbiota and antimicrobials to maintain gut homeostasis by fighting against multi-antibiotic resistance (MAR) bacteria. Further, conventional antibiotics intake creates a synergistic evolutionary pressure for gut AMPs, where antibiotics and gut AMPs fight synergistically against MAR. Overall, gut AMPs are evolving under a complex and highly synergistic co-evolutionary pressure created by the various interactions between gut microbiota, gut AMPs, and antibiotics; however, the complete mechanism is not well understood. The current review explores the synergistic action of gut AMPs and antibiotics along with possibilities to fight against MAR bacteria.
Collapse
Affiliation(s)
- Piyush Baindara
- Radiation Oncology, NextGen Precision Health, School of Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Santi M. Mandal
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India;
| |
Collapse
|
13
|
Ardila CM, Bedoya-García JA, González-Arroyave D. Antimicrobial resistance in patients with endodontic infections: A systematic scoping review of observational studies. AUST ENDOD J 2023; 49:386-395. [PMID: 36054305 DOI: 10.1111/aej.12680] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/31/2022] [Accepted: 08/15/2022] [Indexed: 12/29/2022]
Abstract
The aim of this study was to assess the prevalence and proportions of antimicrobial-resistant species in patients with endodontic infections. A systematic scoping review of scientific evidence was accomplished involving different databases. Nine investigations were selected including 651 patients. Enterococcus faecalis was resistant to tetracycline (30%-70%), clindamycin (100%), erythromycin (10%-20%), ampicillin (9%) and azithromycin (60%). On the contrary, Prevotella spp., Fusobacterium spp., Peptostreptococcus spp. and Streptococcus spp. were resistant to penicillin, tetracycline, doxycycline, ciprofloxacin, amoxicillin, erythromycin, metronidazole and clindamycin in different proportions. Fusobacterium nucleatum showed high resistance to amoxicillin, amoxicillin plus clavulanate and erythromycin. Prevotella oralis presented a predisposition to augment its resistance to clindamycin over time. Tanerella forsythia exhibited resistance to ciprofloxacin and rifampicin. Lactococcus lactis presented robust resistance to cephalosporins, metronidazole, penicillin, amoxicillin and amoxicillin-clavulanic acid. It was observed high levels of resistance to antimicrobials that have been utilised in the local and systemic treatment of oral cavity infections.
Collapse
Affiliation(s)
- Carlos-M Ardila
- Universidad de Antioquia U de A, Medellín, Colombia
- Biomedical Stomatology Research Group, Universidad de Antioquia U de A, Medellín, Colombia
| | | | | |
Collapse
|
14
|
Combating food spoilage and pathogenic microbes via bacteriocins: A natural and eco-friendly substitute to antibiotics. Food Control 2023. [DOI: 10.1016/j.foodcont.2023.109710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
|
15
|
Yazıcı BC, Bakhedda N, Akçelik N. Effect of nisin and p-coumaric acid on autoinducer-2 activity, biofilm formation, and sprE expression of Enterococcus faecalis. Braz J Microbiol 2023; 54:601-608. [PMID: 36930448 PMCID: PMC10234943 DOI: 10.1007/s42770-023-00946-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 03/02/2023] [Indexed: 03/18/2023] Open
Abstract
Quorum sensing (QS) is an inter- and intracellular communication mechanism that regulates gene expression in response to population size. Autoinducer-2 (AI-2) signaling is a QS signaling molecule common to both Gram-negative and Gram-positive bacteria. Enterococcus faecalis is one of the leading causes of nosocomial infections worldwide. There has been an increasing interest in controlling infectious diseases through targeting the QS mechanism using natural compounds. This study aimed to investigate the effect of nisin and p-coumaric acid (pCA), on biofilm formation and AI-2 signaling in E. faecalis. Their effect on the expression of the QS-regulated virulence encoding gene sprE was also investigated. Nisin exhibited a MIC ranging from 0.25 to 0.5 mg/mL, while the MIC of pCA was 1 mg/mL. The luminescence-based response of the reporter strain Vibrio harveyi BB170 was used to determine AI-2 activity in E. faecalis strains. Nisin was not effective in inhibiting AI-2 activity, while pCA reduced AI-2 activity by ≥ 60%. Moreover, pCA and nisin combination showed higher inhibitory effect on biofilm formation of E. faecalis, compared to the treatment of pCA or nisin alone. qRT-PCR analysis showed that nisin alone and the combination of nisin and pCA, at their MIC values, led to a 32.78- and 40.22-fold decrease in sprE gene expression, respectively, while pCA alone did not have a significant effect. Considering the demand to explore new therapeutic avenues for infectious bacteria, this study was the first to report that pCA can act like a quorum sensing inhibitor (QSI) against AI-2 signaling in E. faecalis.
Collapse
Affiliation(s)
| | | | - Nefise Akçelik
- Biotechnology Institute, Ankara University, Ankara, Turkey.
| |
Collapse
|
16
|
Isolation, Characterization, and Effect on Biofilm Formation of Bacteriocin Produced by Lactococcus lactis F01 Isolated from Cyprinus carpio and Application for Biopreservation of Fish Sausage. BIOMED RESEARCH INTERNATIONAL 2022; 2022:8437926. [DOI: 10.1155/2022/8437926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 10/18/2022] [Accepted: 11/03/2022] [Indexed: 11/23/2022]
Abstract
The aim of this work was the screening of bacteriocin-producing LABs isolated from fish, the selection of promising/prominent strain(s), the characterization of the bacteriocin produced, and the evaluation of its potential to be used as biopreservative(s). Amplification and sequencing of the 16S rRNA gene of the bacteriocin-producing strain was performed. Then a partial purification of the produced bacteriocin, using a combination of ammonium sulfate and chloroform-methanol precipitation, was done. Its molecular weight was determined by SDS-PAGE. In addition, the action spectrum, the hemolysis test, and its ability to inhibit biofilm formation were analyzed. A total of 88 isolates of lactic acid bacteria (LAB) including one bacteriocin producer, which was identified as Lactococcus lactis F01, were collected. The bacteriocin was partially purified with an estimated yield of 40%. Regarding the SDS-PAGE profile, the secreted bacteriocin has molecular weight of about 3.5 kDa and was identified as class I bacteriocin. The antimicrobial test showed that the bacteriocin inhibits pathogenic and/or spoilage bacteria, 10 Gram-positive and 16 Gram-negative bacterial species. Moreover, it can inhibit biofilm formation from 1.3% (Escherichia coli) to 63.92% (Pseudomonas aeruginosa ATCC15692) depending on the strain. The hemolytic activity of novel bacteriocin was observed at the concentration of 10 μg/ml of bacteriocin crude extract, which was
. In addition, it exhibited good thermal and pH stability with retained antibacterial activity of 85.25% after treatment at 121°C for 20 min, as well as at a pH range between 2.0 and 10.0. Moreover, this bacteriocin showed the ability to inhibit the growth of bacterial culture load in fish sausage stored at 8°C for 28 days. Considering the results obtained, bacteriocin could be potentially exploited as an alternative to chemical preservatives or as a substitute for antibiotics.
Collapse
|
17
|
Desmond A, O’Halloran F, Cotter L, Hill C, Field D. Bioengineered Nisin A Derivatives Display Enhanced Activity against Clinical Neonatal Pathogens. Antibiotics (Basel) 2022; 11:1516. [PMID: 36358171 PMCID: PMC9686653 DOI: 10.3390/antibiotics11111516] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 08/27/2023] Open
Abstract
Neonatal infection is a significant cause of mortality and morbidity in infants. The global incidence of multi-drug resistance continues to rise among neonatal pathogens, indicating a need for alternative treatment strategies. Nisin is an antimicrobial peptide that exhibits broad-spectrum activity against a wide variety of clinical pathogens and can be used in combination with antibiotics to improve their effectiveness. This study examined the activity of nisin and bioengineered derivatives against multi-drug resistant Streptococcus agalactiae and Staphylococcus capitis isolates and investigated the potential synergy between nisin peptides and selected antibiotics. Whole genome sequence analysis of the strains revealed the presence of multi-drug resistant determinants, e.g., macrolide, tetracycline, β-lactam, aminoglycoside, while the S. agalactiae strains all possessed both nsr and nsrFP genes and the S. capitis strains were found to encode the nsr gene alone. Deferred antagonism assays demonstrated that nisin PV had improved antimicrobial activity against all strains tested (n = 10). The enhanced specific activity of this peptide was confirmed using minimum inhibitory concentrations (MIC) (0-4-fold lower MIC for nisin PV) and broth-based survival assays. Combinations of nisin peptides with antibiotics were assessed for enhanced antimicrobial activity using growth and time-kill assays and revealed a more effective nisin PV/ampicillin combination against one S. capitis strain while a nisin A/erythromycin combination displayed a synergistic effect against one S. agalactiae strain. The findings of this study suggest that nisin derivatives alone and in combination with antibiotics have potential as alternative antimicrobial strategies to target neonatal pathogens.
Collapse
Affiliation(s)
- Anna Desmond
- Department of Biological Sciences, Munster Technological University, T12 P928 Cork, Ireland
| | - Fiona O’Halloran
- Department of Biological Sciences, Munster Technological University, T12 P928 Cork, Ireland
| | - Lesley Cotter
- Department of Biological Sciences, Munster Technological University, T12 P928 Cork, Ireland
| | - Colin Hill
- School of Microbiology, University College Cork, T12 YN60 Cork, Ireland
- APC Microbiome Ireland, University College Cork, T12 YN60 Cork, Ireland
| | - Des Field
- School of Microbiology, University College Cork, T12 YN60 Cork, Ireland
- APC Microbiome Ireland, University College Cork, T12 YN60 Cork, Ireland
| |
Collapse
|
18
|
The Effect of Indole-3-Lactic Acid from Lactiplantibacillus plantarum ZJ316 on Human Intestinal Microbiota In Vitro. Foods 2022; 11:3302. [PMCID: PMC9601829 DOI: 10.3390/foods11203302] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Microbiota-derived tryptophan metabolites are essential signals for maintaining gut homeostasis, yet the potential contribution to modulating gut microbiota has been rarely investigated. In this study, Lactiplantibacillus plantarum ZJ316 (CCTCC No. M 208077) with a high production (43.14 μg/mL) of indole-3-lactic acid (ILA) was screened. ILA with 99.00% purity was prepared by macroporous resin, Sephadex G–25 and reversed-phase high-performance liquid chromatography. Purified ILA can effectively inhibit foodborne pathogens such as Salmonella spp., Staphylococcus spp., Escherichia coli and Listeria monocytogenes. In an in vitro model of the human gut microbiota, a medium-dose ILA (172 mg/L) intervention increased the average relative abundance of phyla Firmicutes and Bacteroidota by 9.27% and 15.38%, respectively, while Proteobacteria decreased by 14.36% after 24 h fermentation. At the genus level, the relative abundance of Bifidobacterium and Faecalibacterium significantly increased to 5.36 ± 2.31% and 2.19 ± 0.77% (p < 0.01), respectively. Escherichia and Phascolarctobacterium decreased to 16.41 ± 4.81% (p < 0.05) and 2.84 ± 1.02% (p < 0.05), respectively. Intestinal short-chain fatty acids, especially butyric acid, were significantly increased (2.98 ± 0.72 µmol/mL, p < 0.05) and positively correlated with Oscillospira and Collinsella. Overall, ILA has the potential to regulate the gut microbiota, and an in-depth understanding of the relationship between tryptophan metabolites and gut microbiota is needed in the future.
Collapse
|
19
|
NISIN and gilaburu (Viburnum opulus L.) combination is a cost-effective way to control foodborne Staphylococcus aureus. Food Control 2022. [DOI: 10.1016/j.foodcont.2022.109213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
20
|
Jarkhi A, Lee AHC, Sun Z, Hu M, Neelakantan P, Li X, Zhang C. Antimicrobial Effects of L-Chg10-Teixobactin against Enterococcus faecalis In Vitro. Microorganisms 2022; 10:microorganisms10061099. [PMID: 35744616 PMCID: PMC9228388 DOI: 10.3390/microorganisms10061099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/22/2022] [Accepted: 05/22/2022] [Indexed: 11/18/2022] Open
Abstract
Objective: Teixobactin and its analogues are a new class of antibiotics that have no detectable bacterial resistance. This study was designed to determine the antibacterial and antibiofilm activities of a novel teixobactin analogue, L-Chg10-teixobactin, against two strains of Enterococcus faecalis (E. faecalis). Materials and Methods: The efficacy of L-Chg10-teixobactin against two strains of E. faecalis (ATCC 29212 and 47077) was determined using Clinical and Laboratory Standards Institute methods. L-Chg10-teixobactin was prepared at a stock concentration of 1 mg/mL in 5% DMSO. The minimum inhibitory concentration (MIC) was calculated using a two-fold serial broth dilution method, utilizing a 96-well plate. The minimum bactericidal concentration (MBC) was determined by plating the bacteria onto agar to define the concentration that resulted in 99.9% of bacterial death. Ampicillin was used as the control. The effect of L-Chg10-teixobactin on the inhibition of ATCC 47077 strain biofilm formation was determined by measuring the minimum biofilm inhibitory concentration (MBIC) using the safranin assay, while the eradication of the preformed biofilm was determined by measuring the minimum biofilm eradication concentration (MBEC) using the XTT assay. For nonlinear data, the log dose–response curve was plotted to calculate the optimum concentration using Excel (version 16.51, Microsoft® excel. 2021, Microsoft Corporation, Reymond, WA, USA). The data are presented as mean ± standard deviation (SD). Results: The MIC and MBC values of L-Chg10-teixobactin against both strains of E. faecalis were 0.8 μg/mL. The MIC of ampicillin was 1.25 μg/mL for ATCC 29212 and ranged from 1.25 to 5 μg/mL for ATCC 47077. The MBC of ampicillin for ATCC 29212 and ATCC 47077 was 10 and 20 μg/mL, respectively. The MIC and MBC of ampicillin were much higher compared with those of L-Chg10-teixobactin. The MBEC80 of L-Chg10-teixobactin was 4.60 μg/mL for ATCC 47077, which was much lower than that of ampicillin (20 μg/mL). Conclusions:L-Chg10-teixobactin demonstrated potent antibacterial and antibiofilm effects against E. faecalis, suggesting its potential role an effective antibacterial and antibiofilm agent in endodontic treatment.
Collapse
|
21
|
Hawas S, Verderosa AD, Totsika M. Combination Therapies for Biofilm Inhibition and Eradication: A Comparative Review of Laboratory and Preclinical Studies. Front Cell Infect Microbiol 2022; 12:850030. [PMID: 35281447 PMCID: PMC8915430 DOI: 10.3389/fcimb.2022.850030] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/04/2022] [Indexed: 12/26/2022] Open
Abstract
Microbial biofilms are becoming increasingly difficult to treat in the medical setting due to their intrinsic resistance to antibiotics. To combat this, several biofilm dispersal agents are currently being developed as treatments for biofilm infections. Combining biofilm dispersal agents with antibiotics is emerging as a promising strategy to simultaneously disperse and eradicate biofilms or, in some cases, even inhibit biofilm formation. Here we review studies that have investigated the anti-biofilm activity of some well-studied biofilm dispersal agents (e.g., quorum sensing inhibitors, nitric oxide/nitroxides, antimicrobial peptides/amino acids) in combination with antibiotics from various classes. This review aims to directly compare the efficacy of different combination strategies against microbial biofilms and highlight synergistic treatments that warrant further investigation. By comparing across studies that use different measures of efficacy, we can conclude that treating biofilms in vitro and, in some limited cases in vivo, with a combination of an anti-biofilm agent and an antibiotic, appears overall more effective than treating with either compound alone. The review identifies the most promising combination therapies currently under development as biofilm inhibition and eradication therapies.
Collapse
Affiliation(s)
- Sophia Hawas
- Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane, QLD, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Anthony D. Verderosa
- Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane, QLD, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Makrina Totsika
- Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane, QLD, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
- *Correspondence: Makrina Totsika,
| |
Collapse
|
22
|
Bacteriocin-Based Synergetic Consortia: a Promising Strategy to Enhance Antimicrobial Activity and Broaden the Spectrum of Inhibition. Microbiol Spectr 2022; 10:e0040621. [PMID: 35170996 PMCID: PMC8849083 DOI: 10.1128/spectrum.00406-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Bacteria-derived natural antimicrobial compounds such as bacteriocins, reruterin, and organic acids have recently received substantial attention as food preservatives or therapeutic alternatives in human or animal sectors. This study aimed to evaluate the antimicrobial activity of different bacteria-derived antimicrobials, alone or in combination, against a large panel of Gram-negative and Gram-positive bacteria. Bacteriocins, including microcin J25, pediocin PA-1, nisin Z, and reuterin, were investigated alone or in combination with lactic acid and citric acid, using a checkerboard assay. Concentrations were selected based on predetermined MICs against Salmonella enterica subsp. enterica serovar Newport ATCC 6962 and Listeria ivanovii HPB28 as Gram-negative and Gram-positive indicator strains, respectively. The results demonstrated that the combination of microcin J25 + citric acid + lactic acid; microcin J25 + reuterin + citric acid; and microcin J25 + reuterin + lactic acid tested against S. Newport ATCC 6962 showed synergistic effects (FIC index = 0.5). Moreover, a combination of pediocin PA-1 + citric acid + lactic acid; and reuterin + citric acid + lactic acid against L. ivanovii HPB28 showed a partially synergistic interactions (FIC index = 0.75). Nisin Z exerted a partially synergistic effect in combination with acids (FIC index = 0.625 -0.75), whereas when it was combined with reuterin or pediocin PA-1, it showed additive effects (FIC index = 1) against L. ivanovii HPB28. The inhibitory activity of synergetic consortia were tested against a large panel of Gram-positive and Gram-negative bacteria. According to our results, combining different antimicrobials with different mechanisms of action led to higher potency and a broad spectrum of inhibition, including multidrug-resistance pathogens. IMPORTANCE Reuterin and bacteriocins, including microcin J25, pediocin PA-1, nisin were produced and purified with >90% purity. Using the broth-based checkerboard assay the interaction between these compounds (synergetic, additive, or antagonistic) was assessed. By combining different natural antimicrobials with different modes of action and structure (reuteirn, microcin J25, pediocin PA-1, and organic acids), we successfully developed five different synergetic consortia with improved antimicrobial activity and a broad spectrum of inhibition. These consortia were shown to be effective against a large panel of pathogenic and spoilage microorganisms as well as clinically important multidrug-resistance bacteria. Moreover, because the lower concentrations of bacteriocins and reuterin are used in the synergetic consortia, there is a limited risk of toxicity and resistance development for these compounds.
Collapse
|
23
|
Yadav MK, Yadav P, Dhiman M, Tewari S, Tiwari SK. Plantaricin LD1 purified from Lactobacillus plantarum LD1 inhibits biofilm formation of Enterococcus faecalis ATCC 29212 in tooth model. Lett Appl Microbiol 2022; 75:623-631. [PMID: 35146783 DOI: 10.1111/lam.13668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 01/14/2022] [Accepted: 02/04/2022] [Indexed: 11/28/2022]
Abstract
Plantaricin LD1 was purified to homogeneity using activity-guided chromatography. Enterococcus faecalis ATCC 29212 was found to be sensitive to plantaricin LD1 showing 13 ± 0.21 mm zone of growth inhibition. The minimum inhibitory concentration (MIC) was found to be 50 µg ml-1 against Ent. faecalis ATCC 29212. The in vitro biofilm formation by Ent. faecalis ATCC 29212 was observed which was completely inhibited in the presence of bacteriocin. Similarly, biofilm formation was also observed on the teeth surface showing purple colour whereas, treated-teeth were clean indicated no biofilm formation. Further, untreated cells of Ent. faecalis ATCC 29212 were found normal and plantaricin LD1-treated cells were ruptured seen under light microscope suggesting killing of target cells. These findings have proven the initial leads for antimicrobial and anti-biofilm activity of plantaricin LD1 against Ent. faecalis and its possible application for the treatment of endodontic diseases.
Collapse
Affiliation(s)
- Manoj Kumar Yadav
- Department of Genetics, Maharshi Dayanand University, Rohtak-124001, Haryana, India
| | - Priyanka Yadav
- Department of Genetics, Maharshi Dayanand University, Rohtak-124001, Haryana, India
| | - Meenu Dhiman
- Department of Conservative Dentistry and Endodontics, Post Graduate Institute of Dental Sciences, Rohtak-124001, Haryana, India
| | - Sanjay Tewari
- Department of Conservative Dentistry and Endodontics, Post Graduate Institute of Dental Sciences, Rohtak-124001, Haryana, India
| | - Santosh Kumar Tiwari
- Department of Genetics, Maharshi Dayanand University, Rohtak-124001, Haryana, India
| |
Collapse
|
24
|
Wei DX, Zhang XW. Biosynthesis, Bioactivity, Biosafety and Applications of Antimicrobial Peptides for Human Health. BIOSAFETY AND HEALTH 2022. [DOI: 10.1016/j.bsheal.2022.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
25
|
Flynn J, Ryan A, Hudson SP. Synergistic antimicrobial interactions of nisin A with biopolymers and solubilising agents for oral drug delivery. Eur J Pharm Biopharm 2022; 171:29-38. [PMID: 34986413 DOI: 10.1016/j.ejpb.2021.12.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/17/2021] [Accepted: 12/22/2021] [Indexed: 01/15/2023]
Abstract
In order to develop bacteriocins, like the lantibiotic nisin A, into effective alternatives to existing antibiotics, their biophysical and physicochemical properties must first be assessed, from solubility, to susceptibility and absorption. It has been well established that formulation strategies at early drug development stages can be crucial for successful outcomes during preclinical and clinical phases of development, particularly for molecules with challenging physicochemical properties. This work elucidates the physicochemical challenges of nisin A in terms of its susceptibility to digestive enzymes like pepsin, pancreatin and proteinase K and its poor solubility at physiological pHs. Low solution concentrations, below the minimum inhibitory concentration against Staphylococcus aureus, were obtained in phosphate buffered saline (PBS, pH 7.4) and in fasted state simulated intestinal fluid (FaSSIF, pH 6.5), while higher solubilities at more acidic pH's such as in a KCl/HCl buffer (pH 2) and in fasted state simulated gastric fluid (FaSSGF, pH 1.6) are observed. Tween® 80 (0.01% v/v) significantly increased the solution concentration of nisin A in PBS (pH 7.4, 24 hr). Pancreatin doubled nisin A's solution concentration at pH 7.4 (PBS) but reduced its' inhibitory activity to ∼ 20%, and pepsin almost completely degraded nisin (after 24 hr), but retained activity at biologically relevant exposure times (∼ 15 min). Harnessing synergism between nisin A and either glycol chitosan or ε-poly lysine, combined with the solubilizing effect of Tween®, increased the antimicrobial activity of nisin A six fold in an in vitro oral administration model.
Collapse
Affiliation(s)
- James Flynn
- SSPC, the Science Foundation Ireland Research Centre for Pharmaceuticals, Department of Chemical Sciences, Bernal Institute, University of Limerick, Ireland
| | - Aoibhín Ryan
- SSPC, the Science Foundation Ireland Research Centre for Pharmaceuticals, Department of Chemical Sciences, Bernal Institute, University of Limerick, Ireland
| | - Sarah P Hudson
- SSPC, the Science Foundation Ireland Research Centre for Pharmaceuticals, Department of Chemical Sciences, Bernal Institute, University of Limerick, Ireland.
| |
Collapse
|
26
|
Masterson K, Meade E, Garvey M, Lynch M, Major I, Rowan NJ. Development of a low-temperature extrusion process for production of GRAS bioactive-polymer loaded compounds for targeting antimicrobial-resistant (AMR) bacteria. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 800:149545. [PMID: 34399333 DOI: 10.1016/j.scitotenv.2021.149545] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/05/2021] [Accepted: 08/05/2021] [Indexed: 06/13/2023]
Abstract
Antimicrobial resistance (AMR) is recognised globally as one of the greatest threats to human and animal health; thus, discovery of alternative antibacterial agents to address AMR is a priority challenge. This study constitutes the first report of a low-melting temperature, polymer- extrusion process for the smart delivery of thermally-sensitive antimicrobial bioactives, including generally-regarded-as-safe (GRAS) bioactives derived from various sources. Bioactives were assessed before and after extrusion by determining their respective minimum inhibitory concentrations (MIC). WHO-priority AMR-bacterial isolates causing zoonotic infections were evaluated along with use of standard ATCC strains. Findings revealed that this copolymer method was capable of delivering thermally-sensitive bioactives with varying degrees of growth inhibition against the AMR-bacterial strains. The extrusion process was found to increase the effect of nisin against MRSA (4-fold increase) and L. monocytogenes (6.4-fold increase), silver nitrate (AgNO3) against E. coli (3.6-fold increase) and S. epidermidis (1.25-fold increase), and chitosan against S. aureus (1.25-fold). Findings show the potential applicability of this polymer extrusion process for developing future bioactive-loaded polymer compounds; thus, highlighting the potential of converging bio-based industry with novel materials for enabling 'One-Health' solutions.
Collapse
Affiliation(s)
- Kevin Masterson
- Bioscience Research Institute, Athlone Institute of Technology, Ireland.
| | - Elaine Meade
- Department of Life Science, Sligo Institute of Technology, Ash Lane, Sligo, Ireland
| | - Mary Garvey
- Department of Life Science, Sligo Institute of Technology, Ash Lane, Sligo, Ireland
| | - Mark Lynch
- Bioscience Research Institute, Athlone Institute of Technology, Ireland
| | - Ian Major
- Materials Research Institute, Athlone Institute of Technology, Ireland
| | - Neil J Rowan
- Bioscience Research Institute, Athlone Institute of Technology, Ireland; Centre for Disinfection, Sterilization and Biosecurity, Athlone Institute of Technology, Ireland
| |
Collapse
|
27
|
Lactic Acid Bacteria Bacteriocin, an Antimicrobial Peptide Effective Against Multidrug Resistance: a Comprehensive Review. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-021-10317-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
28
|
Zhang QY, Yan ZB, Meng YM, Hong XY, Shao G, Ma JJ, Cheng XR, Liu J, Kang J, Fu CY. Antimicrobial peptides: mechanism of action, activity and clinical potential. Mil Med Res 2021; 8:48. [PMID: 34496967 PMCID: PMC8425997 DOI: 10.1186/s40779-021-00343-2] [Citation(s) in RCA: 210] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 08/30/2021] [Indexed: 12/15/2022] Open
Abstract
The management of bacterial infections is becoming a major clinical challenge due to the rapid evolution of antibiotic resistant bacteria. As an excellent candidate to overcome antibiotic resistance, antimicrobial peptides (AMPs) that are produced from the synthetic and natural sources demonstrate a broad-spectrum antimicrobial activity with the high specificity and low toxicity. These peptides possess distinctive structures and functions by employing sophisticated mechanisms of action. This comprehensive review provides a broad overview of AMPs from the origin, structural characteristics, mechanisms of action, biological activities to clinical applications. We finally discuss the strategies to optimize and develop AMP-based treatment as the potential antimicrobial and anticancer therapeutics.
Collapse
Affiliation(s)
- Qi-Yu Zhang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, No. 928, Street 2, Xiasha Higher Education Zone, Hangzhou, 310018, Zhejiang, China
| | - Zhi-Bin Yan
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, No. 928, Street 2, Xiasha Higher Education Zone, Hangzhou, 310018, Zhejiang, China
| | - Yue-Ming Meng
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, No. 928, Street 2, Xiasha Higher Education Zone, Hangzhou, 310018, Zhejiang, China
| | - Xiang-Yu Hong
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, No. 928, Street 2, Xiasha Higher Education Zone, Hangzhou, 310018, Zhejiang, China
| | - Gang Shao
- Department of Oncology, The 903rd Hospital of PLA, Hangzhou, 310013, Zhejiang, China
| | - Jun-Jie Ma
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, No. 928, Street 2, Xiasha Higher Education Zone, Hangzhou, 310018, Zhejiang, China
| | - Xu-Rui Cheng
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, No. 928, Street 2, Xiasha Higher Education Zone, Hangzhou, 310018, Zhejiang, China
| | - Jun Liu
- Department of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California San Francisco, 555 Mission Bay Blvd. South, San Francisco, CA, 94158, USA
| | - Jian Kang
- Oncogenic Signaling and Growth Control Program, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, VIC, 3000, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Cai-Yun Fu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, No. 928, Street 2, Xiasha Higher Education Zone, Hangzhou, 310018, Zhejiang, China.
| |
Collapse
|
29
|
Purification, characterization, and mode of action of Paracin 54, a novel bacteriocin against Staphylococci. Appl Microbiol Biotechnol 2021; 105:6735-6748. [PMID: 34453561 DOI: 10.1007/s00253-021-11505-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/05/2021] [Accepted: 08/05/2021] [Indexed: 10/20/2022]
Abstract
Staphylococci belong to conditionally pathogenic bacteria, and the pathogenicity of Staphylococcus aureus is the strongest among them. Enterotoxin produced by it can contaminate food and cause food poisoning. Bacteriocin is a kind of polypeptide with antibacterial activity synthesized by some bacteria during metabolism. In this study, we report on purification, characterization, and mode of action of the bacteriocin named Paracin 54, produced by Lactobacillus paracasei ZFM54. Paracin 54 was purified by precipitation with 80% ammonium sulfate, strong cation-exchange chromatography, G-25 gel column, and reversed-phase high-performance liquid chromatography (HPLC). The molecular weight of Paracin 54 (5718.1843 Da) was determined by matrix-assisted laser desorption ionization time-of-flight mass spectrometry. Paracin 54 showed broad-spectrum inhibitory activity. It had a strong inhibitory effect on Staphylococci with minimum inhibitory concentration values of 3.00-4.50 μg/mL. Paracin 54 was heat-stable and active only in acidic pH range (2-6). After treatment with proteases, the activity was lost. The results of mode of action showed Paracin 54 damaged the cell membrane and cell wall of Staphylococcus aureus, and then the cytoplasm leaked out, leading to death of the bacteria. These properties make Paracin 54 a promising candidate to prevent the growth of spoilage bacteria and control food poisoning caused by Staphylococci. KEY POINTS: • Paracin 54 was purified from Lactobacillus paracasei ZFM54 with good biochemical characteristics. • Paracin 54 had a strong effect against Staphylococci, making it a promising preservative to prevent the growth of Staphylococci in food. • The mode of action of Paracin 54 on Staphylococcus aureus was revealed.
Collapse
|
30
|
Hemmati F, Rezaee MA, Ebrahimzadeh S, Yousefi L, Nouri R, Kafil HS, Gholizadeh P. Novel Strategies to Combat Bacterial Biofilms. Mol Biotechnol 2021; 63:569-586. [PMID: 33914260 DOI: 10.1007/s12033-021-00325-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 04/09/2021] [Indexed: 12/15/2022]
Abstract
Biofilms are considered as a severe problem in the treatment of bacterial infections; their development causes some noticeable resistance to antibacterial agents. Biofilms are responsible for at least two-thirds of all infections, displaying promoted resistance to classical antibiotic treatments. Therefore, finding new alternative therapeutic approaches is essential for the treatment and inhibition of biofilm-related infections. Therefore, this review aims to describe the potential therapeutic strategies that can inhibit bacterial biofilm development; these include the usage of antiadhesion agents, AMPs, bacteriophages, QSIs, aptamers, NPs and PNAs, which can prevent or eradicate the formation of biofilms. These antibiofilm agents represent a promising therapeutic target in the treatment of biofilm infections and development of a strong capability to interfere with different phases of the biofilm development, including adherence, polysaccharide intercellular adhesion (PIA), quorum sensing molecules and cell-to-cell connection, bacterial aggregation, planktonic bacteria killing and host-immune response modulation. In addition, these components, in combination with antibiotics, can lead to the development of some kind of powerful combined therapy against bacterial biofilm-related infections.
Collapse
Affiliation(s)
- Fatemeh Hemmati
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Saba Ebrahimzadeh
- Department of Food Science and Technology, Faculty of Agriculture and Natural Resources, Urmia University, Urmia, Iran
| | - Leila Yousefi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roghayeh Nouri
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Samadi Kafil
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pourya Gholizadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
31
|
Wei L, Li M, Xia F, Wang J, Ran S, Huang Z, Liang J. Phosphate transport system mediates the resistance of Enterococcus faecalis to multidrug. Microbiol Res 2021; 249:126772. [PMID: 33930841 DOI: 10.1016/j.micres.2021.126772] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 03/04/2021] [Accepted: 04/20/2021] [Indexed: 01/12/2023]
Abstract
Enterococcus faecalis, a severe nosocomial and community opportunistic pathogen, is difficult to control due to its multidrug resistance. Through heredity and the recombination of intrinsic resistance genes and horizontally acquired resistance genes, E. faecalis can rapidly evolve drug resistance. Nisin, an important antimicrobial peptide, is extensively employed in the healthcare and food industries to inhibit Gram-positive bacteria and may induce the emergence of nisin-resistant bacteria worldwide. However, the mechanism governing nisin resistance in E. faecalis has not been fully elucidated. This study utilizes transposon insertion sequencing (TIS) to comprehensively explore novel genes related to nisin resistance. According to the analysis of TIS results, hundreds of genes appear to be essential for nisin resistance in E. faecalis. The phosphate transport system (OG1RF_10018-10021, named PTS), which is screened by TIS results, enhances the resistance of E. faecalis to nisin, the mechanism of which may be involved in potA and/or OG1RF_10526 (hypothetical gene). Meanwhile, PTS also strongly represses the biosynthesis of ribosomes to increase the sensitivity of E. faecalis to gentamycin. In addition, the overexpression of PTS increases the sensitivity of E. faecalis to daptomycin, the mechanism of which is independent of the LiaFSR system. This study first demonstrated that E. faecalis utilizes PTS to mediate the resistance to multidrug, which may help to elucidate the mechanism governing drug resistance and to establish guidelines for the treatment of infectious diseases caused by E. faecalis.
Collapse
Affiliation(s)
- Lifan Wei
- Department of Endodontics and Operative Dentistry, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Mingwei Li
- Department of Pediatric Dentistry, Nanjing Stomatological Hospital, Medical School of Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Feng Xia
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Jia Wang
- Department of Endodontics and Operative Dentistry, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Shujun Ran
- Department of Endodontics and Operative Dentistry, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Zhengwei Huang
- Department of Endodontics and Operative Dentistry, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Jingping Liang
- Department of Endodontics and Operative Dentistry, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China.
| |
Collapse
|
32
|
Barbosa AAT, de Melo MR, da Silva CMR, Jain S, Dolabella SS. Nisin resistance in Gram-positive bacteria and approaches to circumvent resistance for successful therapeutic use. Crit Rev Microbiol 2021; 47:376-385. [PMID: 33689548 DOI: 10.1080/1040841x.2021.1893264] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Antibiotic resistance among bacterial pathogens is one of the most worrying problems in health systems today. To solve this problem, bacteriocins from lactic acid bacteria, especially nisin, have been proposed as an alternative for controlling multidrug-resistant bacteria. Bacteriocins are antimicrobial peptides that have activity mainly against Gram-positive strains. Nisin is one of the most studied bacteriocins and is already approved for use in food preservation. Nisin is still not approved for human clinical use, but many in vitro studies have shown its therapeutic effectiveness, especially for the control of antibiotic-resistant strains. Results from in vitro studies show the emergence of nisin-resistant bacteria after exposure to nisin. Considering that nisin has shown promising results for clinical use, studies to elucidate nisin-resistant mechanisms and the development of approaches to circumvent nisin-resistance are important. Thus, the objectives of this review are to identify the Gram-positive bacterial strains that have shown resistance to nisin, describe their resistance mechanisms and propose ways to overcome the development of nisin-resistance for its successful clinical application.
Collapse
Affiliation(s)
| | | | | | - Sona Jain
- Programa de Pós-Graduação em Biotecnologia Industrial, Universidade Tiradentes, Sergipe, Brasil
| | - Silvio Santana Dolabella
- Programa de Pós-Graduação em Biologia Parasitária, Universidade Federal de Sergipe, São Cristóvão, Brasil
| |
Collapse
|
33
|
In vitro and In vivo Antibacterial Effects of Nisin Against Streptococcus suis. Probiotics Antimicrob Proteins 2021; 13:598-610. [PMID: 33404866 DOI: 10.1007/s12602-020-09732-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2020] [Indexed: 01/01/2023]
Abstract
Nisin is a promising therapeutic candidate because of its potent activity against Gram-positive bacteria. The present study aimed to describe the in vitro and in vivo antibacterial effects of nisin against Streptococcus suis, an important zoonotic pathogen. The minimal inhibitory concentrations (MICs) and minimal bactericidal concentrations (MBCs) of nisin against different S. suis strains ranged from 0.12 to 4.0 μg/mL and from 0.25 to 8.0 μg/mL, respectively. Time-killing curve assays illustrated that nisin killed 100% of tested virulent S. suis strains within 4 h when used at 2× MIC, which indicates the rapid bactericidal activity of nisin against the bacteria. Transmission and scanning electron microscopy revealed that nisin destroyed S. suis cell membrane integrity and affected its cellular ultrastructure, including a significantly wrinkled surface, intracellular content leakage, and cell lysis. In addition, nisin inhibited biofilm formation by S. suis in a concentration-dependent manner and exhibited strong degrading activities against preformed biofilms. More importantly, nisin displayed antimicrobial activity against S. suis infection in vivo. Upon treatment with 5.0-10 mg/kg nisin solution, the survival rates of mice challenged with a lethal dose of virulent S. suis virulent ranged 87.5-100%. Nisin significantly decreased bacterial proliferation and translocation in the mouse spleen, brain, and blood. These results indicate that nisin has potential as a novel antimicrobial agent for the clinical treatment and prevention of infection caused by S. suis in animals.
Collapse
|
34
|
Daba GM, Elkhateeb WA. Bacteriocins of lactic acid bacteria as biotechnological tools in food and pharmaceuticals: Current applications and future prospects. BIOCATALYSIS AND AGRICULTURAL BIOTECHNOLOGY 2020. [DOI: 10.1016/j.bcab.2020.101750] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
35
|
Isayenko OY, Knysh OV, Kotsar OV, Ryzhkova TN, Dyukareva GI. Simultaneous and sequential influence of metabolite complexes of Lactobacillus rhamnosus and Saccharomyces boulardii and antibiotics against poly-resistant Gram-negative bacteria. REGULATORY MECHANISMS IN BIOSYSTEMS 2020. [DOI: 10.15421/022021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
For the first time the poly-resistant strains of Gram-negative microorganisms were studied for the sensitivity to combined simultaneous and sequential influence of metabolic complexes of Lactobacillus rhamnosus GG and Saccharomyces boulardii, obtained by the author’s method without using the growth media, with antibiotics. The synergic activity of antibacterial preparations and metabolic complexes of L. rhamnosus GG and S. boulardii were studied using modified disk-diffusive method of Kirby-Bauer. During the sequential method of testing (at first the microorganisms were incubated with structural components and metabolites, then their sensitivity to the antibacterial preparations was determined), we observed increase in the diameters of the zones of growth inhibition of Pseudomonas aeruginosa PR to the typical antibiotics (gentamicin, amіcyl, ciprofloxacin, сefotaxime) and non-typical (lincomycin, levomycetin) depending on the tested combinations. Acinetobacter baumannii PR exhibited lower susceptibility: growth inhibition was seen for the combination with ciprofloxacin, сefotaxime, levomycetin. Susceptibility of Lelliottia amnigena (Enterobacter amnigenus) PR increased to levofloxacin, lincomycin. The zones of growth inhibition of Klebsiella pneumoniae PR increased to gentamicin, amіcyl, tetracycline, сeftriaxone. Maximum efficiency was determined during sequential combination of antibiotics with separate metabolic complexes of L. rhamnosus and S. boulardii, and also their combination (to 15.2, 20.2 and 15.4 mm respectively) compared with their simultaneous use (to 12.2, 15.2 and 13.0 mm respectively) for all the tested poly-resistant pathogens, regardless of the mechanism of action of antibacterial preparation. Metabolic complexes of L. rhamnosus GG and S. boulardii, due to increase in the susceptibility of microorganisms, can decrease the therapeutic concentration of antibiotic, slow the probability of the development of resistance of microorganisms, and are therefore promising candidates for developing “accompanying medications” to antibiotics and antimicrobial preparations of new generation.
Collapse
|
36
|
El-Kazzaz SS, Abou El-Khier NT. Effect of the lantibiotic nisin on inhibitory and bactericidal activities of antibiotics used against vancomycin-resistant enterococci. J Glob Antimicrob Resist 2020; 22:263-269. [PMID: 32169681 DOI: 10.1016/j.jgar.2020.02.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 02/19/2020] [Accepted: 02/23/2020] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVES Antibiotic resistance is a serious issue facing clinicians all over the world. Vancomycin-resistant enterococci (VRE) are amongst the most common resistant pathogens that are isolated from patients suffering from infections in our locality. New antimicrobial agents such as the lantibiotic nisin have been previously examined against resistant bacteria as it has strong antibacterial action with no chance of resistance development. This study aimed to explore the effect of nisin in combination with the conventional antibiotics against VRE, with a view to using it as an auxiliary therapy with such antibiotics for combating resistant isolates. METHODS Twenty-three VRE had been examined for the combined effect of nisin with the routine sets of antibiotics using the microplate dilution technique for minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) testing. Checkerboard microbroth assay was conducted for inspection of synergism between nisin and either ampicillin or chloramphenicol. RESULTS An obvious improvement of inhibitory and bactericidal activities of the tested antibiotics after addition of lantibiotic nisin was observed, with a remarkable reduction in the MIC values of vancomycin against all of the isolates. Nisin recorded a synergistic outcome when combined with either ampicillin or chloramphenicol using the checkerboard assay. CONCLUSION Nisin could be effectively considered as a supplementary agent to traditional antibiotics in the management of VRE-associated infections, as it had a synergistic outcome with commonly prescribed antibiotics such as ampicillin and chloramphenicol.
Collapse
Affiliation(s)
- Samah Sabry El-Kazzaz
- Medical Microbiology and Immunology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Noha Tharwat Abou El-Khier
- Medical Microbiology and Immunology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| |
Collapse
|
37
|
Alves FCB, Albano M, Andrade BFMT, Chechi JL, Pereira AFM, Furlanetto A, Rall VLM, Fernandes AAH, dos Santos LD, Barbosa LN, Fernandes Junior A. Comparative Proteomics of Methicillin-Resistant Staphylococcus aureus Subjected to Synergistic Effects of the Lantibiotic Nisin and Oxacillin. Microb Drug Resist 2020; 26:179-189. [DOI: 10.1089/mdr.2019.0038] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Fernanda Cristina Bergamo Alves
- Department of Microbiology and Immunology, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu, Brazil
| | - Mariana Albano
- Department of Microbiology and Immunology, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu, Brazil
| | | | - Jéssica Luana Chechi
- Department of Microbiology and Immunology, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu, Brazil
| | - Ana Flávia Marques Pereira
- Department of Microbiology and Immunology, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu, Brazil
| | - Alessandra Furlanetto
- Department of Microbiology and Immunology, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu, Brazil
| | - Vera Lúcia Mores Rall
- Department of Microbiology and Immunology, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu, Brazil
| | - Ana Angélica Henrique Fernandes
- Department of Chemistry and Biochemistry, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu, Brazil
| | - Lucilene Delazari dos Santos
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
- Center for the Study of Venom and Venomous Animals (CEVAP), São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Lidiane Nunes Barbosa
- Graduate Program in Animal Sciences with Emphasis on Bioactive Products, Universidade Paranaense (UNIPAR), Umuarama, Brazil
| | - Ary Fernandes Junior
- Department of Microbiology and Immunology, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu, Brazil
- Electronic Microscopy Center, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu, Brazil
| |
Collapse
|
38
|
Ellis JC, Ross RP, Hill C. Nisin Z and lacticin 3147 improve efficacy of antibiotics against clinically significant bacteria. Future Microbiol 2020; 14:1573-1587. [PMID: 32019322 DOI: 10.2217/fmb-2019-0153] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Aim: To determine if bacteriocins improve antibiotic efficacy. Materials & methods: Deferred antagonism assays identified bacteriocins with activity. Growth curves and time kill assays demonstrated bactericidal activity of antimicrobial combinations, and checkerboard assays confirmed synergy. Methicillin-resistant Staphylococcus aureus-infected porcine skin model determined ex vivo efficacy. Results: Subinhibitory concentrations of lacticin with penicillin or vancomycin resulted in complete growth inhibition of strains and the improved inhibitory effect was apparent after 1 h. Nisin with methicillin proved more effective against methicillin-resistant Staphylococcus aureus than either antimicrobial alone, revealing partial synergy and significantly reduced pathogen numbers on porcine skin after 3 h compared with minimal inhibition for either antimicrobial alone. Conclusion: Nisin Z and lacticin 3147 may support the use of certain antibiotics and revive ineffective antibiotics.
Collapse
Affiliation(s)
| | - Reynolds P Ross
- School of Microbiology, University College Cork, T12 K8AF, Ireland.,APC Microbiome Ireland, University College Cork, T12 YT20, Ireland
| | - Colin Hill
- School of Microbiology, University College Cork, T12 K8AF, Ireland.,APC Microbiome Ireland, University College Cork, T12 YT20, Ireland
| |
Collapse
|
39
|
Meade E, Slattery MA, Garvey M. Bacteriocins, Potent Antimicrobial Peptides and the Fight against Multi Drug Resistant Species: Resistance Is Futile? Antibiotics (Basel) 2020; 9:antibiotics9010032. [PMID: 31963311 PMCID: PMC7168330 DOI: 10.3390/antibiotics9010032] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 12/12/2022] Open
Abstract
Despite highly specialized international interventions and policies in place today, the rapid emergence and dissemination of resistant bacterial species continue to occur globally, threatening the longevity of antibiotics in the medical sector. In particular, problematic nosocomial infections caused by multidrug resistant Gram-negative pathogens present as a major burden to both patients and healthcare systems, with annual mortality rates incrementally rising. Bacteriocins, peptidic toxins produced by bacteria, offer promising potential as substitutes or conjugates to current therapeutic compounds. These non-toxic peptides exhibit significant potency against certain bacteria (including multidrug-resistant species), while producer strains remain insusceptible to the bactericidal peptides. The selectivity and safety profile of bacteriocins have been highlighted as superior advantages over traditional antibiotics; however, many aspects regarding their efficacy are still unknown. Although active at low concentrations, bacteriocins typically have low in vivo stability, being susceptible to degradation by proteolytic enzymes. Another major drawback lies in the feasibility of large-scale production, with these key features collectively limiting their current clinical application. Though such limitations require extensive research, the concept of expanding bacteriocins from food preservation to human health opens many fascinating doors, including novel drug delivery systems and anticancer treatment applications.
Collapse
Affiliation(s)
- Elaine Meade
- Department of Life Science, Sligo Institute of Technology, F91 YW50 Sligo, Ireland;
| | - Mark Anthony Slattery
- Mark Anthony Slattery MVB, Veterinary Practice, Manorhamilton, F91 DP62 Leitrim, Ireland;
| | - Mary Garvey
- Department of Life Science, Sligo Institute of Technology, F91 YW50 Sligo, Ireland;
- Mark Anthony Slattery MVB, Veterinary Practice, Manorhamilton, F91 DP62 Leitrim, Ireland;
- Correspondence: ; Tel.: +353-071-9305529
| |
Collapse
|
40
|
Cao Y, Yin H, Wang W, Pei P, Wang Y, Wang X, Jiang J, Luo SZ, Chen L. Killing Streptococcus mutans in mature biofilm with a combination of antimicrobial and antibiofilm peptides. Amino Acids 2019; 52:1-14. [PMID: 31797056 DOI: 10.1007/s00726-019-02804-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 11/11/2019] [Indexed: 10/25/2022]
Abstract
Biofilm poses a serious challenge for the treatment of bacterial infections, as it endows bacteria a pronounced resistance to traditional antibiotics. Antimicrobial peptides (AMPs) are considered potential substitutes for antibiotics. Combinational use of AMPs with other compounds to exert antibiofilm effects has been proved to be an effective means to reduce their toxicity and maximize their antimicrobial activity. However, the combination of various AMPs with different action mechanisms is rarely investigated. A newly designed lytic AMP ZXR-2.3 combined with antibiofilm peptide IDR-1018 or KT2 was tested for the antibiofilm effect on mature Streptococcus mutans biofilms. In general, the combination of ZXR-2.3 + IDR-1018 displayed synergistic effect on both biofilm eradication and bacterial killing, while ZXR-2.3 + KT2 showed no obvious synergism. The confocal images of preformed S. mutans biofilms confirmed the effective bactericidal activity of ZXR-2.3 + IDR-1018. A tube system was applied to investigate the biofilm infection under a flow of medium and SEM images indicated the biofilm disruption and bacterial killing effects of ZXR-2.3 + IDR-1018. Quantitative RT-PCR analysis showed that IDR-1018 induced dramatic changes in the mRNA expressions of the quorum sensing (QS) related genes comC, comD, vicR, and vicK of S. mutans in mature biofilms, whereas the other peptides and ciprofloxacin did not cause obvious changes in these genes. This might explain the better synergism of ZXR-2.3 and IDR-1018. The results of this study provide a potential application using the combination of different AMPs in the treatment of mature biofilm infection.
Collapse
Affiliation(s)
- Yimeng Cao
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Huiwei Yin
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Wujun Wang
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Pengfei Pei
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Yin Wang
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Xinyu Wang
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Jianhui Jiang
- Engineering Laboratory of Chemical Resources Utilization in South Xinjiang of Xinjiang Production and Construction Corps, College of Life Sciences, Tarim University, Alar, Xinjiang, 843300, China
| | - Shi-Zhong Luo
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China.
| | - Long Chen
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China.
| |
Collapse
|
41
|
Isayenko OY. Synergistic activity of filtrates of Lactobacillus rhamnosus and Saccharomyces boulardii and antibacterial preparations against Corynebacterium spp. REGULATORY MECHANISMS IN BIOSYSTEMS 2019. [DOI: 10.15421/021966] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
We present the results of the first study of the combined influence of the biologically active substances Lactobacillus rhamnosus GG ATCC 53103 and Saccharomyces boulardii, obtained by the author’s method, and antibacterial agents on Corynebacterium spp. The first area of research was the study of increasing the sensitivity of toxigenic microorganisms to antimicrobial drugs due to the consecutive effects of the structural components and metabolites of L. rhamnosus GG and S. boulardii and antibacterial drugs on Corynebacterium spp. tox+. The greatest increase in the sensitivity of test-cultures of corynebacteria to penicillin (by 19.4 mm), imipenem (by 15.0 mm), vancomycin (by 12.0 mm), gentamicin (by 11.0 mm), ciprofloxacin (by 9.8 mm), erythromycin (by 9.6 mm), cefotaxime (by 9.5 mm) occurred due to the products of lactobacteria and a combination of metabolites of lactobacteria and saccharomycetes. The second area of research was the study of the synergic activity of substances L. rhamnosus GG and S. boulardii and traditional antibacterial drugs manifested by their simultaneous effect on Corynebacterium spp. Maximum potentiation of azithromycin (by 4.6 mm), erythromycin (by 4.5 mm), cefotaxime (by 2.2 mm), ceftriaxone (by 1.6 mm) and ampicillin (by 1.0 mm) relative to corynebacteria was also observed under the influence of lactobacteria metabolites and a combination of lactobacteria and saccharomycetes metabolites. Different degrees of manifestation of the combined action of biologically active substances L. rhamnosus GG and S. boulardii with antibiotics were determined, which depended on the selected combinations, the method of influence on the microorganism, the individual sensitivity of the test-cultures, the activity of the test filtrates and the initial concentration of the producers used to obtain the products of vital activity of lactobacteria and saccharomyces. The presented complexes of structural components and metabolites of L. rhamnosus GG and S. boulardii, obtained without the use of traditional nutrient media, by increasing the bioavailability of pathogenic pathogens can reduce the required concentration of the antibiotic, continuing their use, and suspend the likelihood of pathogens developing resistance to microorganisms. This makes them promising candidates both for the development of "accompaniment-preparations" for antibiotics for the additional therapy of infectious diseases of different etiology, and for the creation of a new direction of antimicrobial agents with multifunctional capabilities. Synergistic activity of filtrates L. rhamnosus GG and S. boulardii and antibacterial preparations against Corynebacterium spp.
Collapse
|
42
|
Hayes K, Cotter L, O'Halloran F. In vitro synergistic activity of erythromycin and nisin against clinical Group B Streptococcus isolates. J Appl Microbiol 2019; 127:1381-1390. [PMID: 31342602 DOI: 10.1111/jam.14400] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/04/2019] [Accepted: 07/18/2019] [Indexed: 12/12/2022]
Abstract
AIMS This study investigated the potential synergy between erythromycin and nisin against clinical Group B Streptococcus (GBS) strains. METHODS AND RESULTS The combination of erythromycin and nisin was examined for synergistic activity using checkerboard and time-kill assays against invasive and colonizing GBS strains. Additionally, the immunological effect of the antibiotic combination was investigated in vitro using human U937 cells and ELISA analysis. Checkerboard assays confirmed an additive effect when the antimicrobials were combined, while time-kill assays demonstrated a synergistic effect when antimicrobials were combined for invasive GBS isolates. Furthermore, a significantly lower TNF-alpha response (P < 0·05) was observed in U937 cells challenged with GBS when erythromycin and nisin were used in combination. CONCLUSIONS The results suggest that erythromycin and nisin can act synergistically to inhibit the growth of GBS. SIGNIFICANCE AND IMPACT OF THE STUDY Group B Streptococcus is the leading cause of invasive neonatal disease worldwide and is becoming increasingly more prevalent in adults. Resistance to some conventionally used antibiotics, such as erythromycin and clindamycin, continue to rise among GBS, indicating a need for alternative treatments. This study demonstrates the potential of an erythromycin-nisin combination for treatment of GBS infections and encourages further investigation of this treatment option.
Collapse
Affiliation(s)
- K Hayes
- Department of Biological Sciences, Cork Institute of Technology, Bishopstown, Cork, Ireland
| | - L Cotter
- Department of Biological Sciences, Cork Institute of Technology, Bishopstown, Cork, Ireland
| | - F O'Halloran
- Department of Biological Sciences, Cork Institute of Technology, Bishopstown, Cork, Ireland
| |
Collapse
|
43
|
Luther MK, Mermel LA, LaPlante KL. Comparison of linezolid and vancomycin lock solutions with and without heparin against biofilm-producing bacteria. Am J Health Syst Pharm 2019; 74:e193-e201. [PMID: 28438824 DOI: 10.2146/ajhp150804] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PURPOSE The activity of linezolid and vancomycin lock solutions against biofilm-producing strains of Staphylococcus aureus, S. epidermidis, and Enterococcus faecalis was studied. METHODS Two strains each of methicillin-susceptible S. aureus (MSSA), methicillin-resistant S. aureus (MRSA), and S. epidermidis, and 1 strain of vancomycin-susceptible E. faecalis and vancomycin-resistant E. faecalis were tested against vancomycin and linezolid to assess prevention of biofilm formation and eradication of these pathogens within a formed biofilm. Activity was also tested in a 72-hour in vitro central venous catheter (CVC) model. After 24 hours of biofilm growth in a CVC, a lock solution containing vancomycin (2 or 5 mg/mL) or linezolid (1 or 2 mg/mL) alone or in combination with heparin sodium (5,000 units/mL with benzyl alcohol 0.45%) was instilled and incubated at 35 °C for 72 hr. Heparin and 0.9% sodium chloride injection were also tested. RESULTS Linezolid and vancomycin prevented biofilm formation below the minimum inhibitory concentration for 88% and 25% of isolates tested, respectively. The addition of preservative-containing heparin decreased the activity of vancomycin and linezolid lock solutions against all strains. Vancomycin 2- and 5-mg/mL lock solutions had the most activity against MSSA and E. faecalis strains (p < 0.01). Linezolid 2 mg/mL was the most active lock solution against the MRSA strains tested (p < 0.01). There were no significant differences in vancomycin or linezolid lock solution activity against S. epidermidis. CONCLUSION Heparin reduced activity of vancomycin and linezolid lock solutions against S. aureus, S. epidermidis, and E. faecalis biofilms. While linezolid or vancomycin lock solution reduced overall biofilm burden, it did not completely eradicate the bacteria at tested concentrations.
Collapse
Affiliation(s)
- Megan K Luther
- Rhode Island Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI.,Department of Pharmacy Practice, University of Rhode Island, Kingston, RI
| | - Leonard A Mermel
- Department of Medicine, Warren Alpert Medical School, Brown University, Providence, RI.,Division of Infectious Diseases, Rhode Island Hospital, Providence, RI
| | - Kerry L LaPlante
- Department of Pharmacy Practice, University of Rhode Island, Kingston, RI .,Rhode Island Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI.
| |
Collapse
|
44
|
Hols P, Ledesma-García L, Gabant P, Mignolet J. Mobilization of Microbiota Commensals and Their Bacteriocins for Therapeutics. Trends Microbiol 2019; 27:690-702. [PMID: 30987817 DOI: 10.1016/j.tim.2019.03.007] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/12/2019] [Accepted: 03/19/2019] [Indexed: 01/21/2023]
Abstract
With the specter of resurgence of pathogens due to the propagation of antibiotic-resistance genes, innovative antimicrobial strategies are needed. In this review, we summarize the beneficial aspects of bacteriocins, a set of miscellaneous peptide-based bacterium killers, compared with classical antibiotics, and emphasize their use in cocktails to curb the emergence of new resistance. We highlight that their prey spectrum, their molecular malleability, and their multiple modes of production might lead to specific and personalized treatments to prevent systemic disorders. Complementarily, we discuss how we might exploit prevailing bacterial commensals, such as Streptococcus salivarius, and deliberately mobilize their bacteriocin arsenal 'on site' to cure multiresistant infections or finely reshape the endogenous microbiota for prophylaxis purposes.
Collapse
Affiliation(s)
- Pascal Hols
- Biochemistry and Genetics of Microorganisms (BGM), Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, 1348 Louvain-la-Neuve, Belgium
| | - Laura Ledesma-García
- Biochemistry and Genetics of Microorganisms (BGM), Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, 1348 Louvain-la-Neuve, Belgium
| | - Philippe Gabant
- Syngulon, rue du Bois Saint-Jean 15/1, 4102, Seraing, Belgium
| | - Johann Mignolet
- Biochemistry and Genetics of Microorganisms (BGM), Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, 1348 Louvain-la-Neuve, Belgium; Syngulon, rue du Bois Saint-Jean 15/1, 4102, Seraing, Belgium.
| |
Collapse
|
45
|
Bacteriocin encapsulation for food and pharmaceutical applications: advances in the past 20 years. Biotechnol Lett 2019; 41:453-469. [PMID: 30739282 DOI: 10.1007/s10529-018-02635-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 12/05/2018] [Indexed: 01/02/2023]
Abstract
The encapsulation of bacteriocins from lactic acid bacteria has involved several methods to protect them from unfavourable environmental conditions and incompatibilities. This review encompasses different methods for the encapsulation of bacteriocins and their applications in both food and pharmaceutical fields. Based on the bibliometric analysis of publications from well-reputed journals including different available patents during the period from 1996 to 2017, 135 articles and 60 patents were collected. Continent-wise contributions to the bacteriocins encapsulation research were carried out by America (52%), Asia (29%) and Europe (19%); with the United States of America, Brazil, Thailand and Italy the countries with major contributions. Till date, different methods proposed for encapsulation have been (i) Film coatings (50%), (ii) Liposomes (23%), (iii) Nanofibers (22%) and (iv) Nanoparticles (4%). Bacteriocins encapsulation methods frequently carried out in food protection (70%); while in the pharmaceutical field, 30% of the research was conducted on multi drug resistant therapy.
Collapse
|
46
|
Wang J, Wei R, Song R. Novel Antibacterial Peptides Isolated from the Maillard Reaction Products of Half-Fin Anchovy (Setipinna taty) Hydrolysates/Glucose and Their Mode of Action in Escherichia coli. Mar Drugs 2019; 17:E47. [PMID: 30634704 PMCID: PMC6356202 DOI: 10.3390/md17010047] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/07/2019] [Accepted: 01/08/2019] [Indexed: 11/16/2022] Open
Abstract
The Maillard reaction products (MRPs) of half-fin anchovy hydrolysates and glucose, named as HAHp(9.0)-G MRPs, were fractionated by size exclusion chromatography into three major fractions (F1⁻F3). F2, which demonstrated the strongest antibacterial activity against Escherichia coli (E. coli) and showed self-production of hydrogen peroxide (H₂O₂), was extracted by solid phase extraction. The hydrophobic extract of F2 was further isolated by reverse phase-high performance liquid chromatography into sub-fractions HE-F2-1 and HE-F2-2. Nine peptides were identified from HE-F2-1, and two peptides from HE-F2-2 using liquid chromatography-electrospray ionization/multi-stage mass spectrometry. Three peptides, FEDQLR (HGM-Hp1), ALERTF (HGM-Hp2), and RHPEYAVSVLLR (HGM-Hp3), with net charges of -1, 0, and +1, respectively, were synthesized. The minimal inhibitory concentration of these synthetic peptides was 2 mg/mL against E. coli. Once incubated with logarithmic growth phase of E. coli, HGM-Hp1 and HGM-Hp2 induced significant increases of both extracellular and intracellular H₂O₂ formation. However, HGM-Hp3 only dramatically enhanced intracellular H₂O₂ production in E. coli. The increased potassium ions in E. coli suspension after addition of HGM-Hp1 or HGM-Hp2 indicated the destruction of cell integrity via irreversible membrane damage. It is the first report of hydrolysates MRPs-derived peptides that might perform the antibacterial activity via inducing intracellular H₂O2 production.
Collapse
Affiliation(s)
- Jiaxing Wang
- Key Laboratory of Health Risk Factors for Seafood of Zhejiang Province, School of Food Science and Pharmacy, Zhejiang Ocean University, Zhoushan 316000, China.
| | - Rongbian Wei
- School of Marine Science and Technology, Zhejiang Ocean University, Zhoushan 316000, China.
| | - Ru Song
- Key Laboratory of Health Risk Factors for Seafood of Zhejiang Province, School of Food Science and Pharmacy, Zhejiang Ocean University, Zhoushan 316000, China.
| |
Collapse
|
47
|
Shahrour H, Ferrer-Espada R, Dandache I, Bárcena-Varela S, Sánchez-Gómez S, Chokr A, Martinez-de-Tejada G. AMPs as Anti-biofilm Agents for Human Therapy and Prophylaxis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1117:257-279. [PMID: 30980362 DOI: 10.1007/978-981-13-3588-4_14] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Microbial cells show a strong natural tendency to adhere to surfaces and to colonize them by forming complex communities called biofilms. In this growth mode, biofilm-forming cells encase themselves inside a dense matrix which efficiently protects them against antimicrobial agents and effectors of the immune system. Moreover, at the physiological level, biofilms contain a very heterogeneous cell population including metabolically inactive organisms and persisters, which are highly tolerant to antibiotics. The majority of human infectious diseases are caused by biofilm-forming microorganisms which are responsible for pathologies such as cystic fibrosis, infective endocarditis, pneumonia, wound infections, dental caries, infections of indwelling devices, etc. AMPs are well suited to combat biofilms because of their potent bactericidal activity of broad spectrum (including resting cells and persisters) and their ability to first penetrate and then to disorganize these structures. In addition, AMPs frequently synergize with antimicrobial compounds and were recently reported to repress the molecular pathways leading to biofilm formation. Finally, there is a very active research to develop AMP-containing coatings that can prevent biofilm formation by killing microbial cells on contact or by locally releasing their active principle. In this chapter we will describe these strategies and discuss the perspectives of the use of AMPs as anti-biofilm agents for human therapy and prophylaxis.
Collapse
Affiliation(s)
- Hawraa Shahrour
- Department of Microbiology and Parasitology, University of Navarra, Pamplona, Spain.,Laboratory of Microbiology, Department of Life & Earth Sciences, Faculty of Sciences I, Lebanese University, Hadat campus, Beirut, Lebanon.,Platform of Research and Analysis in Environmental Sciences (PRASE), Doctoral School of Sciences and Technologies, Lebanese University, Hadat Campus, Beirut, Lebanon
| | - Raquel Ferrer-Espada
- Department of Microbiology and Parasitology, University of Navarra, Pamplona, Spain.,Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Israa Dandache
- Laboratory of Microbiology, Department of Life & Earth Sciences, Faculty of Sciences I, Lebanese University, Hadat campus, Beirut, Lebanon.,Platform of Research and Analysis in Environmental Sciences (PRASE), Doctoral School of Sciences and Technologies, Lebanese University, Hadat Campus, Beirut, Lebanon
| | | | | | - Ali Chokr
- Laboratory of Microbiology, Department of Life & Earth Sciences, Faculty of Sciences I, Lebanese University, Hadat campus, Beirut, Lebanon.,Platform of Research and Analysis in Environmental Sciences (PRASE), Doctoral School of Sciences and Technologies, Lebanese University, Hadat Campus, Beirut, Lebanon
| | | |
Collapse
|
48
|
Acquired Nisin Resistance in Staphylococcus aureus Involves Constitutive Activation of an Intrinsic Peptide Antibiotic Detoxification Module. mSphere 2018; 3:3/6/e00633-18. [PMID: 30541781 PMCID: PMC6291627 DOI: 10.1128/mspheredirect.00633-18] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
NIS and related bacteriocins are of interest as candidates for the treatment of human infections caused by Gram-positive pathogens such as Staphylococcus aureus. An important liability of NIS in this regard is the ease with which S. aureus acquires resistance. Here we establish that this organism naturally possesses the cellular machinery to detoxify NIS but that the ABC transporter responsible (VraDE) is not ordinarily produced to a degree sufficient to yield substantial resistance. Acquired NIS resistance mutations prompt activation of the regulatory circuit controlling expression of vraDE, thereby unmasking an intrinsic resistance determinant. Our results provide new insights into the complex mechanism by which expression of vraDE is regulated and suggest that a potential route to overcoming the resistance liability of NIS could involve chemical modification of the molecule to prevent its recognition by the VraDE transporter. Resistance to the lantibiotic nisin (NIS) arises readily in Staphylococcus aureus as a consequence of mutations in the nsaS gene, which encodes the sensor kinase of the NsaRS two-component regulatory system. Here we present a series of studies to establish how these mutational changes result in reduced NIS susceptibility. Comparative transcriptomic analysis revealed upregulation of the NsaRS regulon in a NIS-resistant mutant of S. aureus versus its otherwise-isogenic progenitor, indicating that NIS resistance mutations prompt gain-of-function in NsaS. Two putative ABC transporters (BraDE and VraDE) encoded within the NsaRS regulon that have been reported to provide a degree of intrinsic protection against NIS were shown to be responsible for acquired NIS resistance; as is the case for intrinsic NIS resistance, NIS detoxification was ultimately mediated by VraDE, with BraDE participating in the signaling cascade underlying VraDE expression. Our study revealed new features of this signal transduction pathway, including that BraDE (but not VraDE) physically interacts with NsaRS. Furthermore, while BraDE has been shown to sense stimuli and signal to NsaS in a process that is contingent upon ATP hydrolysis, we established that this protein complex is also essential for onward transduction of the signal from NsaS through energy-independent means. NIS resistance in S. aureus therefore joins the small number of documented examples in which acquired antimicrobial resistance results from the unmasking of an intrinsic detoxification mechanism through gain-of-function mutation in a regulatory circuit. IMPORTANCE NIS and related bacteriocins are of interest as candidates for the treatment of human infections caused by Gram-positive pathogens such as Staphylococcus aureus. An important liability of NIS in this regard is the ease with which S. aureus acquires resistance. Here we establish that this organism naturally possesses the cellular machinery to detoxify NIS but that the ABC transporter responsible (VraDE) is not ordinarily produced to a degree sufficient to yield substantial resistance. Acquired NIS resistance mutations prompt activation of the regulatory circuit controlling expression of vraDE, thereby unmasking an intrinsic resistance determinant. Our results provide new insights into the complex mechanism by which expression of vraDE is regulated and suggest that a potential route to overcoming the resistance liability of NIS could involve chemical modification of the molecule to prevent its recognition by the VraDE transporter.
Collapse
|
49
|
Cunha E, Trovão T, Pinheiro A, Nunes T, Santos R, Moreira da Silva J, São Braz B, Tavares L, Veiga AS, Oliveira M. Potential of two delivery systems for nisin topical application to dental plaque biofilms in dogs. BMC Vet Res 2018; 14:375. [PMID: 30497466 PMCID: PMC6267012 DOI: 10.1186/s12917-018-1692-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/09/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Periodontal disease (PD) is caused by the development of a microbial biofilm (dental plaque) in the periodontium, affecting approximately 80% of dogs. Several bacterial species present in the canine oral cavity can be implicated in the development of this disease, including Enterococcus spp. To decrease antibiotic administration, a possible control strategy for dog's enterococcal PD may involve the use of the antimicrobial peptide (AMP) nisin. Nisin's inhibitory activity was evaluated against a collection of previously characterized enterococci obtained from the oral cavity of dogs with PD (n = 20), as well as the potential of a guar-gum gel and a veterinary toothpaste as topical delivery systems for this AMP. The Minimum Inhibitory (MIC) and Bactericidal Concentrations (MBC) and the Minimum Biofilm Eradication (MBEC) and Inhibitory Concentrations (MBIC) were determined for nisin and for the supplemented guar-gum gel. For the supplemented veterinary toothpaste an agar-well diffusion assay was used to evaluate its inhibitory potential. RESULTS Nisin was effective against all isolates. Independently of being or not incorporated in the guar-gum gel, its inhibitory activity on biofilms was higher, with MBIC (12.46 ± 5.16 and 13.60 ± 4.31 μg/mL, respectively) and MBEC values (21.87 ± 11.33 and 42.34 ± 16.61 μg/mL) being lower than MIC (24.61 ± 4.64 and 14.90 ± 4.10 μg/mL) and MBC (63.09 ± 13.22 and 66.63 ± 19.55 μg/mL) values. The supplemented toothpaste was also effective, showing inhibitory activity against 95% of the isolates. CONCLUSIONS The inhibitory ability of nisin when incorporated in the two delivery systems was maintained or increased, demonstrating the potential of these supplemented vehicles to be applied to PD control in dogs.
Collapse
Affiliation(s)
- Eva Cunha
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477, Lisboa, Portugal
| | - Tiago Trovão
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477, Lisboa, Portugal
| | - Ana Pinheiro
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477, Lisboa, Portugal
| | - Telmo Nunes
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477, Lisboa, Portugal
| | - Raquel Santos
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477, Lisboa, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Jorge Moreira da Silva
- Virbac de Portugal Laboratórios, Lda, Rua do Centro Empresarial, Quinta da Beloura, 2710-693, Sintra, Portugal
| | - Berta São Braz
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477, Lisboa, Portugal
| | - Luís Tavares
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477, Lisboa, Portugal
| | - Ana Salomé Veiga
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Manuela Oliveira
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477, Lisboa, Portugal.
| |
Collapse
|
50
|
Straw Wine Melanoidins as Potential Multifunctional Agents: Insight into Antioxidant, Antibacterial, and Angiotensin-I-Converting Enzyme Inhibition Effects. Biomedicines 2018; 6:biomedicines6030083. [PMID: 30072595 PMCID: PMC6163464 DOI: 10.3390/biomedicines6030083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 07/23/2018] [Accepted: 07/25/2018] [Indexed: 01/17/2023] Open
Abstract
Numerous studies provide robust evidence for a protective effect of red wine against many diseases. This bioactivity has been mainly associated with phenolic fractions of wines. However, the health effects of melanoidins in red sweet wines has been ignored. The goal of the present work was to unravel the antioxidant, antimicrobial, and angiotensin-I-converting enzyme (ACE) inhibitory properties of straw sweet wine melanoidins. Results demonstrated that melanoidins have a potential antioxidant activity, determined by 2,2-Diphenyl-1-picrylhydrazyl (DPPH) and Ferric reducing antioxidant power (FRAP) assays. The antimicrobial activity of melanoidins was also tested against Listeria monocytogenes, Salmonella Enteritidis, and Escherichia coli. Minimum inhibitory concentration (MIC) of isolated melanoidins against three bacterial strains ranged from 5 mg mL−1 to 10 mg mL−1. Finally, the ACE inhibitory effect of isolated melanoidins was evaluated, as it is linked with antihypertensive activity. Results showed that they have ACE-inhibitory activity ranging from 58.2 ± 5.4% to 75.3 ± 6.4% at a concentration level of 2 mg mL−1. Furthermore, the chemical properties of isolated melanoidins were determined. Results demonstrated that the skeleton of straw wine melanoidins is mainly composed of carbohydrates, and bear significant numbers of phenolic compounds that may play critical roles in their functional properties. Overall, this study describing the chemical composition and functional properties of melanoidin fractions isolated from a straw wine highlights that they can be exploited as functional agents for multiple purposes. Finally, melanoidins are an unexplored source of bioactive molecules in straw wines except from polyphenols that contribute to the health effects.
Collapse
|