1
|
Dai C, Liu Y, Lv F, Cheng P, Qu S. An alternative approach to combat multidrug-resistant bacteria: new insights into traditional Chinese medicine monomers combined with antibiotics. ADVANCED BIOTECHNOLOGY 2025; 3:6. [PMID: 39918653 PMCID: PMC11805748 DOI: 10.1007/s44307-025-00059-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/11/2025]
Abstract
Antibiotic treatment is crucial for controlling bacterial infections, but it is greatly hindered by the global prevalence of multidrug-resistant (MDR) bacteria. Although traditional Chinese medicine (TCM) monomers have shown high efficacy against MDR infections, the inactivation of bacteria induced by TCM is often incomplete and leads to infection relapse. The synergistic combination of TCM and antibiotics emerges as a promising strategy to mitigate the limitations inherent in both treatment modalities when independently administered. This review begins with a succinct exploration of the molecular mechanisms such as the antibiotic resistance, which informs the antibiotic discovery efforts. We subsequently provide an overview of the therapeutic effects of TCM/antibiotic combinations that have been developed. Finally, the factors that affect the therapeutic outcomes of these combinations and their underlying molecular mechanisms are systematically summarized. This overview offers insights into alternative strategies to treat clinical infections associated with MDR bacteria and the development of novel TCM/antibiotic combination therapies, with the goal of guiding their appropriate usage and further development.
Collapse
Affiliation(s)
- Cunchun Dai
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
- College of Materials and Chemistry, Anhui Agricultural University, Hefei, 230036, China
| | - Ying Liu
- College of Materials and Chemistry, Anhui Agricultural University, Hefei, 230036, China
| | - Fan Lv
- College of Materials and Chemistry, Anhui Agricultural University, Hefei, 230036, China
| | - Ping Cheng
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China.
| | - Shaoqi Qu
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China.
| |
Collapse
|
2
|
Yang S, Su P, Li L, Liu S, Wang Y. Advances and mechanisms of traditional Chinese medicine and its active ingredients against antibiotic-resistant Escherichia coli infections. J Pharm Anal 2025; 15:101117. [PMID: 40026356 PMCID: PMC11871446 DOI: 10.1016/j.jpha.2024.101117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 08/28/2024] [Accepted: 09/30/2024] [Indexed: 03/05/2025] Open
Abstract
In clinical practice, antibiotics have historically been utilized for the treatment of pathogenic bacteria. However, the gradual emergence of antibiotic resistance among bacterial strains has posed a significant challenge to this approach. In 2022, Escherichia coli, a Gram-negative bacterium renowned for its widespread pathogenicity and high virulence, emerged as the predominant pathogenic bacterium in China. The rapid emergence of antibiotic-resistant E. coli strains has rendered antibiotics insufficient to fight E. coli infections. Traditional Chinese medicine (TCM) has made remarkable contributions to the health of Chinese people for thousands of years, and its significant therapeutic effects have been proven in clinical practice. In this paper, we provide a comprehensive review of the advances and mechanisms of TCM and its active ingredients against antibiotic-resistant E. coli infections. First of all, this review introduces the classification, antibiotic resistance characteristics and mechanisms of E. coli. Then, the TCM formulas and extracts are listed along with their active ingredients against E. coli, including extraction solution, minimum inhibitory concentration (MIC), and the antibacterial mechanisms. In addition, there is growing evidence supporting the synergistic therapeutic strategy of combining TCM with antibiotics for the treatment of antibiotic-resistant E. coli infections, and we provide a summary of this evidence and its underlying mechanisms. In conclusion, we present a comprehensive review of TCM and highlight its potential and advantages in the prevention and treatment of E. coli infections. We hold the opinion that TCM will play an important role in global health, pharmaceutical development, and livestock farming in the future.
Collapse
Affiliation(s)
- Shuo Yang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Ping Su
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Lu Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Shuang Liu
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yi Wang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| |
Collapse
|
3
|
Yang N, Wu T, Li M, Hu X, Ma R, Jiang W, Su Z, Yang R, Zhu C. Silver-quercetin-loaded honeycomb-like Ti-based interface combats infection-triggered excessive inflammation via specific bactericidal and macrophage reprogramming. Bioact Mater 2025; 43:48-66. [PMID: 39318638 PMCID: PMC11421951 DOI: 10.1016/j.bioactmat.2024.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/26/2024] [Accepted: 09/08/2024] [Indexed: 09/26/2024] Open
Abstract
Excessive inflammation caused by bacterial infection is the primary cause of implant failure. Antibiotic treatment often fails to prevent peri-implant infection and may induce unexpected drug resistance. Herein, a non-antibiotic strategy based on the synergy of silver ion release and macrophage reprogramming is proposed for preventing infection and bacteria-induced inflammation suppression by the organic-inorganic hybridization of silver nanoparticle (AgNP) and quercetin (Que) into a polydopamine (PDA)-based coating on the 3D framework of porous titanium (SQPdFT). Once the planktonic bacteria (e.g., Escherichia coli, Staphylococcus aureus) reach the surface of SQPdFT, released Que disrupts the bacterial membrane. Then, AgNP can penetrate the invading bacterium and kill them, which further inhibits the biofilm formation. Simultaneously, released Que can regulate macrophage polarization homeostasis via the peroxisome proliferators-activated receptors gamma (PPARγ)-mediated nuclear factor kappa-B (NF-κB) pathway, thereby terminating excessive inflammatory responses. These advantages facilitate the adhesion and osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs), concomitantly suppressing osteoclast maturation, and eventually conferring superior mechanical stability to SQPdFT within the medullary cavity. In summary, owing to its excellent antibacterial effect, immune remodeling function, and pro-osteointegration ability, SQPdFT is a promising protective coating for titanium-based implants used in orthopedic replacement surgery.
Collapse
Affiliation(s)
- Ning Yang
- Department of Orthopaedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Ting Wu
- CAS Key Lab for Biomedical Effects of Nanomaterials and Nanosafety, Center of Materials Science and Optoelectronics Engineering, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Meng Li
- Department of Orthopaedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Xianli Hu
- Department of Orthopaedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Ruixiang Ma
- Department of Orthopaedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Wei Jiang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Zheng Su
- Department of Orthopaedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Rong Yang
- CAS Key Lab for Biomedical Effects of Nanomaterials and Nanosafety, Center of Materials Science and Optoelectronics Engineering, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Chen Zhu
- Department of Orthopaedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| |
Collapse
|
4
|
Mohamed AA, Saed S, El-Sayed SR, Yassin MT, Gad M, Tartour E, Fathey HA, Taha AS, Mohamed AH, Al-Otibi FO, AbdelGawwad MR, Ahmed MMS, Almalki SA, Abdel-Haleem M. A combined therapy of meropenem-ZnO nanoparticles efficiently eliminates carbapenem-resistant Klebsiella pneumoniae biofilms, with reduced nephrotoxicity (in vitro). Lett Appl Microbiol 2024; 77:ovae136. [PMID: 39701814 DOI: 10.1093/lambio/ovae136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 12/21/2024]
Abstract
In response to the World Health Organization's research agenda of antimicrobial resistance in human health, this study appraised the antibacterial and antibiofilm synergistic activity of meropenem and ZnO nanoparticles (ZnO-NPs) combination against carbapenem-resistant Klebsiella pneumoniae (CRKP). The minimum inhibitory concentration (MIC) of meropenem in combination was found to be ~1/12 of its MIC alone. The results of microtiter dilution assay showed that the combination was more efficient in reducing the biofilm biomass than meropenem alone or ZnO-NPs alone. The scanning-electron-microscopy micrographs elucidated that the combination of meropenem with ZnO-NPs has significantly enhanced its competence in eradicating the preformed biofilms of CRKP strains. In addition, the relative gene expression results showed that the combination compared to the meropenem alone and ZnO-NPs alone eloquently down-regulated the expression of biofilm genes (mrkA, fimA, and ecpA). Besides, the MTT-assay demonstrated that the combination has limited cytotoxicity against Vero-cells (in vitro). Overall, this study represents an efficient safe enhancement of meropenem to tackle the growing health threat of CRKP and carbapenem-resistant Enterobacterals prevalence.
Collapse
Affiliation(s)
- Alzhraa Ali Mohamed
- Microbiology and Botany Department, Faculty of Science, Zagazig University,Zagazig 44519, Egypt
- Graduate School of Natural and Applied Science, Ege University, Izmir, Türkiye
| | - Safaa Saed
- Microbiology and Botany Department, Faculty of Science, Zagazig University,Zagazig 44519, Egypt
| | - Sara Ramadan El-Sayed
- Microbiology and Botany Department, Faculty of Science, Zagazig University,Zagazig 44519, Egypt
| | - Mohamed Taha Yassin
- Botany and Microbiology Department, College of Science, King Saud University, 2455 Riyadh, Saudi Arabia
- King Salman Center for Disability Research, Riyadh 11614, Saudi Arabia
| | - Mohamed Gad
- Zoology Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
| | - Eman Tartour
- Microbiology and Botany Department, Faculty of Science, Zagazig University,Zagazig 44519, Egypt
| | - Hoda A Fathey
- Microbiology and Botany Department, Faculty of Science, Zagazig University,Zagazig 44519, Egypt
| | - Asmaa S Taha
- Microbiology and Botany Department, Faculty of Science, Zagazig University,Zagazig 44519, Egypt
| | - Asmaa H Mohamed
- Microbiology and Botany Department, Faculty of Science, Zagazig University,Zagazig 44519, Egypt
| | - Fatimah Olyan Al-Otibi
- Botany and Microbiology Department, College of Science, King Saud University, 2455 Riyadh, Saudi Arabia
| | - Mohamed Ragab AbdelGawwad
- Genetics and Bioengineering Department, Faculty of Engineering and Natural Sciences, International University of Sarajevo, 71210 Sarajevo, Bosnia and Herzegovina
| | - Mohamed M Sayed Ahmed
- Microbiology and Botany Department, Faculty of Science, Zagazig University,Zagazig 44519, Egypt
| | - Susan Ahmed Almalki
- Laboratory medicine department, Faculty of Applied Medical Sciences, Al-Baha University, 65779, Saudi Arabia
| | - Mohamed Abdel-Haleem
- Microbiology and Botany Department, Faculty of Science, Zagazig University,Zagazig 44519, Egypt
| |
Collapse
|
5
|
Wang Z, Shen W, Li Y, Wang X, Zhong X, Wang X. Multi-omics Analysis of Klebsiella pneumoniae Revealed Opposing Effects of Rutin and Luteolin on Strain Growth. Curr Microbiol 2024; 82:9. [PMID: 39585437 DOI: 10.1007/s00284-024-03982-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 11/11/2024] [Indexed: 11/26/2024]
Abstract
The emergence of pathogenic bacteria resistant to conventional antibiotics is becoming increasingly challenging. Plant-derived flavonoids are potential alternatives to antibiotics, owing to their antimicrobial properties. However, the molecular mechanisms through which they inhibit the growth of pathogenic microorganisms remain unclear. Therefore, Klebsiella pneumoniae ATCC700603 was separately incubated in two flavonoids to elucidate their inhibitory mechanism. Metabolomic and transcriptomic analyses were performed after 4-h incubation. In total, 5483 genes and 882 metabolites were identified. Compared to the untreated control, rutin and luteolin activated 507 and 374 differentially expressed genes (DEGs), respectively. However, the number of differential abundant metabolites (DAMs) remained the same. The top 10 correlated DEGs and DAMs were identified within each comparative group after a correlation analysis. Rutin induced the accumulation of unique metabolites and suppressed gene expression whereas luteolin did not. Our results explain the disparate effects of these two flavonoids and demonstrate the inhibitory mechanism of rutin on strain growth.
Collapse
Affiliation(s)
- Zhibin Wang
- Inflammation & Allergic Diseases Research Unit, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Wanxia Shen
- Citrus Research Institute, Southwest University, Beibei, Chongqing, 400715, China
| | - Yuejiao Li
- Inflammation & Allergic Diseases Research Unit, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xiaoyun Wang
- Inflammation & Allergic Diseases Research Unit, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xiaolin Zhong
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xing Wang
- Inflammation & Allergic Diseases Research Unit, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Department of Respiratory and Critical Care Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
6
|
Frenț OD, Stefan L, Morgovan CM, Duteanu N, Dejeu IL, Marian E, Vicaș L, Manole F. A Systematic Review: Quercetin-Secondary Metabolite of the Flavonol Class, with Multiple Health Benefits and Low Bioavailability. Int J Mol Sci 2024; 25:12091. [PMID: 39596162 PMCID: PMC11594109 DOI: 10.3390/ijms252212091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
The main goal of this systematic review on the flavonol class secondary metabolite quercetin is to evaluate and summarize the existing research on quercetin's potential health benefits, therapeutic properties, and effectiveness in disease prevention and treatment. In addition to evaluating quercetin's potential for drug development with fewer side effects and lower toxicity, this type of review attempts to collect scientific evidence addressing quercetin's roles as an antioxidant, anti-inflammatory, antibacterial, and anticancer agent. In the first part, we analyze various flavonoid compounds, focusing on their chemical structure, classification, and natural sources. We highlight their most recent biological activities as reported in the literature. Among these compounds, we pay special attention to quercetin, detailing its chemical structure, physicochemical properties, and process of biosynthesis in plants. We also present natural sources of quercetin and emphasize its health benefits, such as its antioxidant and anti-inflammatory effects. Additionally, we discuss methods to enhance its bioavailability, analyzing the latest and most effective delivery systems based on quercetin.
Collapse
Affiliation(s)
- Olimpia-Daniela Frenț
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, No. 29 Nicolae Jiga Street, 410028 Oradea, Romania; (O.-D.F.); (E.M.); (L.V.)
| | - Liana Stefan
- Department of Surgical Discipline, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Claudia Mona Morgovan
- Department of Chemistry, Faculty of Informatics and Sciences, University of Oradea, No 1 University Street, 410087 Oradea, Romania
| | - Narcis Duteanu
- Faculty of Chemical Engineering, Biotechnologies, and Environmental Protection, Politehnica University of Timisoara, No. 2 Victoriei Square, 300006 Timişoara, Romania
- National Institute of Research and Development for Electrochemistry and Condensed Matter, 144 Dr. A. P. Podeanu, 300569 Timisoara, Romania
| | - Ioana Lavinia Dejeu
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, No. 29 Nicolae Jiga Street, 410028 Oradea, Romania; (O.-D.F.); (E.M.); (L.V.)
| | - Eleonora Marian
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, No. 29 Nicolae Jiga Street, 410028 Oradea, Romania; (O.-D.F.); (E.M.); (L.V.)
| | - Laura Vicaș
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, No. 29 Nicolae Jiga Street, 410028 Oradea, Romania; (O.-D.F.); (E.M.); (L.V.)
| | - Felicia Manole
- Department of Surgical Discipline, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| |
Collapse
|
7
|
Macêdo HLRDQ, de Oliveira LL, de Oliveira DN, Lima KFA, Cavalcanti IMF, Campos LADA. Nanostructures for Delivery of Flavonoids with Antibacterial Potential against Klebsiella pneumoniae. Antibiotics (Basel) 2024; 13:844. [PMID: 39335017 PMCID: PMC11428843 DOI: 10.3390/antibiotics13090844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/31/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Flavonoids are secondary metabolites that exhibit remarkable biological activities, including antimicrobial properties against Klebsiella pneumoniae, a pathogen responsible for several serious nosocomial infections. However, oral administration of these compounds faces considerable challenges, such as low bioavailability and chemical instability. Thus, the encapsulation of flavonoids in nanosystems emerges as a promising strategy to mitigate these limitations, offering protection against degradation; greater solubility; and, in some cases, controlled and targeted release. Different types of nanocarriers, such as polymeric nanoparticles, liposomes, and polymeric micelles, among others, have shown potential to increase the antimicrobial efficacy of flavonoids by reducing the therapeutic dose required and minimizing side effects. In addition, advances in nanotechnology enable co-encapsulation with other therapeutic agents and the development of systems responsive to more specific stimuli, optimizing treatment. In this context, the present article provides an updated review of the literature on flavonoids and the main nanocarriers used for delivering flavonoids with antibacterial properties against Klebsiella pneumoniae.
Collapse
Affiliation(s)
- Hanne Lazla Rafael de Queiroz Macêdo
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Recife 50670-901, PE, Brazil; (H.L.R.d.Q.M.); (L.L.d.O.); (D.N.d.O.); (K.F.A.L.); (L.A.d.A.C.)
| | - Lara Limeira de Oliveira
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Recife 50670-901, PE, Brazil; (H.L.R.d.Q.M.); (L.L.d.O.); (D.N.d.O.); (K.F.A.L.); (L.A.d.A.C.)
| | - David Nattan de Oliveira
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Recife 50670-901, PE, Brazil; (H.L.R.d.Q.M.); (L.L.d.O.); (D.N.d.O.); (K.F.A.L.); (L.A.d.A.C.)
| | - Karitas Farias Alves Lima
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Recife 50670-901, PE, Brazil; (H.L.R.d.Q.M.); (L.L.d.O.); (D.N.d.O.); (K.F.A.L.); (L.A.d.A.C.)
| | - Isabella Macário Ferro Cavalcanti
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Recife 50670-901, PE, Brazil; (H.L.R.d.Q.M.); (L.L.d.O.); (D.N.d.O.); (K.F.A.L.); (L.A.d.A.C.)
- Academic Center of Vitória (CAV), Federal University of Pernambuco (UFPE), Vitória de Santo Antão 50670-901, PE, Brazil
| | - Luís André de Almeida Campos
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Recife 50670-901, PE, Brazil; (H.L.R.d.Q.M.); (L.L.d.O.); (D.N.d.O.); (K.F.A.L.); (L.A.d.A.C.)
| |
Collapse
|
8
|
Saini R, Kumar V, Sourirajan A, Dev K. Fruit Extract and Phenolic Compounds of Phyllanthus emblica Fruits as Bioactivity Enhancer of Chloramphenicol Against Bacterial Species. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2024; 79:656-661. [PMID: 38951374 DOI: 10.1007/s11130-024-01206-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/16/2024] [Indexed: 07/03/2024]
Abstract
Phyllanthus emblica L. (syn. Emblica officinalis) fruits have been traditionally exploited to enhance the immune system and provide protection against bacterial and fungal diseases. The present study aimed to evaluate the synergistic interactions between chloramphenicol and several phenolic compounds found in P. emblica fruits against bacterial strains. The combination of P. emblica fruit extracts and its phenolic compounds demonstrated synergistic antibacterial activity when used in conjunction with chloramphenicol against both Gram-positive and Gram-negative bacteria. The combination of MICGA with ½MICChl exhibited a significant increase in bioactivity, with a 333.33-fold enhancement against B. subtilis. Similarly, the combination of MICGA with 2MICChl displayed a bioactivity enhancement of 16.02 folds against S. aureus. The co-administration of ½MICQ and ½MICChl resulted in a significant 35.71-fold increase in bioactivity against P. aeruginosa. Similarly, the combination of MIC GA and ½MICChl exhibited a remarkable 166.66-fold enhancement in bioactivity against E. coli. The combinations of 2MICFPE and ½MICChloramphenicol, as well as ½MICGA and ½MICChl demonstrated the highest bioactivity enhancement of 17.85 folds for K. pneumoniae. This study claimed that the fruit extracts of P. emblica and its phenolic compounds could be utilized to augment the effectiveness of conventional antibiotics, which have acquired resistance to bacterial infections.
Collapse
Affiliation(s)
- Rakshandha Saini
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, PO Sultanpur, Distt. Solan-173229 HP, Bajhol, India
| | - Vikas Kumar
- University Institute of Biotechnology, Chandigarh University, Gharuan, Mohali, Punjab, 140413, India.
| | - Anuradha Sourirajan
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, PO Sultanpur, Distt. Solan-173229 HP, Bajhol, India
| | - Kamal Dev
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, PO Sultanpur, Distt. Solan-173229 HP, Bajhol, India.
- Department of Pharmacology and Toxicology, Wright State University, Dayton, OH, 45435, USA.
| |
Collapse
|
9
|
Hurtová M, Brdová D, Křížkovská B, Tedeschi G, Nejedlý T, Strnad O, Dobiasová S, Osifová Z, Kroneislová G, Lipov J, Valentová K, Viktorová J, Křen V. Nitrogen-Containing Flavonoids-Preparation and Biological Activity. ACS OMEGA 2024; 9:34938-34950. [PMID: 39157108 PMCID: PMC11325505 DOI: 10.1021/acsomega.4c04627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 08/20/2024]
Abstract
In this work, we report the application of Buchwald-Hartwig amination for the preparation of new derivatives of quercetin and luteolin. Our investigation delves into the impact of aniline moiety on antioxidant, and anti-inflammatory activity, cytotoxicity, and the ability of flavonoids to modulate drug-resistance mechanisms in bacteria. The anti-inflammatory activity disappeared after the introduction of aniline into the flavonoids and the cytotoxicity remained low. Although the ability of quercetin and luteolin to modulate bacterial resistance to antibiotics has already been published, this is the first report on the molecular mechanism of this process. Both flavonoids attenuate erythromycin resistance by suppressing the ribosomal methyltransferase encoded by the ermA gene in Staphylococcus aureus. Notably, 4-(trifluoromethyl)anilino quercetin emerged as a potent ErmA inhibitor, likely by interacting with the RNA-binding pocket of ErmA. Additionally, both 4-fluoroanilino derivatives effectively impended the staphylococcal efflux system. All the prepared derivatives exhibited superior activity in modulating gentamicin resistance in S. aureus compared to the parent compounds. Overall, the incorporation of substituted anilines into the flavonoid core significantly enhanced its ability to combat multidrug resistance in bacteria.
Collapse
Affiliation(s)
- Martina Hurtová
- Institute
of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 142 00, Czech Republic
| | - Daniela Brdová
- Department
of Biochemistry and Microbiology, University
of Chemistry and Technology Prague, Technická 5, Prague 166 28, Czech Republic
| | - Bára Křížkovská
- Department
of Biochemistry and Microbiology, University
of Chemistry and Technology Prague, Technická 5, Prague 166 28, Czech Republic
| | - Guglielmo Tedeschi
- Department
of Biochemistry and Microbiology, University
of Chemistry and Technology Prague, Technická 5, Prague 166 28, Czech Republic
| | - Tomáš Nejedlý
- Department
of Biochemistry and Microbiology, University
of Chemistry and Technology Prague, Technická 5, Prague 166 28, Czech Republic
| | - Ondřej Strnad
- Department
of Biochemistry and Microbiology, University
of Chemistry and Technology Prague, Technická 5, Prague 166 28, Czech Republic
| | - Simona Dobiasová
- Department
of Biochemistry and Microbiology, University
of Chemistry and Technology Prague, Technická 5, Prague 166 28, Czech Republic
| | - Zuzana Osifová
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 542, Prague 160 00, Czech Republic
| | - Gabriela Kroneislová
- Department
of Biochemistry and Microbiology, University
of Chemistry and Technology Prague, Technická 5, Prague 166 28, Czech Republic
- Department
of Clinical Microbiology and ATB Center, Institute of Medical Biochemistry and Laboratory Diagnostics of the
General University Hospital and of The First Faculty of Medicine of
Charles University, U
Nemocnice 2, Prague 2 128
08, Czech Republic
| | - Jan Lipov
- Department
of Biochemistry and Microbiology, University
of Chemistry and Technology Prague, Technická 5, Prague 166 28, Czech Republic
| | - Kateřina Valentová
- Institute
of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 142 00, Czech Republic
| | - Jitka Viktorová
- Department
of Biochemistry and Microbiology, University
of Chemistry and Technology Prague, Technická 5, Prague 166 28, Czech Republic
| | - Vladimír Křen
- Institute
of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 142 00, Czech Republic
| |
Collapse
|
10
|
Lee T, Lee S, Kim MK, Ahn JH, Park JS, Seo HW, Park KH, Chong Y. 3- O-Substituted Quercetin: an Antibiotic-Potentiating Agent against Multidrug-Resistant Gram-Negative Enterobacteriaceae through Simultaneous Inhibition of Efflux Pump and Broad-Spectrum Carbapenemases. ACS Infect Dis 2024; 10:1624-1643. [PMID: 38652574 DOI: 10.1021/acsinfecdis.3c00715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
The discovery of safe and efficient inhibitors against efflux pumps as well as metallo-β-lactamases (MBL) is one of the main challenges in the development of multidrug-resistant (MDR) reversal agents which can be utilized in the treatment of carbapenem-resistant Gram-negative bacteria. In this study, we have identified that introduction of an ethylene-linked sterically demanding group at the 3-OH position of the previously reported MDR reversal agent di-F-Q endows the resulting compounds with hereto unknown multitarget inhibitory activity against both efflux pumps and broad-spectrum β-lactamases including difficult-to-inhibit MBLs. A molecular docking study of the multitarget inhibitors against efflux pump, as well as various classes of β-lactamases, revealed that the 3-O-alkyl substituents occupy the novel binding sites in efflux pumps as well as carbapenemases. Not surprisingly, the multitarget inhibitors rescued the antibiotic activity of a carbapenem antibiotic, meropenem (MEM), in NDM-1 (New Delhi Metallo-β-lactamase-1)-producing carbapenem-resistant Enterobacteriaceae (CRE), and they reduced MICs of MEM more than four-fold (synergistic effect) in 8-9 out of 14 clinical strains. The antibiotic-potentiating activity of the multitarget inhibitors was also demonstrated in CRE-infected mouse model. Taken together, these results suggest that combining inhibitory activity against two critical targets in MDR Gram-negative bacteria, efflux pumps, and β-lactamases, in one molecule is possible, and the multitarget inhibitors may provide new avenues for the discovery of safe and efficient MDR reversal agents.
Collapse
Affiliation(s)
- Taegum Lee
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 05029, Korea
| | - Seongyeon Lee
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 05029, Korea
| | - Mi Kyoung Kim
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 05029, Korea
| | - Joong Hoon Ahn
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 05029, Korea
| | - Ji Sun Park
- Infectious Disease Research Center, Korea Research Institute of Bioscience & Biotechnology, Yuseong-gu, Daejeon 34141, Korea
| | - Hwi Won Seo
- Infectious Disease Research Center, Korea Research Institute of Bioscience & Biotechnology, Yuseong-gu, Daejeon 34141, Korea
| | - Ki-Ho Park
- Department of Infectious Disease, Kyung Hee University School of Medicine, Seoul 02447, Korea
| | - Youhoon Chong
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 05029, Korea
| |
Collapse
|
11
|
Saifi S, Ashraf A, Hasan GM, Shamsi A, Hassan MI. Insights into the preventive actions of natural compounds against Klebsiella pneumoniae infections and drug resistance. Fitoterapia 2024; 173:105811. [PMID: 38168570 DOI: 10.1016/j.fitote.2023.105811] [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: 09/29/2023] [Revised: 12/27/2023] [Accepted: 12/29/2023] [Indexed: 01/05/2024]
Abstract
Klebsiella pneumoniae is a type of Gram-negative bacteria that causes a variety of infections, including pneumonia, bloodstream infections, wound infections, and meningitis. The treatment of K. pneumoniae infection depends on the type of infection and the severity of the symptoms. Antibiotics are generally used to treat K. pneumoniae infections. However, some strains of K. pneumoniae have become resistant to antibiotics. This comprehensive review examines the potential of natural compounds as effective strategies against K. pneumonia infections. The alarming rise in antibiotic resistance underscores the urgent need for alternative therapies. This article represents current research on the effects of diverse natural compounds, highlighting their anti-microbial and antibiofilm properties against K. pneumonia. Notably, compounds such as andrographolide, artemisinin, baicalin, berberine, curcumin, epigallocatechin gallate, eugenol, mangiferin, piperine, quercetin, resveratrol, and thymol have been extensively investigated. These compounds exhibit multifaceted mechanisms, including disruption of bacterial biofilms, interference with virulence factors, and augmentation of antibiotic effectiveness. Mechanistic insights into their actions include membrane perturbation, oxidative stress induction, and altered gene expression. While promising, challenges such as limited bioavailability and varied efficacy across bacterial strains are addressed. This review further discusses the potential of natural compounds as better alternatives in combating K. pneumonia infection and emphasizes the need for continued research to harness their full therapeutic potential. As antibiotic resistance persists, these natural compounds offer a promising avenue in the fight against K. pneumonia and other multidrug-resistant pathogens.
Collapse
Affiliation(s)
- Sana Saifi
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Anam Ashraf
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Anas Shamsi
- Center for Medical and Bio-Allied Health Sciences Research, Ajman University, United Arab Emirates
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India.
| |
Collapse
|
12
|
Lu L, Wang J, Wang C, Zhu J, Wang H, Liao L, Zhao Y, Wang X, Yang C, He Z, Li M. Plant-derived virulence arresting drugs as novel antimicrobial agents: Discovery, perspective, and challenges in clinical use. Phytother Res 2024; 38:727-754. [PMID: 38014754 DOI: 10.1002/ptr.8072] [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: 06/21/2023] [Revised: 09/23/2023] [Accepted: 11/05/2023] [Indexed: 11/29/2023]
Abstract
Antimicrobial resistance (AMR) emerges as a severe crisis to public health and requires global action. The occurrence of bacterial pathogens with multi-drug resistance appeals to exploring alternative therapeutic strategies. Antivirulence treatment has been a positive substitute in seeking to circumvent AMR, which aims to target virulence factors directly to combat bacterial infections. Accumulated evidence suggests that plant-derived natural products, which have been utilized to treat infectious diseases for centuries, can be abundant sources for screening potential virulence-arresting drugs (VADs) to develop advanced therapeutics for infectious diseases. This review sums up some virulence factors and their actions in various species of bacteria, as well as recent advances pertaining to plant-derived natural products as VAD candidates. Furthermore, we also discuss natural VAD-related clinical trials and patents, the perspective of VAD-based advanced therapeutics for infectious diseases and critical challenges hampering clinical use of VADs, and genomics-guided identification for VAD therapeutic. These newly discovered natural VADs will be encouraging and optimistic candidates that may sustainably combat AMR.
Collapse
Affiliation(s)
- Lan Lu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, Sichuan, P.R. China
| | - Jingya Wang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, Sichuan, P.R. China
| | - Chongrui Wang
- Faculty of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, P.R. China
| | - Jie Zhu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, Sichuan, P.R. China
| | - Hongping Wang
- Safety Evaluation Center, Sichuan Institute for Drug Control (Sichuan Testing Center of Medical Devices), Chengdu, Sichuan, P.R. China
| | - Li Liao
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, Sichuan, P.R. China
| | - Yuting Zhao
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, Sichuan, P.R. China
| | - Xiaobo Wang
- Department of Hepatobiliary Surgery, Langzhong People's Hospital, Langzhong, Sichuan, P.R. China
| | - Chen Yang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, Sichuan, P.R. China
| | - Zhengyou He
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, Sichuan, P.R. China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
| |
Collapse
|
13
|
Lee S, Lee T, Kim MK, Ahn JH, Jeong S, Park KH, Chong Y. Potentiation of Antibiotic Activity of Aztreonam against Metallo-β-Lactamase-Producing Multidrug-Resistant Pseudomonas aeruginosa by 3- O-Substituted Difluoroquercetin Derivatives. Pharmaceutics 2024; 16:185. [PMID: 38399246 PMCID: PMC10892423 DOI: 10.3390/pharmaceutics16020185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
The combination of aztreonam (ATM) and ceftazidime-avibactam (CAZ-AVI; CZA) has shown therapeutic potential against serine-β-lactamase (SBL)- and metallo-β-lactamase (MBL)-producing Enterobacterales. However, the ability of CZA to restore the antibiotic activity of ATM is severely limited in MBL-producing multidrug-resistant (MDR) Pseudomonas aeruginosa strains because of the myriad of intrinsic and acquired resistance mechanisms associated with this pathogen. We reasoned that the simultaneous inhibition of multiple targets associated with multidrug resistance mechanisms may potentiate the antibiotic activity of ATM against MBL-producing P. aeruginosa. During a search for the multitarget inhibitors through a molecular docking study, we discovered that di-F-Q, the previously reported efflux pump inhibitor of MDR P. aeruginosa, binds to the active sites of the efflux pump (MexB), as well as various β-lactamases, and these sites are open to the 3-O-position of di-F-Q. The 3-O-substituted di-F-Q derivatives were thus synthesized and showed hereto unknown multitarget MDR inhibitory activity against various ATM-hydrolyzing β-lactamases (AmpC, KPC, and New Delhi metallo-β-lactamase (NDM)) and the efflux pump of P. aeruginosa, presumably by forming additional hydrophobic contacts with the targets. The multitarget MDR inhibitor 27 effectively potentiated the antimicrobial activity of ATM and reduced the MIC of ATM more than four-fold in 19 out of 21 MBL-producing P. aeruginosa clinical strains, including the NDM-producing strains which were highly resistant to various combinations of ATM with β-lactamase inhibitors and/or efflux pump inhibitors. Our findings suggest that the simultaneous inhibition of multiple MDR targets might provide new avenues for the discovery of safe and efficient MDR reversal agents which can be used in combination with ATM against MBL-producing MDR P. aeruginosa.
Collapse
Affiliation(s)
- Seongyeon Lee
- Department of Bioscience and Biotechnology, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea; (S.L.); (T.L.)
- Bio/Molecular Informatics Center, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea; (M.K.K.); (J.H.A.)
| | - Taegum Lee
- Department of Bioscience and Biotechnology, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea; (S.L.); (T.L.)
- Bio/Molecular Informatics Center, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea; (M.K.K.); (J.H.A.)
| | - Mi Kyoung Kim
- Bio/Molecular Informatics Center, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea; (M.K.K.); (J.H.A.)
| | - Joong Hoon Ahn
- Bio/Molecular Informatics Center, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea; (M.K.K.); (J.H.A.)
- Department of Integrative Bioscience and Biotechnology, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Seri Jeong
- Department of Laboratory Medicine, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea;
| | - Ki-Ho Park
- Department of Infectious Disease, Kyung Hee University School of Medicine, Seoul 02447, Republic of Korea
| | - Youhoon Chong
- Bio/Molecular Informatics Center, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea; (M.K.K.); (J.H.A.)
- Department of Integrative Bioscience and Biotechnology, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| |
Collapse
|
14
|
Khataybeh B, Jaradat Z, Ababneh Q. Anti-bacterial, anti-biofilm and anti-quorum sensing activities of honey: A review. JOURNAL OF ETHNOPHARMACOLOGY 2023; 317:116830. [PMID: 37400003 DOI: 10.1016/j.jep.2023.116830] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/31/2023] [Accepted: 06/20/2023] [Indexed: 07/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Man has used honey to treat diseases since ancient times, perhaps even before the history of medicine itself. Several civilizations have utilized natural honey as a functional and therapeutic food to ward off infections. Recently, researchers worldwide have been focusing on the antibacterial effects of natural honey against antibiotic-resistant bacteria. AIM OF THE STUDY This review aims to summarize research on the use of honey properties and constituents with their anti-bacterial, anti-biofilm, and anti-quorum sensing mechanisms of action. Further, honey's bacterial products, including probiotic organisms and antibacterial agents which are produced to curb the growth of other competitor microorganisms is addressed. MATERIALS AND METHODS In this review, we have provided a comprehensive overview of the antibacterial, anti-biofilm, and anti-quorum sensing activities of honey and their mechanisms of action. Furthermore, the review addressed the effects of antibacterial agents of honey from bacterial origin. Relevant information on the antibacterial activity of honey was obtained from scientific online databases such as Web of Science, Google Scholar, ScienceDirect, and PubMed. RESULTS Honey's antibacterial, anti-biofilm, and anti-quorum sensing activities are mostly attributed to four key components: hydrogen peroxide, methylglyoxal, bee defensin-1, and phenolic compounds. The performance of bacteria can be altered by honey components, which impact their cell cycle and cell morphology. To the best of our knowledge, this is the first review that specifically summarizes every phenolic compound identified in honey along with their potential antibacterial mechanisms of action. Furthermore, certain strains of beneficial lactic acid bacteria such as Bifidobacterium, Fructobacillus, and Lactobacillaceae, as well as Bacillus species can survive and even grow in honey, making it a potential delivery system for these agents. CONCLUSION Honey could be regarded as one of the best complementary and alternative medicines. The data presented in this review will enhance our knowledge of some of honey's therapeutic properties as well as its antibacterial activities.
Collapse
Affiliation(s)
- Batool Khataybeh
- Department of Nutrition and Food Technology, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Ziad Jaradat
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, 22110, Jordan.
| | - Qutaiba Ababneh
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, 22110, Jordan
| |
Collapse
|
15
|
Liang X, Tu C, Li Y, Sun J, Zhao R, Ran J, Jiao L, Huang J, Li J. Inhibitory mechanism of quercetin on Alicyclobacillus acidoterrestris. Front Microbiol 2023; 14:1286187. [PMID: 38033555 PMCID: PMC10684910 DOI: 10.3389/fmicb.2023.1286187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/12/2023] [Indexed: 12/02/2023] Open
Abstract
In this the antibacterial of quercetin against Alicyclobacillus acidoterrestris was evaluated by measuring the minimal inhibitory concentration (MIC) and minimal bactericidal concentration (MBC). Subsequently, the effect of quercetin on A. acidoterrestris cell membrane was evaluated through scanning electron microscopy (SEM), surface hydrophobicity determination, diacetate fluorescein staining and propidium iodide (PI) staining. Additionally, the effects of quercetin on intracellular macromolecules and cell metabolism were explored by measuring the culture medium protein, bacterial protein and intracellular sodium and potassium adenosine triphosphate (ATP) enzyme activity. The results revealed that quercetin exhibited the MIC and MBC values of 100 ug/mL and 400 ug/mL, respectively, against A. acidoterrestris. The SEM results revealed that quercetin could induce irreversible damage to the cell membrane effectively. Moreover, quercetin could enhance the surface hydrophobicity of A. acidoterrestris. The results of flow cytometry and fluorescence microscopy analyses revealed that quercetin could promote cell damage by altering the cell membrane permeability of A. acidoterrestris, inducing the release of nucleic acid substances from the cells. Furthermore, the determination of protein content in the culture medium, bacterial protein content, and the Na(+)/K(+)-ATPase activity demonstrated that quercetin could reduce the intracellular protein content and impedes protein expression and ATPase synthesis effectively, leading to apoptosis.
Collapse
Affiliation(s)
- Xinhong Liang
- School of Food Science, Henan Institute of Science and Technology, Xinxiang, China
| | - Cunjian Tu
- School of Food Science, Henan Institute of Science and Technology, Xinxiang, China
| | - Yongchao Li
- School of Life Sciences, Henan Institute of Science and Technology, Xinxiang, China
| | - Junliang Sun
- School of Food Science, Henan Institute of Science and Technology, Xinxiang, China
| | - Ruixiang Zhao
- School of Food Science, Henan Institute of Science and Technology, Xinxiang, China
| | - Junjian Ran
- School of Food Science, Henan Institute of Science and Technology, Xinxiang, China
| | - Lingxia Jiao
- School of Food Science, Henan Institute of Science and Technology, Xinxiang, China
| | - Junchao Huang
- School of Food Science, Henan Institute of Science and Technology, Xinxiang, China
| | - Junrui Li
- School of Food Science, Henan Institute of Science and Technology, Xinxiang, China
| |
Collapse
|
16
|
Benin BM, Hillyer T, Crugnale AS, Fulk A, Thomas CA, Crowder MW, Smith MA, Shin WS. Taxifolin as a Metallo-β-Lactamase Inhibitor in Combination with Augmentin against Verona Imipenemase 2 Expressing Pseudomonas aeruginosa. Microorganisms 2023; 11:2653. [PMID: 38004664 PMCID: PMC10673258 DOI: 10.3390/microorganisms11112653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
Among the various mechanisms that bacteria use to develop antibiotic resistance, the multiple expression of β-lactamases is particularly problematic, threatening public health and increasing patient mortality rates. Even if a combination therapy-in which a β-lactamase inhibitor is administered together with a β-lactam antibiotic-has proven effective against serine-β-lactamases, there are no currently approved metallo-β-lactamase inhibitors. Herein, we demonstrate that quercetin and its analogs are promising starting points for the further development of safe and effective metallo-β-lactamase inhibitors. Through a combined computational and in vitro approach, taxifolin was found to inhibit VIM-2 expressing P. aeruginosa cell proliferation at <4 μg/mL as part of a triple combination with amoxicillin and clavulanate. Furthermore, we tested this combination in mice with abrasive skin infections. Together, these results demonstrate that flavonol compounds, such as taxifolin, may be developed into effective metallo-β-lactamase inhibitors.
Collapse
Affiliation(s)
- Bogdan M. Benin
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (B.M.B.); (T.H.); (A.F.); (M.A.S.)
| | - Trae Hillyer
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (B.M.B.); (T.H.); (A.F.); (M.A.S.)
| | - Aylin S. Crugnale
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (B.M.B.); (T.H.); (A.F.); (M.A.S.)
| | - Andrew Fulk
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (B.M.B.); (T.H.); (A.F.); (M.A.S.)
| | - Caitlyn A. Thomas
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH 45056, USA; (C.A.T.); (M.W.C.)
| | - Michael W. Crowder
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH 45056, USA; (C.A.T.); (M.W.C.)
| | - Matthew A. Smith
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (B.M.B.); (T.H.); (A.F.); (M.A.S.)
- Akron Children’s Hospital, Rebecca D. Considine Research Institute, Akron, OH 44302, USA
| | - Woo Shik Shin
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (B.M.B.); (T.H.); (A.F.); (M.A.S.)
| |
Collapse
|
17
|
Sweet R, Booth C, Gotts K, Grove SF, Kroon PA, Webber M. Comparison of Antibacterial Activity of Phytochemicals against Common Foodborne Pathogens and Potential for Selection of Resistance. Microorganisms 2023; 11:2495. [PMID: 37894153 PMCID: PMC10609411 DOI: 10.3390/microorganisms11102495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
Antimicrobial resistance is now commonly observed in bacterial isolates from multiple settings, compromising the efficacy of current antimicrobial agents. Therefore, there is an urgent requirement for efficacious novel antimicrobials to be used as therapeutics, prophylactically or as preservatives. One promising source of novel antimicrobial chemicals is phytochemicals, which are secondary metabolites produced by plants for numerous purposes, including antimicrobial defence. In this report, we compare the bioactivity of a range of phytochemical compounds, testing their ability to directly inhibit growth or to potentiate other antimicrobials against Salmonella enterica Typhimurium, Pseudomonas aeruginosa, Listeria monocytogenes, and Staphylococcus aureus. We found that nine compounds displayed consistent bioactivity either as direct antimicrobials or as potentiators. Thymol at 0.5 mg/mL showed the greatest antimicrobial effect and significantly reduced the growth of all species, reducing viable cell populations by 66.8%, 43.2%, 29.5%, and 70.2% against S. enterica Typhimurium, S. aureus, P. aeruginosa, and L. monocytogenes, respectively. Selection of mutants with decreased susceptibility to thymol was possible for three of the pathogens, at a calculated rate of 3.77 × 10-8, and characterisation of S. enterica Typhimurium mutants showed a low-level MDR phenotype due to over-expression of the major efflux system AcrAB-TolC. These data show that phytochemicals can have strong antimicrobial activity, but emergence of resistance should be evaluated in any further development.
Collapse
Affiliation(s)
- Ryan Sweet
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK; (R.S.); (C.B.); (P.A.K.)
| | - Catherine Booth
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK; (R.S.); (C.B.); (P.A.K.)
| | - Kathryn Gotts
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK; (R.S.); (C.B.); (P.A.K.)
| | | | - Paul A. Kroon
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK; (R.S.); (C.B.); (P.A.K.)
| | - Mark Webber
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK; (R.S.); (C.B.); (P.A.K.)
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| |
Collapse
|
18
|
Miklasińska-Majdanik M, Kępa M, Wąsik TJ, Zapletal-Pudełko K, Klim M, Wojtyczka RD. The Direction of the Antibacterial Effect of Rutin Hydrate and Amikacin. Antibiotics (Basel) 2023; 12:1469. [PMID: 37760765 PMCID: PMC10525965 DOI: 10.3390/antibiotics12091469] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
The aim of the presented study was to examine the in vitro antimicrobial activity of rutin hydrate (RH) alone and in combination with amikacin against 12 reference strains of Gram-positive and Gram-negative bacteria. The antibacterial activity assay was evaluated in the concentration range of 2-2048 µg/mL. A serial microdilution method was used to determine the minimal inhibitory concentration (MIC) of the examined compound against reference strains. RH showed varying potential against the tested strains with MICs ranging from 128 to 1024 µg/mL. In order to examine the combinatory profile of RH and amikacin, the fractional inhibitory concentrations (FICs) were determined. The RH-amikacin combination was more active against Gram-negative bacteria where four synergism and two additive interactions were noted. For four out of six Gram-positive isolates, an indifferent effect of RH and amikacin was demonstrated, and for two strains, the tested combination had an additive effect. The results of this study showed that RH possesses antimicrobial potential in vitro towards the tested reference isolates. Moreover, it shows a promising combined effect with amikacin against Gram-negative bacteria.
Collapse
Affiliation(s)
- Maria Miklasińska-Majdanik
- Department of Microbiology and Virology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Jagiellońska 4, 41-200 Sosnowiec, Poland; (M.K.); (K.Z.-P.); (M.K.); (R.D.W.)
| | - Małgorzata Kępa
- Department of Microbiology and Virology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Jagiellońska 4, 41-200 Sosnowiec, Poland; (M.K.); (K.Z.-P.); (M.K.); (R.D.W.)
| | - Tomasz J. Wąsik
- Department of Medical Microbiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Medyków 18, 40-752 Katowice, Poland;
| | - Karolina Zapletal-Pudełko
- Department of Microbiology and Virology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Jagiellońska 4, 41-200 Sosnowiec, Poland; (M.K.); (K.Z.-P.); (M.K.); (R.D.W.)
| | - Magdalena Klim
- Department of Microbiology and Virology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Jagiellońska 4, 41-200 Sosnowiec, Poland; (M.K.); (K.Z.-P.); (M.K.); (R.D.W.)
| | - Robert D. Wojtyczka
- Department of Microbiology and Virology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Jagiellońska 4, 41-200 Sosnowiec, Poland; (M.K.); (K.Z.-P.); (M.K.); (R.D.W.)
| |
Collapse
|
19
|
Luo S, Kang X, Luo X, Li C, Wang G. Study on the inhibitory effect of quercetin combined with gentamicin on the formation of Pseudomonas aeruginosa and its bioenvelope. Microb Pathog 2023; 182:106274. [PMID: 37516213 DOI: 10.1016/j.micpath.2023.106274] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/13/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023]
Abstract
OBJECTIVE The potential effects of quercetin and gentamicin combination on the bacteriostatic activity and biofilm formation of Pseudomonas aeruginosa (PA) were examined, and the findings provided a theoretical basis for the development of quercetin as a new biofilm inhibitor. METHODS The minimum inhibitory concentration (MIC) of eight PAs was determined by microdilution method and the partial inhibitory concentration index (FICI) of the combined drug was analyzed by micro-dilution method. Thereafter, the lowest film inhibitory concentration (MBIC) of quercetin and gentamicin alone and in combination was evaluated by crystal violet staining. Finally, scanning electron microscopy (SEM) and laser confocal microscopy (CLSM) were used to decipher the inhibitory effect of the combination on biofilm formation. OUTCOME The antibacterial activity of quercetin alone was relatively weak, but after combination with gentamicin, the antibacterial activity was significantly enhanced, as evident by FICI of 0.28 and 0.53 and manifested as synergistic or additive effect, which indicated that quercetin can enhance gentamicin antibacterial activity. The results of crystal violet staining revealed that quercetin and gentamicin alone exhibited a similar biofilm formation inhibitory effect, but the inhibitory effect was substantially weaker, and the antibiofilm activity was stronger and exhibited a dose-dependent response after the combination of the two with 1/2FICI. The results of scanning electron microscopy and laser confocal microscopy also showed that the treatment of PA biofilm after combining quercetin and gentamicin with 1/2FICI could completely destroy the spatial structure of the complete biofilm, significantly reduce the thickness of bacteria, and markedly reduce the proportion of viable bacteria in the membrane. CONCLUSION The combination of quercetin and gentamicin can effectively inhibit the formation of PA as well as its biofilm, and exhibit synergistic and additive effects.
Collapse
Affiliation(s)
- Shuangyan Luo
- School of Agriculture, Ningxia University, 750021, Yinchuan, China
| | - Xinyun Kang
- School of Agriculture, Ningxia University, 750021, Yinchuan, China
| | - Xiaofeng Luo
- School of Agriculture, Ningxia University, 750021, Yinchuan, China
| | - Caixia Li
- School of Agriculture, Ningxia University, 750021, Yinchuan, China
| | - Guiqin Wang
- School of Agriculture, Ningxia University, 750021, Yinchuan, China.
| |
Collapse
|
20
|
Odabaş Köse E, Koyuncu Özyurt Ö, Bilmen S, Er H, Kilit C, Aydemir E. Quercetin: Synergistic Interaction with Antibiotics against Colistin-Resistant Acinetobacter baumannii. Antibiotics (Basel) 2023; 12:antibiotics12040739. [PMID: 37107101 PMCID: PMC10135113 DOI: 10.3390/antibiotics12040739] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
Infections caused by resistant strains of Acinetobacter baumannii are now a global problem that requires the immediate development of new antimicrobial drugs. Combination therapy is one of the strategies used to solve this problem. Based on this information, the purpose of this study was to determine whether quercetin (QUE), in combination with three antibiotics, is effective against colistin-resistant A. baumannii strains (ColR-Ab). The effects of the combination of QUE with colistin (COL), amikacin (AMK), and meropenem (MEM) were evaluated according to the checkerboard synergy test. The combinations of QUE + COL and QUE + AMK showed synergistic activity on ColR-Ab strains with FICI values in the range of 0.1875-0.5 and 0.1875-0.2825, respectively. A 4- to 16-fold decrease in COL MIC and a 16- to 64-fold decrease in AMK MIC values were detected. Synergistic activity was confirmed by the time-kill test, and these combinations were found to be bactericidal at the end of 24 h. According to spectrophotometric measurements, the combinations of QUE + COL and QUE + AMK induced membrane damage, leading to the leakage of nucleic acids. Cell lysis and cell death were confirmed with SEM observations. The detected synergy offers an opportunity for the future development of treatment strategies for potential infections caused by ColR-Ab strains.
Collapse
Affiliation(s)
- Elif Odabaş Köse
- Vocational School of Health Services, Akdeniz University, 07058 Antalya, Turkey
| | - Özlem Koyuncu Özyurt
- Department of Medical Microbiology, Faculty of Medicine, Akdeniz University, 07070 Antalya, Turkey
| | - Süreyya Bilmen
- Vocational School of Health Services, Akdeniz University, 07058 Antalya, Turkey
| | - Hakan Er
- Vocational School of Health Services, Akdeniz University, 07058 Antalya, Turkey
- Department of Biophysics, Faculty of Medicine, Akdeniz University, 07070, Antalya, Turkey
| | - Cansu Kilit
- Department of Biology, Faculty of Science, Akdeniz University, 07070 Antalya, Turkey
| | - Esra Aydemir
- Department of Biology, Faculty of Science, Akdeniz University, 07070 Antalya, Turkey
| |
Collapse
|
21
|
Zhang Y, Chen C, Cheng B, Gao L, Qin C, Zhang L, Zhang X, Wang J, Wan Y. Discovery of Quercetin and Its Analogs as Potent OXA-48 Beta-Lactamase Inhibitors. Front Pharmacol 2022; 13:926104. [PMID: 35814247 PMCID: PMC9258905 DOI: 10.3389/fphar.2022.926104] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Carbapenem resistance in Enterobacteriaceae caused by OXA-48 β-lactamase is a growing global health threat and has rapidly spread in many regions of the world. Developing inhibitors is a promising way to overcome antibiotic resistance. However, there are few options for problematic OXA-48. Here we identified quercetin, fisetin, luteolin, 3′,4′,7-trihydroxyflavone, apigenin, kaempferol, and taxifolin as potent inhibitors of OXA-48 with IC50 values ranging from 0.47 to 4.54 μM. Notably, the structure-activity relationship revealed that the substitute hydroxyl groups in the A and B rings of quercetin and its structural analogs improved the inhibitory effect against OXA-48. Mechanism studies including enzymatic kinetic assay, isothermal titration calorimetry (ITC), and surface plasmon resonance (SPR) analysis demonstrated that quercetin reversibly inhibited OXA-48 through a noncompetitive mode. Molecular docking suggested that hydroxyl groups at the 3′, 4′ and 7 positions in flavonoids formed hydrogen-bonding interactions with the side chains of Thr209, Ala194, and Gln193 in OXA-48. Quercetin, fisetin, luteolin, and 3′,4′,7-trihydroxyflavone effectively restored the antibacterial efficacy of piperacillin or imipenem against E. coli producing OXA-48, resulting in 2–8-fold reduction in MIC. Moreover, quercetin combined with piperacillin showed antimicrobial efficacy in mice infection model. These studies provide potential lead compounds for the development of β-lactamase inhibitors and in combination with β-lactams to combat OXA-48 producing pathogen.
Collapse
Affiliation(s)
- Yuejuan Zhang
- Microbiology Institute of Shaanxi, Xi’an, China
- Engineering Center of Qinling Mountains Natural Products, Shaanxi Academy of Sciences, Xi’an, China
| | - Cheng Chen
- College of Forestry, Northwest A&F University, Yangling, China
| | - Bin Cheng
- MOE Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Lei Gao
- Microbiology Institute of Shaanxi, Xi’an, China
- Engineering Center of Qinling Mountains Natural Products, Shaanxi Academy of Sciences, Xi’an, China
| | - Chuan Qin
- Microbiology Institute of Shaanxi, Xi’an, China
- Engineering Center of Qinling Mountains Natural Products, Shaanxi Academy of Sciences, Xi’an, China
| | - Lixia Zhang
- Clinical Laboratory, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Xu Zhang
- Microbiology Institute of Shaanxi, Xi’an, China
- Engineering Center of Qinling Mountains Natural Products, Shaanxi Academy of Sciences, Xi’an, China
| | - Jun Wang
- Microbiology Institute of Shaanxi, Xi’an, China
- Engineering Center of Qinling Mountains Natural Products, Shaanxi Academy of Sciences, Xi’an, China
| | - Yi Wan
- Microbiology Institute of Shaanxi, Xi’an, China
- Engineering Center of Qinling Mountains Natural Products, Shaanxi Academy of Sciences, Xi’an, China
- *Correspondence: Yi Wan,
| |
Collapse
|
22
|
Li J, Feng S, Liu X, Jia X, Qiao F, Guo J, Deng S. Effects of Traditional Chinese Medicine and its Active Ingredients on Drug-Resistant Bacteria. Front Pharmacol 2022; 13:837907. [PMID: 35721131 PMCID: PMC9204478 DOI: 10.3389/fphar.2022.837907] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
The increasing and widespread application of antibacterial drugs makes antibiotic resistance a prominent and growing concern in clinical practice. The emergence of multidrug-resistant bacteria presents a global threat. However, the development and use of novel antibacterial agents involves time-consuming and costly challenges that may lead to yet further drug resistance. More recently, researchers have turned to traditional Chinese medicine to stem the rise of antibiotic resistance in pathogens. Many studies have shown traditional Chinese medicines to have significant bacteriostatic and bactericidal effects, with the advantage of low drug resistance. Some of which when combined with antibiotics, have also demonstrated antibacterial activity by synergistic effect. Traditional Chinese medicine has a variety of active components, including flavonoids, alkaloids, phenols, and quinones, which can inhibit the growth of drug-resistant bacteria and be used in combination with a variety of antibiotics to treat various drug-resistant bacterial infections. We reviewed the interaction between the active ingredients of traditional Chinese medicines and antibiotic-resistant bacteria. At present, flavonoids and alkaloids are the active ingredients that have been most widely studied, with significant synergistic activity demonstrated when used in combination with antibiotics against drug-resistant bacteria. The reviewed studies show that traditional Chinese medicine and its active ingredients have antimicrobial activity on antibiotic-resistant bacteria, which may enhance the susceptibility of antibiotic-resistant bacteria, potentially reduce the required dosage of antibacterial agents and the rate of drug resistance. Our results provide direction for finding and developing alternative methods to counteract drug-resistant bacteria, offering a new therapeutic strategy for tackling antibiotic resistance.
Collapse
Affiliation(s)
- Jimin Li
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Non-Coding RNA and Drug Discovery Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu, China
| | - Shanshan Feng
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xin Liu
- School of Public Health, Chengdu Medical College, Chengdu, China
| | - Xu Jia
- Non-Coding RNA and Drug Discovery Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu, China.,School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Fengling Qiao
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinlin Guo
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Key Laboratory of Systematic Research of Distinctive Chinese Medicine Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shanshan Deng
- Non-Coding RNA and Drug Discovery Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu, China.,School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| |
Collapse
|
23
|
Sundaramoorthy NS, Shankaran P, Gopalan V, Nagarajan S. New tools to mitigate drug resistance in Enterobacteriaceae - Escherichia coli and Klebsiella pneumoniae. Crit Rev Microbiol 2022:1-20. [PMID: 35649163 DOI: 10.1080/1040841x.2022.2080525] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Treatment to common bacterial infections are becoming ineffective of late, owing to the emergence and dissemination of antibiotic resistance globally. Escherichia coli and Klebsiella pneumoniae are the most notorious microorganisms and are among the critical priority pathogens listed by WHO in 2017. These pathogens are the predominant cause of sepsis, urinary tract infections (UTIs), pneumonia, meningitis and pyogenic liver abscess. Concern arises due to the resistance of bacteria to most of the beta lactam antibiotics like penicillin, cephalosporin, monobactams and carbapenems, even to the last resort antibiotics like colistin. Preventing influx by modulation of porins, extruding the antibiotics by overexpression of efflux pumps, mutations of drug targets/receptors, biofilm formation, altering the drug molecules and rendering them ineffective are few resistance mechanisms that are adapted by Enterobacteriaeceae upon exposure to antibiotics. The situation is exacerbated due to the process of horizontal gene transfer (HGT), wherein the genes encoding resistance mechanisms are transferred to the neighbouring bacteria through plasmids/phages/uptake of free DNA. Carbapenemases, other beta lactamases and mcr genes coding for colistin resistance are widely disseminated leading to limited/no therapeutic options against those infections. Development of new antibiotics can be viewed as a possible solution but it involves major investment, time and labour despite which, the bacteria can easily adapt to the new antibiotic and evolve resistance in a relatively short time. Targeting the resistance mechanisms can be one feasible alternative to tackle these multidrug resistant (MDR) pathogens. Removal of plasmid (plasmid curing) causing resistance, use of bacteriophages and bacteriotherapy can be other potential approaches to combat infections caused by MDR E. coli and K. pneumoniae. The present review discusses the efficacies of these therapies in mitigating these infections, which can be potentially used as an adjuvant therapy along with existing antibiotics.
Collapse
Affiliation(s)
- Niranjana Sri Sundaramoorthy
- Center for Research on Infectious Diseases, School of Chemical and Biotechnology, SASTRA deemed University, Thanjavur, Tamil Nadu, India
| | - Prakash Shankaran
- Center for Research on Infectious Diseases, School of Chemical and Biotechnology, SASTRA deemed University, Thanjavur, Tamil Nadu, India
| | - Vidhya Gopalan
- Department of Virology, Kings Institute of Preventative Medicine, Guindy, Chennai, Tamil Nadu, India
| | - Saisubramanian Nagarajan
- Center for Research on Infectious Diseases, School of Chemical and Biotechnology, SASTRA deemed University, Thanjavur, Tamil Nadu, India
| |
Collapse
|
24
|
Nguyen TLA, Bhattacharya D. Antimicrobial Activity of Quercetin: An Approach to Its Mechanistic Principle. Molecules 2022; 27:molecules27082494. [PMID: 35458691 PMCID: PMC9029217 DOI: 10.3390/molecules27082494] [Citation(s) in RCA: 177] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/04/2022] [Accepted: 04/07/2022] [Indexed: 02/04/2023] Open
Abstract
Quercetin, an essential plant flavonoid, possesses a variety of pharmacological activities. Extensive literature investigates its antimicrobial activity and possible mechanism of action. Quercetin has been shown to inhibit the growth of different Gram-positive and Gram-negative bacteria as well as fungi and viruses. The mechanism of its antimicrobial action includes cell membrane damage, change of membrane permeability, inhibition of synthesis of nucleic acids and proteins, reduction of expression of virulence factors, mitochondrial dysfunction, and preventing biofilm formation. Quercetin has also been shown to inhibit the growth of various drug-resistant microorganisms, thereby suggesting its use as a potent antimicrobial agent against drug-resistant strains. Furthermore, certain structural modifications of quercetin have sometimes been shown to enhance its antimicrobial activity compared to that of the parent molecule. In this review, we have summarized the antimicrobial activity of quercetin with a special focus on its mechanistic principle. Therefore, this review will provide further insights into the scientific understanding of quercetin’s mechanism of action, and the implications for its use as a clinically relevant antimicrobial agent.
Collapse
|
25
|
Alizadeh SR, Ebrahimzadeh MA. O-substituted quercetin derivatives: Structural classification, drug design, development, and biological activities, a review. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
26
|
Alizadeh SR, Ebrahimzadeh MA. O-Glycoside quercetin derivatives: Biological activities, mechanisms of action, and structure-activity relationship for drug design, a review. Phytother Res 2021; 36:778-807. [PMID: 34964515 DOI: 10.1002/ptr.7352] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/27/2021] [Accepted: 11/10/2021] [Indexed: 02/06/2023]
Abstract
Quercetin as a valuable natural flavonoid has shown extensive biological activities, including anticancer, antioxidant, antibacterial, antiinflammatory, anti-Alzheimer, antifungal, antiviral, antithalassemia, iron chelation, antiobesity, antidiabetic, antihypertension, and antiphospholipase A2 (PLA2) activities, by the modulation of various targets and signaling pathways that have attracted much attention. However, the low solubility and poor bioavailability of quercetin have limited its applications; therefore, the researchers have tried to design and synthesize many new derivatives of quercetin through different strategies to modify quercetin restrictions and improve its biological activities. This review categorized the O-glycoside derivatives of Quercetin into two main classes, 3-O-glycoside and other O-glycoside derivatives. Also, it studied biological activities, structure-activity relationship (SAR), and the action mechanism of O-glycoside quercetin derivatives. Overall, we summarized past and present research for discovering new potent lead compounds.
Collapse
Affiliation(s)
- Seyedeh Roya Alizadeh
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Ali Ebrahimzadeh
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
27
|
Alizadeh SR, Ebrahimzadeh MA. Quercetin derivatives: Drug design, development, and biological activities, a review. Eur J Med Chem 2021; 229:114068. [PMID: 34971873 DOI: 10.1016/j.ejmech.2021.114068] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/24/2021] [Accepted: 12/19/2021] [Indexed: 02/08/2023]
Abstract
More studies are needed to develop new drugs for problems associated with drug resistance and unfavorable side effects. The natural flavonoid of quercetin revealed a wide range of biological activities by the modulation of various targets and signaling pathways. However, quercetin's low solubility and poor bioavailability have restricted its applicability; as a result, researchers have attempted to design and synthesize numerous novel quercetin derivatives using various methodologies in order to modify quercetin's constraints; the physico-chemical properties of quercetin's molecular scaffold make it appealing for drug development; low molecular mass and chemical groups are two of these characteristics. Therefore, the biological activities of quercetin derivatives, as well as the relationship between activity and chemical structure and their mechanism of action, were investigated. These quercetin-based molecules could be valuable in the creation and discovery of medications for a number of diseases.
Collapse
Affiliation(s)
- Seyedeh Roya Alizadeh
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Ali Ebrahimzadeh
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
28
|
Wang M, Fu D, Yao L, Li J. Theoretical Study of the Molecular Mechanism of Maxingyigan Decoction Against COVID-19: Network Pharmacology-based Strategy. Comb Chem High Throughput Screen 2021; 24:294-305. [PMID: 32767929 DOI: 10.2174/1386207323666200806164635] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/14/2020] [Accepted: 06/23/2020] [Indexed: 12/15/2022]
Abstract
AIM AND OBJECTIVE Maxingyigan (MXYG) decoction is a traditional Chinese medicine (TCM) prescription. However, how MXYG acts against coronavirus disease 2019 (COVID-19) is not known. We investigated the active ingredients and the therapeutic targets of MXYG decoction against COVID-19. METHODS A network pharmacology strategy involving drug-likeness evaluation, prediction of oral bioavailability, network analyses, and virtual molecular docking was used to predict the mechanism of action of MXYG against COVID-19. RESULTS Thirty-three core COVID-19-related targets were identified from 1023 gene targets through analyses of protein-protein interactions. Eighty-six active ingredients of MXYG decoction hit by 19 therapeutic targets were screened out by analyses of a compound-compound target network. Via network topology, three "hub" gene targets (interleukin (IL-6), caspase-3, IL-4) and three key components (quercetin, formononetin, luteolin) were recognized and verified by molecular docking. Compared with control compounds (ribavirin, arbidol), the docking score of quercetin to the IL-6 receptor was highest, with a score of 5. Furthermore, the scores of three key components to SARS-CoV-2 are large as 4, 5, and 5, respectively, which are even better than those of ribavirin at 3. Bioinformatics analyses revealed that MXYG could prevent and treat COVID-19 through anti-inflammatory and immunity-based actions involving activation of T cells, lymphocytes, and leukocytes, as well as cytokine-cytokine-receptor interaction, and chemokine signaling pathways. CONCLUSION The hub genes of COVID-19 helped to reveal the underlying pathogenesis and therapeutic targets of COVID-19. This study represents the first report on the molecular mechanism of MXYG decoction against COVID-19.
Collapse
Affiliation(s)
- Mingzhu Wang
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Deyu Fu
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Yao
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianhua Li
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
29
|
Vallejo CV, Minahk CJ, Rollán GC, Rodríguez-Vaquero MJ. Inactivation of Listeria monocytogenes and Salmonella Typhimurium in strawberry juice enriched with strawberry polyphenols. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2021; 101:441-448. [PMID: 32648586 DOI: 10.1002/jsfa.10653] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 07/01/2020] [Accepted: 07/10/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Low molecular-weight phenolic fractions (LMPFs) were extracted from Albion (LMPF-A) and Camarosa (LMPF-C) strawberry cultivars. Their antibacterial activity against Listeria monocytogenes and Salmonella Typhimurium cocktails in vitro and in vivo was investigated using strawberry juice as a food model. This study also sought to determine their antibacterial mechanism. RESULTS Quercetin was identified as a principal compound in both phenolic fractions. The minimum bactericide concentration (MBC) values were 750 and 850 μg mL-1 (LMPF-C) and 800 and 950 μg mL-1 (LMPF-A) against S.Typhimurium and L. monocytogenes, respectively. The possible antibacterial activity of the phenolic extracts could be related to the release of phosphate and potassium ions, the effect of the disruption of membrane integrity on L. monocytogenes, and the effect of the inhibition of dihydronicotinamide adenine dinucleotide (NADH) oxidase activity on S. Typhimurium. Quercetin and kaempferol were the most active compounds in producing bacterial damage. Strawberry juice supplemented with the phenolic fractions and incubated at 37, 20, and 4 °C reduced bacterial viability; moreover, after treatment with the phenolic fraction at the lowest temperature, no viable cells were detected after 7 days' incubation. Salmonella was more sensitive to the supplements than Listeria in strawberry juice. CONCLUSIONS This study could form the basis for the development of natural antibacterial agents that could be included in natural juice or used by the pharmaceutical industry. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Claudia Verónica Vallejo
- Institute of Microbiology, Faculty of Biochemistry, Chemistry and Pharmacy (FBQF), National University of Tucumán (UNT), Tucumán, Argentina
- CONICET NOA Sur., Tucumán, Argentina
| | | | | | - María José Rodríguez-Vaquero
- Institute of Microbiology, Faculty of Biochemistry, Chemistry and Pharmacy (FBQF), National University of Tucumán (UNT), Tucumán, Argentina
- CONICET NOA Sur., Tucumán, Argentina
| |
Collapse
|
30
|
Yang D, Wang T, Long M, Li P. Quercetin: Its Main Pharmacological Activity and Potential Application in Clinical Medicine. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8825387. [PMID: 33488935 PMCID: PMC7790550 DOI: 10.1155/2020/8825387] [Citation(s) in RCA: 296] [Impact Index Per Article: 59.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/24/2020] [Accepted: 12/14/2020] [Indexed: 12/23/2022]
Abstract
Quercetin is a flavonoid compound widely present in plants and exhibits a variety of biological activities. Research on quercetin has shown its potential for medical application. In this research, we elucidate its antioxidant mechanism and the broad-spectrum antibacterial and antiparasite properties; summarise its potential application in antioncology and cardiovascular protection and anti-immunosuppression treatment; and demonstrate its ability to alleviate the toxicity of mycotoxins. This research is expected to offer some insights and inspirations for the further study of quercetin, its properties, and the scientific basis for its better application in clinical practice.
Collapse
Affiliation(s)
- Dengyu Yang
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Tiancheng Wang
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Miao Long
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| | - Peng Li
- Key Laboratory of Zoonosis of Liaoning Province, College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
| |
Collapse
|
31
|
Zielińska S, Dziągwa-Becker M, Piątczak E, Jezierska-Domaradzka A, Brożyna M, Junka A, Kucharski M, Çiçek SS, Zidorn C, Matkowski A. Phytochemical Composition and Antimicrobial Activity of Corydalis solida and Pseudofumaria lutea. Molecules 2020; 25:E3591. [PMID: 32784618 PMCID: PMC7464254 DOI: 10.3390/molecules25163591] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/03/2020] [Accepted: 08/06/2020] [Indexed: 12/22/2022] Open
Abstract
Corydalis and Pseudofumaria are two closely related genera from the Papaveraceae subfamily Fumarioideae with Corydalis solida (C. solida) and Pseudofumaria lutea (P. lutea) as two representative species. Phytochemical analysis revealed significant differences in the quality and quantity of isoquinoline alkaloids, phenolic compounds and non-phenolic carboxylic acids between aerial and underground parts of both species. Using the Liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS) technique, 21 compounds were identified: five protoberberine derivatives, three protopine derivatives, four phenanthridine derivatives, as well as three carboxylic acids, two hydroxycinnamic acids, one chlorogenic acid, one phenolic aldehyde, and two flavonoids. Moroever, significant differences in the content of individual compounds were observed between the two studied species. The phytochemical profile of C. solida showed a higher variety of compounds that were present in lower amounts, whereas P. lutea extracts contained fewer compounds but in larger quantities. Protopine was one of the most abundant constituents in C. solida (440-1125 µg/g d.w.) and in P. lutea (1036-1934 µg/g d.w.). Moreover, considerable amounts of coptisine (1526 µg/g) and quercetin (3247 µg/g) were detected in the aerial parts of P. lutea. Extracts from aerial and underground parts of both species were also examined for the antimicrobial potential against S. aureus, P. aeruginosa and C. albicans. P. lutea herb extract was the most effective (MIC at 0.39 mg/L) against all three pathogens.
Collapse
Affiliation(s)
- Sylwia Zielińska
- Department of Pharmaceutical Biology, Wroclaw Medical University, Borowska 211, 50-556 Wrocław, Poland; (A.J.-D.); (A.M.)
| | - Magdalena Dziągwa-Becker
- Department of Weed Science and Tillage Systems, Institute of Soil Science and Plant, Cultivation State Research Institute, Orzechowa 61, 50-540 Wrocław, Poland; (M.D.-B.); (M.K.)
| | - Ewelina Piątczak
- Department of Biology and Pharmaceutical Botany, Medical University of Łódź, Muszyńskiego 1, 90-151 Łódź, Poland;
| | - Anna Jezierska-Domaradzka
- Department of Pharmaceutical Biology, Wroclaw Medical University, Borowska 211, 50-556 Wrocław, Poland; (A.J.-D.); (A.M.)
- Laboratory of Experimental Cultivation, Botanical Garden of Medicinal Plants, Wroclaw Medical University, Al. Jana Kochanowskiego 14, 50-556 Wroclaw, Poland
| | - Malwina Brożyna
- Pharmaceutical Microbiology and Parasitology, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland; (M.B.); (A.J.)
| | - Adam Junka
- Pharmaceutical Microbiology and Parasitology, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland; (M.B.); (A.J.)
| | - Mariusz Kucharski
- Department of Weed Science and Tillage Systems, Institute of Soil Science and Plant, Cultivation State Research Institute, Orzechowa 61, 50-540 Wrocław, Poland; (M.D.-B.); (M.K.)
| | - Serhat Sezai Çiçek
- Department of Pharmaceutical Biology, Kiel University, Gutenbergstraße 76, 24118 Kiel, Germany; (S.S.Ç.); (C.Z.)
| | - Christian Zidorn
- Department of Pharmaceutical Biology, Kiel University, Gutenbergstraße 76, 24118 Kiel, Germany; (S.S.Ç.); (C.Z.)
| | - Adam Matkowski
- Department of Pharmaceutical Biology, Wroclaw Medical University, Borowska 211, 50-556 Wrocław, Poland; (A.J.-D.); (A.M.)
- Laboratory of Experimental Cultivation, Botanical Garden of Medicinal Plants, Wroclaw Medical University, Al. Jana Kochanowskiego 14, 50-556 Wroclaw, Poland
| |
Collapse
|
32
|
Mohamed EH, Alghamdi YS, Mostafa Abdel-Hafez S, Soliman MM, Alotaibi SH, Hassan MY, Hany NAD, Amer HH. Susceptibility Assessment of Multidrug Resistant Bacteria to Natural Products. Dose Response 2020; 18:1559325820936189. [PMID: 32669983 PMCID: PMC7338649 DOI: 10.1177/1559325820936189] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/22/2020] [Accepted: 05/27/2020] [Indexed: 01/14/2023] Open
Abstract
Objective: The aim of this study was to examine the effect of some natural compounds
against multidrug-resistant bacteria. Methods: Forty-three bacterial strains were collected. Disc diffusion and minimum
inhibitory concentration (MIC) tests were carried out for natural compounds
including quercetin, Acacia nilotica, Syzygium
aromaticum, and Holothuria atra. Scanning
electron microscope analysis and bacterial DNA apoptosis assays were
performed. Results: Staphylococcus aureus strains were resistant to imipenim,
ampicillin, and penicillin. Most Escherichia coli strains
were resistant to amoxicillin, clavulanat, and ampicillin. Finally,
tigecycline was effective with Klebsiella pneumoniae and
was resistant to all antibiotics. Only S aromaticum had an
antibacterial effect on K pneumoniae. Most S
aureus strains were sensitive to S aromaticum,
A nilotica, and quercetin. All examined natural
extracts had no effect on E coli. Holothuria
atra had no effect on any of the strains tested. Minimum
inhibitory concentration and minimum bactericidal concentration values for
examined plants against S aureus were 6.25 to 12, 1.6 to
3.2, and 9.12 to 18.24 mg/mL, respectively. Syzygium
aromaticum was active against K pneumoniae
with an MIC of 12.5 mg/mL. Scanning electron microscope analysis performed
after 24 and 48 hours of incubation showed bacterial strains with distorted
shapes and severe cell wall damage. Syzygium aromaticum,
quercetin, and A nilotica showed clear fragmentations of
S aureus DNA. Conclusions: Current findings confirmed the beneficial effect of using natural products
such as clove (S aromaticum), quercetin, and A
nilotica as a promising therapy to overcome multidrug resistant
bacteria.
Collapse
Affiliation(s)
- Essam Hassan Mohamed
- Department of Biology, Turabah University College, Taif University, Saudi Arabia.,Department of Microbiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | | | - Salama Mostafa Abdel-Hafez
- Department of Biology, Turabah University College, Taif University, Saudi Arabia.,Immunobiology and Immunopharmacology Unit, Animal Reproduction Research Institute, Giza, Egypt
| | - Mohamed Mohamed Soliman
- Clinical Laboratories Sciences, Turabah University College, Taif University, Turabah, Saudi Arabia.,Biochemistry Department, Faculty of Veterinary Medicine, Benha University, Benha, Egypt
| | - Saad H Alotaibi
- Chemistry Department, Turabah University College, Turabah, Taif University, Saudi Arabia
| | - Magdy Yassin Hassan
- Department of Biology, Turabah University College, Taif University, Saudi Arabia.,Reproductive Disease Department, Animal Reproduction Research Institute, Giza, Egypt
| | | | - Hamada H Amer
- Chemistry Department, Turabah University College, Turabah, Taif University, Saudi Arabia.,Animal Medicine and Infectious Diseases Department, Faculty of Veterinary Medicine, University of Sadat City, Egypt
| |
Collapse
|