1
|
Okpala OE, Rondevaldova J, Kokoska L. Anti-inflammatory drugs as potential antimicrobial agents: a review. Front Pharmacol 2025; 16:1557333. [PMID: 40264668 PMCID: PMC12011823 DOI: 10.3389/fphar.2025.1557333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/13/2025] [Indexed: 04/24/2025] Open
Abstract
The association and causal role of infectious agents in chronic inflammatory diseases have major implications for public health, treatment, and prevention. Pharmacological treatment of combined infectious and inflammatory diseases requires the administration of multiple drugs, including antibiotics and anti-inflammatory drugs. However, this can cause adverse effects, and therefore, dual-action drugs need to be developed. Anti-inflammatory drugs that have already shown antimicrobial properties appear to be promising candidates. NSAIDs, namely aceclofenac, diclofenac, and ibuprofen, were tested in clinical trials with patients diagnosed with uncomplicated urinary tract infections (UTIs) and cellulitis. The administration of ibuprofen, a drug tested in the highest number of studies, resulted in symptom resolution in patients with UTIs. Additionally, ibuprofen caused a high survival rate in mice infected with Pseudomonas aeruginosa and demonstrated potent in vitro antibacterial effects against Bacillus cereus, Escherichia coli, and Staphylococcus aureus, including methicillin-resistant S. aureus (MRSA) (MIC 0.625-2.5 mg/L). For most anti-inflammatory drugs, only data showing their in vitro and in vivo antimicrobial effects are available. Among these, auranofin caused a high survival rate in mice infected with Enterococcus faecium, S. aureus, and Clostridioides difficile. It also produced a strong in vitro growth-inhibitory effect against Streptococcus agalactiae, S. pneumoniae, S. aureus, S. epidermidis, Bacillus subtilis, C. difficile, E. faecalis, E. faecium, and Mycobacterium tuberculosis (MIC 0.0015-5 mg/L). Similarly, aspirin caused a high survival rate in M. tuberculosis-infected mice and strong to moderate in vitro activity against E. coli, B. cereus, P. aeruginosa, Enterobacter aerogenes, Klebsiella pneumoniae and Salmonella choleraesuis (MIC 1.2-5 mg/L). Moreover, topical application of celecoxib resulted in a high reduction in MRSA burden in mice. However, it only caused moderate in vitro effects against S. epidermidis, S. aureus and Bacillus subitilis (MIC 16-64 mg/L). These data suggest that certain non-steroidal anti-inflammatory drugs (NSAIDs) are promising drug candidates for the development of dual-action drugs for the potential treatment of combined infectious and inflammatory diseases such as tuberculosis, musculoskeletal infections and UTIs. Nevertheless, future clinical trials must be conducted to ascertain the antibacterial effect of these NSAIDs before their practical use.
Collapse
Affiliation(s)
| | | | - Ladislav Kokoska
- Department of Crop Sciences and Agroforestry, Faculty of Tropical AgriSciences, Czech University of Life Sciences Prague, Prague, Czechia
| |
Collapse
|
2
|
Pérez-Lavalle L, Borges A, Gomes IB, Carrasco E, Valero A, Simões M. The ability of Salmonella enterica subsp. enterica strains to form biofilms on abiotic surfaces and their susceptibility to selected essential oil components. Lett Appl Microbiol 2025; 78:ovaf032. [PMID: 40053511 DOI: 10.1093/lambio/ovaf032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 02/14/2025] [Accepted: 03/06/2025] [Indexed: 03/09/2025]
Abstract
The ability of Salmonella enterica subsp. enterica to persist and form biofilms on different surfaces can constitute a source of food contamination, being an issue of global concern. The objective of this study was to understand the biofilm formation profile of 14 S. enterica strains among different serovars and sources and to evaluate the ability of essential oil (EO) components (carveol, citronellol, and citronellal) to disinfect the biofilms formed on stainless steel and polypropylene surfaces. All the strains were able to form biofilms with counts between 5.34 to 6.78 log CFU cm-2. Then, the minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of EO components were evaluated on two selected strains. All compounds inhibited the growth of Salmonella Typhimurium (strain 1; MIC = 800-1000 µg ml-1) and Salmonella Enteritidis (strain 5; MIC = 400-1000 µg ml-1) and only carveol showed bactericidal activity against strains 1 and 5 (MBC = 1200 µg ml-1). Biofilms were exposed to the EO components at 10 × MIC for 30 min and polypropylene surfaces were more difficult to disinfect showing reductions between 0.9 and <1.2 log CFU cm-2. In general, the S. enterica biofilms demonstrated a significant tolerance to disinfection, demonstrating their high degree of recalcitrance on food processing surfaces.
Collapse
Affiliation(s)
- Liliana Pérez-Lavalle
- Faculty of Basic and Biomedical Sciences, Centro de Investigación e Innovación en Biodiversidad y Cambio Climático (Adaptia), Universidad Simón Bolívar, 080002 Barranquilla, Colombia
| | - Anabela Borges
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Inês B Gomes
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Elena Carrasco
- Department of Food Science and Technology, UIC Zoonosis y Enfermedades Emergentes (ENZOEM), CeiA3, Universidad de Córdoba, Campus Rabanales, 14014 Córdoba, Spain
| | - Antonio Valero
- Department of Food Science and Technology, UIC Zoonosis y Enfermedades Emergentes (ENZOEM), CeiA3, Universidad de Córdoba, Campus Rabanales, 14014 Córdoba, Spain
| | - Manuel Simões
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| |
Collapse
|
3
|
Abavisani M, Khoshrou A, Eshaghian S, Karav S, Sahebkar A. Overcoming antibiotic resistance: the potential and pitfalls of drug repurposing. J Drug Target 2025; 33:341-367. [PMID: 39485073 DOI: 10.1080/1061186x.2024.2424895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 10/18/2024] [Accepted: 10/27/2024] [Indexed: 11/03/2024]
Abstract
Since its emergence shortly after the discovery of penicillin, antibiotic resistance has escalated dramatically, posing a significant health threat and economic burden. Drug repositioning, or drug repurposing, involves identifying new therapeutic applications for existing drugs, utilising their established safety profiles and pharmacological data to swiftly provide effective treatments against resistant pathogens. Several drugs, including otilonium bromide, penfluridol, eltrombopag, ibuprofen, and ceritinib, have demonstrated potent antibacterial activity against multidrug-resistant (MDR) bacteria. These drugs can disrupt biofilms, damage bacterial membranes, and inhibit bacterial growth. The combination of repurposed drugs with conventional antibiotics can reduce the required dosage of individual drugs, mitigate side effects, and delay the development of resistance, making it a promising strategy against MDR bacteria such as Staphylococcus aureus, Klebsiella pneumoniae, Pseudomonas aeruginosa, and Escherichia coli. Despite its promise, drug repurposing faces challenges such as potential off-target effects, toxicity, and regulatory and intellectual property issues, necessitating rigorous evaluations and strategic solutions. This article aims to explore the potential of drug repurposing as a strategy to combat antibiotic resistance, examining its benefits, challenges, and future prospects. We address the legal, economic, and practical challenges associated with repurposing existing drugs, highlight successful examples, and propose solutions to enhance the efficacy and viability of this approach in combating MDR bacterial infections.
Collapse
Affiliation(s)
- Mohammad Abavisani
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Khoshrou
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Souzan Eshaghian
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale, Turkey
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Ferreira da Silva B, Gomes Pereira AM, Frota Araújo IM, Carvalho Aguiar FK, Mesquita Cajazeiras FF, Onassis Cardoso Viana Gomes A, Marinho MM, Rodrigues Teixeira AM, Marinho ES, Costa RA, Carneiro VA, Santos HSD. Structural characterization, cytotoxicity, antibiofilm activity, and synergistic potential with molecular docking analysis of ibuprofen-derived hydrazide against bacterial pathogens. Microb Pathog 2025; 199:107230. [PMID: 39689746 DOI: 10.1016/j.micpath.2024.107230] [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: 10/02/2024] [Revised: 11/29/2024] [Accepted: 12/12/2024] [Indexed: 12/19/2024]
Abstract
The study investigates the synthesis, characterization, and antibacterial activity of an ibuprofen-derived hydrazide (HIDZ). It was synthesized and characterized using NMR spectroscopy, DFT Calculations, and ADMET studies. Furthermore, HIDZ cytotoxicity on L929 cells was evaluated using the MTT reduction assay. Antibacterial activity was assessed against Gram-positive and Gram-negative strains through the microdilution method. The combinatory potential of HIDZ was performed using the checkerboard test with β-lactam antibiotics, oxacillin (OXA), meropenem (MER), and cefepime (CPM). Antibiofilm activity was evaluated for biofilm inhibition and disruption, particularly in combination with OXA. Molecular docking analysis examined HIDZ interactions with Thymidylate kinase, DNA Gyrase B, and DNA Topoisomerase IV subunit B. The global chemical reactivity descriptors analysis revealed significant variations in the atomic centers' susceptibility, highlighting the environment's importance in determining the reactive behavior of HIDZ. Pharmacokinetic predictions indicated efficient permeability across biological membranes, suggesting favorable bioavailability. MTT experiment showed that HIDZ caused cytotoxicity on higher concentrations over L929 fibroblasts. HIDZ exhibited superior activity against Gram-positive strains compared to ibuprofen, with lower MIC and MBC values. Both compounds were ineffective against Gram-negative strains. However, HIDZ was able to inhibit the biofilm formation of the most tested strains. The combinatory effect shows an additive effect between HIDZ and β-lactams. However, the HIDZ/OXA combination improved biofilm disruption, achieving up to a 92 % reduction in residual biofilm and cell viability compared to the control. Molecular docking simulations showed that HIDZ may interact with bacterial enzymes, improving antibiotic efficiency. The study suggests that HIDZ has promising potential as an antibacterial and antibiofilm agent, particularly against Gram-positive bacteria and in combination with β-lactam antibiotics.
Collapse
Affiliation(s)
- Benise Ferreira da Silva
- State University of Ceará, Northeast Network of Biotechnology Program (RENORBIO), Campus Itaperi, Fortaleza, Brazil; University Center INTA - UNINTA, Center for Bioprospecting and Applied Molecular Experimentation (NUBEM), Sobral, Brazil
| | - Antonio Mateus Gomes Pereira
- State University of Ceará, Northeast Network of Biotechnology Program (RENORBIO), Campus Itaperi, Fortaleza, Brazil; University Center INTA - UNINTA, Center for Bioprospecting and Applied Molecular Experimentation (NUBEM), Sobral, Brazil
| | - Ingrid Maria Frota Araújo
- University Center INTA - UNINTA, Center for Bioprospecting and Applied Molecular Experimentation (NUBEM), Sobral, Brazil
| | - Francisco Kauê Carvalho Aguiar
- University Center INTA - UNINTA, Center for Bioprospecting and Applied Molecular Experimentation (NUBEM), Sobral, Brazil
| | | | | | | | - Alexandre Magno Rodrigues Teixeira
- State University of Ceará, Northeast Network of Biotechnology Program (RENORBIO), Campus Itaperi, Fortaleza, Brazil; Postgraduate in Natural Sciences, Sciences and Technology Center, State University of Ceará, Fortaleza, CE, Brazil
| | - Emmanuel Silva Marinho
- Postgraduate in Natural Sciences, Sciences and Technology Center, State University of Ceará, Fortaleza, CE, Brazil
| | - Renata Albuquerque Costa
- University Center INTA - UNINTA, Center for Bioprospecting and Applied Molecular Experimentation (NUBEM), Sobral, Brazil
| | - Victor Alves Carneiro
- University Center INTA - UNINTA, Center for Bioprospecting and Applied Molecular Experimentation (NUBEM), Sobral, Brazil
| | - Hélcio Silva Dos Santos
- State University of Ceará, Northeast Network of Biotechnology Program (RENORBIO), Campus Itaperi, Fortaleza, Brazil; Course of Chemistry, State University of Vale Acaraú, Sobral, Ceará, Brazil; Postgraduate in Natural Sciences, Sciences and Technology Center, State University of Ceará, Fortaleza, CE, Brazil.
| |
Collapse
|
5
|
Soni S, De UK, Gandhar JS, Sarkar VK, Saminathan M, Agarwal RK, Patra MK, Singh BR, Kumar A, Bandyopadhyay S. Diflunisal attenuates acute inflammatory responses through inhibition of NF-κB signaling pathway in Staphylococcus aureus-induced mastitis of lactating mouse model. Microb Pathog 2025; 199:107260. [PMID: 39736343 DOI: 10.1016/j.micpath.2024.107260] [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: 10/04/2024] [Revised: 12/06/2024] [Accepted: 12/24/2024] [Indexed: 01/01/2025]
Abstract
The cure rate of Staphylococcus aureus mastitis by conventional antibiotic therapy is very poor. Diflunisal (DIF), a difluorophenol derivative of salicylic acid, is reported to have strong anti-bacterial and anti-inflammatory effects against S. aureus infection. The present study aimed to evaluate the potential therapeutic effect of DIF administration against S. aureus-induced mastitis in mouse model by assessing the bacterial load, inflammation and histopathological changes in mammary gland. Eighteen lactating Swiss albino mice were divided into four groups: uninfected control, S. aureus-induced mastitis model, antibiotic (ceftriaxone)-treatment and diflunisal (DIF)-treatment. In S. aureus-induced mastitis mice, markedly increased bacterial load, myeloperoxidase, NF-κB and nitric oxide (NO) levels and up regulations of IL-1β, NF-κB and TNF-α mRNA expressions in mammary tissues with severe necrosis, marked infiltration of neutrophils and fibrosis in histopathology were noticed. Intramammary administration of DIF in S. aureus-induced mastitis mice showed a significant reduction in bacterial load, myeloperoxidase, NF-κB and NO concentrations in mammary tissues. The DIF treatment also suppressed the inflammatory NF-κB signaling in the inflamed mammary tissues by downregulation of IL-1β, NF-κB and TNF-α mRNA expressions. Further, the histopathology of mammary tissues showed mild necrosis with mild inflammatory cells infiltration, few bacterial colonies, moderate fibrosis, and marked regenerative changes with near to normal histological architecture. The findings of the study provide the evidence of therapeutic potential of DIF in S. aureus-induced mastitis by promising antibacterial and anti-inflammatory activities along with ameliorative impact against the histopathological alterations in mammary tissues.
Collapse
Affiliation(s)
- Srishti Soni
- Division of Medicine, ICAR-Indian Veterinary Research Institute, Izatnagar, 243 122, Bareilly, Uttar Pradesh, India
| | - Ujjwal Kumar De
- Division of Medicine, ICAR-Indian Veterinary Research Institute, Izatnagar, 243 122, Bareilly, Uttar Pradesh, India.
| | - Jitendra Singh Gandhar
- Division of Medicine, ICAR-Indian Veterinary Research Institute, Izatnagar, 243 122, Bareilly, Uttar Pradesh, India
| | - Varun Kumar Sarkar
- Division of Medicine, ICAR-Indian Veterinary Research Institute, Izatnagar, 243 122, Bareilly, Uttar Pradesh, India
| | - M Saminathan
- Centre for Animal Disease Research and Diagnosis, ICAR-Indian Veterinary Research Institute, Izatnagar, 243 122, Bareilly, Uttar Pradesh, India.
| | - Ravi Kant Agarwal
- Division of Biological Products, ICAR-Indian Veterinary Research Institute, Izatnagar, 243 122, Bareilly, Uttar Pradesh, India
| | - Manas Kumar Patra
- Livestock Production and Management Section, ICAR-Indian Veterinary Research Institute, Izatnagar, 243 122, Bareilly, Uttar Pradesh, India
| | - Bhoj Raj Singh
- Division of Epidemiology, ICAR-Indian Veterinary Research Institute, Izatnagar, 243 122, Bareilly, Uttar Pradesh, India
| | - Amit Kumar
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, 243 122, Bareilly, Uttar Pradesh, India
| | - Samiran Bandyopadhyay
- Eastern Regional Station, ICAR-Indian Veterinary Research Institute, Kolkata, 700037, West Bengal, India
| |
Collapse
|
6
|
Aggarwal M, Patra A, Awasthi I, George A, Gagneja S, Gupta V, Capalash N, Sharma P. Drug repurposing against antibiotic resistant bacterial pathogens. Eur J Med Chem 2024; 279:116833. [PMID: 39243454 DOI: 10.1016/j.ejmech.2024.116833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/22/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024]
Abstract
The growing prevalence of MDR and XDR bacterial pathogens is posing a critical threat to global health. Traditional antibiotic development paths have encountered significant challenges and are drying up thus necessitating innovative approaches. Drug repurposing, which involves identifying new therapeutic applications for existing drugs, offers a promising alternative to combat resistant pathogens. By leveraging pre-existing safety and efficacy data, drug repurposing accelerates the development of new antimicrobial therapy regimes. This review explores the potential of repurposing existing FDA approved drugs against the ESKAPE and other clinically relevant bacterial pathogens and delves into the identification of suitable drug candidates, their mechanisms of action, and the potential for combination therapies. It also describes clinical trials and patent protection of repurposed drugs, offering perspectives on this evolving realm of therapeutic interventions against drug resistance.
Collapse
Affiliation(s)
- Manya Aggarwal
- Departmen of Microbiology, Panjab University, Chandigarh, India
| | - Anushree Patra
- Departmen of Microbiology, Panjab University, Chandigarh, India
| | - Ishita Awasthi
- Departmen of Microbiology, Panjab University, Chandigarh, India
| | - Annu George
- Departmen of Microbiology, Panjab University, Chandigarh, India
| | - Simran Gagneja
- Departmen of Microbiology, Panjab University, Chandigarh, India
| | - Varsha Gupta
- Department of Microbiology, Government Multi-speciality hospital, Sector 16, Chandigarh, India
| | - Neena Capalash
- Department of Biotechnology, Panjab University, Chandigarh, India
| | - Prince Sharma
- Departmen of Microbiology, Panjab University, Chandigarh, India.
| |
Collapse
|
7
|
Ma Y, Kang X, Wang G, Luo S, Luo X, Wang G. Inhibition of Staphylococcus aureus biofilm by quercetin combined with antibiotics. BIOFOULING 2024; 40:996-1011. [PMID: 39639551 DOI: 10.1080/08927014.2024.2435027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 11/13/2024] [Accepted: 11/20/2024] [Indexed: 12/07/2024]
Abstract
This study aimed to investigate the effects of combined quercetin and antibiotics on the bacteriostatic activity and biofilm formation of Staphylococcus aureus. Optimal concentrations of quercetin and antibiotics (tetracycline and doxycycline) for inhibiting biofilm formation were determined using the Fractional Inhibitory Concentration Index and Minimum Biofilm Inhibitory Concentration assays. The impact of the drug combinations on biofilm clearance at various formation stages was determined using crystal violet staining, scanning electron microscopy and confocal laser microscopy. The results indicated that quercetin enhanced the bactericidal effect of tetracycline antibiotics against S. aureus. The combination significantly reduced both the metabolic activity within S. aureus biofilms and the production of biofilm matrix components. Scanning electron microscopy and confocal laser microscopy confirmed that the combination treatment significantly reduced bacterial cell counts within the biofilm. Quercetin treatment significantly increased the sensitivity of biofilms to antibiotics, supporting its potential application as a novel antibiotic synergist.
Collapse
Affiliation(s)
- Yanjun Ma
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Xinyun Kang
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Guiqin Wang
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Shuangyan Luo
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Xiaofeng Luo
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Guilai Wang
- Yinchuan Hospital of Traditional Chinese Medicine, Yinchuan, China
| |
Collapse
|
8
|
Gonzalez-Prada I, Borges A, Santos-Torres B, Magariños B, Simões M, Concheiro A, Alvarez-Lorenzo C. Antimicrobial cyclodextrin-assisted electrospun fibers loaded with carvacrol, citronellol and cinnamic acid for wound healing. Int J Biol Macromol 2024; 277:134154. [PMID: 39116822 DOI: 10.1016/j.ijbiomac.2024.134154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/14/2024] [Accepted: 07/23/2024] [Indexed: 08/10/2024]
Abstract
This work aimed to explore an alternative to the use of antibiotics for prevention and treatment of wounds infection caused by two common bacterial pathogens Staphylococcus aureus and Pseudomonas aeruginosa. For this purpose, three different essential oil components (EOCs), namely carvacrol, citronellol and cinnamic acid, were loaded into electrospun fibers of poly-ε-caprolactone (PCL) aided by alpha-cyclodextrin (αCD) and hydroxypropyl-β-cyclodextrin (HPβCD). Electrospun-fibers prepared with each EOC and their mixtures were screened for antimicrobial capability and characterized regarding morphological, mechanical, thermal, surface polarity, antibiofilm and antioxidant properties. αCD formed poly(pseudo)rotaxanes with PCL and weakly interacted with EOCs, while HPβCD facilitated EOC encapsulation and formation of homogeneous fibers (500-1000 nm diameter) without beads. PCL/HPβCD fibers with high concentration of EOCs (mainly carvacrol and cinnamic acid) showed strong antibiofilm (>3 log CFU reduction) and antioxidant activity (10-50% DPPH scavenging effects). Different performances were recorded for the EOCs and their mixtures; cinnamic acid migrated to fiber surface and was released faster. Fibers biocompatibility was verified using hemolysis tests and in ovo tissue integration and angiogenesis assays. Overall, HPβCD facilitates complete release of EOCs from the fibers to the aqueous medium, being an environment-friendly and cost-effective strategy for the treatment of infected wounds.
Collapse
Affiliation(s)
- Iago Gonzalez-Prada
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Faculty of Pharmacy, Institute of Materials (iMATUS), and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain
| | - Anabela Borges
- LEPABE - Department of Chemical Engineering, Faculty of Engineering, University of Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Portugal
| | - Beatriz Santos-Torres
- Departamento de Microbiología y Parasitología, Facultad de Biología, CIBUS, Universidade de Santiago de Compostela, Spain
| | - Beatriz Magariños
- Departamento de Microbiología y Parasitología, Facultad de Biología, CIBUS, Universidade de Santiago de Compostela, Spain
| | - Manuel Simões
- LEPABE - Department of Chemical Engineering, Faculty of Engineering, University of Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Portugal
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Faculty of Pharmacy, Institute of Materials (iMATUS), and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Faculty of Pharmacy, Institute of Materials (iMATUS), and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Spain.
| |
Collapse
|
9
|
Bakeer W, Gaafar M, El-Gendy AO, El Badry MA, Khalil MG, Mansour AT, Alharbi NK, Selim HMRM, Bendary MM. Proven anti-virulence therapies in combating methicillin- and vancomycin-resistant Staphylococcus aureus infections. Front Cell Infect Microbiol 2024; 14:1403219. [PMID: 39253327 PMCID: PMC11381379 DOI: 10.3389/fcimb.2024.1403219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/04/2024] [Indexed: 09/11/2024] Open
Abstract
Introduction Despite years of efforts to develop new antibiotics for eradicating multidrug-resistant (MDR) and multi-virulent Methicillin-Resistant Staphylococcus aureus (MRSA) and Vancomycin-Resistant Staphylococcus aureus (VRSA) infections, treatment failures and poor prognoses in most cases have been common. Therefore, there is an urgent need for new therapeutic approaches targeting virulence arrays. Our aim is to discover new anti-virulence therapies targeting MRSA and VRSA virulence arrays. Methodology We employed phenotypic, molecular docking, and genetic studies to screen for anti-virulence activities among selected promising compounds: Coumarin, Simvastatin, and Ibuprofen. Results We found that nearly all detected MRSA and VRSA strains exhibited MDR and multi-virulent profiles. The molecular docking results aligned with the phenotypic and genetic assessments of virulence production. Biofilm and hemolysin productions were inhibited, and all virulence genes were downregulated upon treatment with sub-minimum inhibitory concentration (sub-MIC) of these promising compounds. Ibuprofen was the most active compound, exhibiting the highest inhibition and downregulation of virulence gene products. Moreover, in vivo and histopathological studies confirmed these results. Interestingly, we observed a significant decrease in wound area and improvements in re-epithelialization and tissue organization in the Ibuprofen and antimicrobial treated group compared with the group treated with antimicrobial alone. These findings support the idea that a combination of Ibuprofen and antimicrobial drugs may offer a promising new therapy for MRSA and VRSA infections. Conclusion We hope that our findings can be implemented in clinical practice to assist physicians in making the most suitable treatment decisions.
Collapse
Affiliation(s)
- Walid Bakeer
- Department of Microbiology and Immunology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Marwa Gaafar
- Department of Microbiology and Immunology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
- Quality Control Specialist at Egyptian Drug Authority (EDA), Cairo, Egypt
| | - Ahmed O El-Gendy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Mohamed A El Badry
- Department of Botany and Microbiology, Faculty of Sciences, Al- Azhar University, Cairo, Egypt
| | - Mona G Khalil
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | - Abdallah Tageldein Mansour
- Department of Fish and Animal Production and Aquaculture, College of Agriculture and Food Sciences, King Faisal University, Al-Ahsa, Saudi Arabia
- Department of Fish and Animal Production, Faculty of Agriculture (Saba Basha), Alexandria University, Alexandria, Egypt
| | - Nada K Alharbi
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Heba M R M Selim
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Riyadh, Saudi Arabia
| | - Mahmoud M Bendary
- Department of Microbiology and Immunology, Faculty of Pharmacy, Port Said University, Port Said, Egypt
| |
Collapse
|
10
|
Xi H, Luo Z, Liu MF, Chen Q, Zhu Q, Yuan L, Sheng YY, Zhao R. Diclofenac sodium effectively inhibits the biofilm formation of Staphylococcus epidermidis. Arch Microbiol 2024; 206:289. [PMID: 38847838 DOI: 10.1007/s00203-024-04020-5] [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: 04/23/2024] [Accepted: 05/27/2024] [Indexed: 08/07/2024]
Abstract
Staphylococcus epidermidis is an opportunistic pathogen commonly implicated in medical device-related infections. Its propensity to form biofilms not only leads to chronic infections but also exacerbates the issue of antibiotic resistance, necessitating high-dose antimicrobial treatments. In this study, we explored the use of diclofenac sodium, a non-steroidal anti-inflammatory drug, as an anti-biofilm agent against S. epidermidis. In this study, crystal violet staining and confocal laser scanning microscope analysis showed that diclofenac sodium, at subinhibitory concentration (0.4 mM), significantly inhibited biofilm formation in both methicillin-susceptible and methicillin-resistant S. epidermidis isolates. MTT assays demonstrated that 0.4 mM diclofenac sodium reduced the metabolic activity of biofilms by 25.21-49.01% compared to untreated controls. Additionally, the treatment of diclofenac sodium resulted in a significant decrease (56.01-65.67%) in initial bacterial adhesion, a crucial early phase of biofilm development. Notably, diclofenac sodium decreased the production of polysaccharide intercellular adhesin (PIA), a key component of the S. epidermidis biofilm matrix, in a dose-dependent manner. Real-time quantitative PCR analysis revealed that diclofenac sodium treatment downregulated biofilm-associated genes icaA, fnbA, and sigB and upregulated negative regulatory genes icaR and luxS, providing potential mechanistic insights. These findings indicate that diclofenac sodium inhibits S. epidermidis biofilm formation by affecting initial bacterial adhesion and the PIA synthesis. This underscores the potential of diclofenac sodium as a supplementary antimicrobial agent in combating staphylococcal biofilm-associated infections.
Collapse
Affiliation(s)
- Huimin Xi
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, PR China
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, PR China
| | - Zhaoxia Luo
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, PR China
| | - Mei-Fang Liu
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, PR China
| | - Qiang Chen
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, PR China
| | - Qing Zhu
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, PR China
| | - Lei Yuan
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, PR China
| | - Yi-Yun Sheng
- Department of Pathology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, PR China.
| | - Rui Zhao
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, PR China.
| |
Collapse
|
11
|
Said G, Ali A, Ahmad L. Design, semi-synthesis of soft coral-derived Aspergillus sp. secondary metabolite geodin derivatives and their antibacterial activities. Nat Prod Res 2024:1-9. [PMID: 38462767 DOI: 10.1080/14786419.2024.2326990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/27/2024] [Indexed: 03/12/2024]
Abstract
A series of novel ester derivatives 2 - 7, of natural product geodin 1, isolated from the soft coral-derived fungus Aspergillus sp., were designed and semi-synthesised through one step reaction with high yield. Compound 5 showed strong antifouling inhibitory activities with MIC of 4.80 μM while compound 4 showed selective inhibitory activities with MICs values 8.59 μM against Aeromonas salmonicida and Pseudomonas aeruginosa (Sea-Nine 211, MIC = 0.27 μM). Compounds 3, 4 and 6 showed potent anti-pathogenic inhibitory activities with MICs of 2.29 μM, 4.29 μM and 4.56 μM respectively against Staphylococcus aureus (Ciprofloxacin, MIC = 0.156 μM). Compound 2 showed weak inhibitory activity against A. salmonicida with MIC 18.75 μM (Sea-Nine 211, MIC = 0.27 μM) and with MICs 9.38 μM against S. aureus (ciprofloxacin, MIC = 0.156 μM). However, compound 7 showed very low antibacterial activities with MIC = >20 μM. The preliminary structure-activity relationships of compounds 2 - 7 further prove that the modification of 4-OH group of natural product geodin 1 improves the antibacterial activities such as antifouling and anti-pathogenic activities.
Collapse
Affiliation(s)
- Gulab Said
- Department of Chemistry, Women University Swabi, Swabi, Pakistan
| | - Amjad Ali
- Center of Excellence in Marine Biology, University of Karachi, Karachi, Pakistan
| | - Latif Ahmad
- Department of Botany, Women University Swabi, Swabi, Pakistan
| |
Collapse
|
12
|
Nunes B, Cagide F, Fernandes C, Borges A, Borges F, Simões M. Efficacy of Novel Quaternary Ammonium and Phosphonium Salts Differing in Cation Type and Alkyl Chain Length against Antibiotic-Resistant Staphylococcus aureus. Int J Mol Sci 2023; 25:504. [PMID: 38203676 PMCID: PMC10778626 DOI: 10.3390/ijms25010504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
Antibacterial resistance poses a critical public health threat, challenging the prevention and treatment of bacterial infections. The search for innovative antibacterial agents has spurred significant interest in quaternary heteronium salts (QHSs), such as quaternary ammonium and phosphonium compounds as potential candidates. In this study, a library of 49 structurally related QHSs was synthesized, varying the cation type and alkyl chain length. Their antibacterial activities against Staphylococcus aureus, including antibiotic-resistant strains, were evaluated by determining minimum inhibitory/bactericidal concentrations (MIC/MBC) ≤ 64 µg/mL. Structure-activity relationship analyses highlighted alkyl-triphenylphosphonium and alkyl-methylimidazolium salts as the most effective against S. aureus CECT 976. The length of the alkyl side chain significantly influenced the antibacterial activity, with optimal chain lengths observed between C10 and C14. Dose-response relationships were assessed for selected QHSs, showing dose-dependent antibacterial activity following a non-linear pattern. Survival curves indicated effective eradication of S. aureus CECT 976 by QHSs at low concentrations, particularly compounds 1e, 3e, and 5e. Moreover, in vitro human cellular data indicated that compounds 2e, 4e, and 5e showed favourable safety profiles at concentrations ≤ 2 µg/mL. These findings highlight the potential of these QHSs as effective agents against susceptible and resistant bacterial strains, providing valuable insights for the rational design of bioactive QHSs.
Collapse
Affiliation(s)
- Bárbara Nunes
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal; (B.N.); (A.B.)
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- CIQUP-IMS, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal (C.F.); (F.B.)
| | - Fernando Cagide
- CIQUP-IMS, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal (C.F.); (F.B.)
| | - Carlos Fernandes
- CIQUP-IMS, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal (C.F.); (F.B.)
| | - Anabela Borges
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal; (B.N.); (A.B.)
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Fernanda Borges
- CIQUP-IMS, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre, s/n, 4169-007 Porto, Portugal (C.F.); (F.B.)
| | - Manuel Simões
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal; (B.N.); (A.B.)
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| |
Collapse
|
13
|
Sousa M, Afonso AC, Saavedra MJ, Simões LC, Simões M. Hydrocinnamic acid and perillyl alcohol are effective against Escherichia coli biofilms when used alone and combined with antibiotics. J Appl Microbiol 2023; 134:lxad234. [PMID: 37827567 DOI: 10.1093/jambio/lxad234] [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: 08/11/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 10/14/2023]
Abstract
AIMS The use of phytochemicals to improve the effectiveness of antibiotics is a promising strategy for the development of novel antimicrobials. In this study, the antibiofilm activity of perillyl alcohol and hydrocinnamic acid, both phytochemicals present in several plants, and two antibiotics from different classes (amoxicillin and chloramphenicol) was tested, alone and in combination, against Escherichia coli. METHODS AND RESULTS Each molecule was tested at the minimum inhibitory concentration (MIC), 5 × MIC, and 10 × MIC, and characterized concerning biomass removal, metabolic inactivation, and cellular culturability. The highest percentages of metabolic inactivation (88.5% for 10 × MIC) and biomass reduction (61.7% for 10 × MIC) were obtained with amoxicillin. Interestingly, for 5 × MIC and 10 × MIC, phytochemicals provided a total reduction of colony-forming units (CFUs). Dual and triple combinations of phytochemicals and antibiotics (at MIC and 5 × MIC) demonstrated high efficacy in metabolic inactivation, moderate efficacy in terms of biomass reduction, and total reduction of cellular culturability for 5 × MIC. CONCLUSIONS The results demonstrated the antibiofilm potential of phytochemicals, highlighting the advantage of phytochemical/antibiotic combinations for biofilm control.
Collapse
Affiliation(s)
- Mariana Sousa
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, Department of Chemical Engineering, University of Porto, 4200-465 Porto, Portugal
- ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
| | - Ana Cristina Afonso
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, Department of Chemical Engineering, University of Porto, 4200-465 Porto, Portugal
- ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
- CITAB-Centre for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
- CEB, LABBELS-Centre of Biological Engineering, Associate Laboratory on Biotechnology and Bioengineering, and Electromechanical Systems, School of Engineering, University of Minho, 4710-057 Braga, Portugal
| | - Maria José Saavedra
- CITAB-Centre for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
| | - Lúcia Chaves Simões
- CEB, LABBELS-Centre of Biological Engineering, Associate Laboratory on Biotechnology and Bioengineering, and Electromechanical Systems, School of Engineering, University of Minho, 4710-057 Braga, Portugal
| | - Manuel Simões
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, Department of Chemical Engineering, University of Porto, 4200-465 Porto, Portugal
- ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
| |
Collapse
|
14
|
Batista S, Fernandez-Pittol M, Nicolás LS, Martínez D, Rubio M, Garrigo M, Vila J, Tudó G, González-Martin J. In Vitro Effect of Three-Antibiotic Combinations plus Potential Antibiofilm Agents against Biofilm-Producing Mycobacterium avium and Mycobacterium intracellulare Clinical Isolates. Antibiotics (Basel) 2023; 12:1409. [PMID: 37760706 PMCID: PMC10526108 DOI: 10.3390/antibiotics12091409] [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: 08/04/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Patients with chronic pulmonary diseases infected by Mycobacterium avium complex (MAC) often develop complications and suffer from treatment failure due to biofilm formation. There is a lack of correlation between in vitro susceptibility tests and the treatment of clinical isolates producing biofilm. We performed susceptibility tests of 10 different three-drug combinations, including two recommended in the guidelines, in biofilm forms of eight MAC clinical isolates. Biofilm developed in the eight isolates following incubation of the inoculum for 3 weeks. Then, the biofilm was treated with three-drug combinations with and without the addition of potential antibiofilm agents (PAAs). Biofilm bactericidal concentrations (BBCs) were determined using the Vizion lector system. All selected drug combinations showed synergistic activity, reducing BBC values compared to those treated with single drugs, but BBC values remained high enough to treat patients. However, with the addition of PAAs, the BBCs steadily decreased, achieving similar values to the combinations in planktonic forms and showing synergistic activity in all the combinations and in both species. In conclusion, three-drug combinations with PAAs showed synergistic activity in biofilm forms of MAC isolates. Our results suggest the need for clinical studies introducing PAAs combined with antibiotics for the treatment of patients with pulmonary diseases infected by MAC.
Collapse
Affiliation(s)
- Sara Batista
- Unitat de Microbiologia, Department de Fonaments Clínics, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, c/Casanova 143, 08036 Barcelona, Spain; (S.B.); (M.F.-P.); (J.V.)
- ISGlobal Barcelona, Institute for Global Health, c/Rosselló 132, 08036 Barcelona, Spain
- Servei de Microbiologia, CDB, Hospital Clínic de Barcelona, c/Villarroel 170, 08036 Barcelona, Spain; (L.S.N.); (D.M.)
| | - Mariana Fernandez-Pittol
- Unitat de Microbiologia, Department de Fonaments Clínics, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, c/Casanova 143, 08036 Barcelona, Spain; (S.B.); (M.F.-P.); (J.V.)
- ISGlobal Barcelona, Institute for Global Health, c/Rosselló 132, 08036 Barcelona, Spain
- Servei de Microbiologia, CDB, Hospital Clínic de Barcelona, c/Villarroel 170, 08036 Barcelona, Spain; (L.S.N.); (D.M.)
| | - Lorena San Nicolás
- Servei de Microbiologia, CDB, Hospital Clínic de Barcelona, c/Villarroel 170, 08036 Barcelona, Spain; (L.S.N.); (D.M.)
| | - Diego Martínez
- Servei de Microbiologia, CDB, Hospital Clínic de Barcelona, c/Villarroel 170, 08036 Barcelona, Spain; (L.S.N.); (D.M.)
| | - Marc Rubio
- Servei de Microbiologia, Fundació de Gestió de l’Hospital de la Santa Creu i Sant Pau, c/Sant Quintí 89, 08026 Barcelona, Spain; (M.R.); (M.G.)
- Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), c/Sant Quintí, 89, 08026 Barcelona, Spain
| | - Montserrat Garrigo
- Servei de Microbiologia, Fundació de Gestió de l’Hospital de la Santa Creu i Sant Pau, c/Sant Quintí 89, 08026 Barcelona, Spain; (M.R.); (M.G.)
- Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), c/Sant Quintí, 89, 08026 Barcelona, Spain
| | - Jordi Vila
- Unitat de Microbiologia, Department de Fonaments Clínics, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, c/Casanova 143, 08036 Barcelona, Spain; (S.B.); (M.F.-P.); (J.V.)
- ISGlobal Barcelona, Institute for Global Health, c/Rosselló 132, 08036 Barcelona, Spain
- Servei de Microbiologia, CDB, Hospital Clínic de Barcelona, c/Villarroel 170, 08036 Barcelona, Spain; (L.S.N.); (D.M.)
- CIBER of Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Griselda Tudó
- Unitat de Microbiologia, Department de Fonaments Clínics, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, c/Casanova 143, 08036 Barcelona, Spain; (S.B.); (M.F.-P.); (J.V.)
- ISGlobal Barcelona, Institute for Global Health, c/Rosselló 132, 08036 Barcelona, Spain
- Servei de Microbiologia, CDB, Hospital Clínic de Barcelona, c/Villarroel 170, 08036 Barcelona, Spain; (L.S.N.); (D.M.)
| | - Julian González-Martin
- Unitat de Microbiologia, Department de Fonaments Clínics, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, c/Casanova 143, 08036 Barcelona, Spain; (S.B.); (M.F.-P.); (J.V.)
- ISGlobal Barcelona, Institute for Global Health, c/Rosselló 132, 08036 Barcelona, Spain
- Servei de Microbiologia, CDB, Hospital Clínic de Barcelona, c/Villarroel 170, 08036 Barcelona, Spain; (L.S.N.); (D.M.)
- CIBER of Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
15
|
Chan LC, Lee HK, Wang L, Chaili S, Xiong YQ, Bayer AS, Proctor RA, Yeaman MR. Diflunisal and Analogue Pharmacophores Mediating Suppression of Virulence Phenotypes in Staphylococcus aureus. Antibiotics (Basel) 2023; 12:1180. [PMID: 37508276 PMCID: PMC10376238 DOI: 10.3390/antibiotics12071180] [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: 05/21/2023] [Revised: 06/26/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023] Open
Abstract
Invasive methicillin-resistant Staphylococcus aureus (MRSA) infections are leading causes of morbidity and mortality that are complicated by increasing resistance to conventional antibiotics. Thus, minimizing virulence and enhancing antibiotic efficacy against MRSA is a public health imperative. We originally demonstrated that diflunisal (DIF; [2-hydroxy-5-(2,4-difluorophenyl) benzoic acid]) inhibits S. aureus virulence factor expression. To investigate pharmacophores that are active in this function, we evaluated a library of structural analogues for their efficacy to modulate virulence phenotypes in a panel of clinically relevant S. aureus isolates in vitro. Overall, the positions of the phenyl, hydroxyl, and carboxylic moieties and the presence or type of halogen (F vs. Cl) influenced the efficacy of compounds in suppressing hemolysis, proteolysis, and biofilm virulence phenotypes. Analogues lacking halogens inhibited proteolysis to an extent similar to DIF but were ineffective at reducing hemolysis or biofilm production. In contrast, most analogues lacking the hydroxyl or carboxylic acid groups did not suppress proteolysis but did mitigate hemolysis and biofilm production to an extent similar to DIF. Interestingly, chirality and the substitution of fluorine with chlorine resulted in a differential reduction in virulence phenotypes. Together, this pattern of data suggests virulence-suppressing pharmacophores of DIF and structural analogues integrate halogen, hydroxyl, and carboxylic acid moiety stereochemistry. The anti-virulence effects of DIF were achieved using concentrations that are safe in humans, do not impair platelet antimicrobial functions, do not affect S. aureus growth, and do not alter the efficacy of conventional antibiotics. These results offer proof of concept for using novel anti-virulence strategies as adjuvants to antibiotic therapy to address the challenge of MRSA infection.
Collapse
Affiliation(s)
- Liana C. Chan
- Division of Molecular Medicine, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (L.C.C.); (H.K.L.); (L.W.)
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (Y.Q.X.); (A.S.B.)
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024, USA
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Hong K. Lee
- Division of Molecular Medicine, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (L.C.C.); (H.K.L.); (L.W.)
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Ling Wang
- Division of Molecular Medicine, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (L.C.C.); (H.K.L.); (L.W.)
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Siyang Chaili
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, 2311 Pierce Ave., Nashville, TN 37232, USA;
| | - Yan Q. Xiong
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (Y.Q.X.); (A.S.B.)
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024, USA
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Arnold S. Bayer
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (Y.Q.X.); (A.S.B.)
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024, USA
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Richard A. Proctor
- Departments of Medical Microbiology & Immunology and Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA;
| | - Michael R. Yeaman
- Division of Molecular Medicine, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (L.C.C.); (H.K.L.); (L.W.)
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (Y.Q.X.); (A.S.B.)
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024, USA
- Institute for Infection and Immunity, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| |
Collapse
|
16
|
Biofilm Formation and Control of Foodborne Pathogenic Bacteria. Molecules 2023; 28:molecules28062432. [PMID: 36985403 PMCID: PMC10058477 DOI: 10.3390/molecules28062432] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/17/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023] Open
Abstract
Biofilms are microbial aggregation membranes that are formed when microorganisms attach to the surfaces of living or nonliving things. Importantly, biofilm properties provide microorganisms with protection against environmental pressures and enhance their resistance to antimicrobial agents, contributing to microbial persistence and toxicity. Thus, bacterial biofilm formation is part of the bacterial survival mechanism. However, if foodborne pathogens form biofilms, the risk of foodborne disease infections can be greatly exacerbated, which can cause major public health risks and lead to adverse economic consequences. Therefore, research on biofilms and their removal strategies are very important in the food industry. Food waste due to spoilage within the food industry remains a global challenge to environmental sustainability and the security of food supplies. This review describes bacterial biofilm formation, elaborates on the problem associated with biofilms in the food industry, enumerates several kinds of common foodborne pathogens in biofilms, summarizes the current strategies used to eliminate or control harmful bacterial biofilm formation, introduces the current and emerging control strategies, and emphasizes future development prospects with respect to bacterial biofilms.
Collapse
|
17
|
Liu S, She P, Li Z, Li Y, Li L, Yang Y, Zhou L, Wu Y. Antibacterial and Antibiofilm Efficacy of Repurposing Drug Hexestrol against Methicillin-resistant Staphylococcus aureus. Int J Med Microbiol 2023; 313:151578. [PMID: 37001448 DOI: 10.1016/j.ijmm.2023.151578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 03/17/2023] [Accepted: 03/22/2023] [Indexed: 03/30/2023] Open
Abstract
There has been an explosion in the prevalence of methicillin-resistant Staphylococcus aureus (MRSA) because of the indiscriminate use of antibiotics. In this study, we repurposed hexestrol (HXS) as an antibacterial agent to fight planktonic and biofilm-related MRSA infections. HXS is a nonsteroidal synthetic estrogen that targets estrogen receptors (ERα and ERβ) and has been used as a hormonal antineoplastic agent. In our work, the minimum inhibitory concentrations (MICs) were determined using the antimicrobial susceptibility of MSSA and MRSA strains. Anti-biofilm activity was evaluated using biofilm inhibition and eradication assays. Biofilm-related genes were analyzed with or without HXS treatment using RTqPCR analysis of S. aureus. HXS was tested using the checkerboard dilution assay to identify antibiotics that may have synergistic effects. Measurement of ATP and detection of ATPase allowed the determination of bacterial energy metabolism. As shown in the results, HXS showed effective antimicrobial activity against S. aureus, including both type strains and clinical isolations, with MICs of 16 µg/mL. Sub-HXS strongly inhibited the adhesion of S. aureus. The content of extracellular polymeric substances (EPS) and the relative transcription levels of eno, sacC, clfA, pls and fnbpB were reduced after HXS treatment. HXS showed antibacterial effects against S. aureus and synergistic activity with aminoglycosides by directly interfering with cellular energy metabolism. HXS inhibits adhesion and biofilm formation and eradicates biofilms formed by MRSA by reducing the expression of related genes. Furthermore, HXS increases the susceptibility of aminoglycosides against MRSA. In conclusion, HXS is a repurposed drug that may be a promising therapeutic option for MRSA infection.
Collapse
|
18
|
Sousa M, Afonso AC, Teixeira LS, Borges A, Saavedra MJ, Simões LC, Simões M. Hydrocinnamic Acid and Perillyl Alcohol Potentiate the Action of Antibiotics against Escherichia coli. Antibiotics (Basel) 2023; 12:antibiotics12020360. [PMID: 36830271 PMCID: PMC9952493 DOI: 10.3390/antibiotics12020360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 01/28/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
The treatment of bacterial infections has been troubled by the increased resistance to antibiotics, instigating the search for new antimicrobial therapies. Phytochemicals have demonstrated broad-spectrum and effective antibacterial effects as well as antibiotic resistance-modifying activity. In this study, perillyl alcohol and hydrocinnamic acid were characterized for their antimicrobial action against Escherichia coli. Furthermore, dual and triple combinations of these molecules with the antibiotics chloramphenicol and amoxicillin were investigated for the first time. Perillyl alcohol had a minimum inhibitory concentration (MIC) of 256 µg/mL and a minimum bactericidal concentration (MBC) of 512 µg/mL. Hydrocinnamic acid had a MIC of 2048 µg/mL and an MBC > 2048 µg/mL. Checkerboard and time-kill assays demonstrated synergism or additive effects for the dual combinations chloramphenicol/perillyl alcohol, chloramphenicol/hydrocinnamic acid, and amoxicillin/hydrocinnamic acid at low concentrations of both molecules. Combenefit analysis showed synergism for various concentrations of amoxicillin with each phytochemical. Combinations of chloramphenicol with perillyl alcohol and hydrocinnamic acid revealed synergism mainly at low concentrations of antibiotics (up to 2 μg/mL of chloramphenicol with perillyl alcohol; 0.5 μg/mL of chloramphenicol with hydrocinnamic acid). The results highlight the potential of combinatorial therapies for microbial growth control, where phytochemicals can play an important role as potentiators or resistance-modifying agents.
Collapse
Affiliation(s)
- Mariana Sousa
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, Department of Chemical Engineering, University of Porto, 4200-465 Porto, Portugal
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
| | - Ana Cristina Afonso
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, Department of Chemical Engineering, University of Porto, 4200-465 Porto, Portugal
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
- CITAB—Centre for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
- CEB, LABBELS—Centre of Biological Engineering, Associate Laboratory on Biotechnology and Bioengineering, and Electromechanical Systems, School of Engineering, University of Minho, 4710-057 Braga, Portugal
| | - Lília Soares Teixeira
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, Department of Chemical Engineering, University of Porto, 4200-465 Porto, Portugal
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
| | - Anabela Borges
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, Department of Chemical Engineering, University of Porto, 4200-465 Porto, Portugal
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
| | - Maria José Saavedra
- CITAB—Centre for the Research and Technology of Agro-Environmental and Biological Sciences, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
| | - Lúcia Chaves Simões
- CEB, LABBELS—Centre of Biological Engineering, Associate Laboratory on Biotechnology and Bioengineering, and Electromechanical Systems, School of Engineering, University of Minho, 4710-057 Braga, Portugal
| | - Manuel Simões
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, Department of Chemical Engineering, University of Porto, 4200-465 Porto, Portugal
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
- Correspondence:
| |
Collapse
|
19
|
Ugrinovic V, Milutinovic M, Bozic B, Petrovic R, Janackovic D, Panic V, Veljovic D. Poly(methacrylic acid)/gelatin interpenetrating network hydrogels reinforced by nano-structured hydroxyapatite particles—improved drug delivery systems. INT J POLYM MATER PO 2023. [DOI: 10.1080/00914037.2022.2164281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Vukasin Ugrinovic
- Innovation Center of Faculty of Technology and Metallurgy, Belgrade, Serbia
| | - Milica Milutinovic
- Department of Biochemical Engineering and Biotechnology, Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| | - Bojan Bozic
- Institute of Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Rada Petrovic
- Department of Inorganic Chemical Technology, Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| | - Djordje Janackovic
- Department of Inorganic Chemical Technology, Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| | - Vesna Panic
- Innovation Center of Faculty of Technology and Metallurgy, Belgrade, Serbia
| | - Djordje Veljovic
- Department of Inorganic Chemical Technology, Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
20
|
Ruiz-Linares M, Monroy-Rojas JF, Solana C, Baca P, Aguado B, Soriano-Lerma A, Arias-Moliz MT, Ferrer-Luque CM. Antimicrobial potential of new diclofenac hydrogels for disinfection in regenerative endodontics: An in vitro and ex vivo study. Int Endod J 2023; 56:103-117. [PMID: 36169591 DOI: 10.1111/iej.13840] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 09/07/2022] [Accepted: 09/22/2022] [Indexed: 12/13/2022]
Abstract
AIM There is a need to explore new alternatives for root canal disinfection in regenerative endodontics, since the current strategies are far from ideal. Currently, the potential use of diclofenac (DC) is being investigated for controlling root canal infections. The objective was to evaluate the antimicrobial efficacy of novel DC-based hydrogels (DCHs) against polymicrobial biofilms grown in radicular dentine and root canals and to compare results with triantibiotic (TAH) and diantibiotic (DAH) hydrogels, and calcium hydroxide (Ca[OH]2 ). METHODOLOGY The in vitro antimicrobial activity of intracanal medicaments was evaluated against 3-week-old polymicrobial root canal biofilms grown on human radicular dentine. Dentine samples were obtained and randomly divided into the study groups (n = 4/group): (1) 1 mg/ml TAH; (2) 1 mg/ml DAH; (3) 5% diclofenac (DCH); (4) 2.5% DCH; (5) 1.25% DCH; (6) 1 mg/ml DAH + 5% DCH; (7) Ca(OH)2 paste; (8) positive control. The microbial viability, in terms of percentage of intact cell membranes, was assessed after 7 days by confocal scanning laser microscopy (CSLM). The ex vivo efficacy of intracanal medications was evaluated in root canals infected with a polymicrobial suspension. Intracanal microbiological samples at baseline (S1) and 7 days post-treatment (S2) were taken; microbial quantification and cell viability were assessed by quantitative polymerase chain reaction (qPCR) and flow cytometry (FC). The mean Log10 of bacterial DNA copies in root canal samples before (S1) and the Log10 reduction of DNA copies S1-S2 in qPCR were recorded. The absolute value of total cells stained, and the percentage reduction of intact membrane cells after treatment (S1-S2), were analysed by FC. Global comparison was done using the Kruskal-Wallis test, whilst the Mann-Whitney U test was used for pair-by-pair comparison. RESULTS Confocal scanning laser microscopy analysis indicated that the greatest effectiveness was obtained with 5% DCH, showing significant differences with respect to the other groups (p < .001). In root canals, the highest Log10 DNA reduction S1-S2 was obtained with 5% DCH and TAH, with no differences between them. The results of FC showed that only 5% DCH proved significantly superior to the other treatments. CONCLUSIONS Sodium DC hydrogels demonstrate antimicrobial efficacy against endodontic biofilms.
Collapse
Affiliation(s)
- Matilde Ruiz-Linares
- Department of Stomatology, School of Dentistry, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Javier F Monroy-Rojas
- Department of Stomatology, School of Dentistry, University of Granada, Granada, Spain
| | - Carmen Solana
- Department of Stomatology, School of Dentistry, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Pilar Baca
- Department of Stomatology, School of Dentistry, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Beatriz Aguado
- Department of Stomatology, School of Dentistry, University of Granada, Granada, Spain
| | - Ana Soriano-Lerma
- Department of Physiology, Faculty of Pharmacy, Institute of Nutrition and Food Technology "José Mataix", University of Granada, Granada, Spain.,GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain
| | - María Teresa Arias-Moliz
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain.,Department of Microbiology, School of Dentistry, University of Granada, Granada, Spain
| | - Carmen María Ferrer-Luque
- Department of Stomatology, School of Dentistry, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| |
Collapse
|
21
|
Jampilek J. Drug repurposing to overcome microbial resistance. Drug Discov Today 2022; 27:2028-2041. [PMID: 35561965 DOI: 10.1016/j.drudis.2022.05.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/02/2022] [Accepted: 05/06/2022] [Indexed: 12/15/2022]
Abstract
Infections are a growing global threat, and the number of resistant species of microbial pathogens is alarming. However, the rapid development of cross-resistant or multidrug-resistant strains and the development of so-called 'superbugs' are in stark contrast to the number of newly launched anti-infectives on the market. In this review, I summarize the causes of antimicrobial resistance, briefly discuss different approaches to the discovery and development of new anti-infective drugs, and focus on drug repurposing strategy, which is discussed from all possible perspectives. A comprehensive overview of drugs of other indications tested for their in vitro antimicrobial activity to support existing anti-infective therapeutics is provided, including several critical remarks on this strategy of repurposing non-antibiotics to antibacterial drugs.
Collapse
Affiliation(s)
- Josef Jampilek
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia; Department of Chemical Biology, Faculty of Science, Palacky University, Slechtitelu 27, 783 71 Olomouc, Czech Republic.
| |
Collapse
|
22
|
Daud S, Abid OUR, Sardar A, Abdullah S, Shahid W, Ashraf M, Ejaz SA, Saeed A, Shah BA, Niaz B. Exploring ibuprofen derivatives as α-glucosidase and lipoxygenase inhibitors: Cytotoxicity and in silico studies. Arch Pharm (Weinheim) 2022; 355:e2200013. [PMID: 35532320 DOI: 10.1002/ardp.202200013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 03/19/2022] [Accepted: 04/06/2022] [Indexed: 01/01/2023]
Abstract
This study reports the synthesis of a series of ibuprofen derivatives, including thiosemicarbazides 4a-f, 1,3,4-oxadiazoles 5a-f, 1,3,4-thiadiazoles 6a-f, 1,2,4-triazoles 7a-f, and their S-alkylated derivatives 8a-d. All of the newly synthesized derivatives were analyzed using 1 H NMR, 13 C NMR spectroscopy, and high-resolution mass spectra (electron ionization) spectrometry. These synthetic molecules were examined for their in vitro baking yeast α-glucosidase and soybean 15-lipoxygenase (15-LOX) inhibition and cell viability studies. The results revealed that the compounds N-(3,4-dichlorophenyl)-5-[1-(4-isobutylphenyl)ethyl]-1,3,4-oxadiazol-2-amine 5f (IC50 3.05 ± 1.23 µM) and N-(3-fluorophenyl)-5-[1-(4-isobutylphenyl)ethyl]-1,3,4-oxadiazol-2-amine 5b (IC50 3.12 ± 1.21 µM) were the most potent with respect to the α-glucosidase enzyme while in case of 15-LOX, the compound 4-(2,4-dichlorophenyl)-1-[2-(4-isobutylphenyl)propanoyl]thiosemicarbazide 4e showed potent inhibition with an IC50 value of 55.41 ± 0.41 µM. All these compounds were found least toxic by displaying a blood mononuclear cell viability value of 69.2%-97.8% by the MTT assay compared to the standards when assayed at 0.25 mM concentration. Molecular docking analyses were conducted to evaluate the inhibition profiles of these derivatives against the said enzymes and the data supported the in vitro profiles.
Collapse
Affiliation(s)
- Saima Daud
- Department of Chemistry, Hazara University, Mansehra, Pakistan
| | | | - Asma Sardar
- Department of Chemistry, Hazara University, Mansehra, Pakistan
| | - Shawana Abdullah
- Institute of Chemistry, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Wardah Shahid
- Institute of Chemistry, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Muhammad Ashraf
- Institute of Chemistry, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Syeda Abida Ejaz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Amna Saeed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Basit Ali Shah
- School of Material Science & Engineering, South China University of Technology, Guangzhou, China
| | - Basit Niaz
- Department of Chemistry, Hazara University, Mansehra, Pakistan
| |
Collapse
|
23
|
Babaei N, Rasti B, Zamani H. Does ibuprofen affect the expression of alginate genes in pathogenic Pseudomonas aeruginosa strains? Folia Microbiol (Praha) 2022; 67:617-623. [PMID: 35325409 DOI: 10.1007/s12223-022-00962-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 03/07/2022] [Indexed: 11/25/2022]
Abstract
Conversion to mucoid form is a crucial step in the pathogenesis of P. aeruginosa in burns and cystic fibrosis (CF) patients. Alginate is considered the major component of biofilm and is highly associated with the formation of mucoid biofilm in this species. Nonsteroid anti-inflammatory drugs (NSAIDs), including ibuprofen, have shown promising antibacterial and antibiofilm potential for bacterial pathogens. In this study, we aimed to evaluate the effect of ibuprofen on the expression of alginate synthetase (alg8), GDP-mannose dehydrogenase (algD), and alginate lyase (algL) genes in multiple drug-resistant (MDR) P. aeruginosa strains. The biofilm formation potential and the expression of alg8, algD, and algL among the bacteria treated with ibuprofen (at sub-inhibitory concentration) were investigated using the crystal violet staining and real-time PCR assays, respectively. The minimum inhibitory concentration of ibuprofen for the studied strains was determined 1024-2048 µg/mL. We observed that ibuprofen was able to reduce bacterial biofilm by 51-77%. Also, the expression of alg8, algD, and algL decreased by 32, 52, and 48%, respectively. The reduction of the genes responsible for alginate synthesis indicates promising antivirulece potential of ibuprofen to combat P. aeruginosa infection, especially in burns and CF patients. Our findings suggest that ibuprofen could be used to reduce the pathogenicity of P. aeruginosa that could be used in combination with antibiotics to treat drug-resistant infections.
Collapse
Affiliation(s)
- Nastaran Babaei
- Department of Microbiology, Faculty of Basic Sciences, Lahijan Branch, Islamic Azad University (IAU), Lahijan, Guilan, Iran
| | - Behnam Rasti
- Department of Microbiology, Faculty of Basic Sciences, Lahijan Branch, Islamic Azad University (IAU), Lahijan, Guilan, Iran
| | - Hojjatolah Zamani
- Department of Biology, Faculty of Science, University of Guilan, Guilan, Iran.
| |
Collapse
|
24
|
Kovacs ED, Silaghi-Dumitrescu L, Roman C, Tian D. Structural and Metabolic Profiling of Lycopersicon esculentum Rhizosphere Microbiota Artificially Exposed at Commonly Used Non-Steroidal Anti-Inflammatory Drugs. Microorganisms 2022; 10:microorganisms10020254. [PMID: 35208709 PMCID: PMC8878439 DOI: 10.3390/microorganisms10020254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/06/2022] [Accepted: 01/18/2022] [Indexed: 12/10/2022] Open
Abstract
In this study, the effect of common non-steroidal anti-inflammatory drugs on Lycopersicon esculentum rhizosphere microbiota was monitored. The experiments were performed with artificially contaminated soil with ibuprofen (0.5 mg·kg−1), ketoprofen (0.2 mg·kg−1) and diclofenac (0.7 mg·kg−1). The results evidenced that the rhizosphere microbiota abundance decreased especially under exposure to diclofenac (187–201 nmol·g−1 dry weight soil) and ibuprofen (166–183 nmol·g−1 dry weight soil) if compared with control (185–240 nmol·g−1 dry weight soil), while the fungal/bacteria ratio changed significantly with exposure to diclofenac (<27%) and ketoprofen (<18%). Compared with control samples, the average amount of the ratio of Gram-negative/Gram-positive bacteria was higher in rhizosphere soil contaminated with ibuprofen (>25%) and lower in the case of diclofenac (<46%) contamination. Carbon source consumption increased with the time of assay in case of the control samples (23%) and those contaminated with diclofenac (8%). This suggests that rhizosphere microbiota under contamination with diclofenac consume a higher amount of carbon, but they do not consume a larger variety of its sources. In the case of contamination with ibuprofen and ketoprofen, the consumption of carbon source presents a decreasing tendency after day 30 of the assay. Rhizosphere microbiota emitting volatile organic compounds were also monitored. Volatile compounds belonging to alcohol, aromatic compounds, ketone, terpene, organic acids, aldehyde, sulphur compounds, esters, alkane, nitrogen compounds, alkene and furans were detected in rhizosphere soil samples. Among these, terpene, ketone, alcohol, aromatic compounds, organic acids and alkane were the most abundant compound classes (>75%), but their percentage changed with exposure to diclofenac, ketoprofen and ibuprofen. Such changes in abundance, structure and the metabolic activity of Lycopersicon esculentum rhizosphere microbiota under exposure to common non-steroidal anti-inflammatory drugs suggest that there is a probability to also change the ecosystem services provided by rhizosphere microbiota.
Collapse
Affiliation(s)
- Emoke Dalma Kovacs
- Research Institute for Analytical Instrumentation, INCDO-INOE 2000, 400293 Cluj-Napoca, Romania;
- Faculty of Chemistry and Chemical Engineering, Babes-Bolyai University, 400028 Cluj-Napoca, Romania;
- Correspondence:
| | | | - Cecilia Roman
- Research Institute for Analytical Instrumentation, INCDO-INOE 2000, 400293 Cluj-Napoca, Romania;
| | - Di Tian
- Research Center of Forest Management Engineering of State Forestry and Grassland Administration, College of Forestry, Beijing Forestry University, Beijing 100083, China;
| |
Collapse
|
25
|
Xu L, She P, Chen L, Li S, Zhou L, Hussain Z, Liu Y, Wu Y. Repurposing Candesartan Cilexetil as Antibacterial Agent for MRSA Infection. Front Microbiol 2021; 12:688772. [PMID: 34589063 PMCID: PMC8473943 DOI: 10.3389/fmicb.2021.688772] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/17/2021] [Indexed: 11/13/2022] Open
Abstract
Staphylococcus aureus is an important pathogen causing hospital-acquired infections. Methicillin-resistant S. aureus (MRSA), biofilms, and persisters are highly tolerant to traditional antibiotics and make it difficult to treat. Therefore, new antimicrobial agents are urgently needed to treat hard-to-eradicate diseases caused by this bacterium. In this study, candesartan cilexetil (CC), an angiotensin hypertension drug, had strong antimicrobial activity against S. aureus with minimal inhibitory concentrations (MICs) and minimal bactericidal concentrations (MBCs) of 8-16 μg/ml and 16-32 μg/ml. CC exhibited limited cytotoxicity and low potential to induce drug resistance. In addition, it showed a synergistic antibacterial effect when combined with gentamicin and tobramycin. The effective concentrations to inhibit MRSA biofilm formation were 16-64 μg/ml, and intractable persisters were killed at 4-8 × MIC. Through the analysis of its mechanism of action, it was evident that the membrane permeability was disrupted as well as the cell structure was damaged. Furthermore, we demonstrated that CC had antibacterial effects in vivo in MRSA-infected murine skin abscess models. In conclusion, these results imply that CC might be a potential antibacterial agent for the treatment of S. aureus-associated infections.
Collapse
Affiliation(s)
- Lanlan Xu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Pengfei She
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Lihua Chen
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Shijia Li
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Linying Zhou
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zubair Hussain
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yaqian Liu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yong Wu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
26
|
Graham GG, Scott KF. Limitations of drug concentrations used in cell culture studies for understanding clinical responses of NSAIDs. Inflammopharmacology 2021; 29:1261-1278. [PMID: 34510275 DOI: 10.1007/s10787-021-00871-2] [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: 06/13/2021] [Accepted: 08/21/2021] [Indexed: 02/02/2023]
Abstract
In this review, the in vitro cellular effects of six nonsteroidal anti-inflammatory drugs (NSAIDs), salicylate, ibuprofen, naproxen, indomethacin, celecoxib and diclofenac, are examined. Inhibition of prostanoid synthesis in vitro generally occurs within the therapeutic range of plasma concentrations that are observed in vivo, consistent with the major action of NSAIDs being inhibition of prostanoid production. An additional probable cellular action of NSAIDs has been discovered recently, viz. decreased oxidation of the endocannabinoids, 2-arachidonoyl glycerol and arachidonyl ethanolamide. Many effects of NSAIDs, other than decreased oxidation of arachidonic acid and endocannabinoids, have been put forward but almost all of these additional processes are observed at supratherapeutic concentrations when the concentration of albumin, the major protein that binds NSAIDs, is taken into account. However, one exception is salicylate, a very potent inhibitor of the neutrophilic enzyme, myeloperoxidase, the inhibition of which leads to reduced production of the inflammatory mediator, hypochlorous acid, and inhibition of the inflammation associated with rheumatoid arthritis.
Collapse
Affiliation(s)
- Garry G Graham
- Department of Clinical Pharmacology, St Vincent's Hospital Sydney, Darlinghurst, NSW, 2010, Australia. .,School of Medical Sciences, University of New South Wales, Kensington, NSW, 2052, Australia.
| | - Kieran F Scott
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia. .,Ingham Institute of Applied Medical Research, 1 Campbell St, Liverpool, NSW, 2170, Australia.
| |
Collapse
|
27
|
Paes Leme RC, da Silva RB. Antimicrobial Activity of Non-steroidal Anti-inflammatory Drugs on Biofilm: Current Evidence and Potential for Drug Repurposing. Front Microbiol 2021; 12:707629. [PMID: 34385992 PMCID: PMC8353384 DOI: 10.3389/fmicb.2021.707629] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/05/2021] [Indexed: 12/20/2022] Open
Abstract
It has been demonstrated that some non-steroidal anti-inflammatory drugs (NSAIDs), like acetylsalicylic acid, diclofenac, and ibuprofen, have anti-biofilm activity in concentrations found in human pharmacokinetic studies, which could fuel an interest in repurposing these well tolerated drugs as adjunctive therapies for biofilm-related infections. Here we sought to review the currently available data on the anti-biofilm activity of NSAIDs and its relevance in a clinical context. We performed a systematic literature review to identify the most commonly tested NSAIDs drugs in the last 5 years, the bacterial species that have demonstrated to be responsive to their actions, and the emergence of resistance to these molecules. We found that most studies investigating NSAIDs' activity against biofilms were in vitro, and frequently tested non-clinical bacterial isolates, which may not adequately represent the bacterial populations that cause clinically-relevant biofilm-related infections. Furthermore, studies concerning NSAIDs and antibiotic resistance are scarce, with divergent outcomes. Although the potential to use NSAIDs to control biofilm-related infections seems to be an exciting avenue, there is a paucity of studies that tested these drugs using appropriate in vivo models of biofilm infections or in controlled human clinical trials to support their repurposing as anti-biofilm agents.
Collapse
Affiliation(s)
- Rodrigo Cuiabano Paes Leme
- Laboratório Especial de Microbiologia Clínica (LEMC), Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, Brazil.,Department of Infectious Diseases, Centro Universitário de Volta Redonda, Volta Redonda, Brazil
| | | |
Collapse
|
28
|
Foletto VS, da Rosa TF, Serafin MB, Bottega A, Hörner R. Repositioning of non-antibiotic drugs as an alternative to microbial resistance: a systematic review. Int J Antimicrob Agents 2021; 58:106380. [PMID: 34166776 DOI: 10.1016/j.ijantimicag.2021.106380] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/10/2021] [Accepted: 06/10/2021] [Indexed: 01/19/2023]
Abstract
The global spread of microbial resistance coupled with high costs and slow pace in the discovery of a new antibiotic have made drug repositioning an attractive and promising alternative in the treatment of infections caused by multidrug resistant (MDR) microorganisms. The reuse involves the production of compounds with lower costs and development time, using diversified production technologies. The present systematic review aimed to present a selection of studies published in the last 20 years, which report the antimicrobial activity of non-antibiotic drugs that are candidates for repositioning, which could be used against the current microbial multidrug resistance. A search was performed in the PubMed, SciELO and Google Scholar databases using the following search strategies: [(drug repurposing) OR (drug repositioning) OR (repositioning) AND (non-antibiotic) AND (antibacterial activity) AND (antimicrobial activity)]. Overall, 112 articles were included, which explored the antimicrobial activity in antidepressants, antihypertensives, anti-inflammatories, antineoplastics, hypoglycemic agents, among other drugs. It was concluded that they have significant antimicrobial activity in vitro and in vivo, against standard strain and clinical isolates (Gram-negative and Gram-positive) and fungi. When associated with antibacterials, most of these drugs had their antibacterial activity enhanced. It was also a consensus of the studies included in this review that the presence of aromatic rings in the molecular structure contributes to antimicrobial activity. This review highlights the potential repositioning of several classes of non-antibiotic drugs as promising candidates for repositioning in the treatment of severe bacterial infections of MDR bacteria, extensively resistant (XDR) and pan-resistant (PDR) to drugs.
Collapse
Affiliation(s)
- Vitória S Foletto
- Universidade Federal de Santa Maria, Programa de Pós-Graduação em Ciências Farmacêuticas, Santa Maria, RS, Brasil
| | - Taciéli F da Rosa
- Universidade Federal de Santa Maria, Programa de Pós-Graduação em Ciências Farmacêuticas, Santa Maria, RS, Brasil
| | - Marissa B Serafin
- Universidade Federal de Santa Maria, Programa de Pós-Graduação em Ciências Farmacêuticas, Santa Maria, RS, Brasil
| | - Angelita Bottega
- Universidade Federal de Santa Maria, Programa de Pós-Graduação em Ciências Farmacêuticas, Santa Maria, RS, Brasil
| | - Rosmari Hörner
- Universidade Federal de Santa Maria, Programa de Pós-Graduação em Ciências Farmacêuticas, Santa Maria, RS, Brasil; Universidade Federal de Santa Maria, Departamento de Análises Clínicas e Toxicológicas, Santa Maria, RS, Brasil.
| |
Collapse
|
29
|
Silva V, Miranda C, Bezerra M, Antão HS, Guimarães J, Prada J, Pires I, Maltez L, Pereira JE, Capelo JL, Igrejas G, Poeta P. Anti-biofilm activity of dalbavancin against methicillin-resistant Staphylococcus aureus (MRSA) isolated from human bone infection. J Chemother 2021; 33:469-475. [PMID: 33904369 DOI: 10.1080/1120009x.2021.1911518] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The presence of methicillin-resistant Staphylococcus aureus (MRSA) in bone infections difficults its treatment and is a sign of concern. The aim of this study was to evaluate in vitro activity of dalbavancin on pre-established adhered cells and 24 h old biofilms of MRSA strains isolated from a human bone infection. Thirty-three MRSA were isolated from osteomyelitis episodes. The antimicrobial susceptibility of these strains was assessed by the Kirby-Bauer disc diffusion method and the presence of resistance genes was screened by PCR. MRSA planktonic minimum inhibitory concentration and minimum bactericidal concentration were assessed. Minimum biofilm eradication concentration (MBEC) was performed by the microtiter biofilm formation assay. All 33 MRSA strains were classified as multidrug-resistant strains and susceptible to dalbavancin. Dalbavancin inhibited the growth of 54.6% and 52% of strains at the concentrations of 0.05 µg/mL and 1 µg/mL, respectively. The MBEC values up to 0.4 µg/mL demonstrated that dalbavancin was active against most strains in pre-established adhered cells and 24 h old biofilms. The current results show that dalbavancin is active against adhered cells and biofilms in vitro, suggesting that this antimicrobial agent may be an option for the treatment of bone infections caused by MRSA.
Collapse
Affiliation(s)
- Vanessa Silva
- Microbiology and Antibiotic Resistance Team (MicroART), Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Functional Genomics and Proteomics Unit, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Associated Laboratory for Green Chemistry (LAQV-REQUIMTE), University NOVA of Lisboa, Lisboa, Caparica, Portugal.,Animal and Veterinary Research Center (CECAV), Universidade de Trás-os-Montes e Alto Douro, Vila Real, Portugal
| | - Carla Miranda
- Microbiology and Antibiotic Resistance Team (MicroART), Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Associated Laboratory for Green Chemistry (LAQV-REQUIMTE), University NOVA of Lisboa, Lisboa, Caparica, Portugal.,Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Mário Bezerra
- Microbiology and Antibiotic Resistance Team (MicroART), Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Functional Genomics and Proteomics Unit, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - H Sofia Antão
- Medical department, Angelini Farmacêutica Lda, Dafundo, Portugal
| | - João Guimarães
- Medical department, Angelini Farmacêutica Lda, Dafundo, Portugal
| | - Justina Prada
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Animal and Veterinary Research Center (CECAV), Universidade de Trás-os-Montes e Alto Douro, Vila Real, Portugal
| | - Isabel Pires
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Animal and Veterinary Research Center (CECAV), Universidade de Trás-os-Montes e Alto Douro, Vila Real, Portugal
| | - Luís Maltez
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Animal and Veterinary Research Center (CECAV), Universidade de Trás-os-Montes e Alto Douro, Vila Real, Portugal
| | - José E Pereira
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Animal and Veterinary Research Center (CECAV), Universidade de Trás-os-Montes e Alto Douro, Vila Real, Portugal
| | - José L Capelo
- BIOSCOPE Group, LAQV@REQUIMTE, Chemistry Deparment, Faculty of Science and Tecnology, Nova University of Lisbon, Almada, Portugal.,PROTEOMASS Scientific Society, Costa de Caparica, Portugal
| | - Gilberto Igrejas
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Functional Genomics and Proteomics Unit, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Associated Laboratory for Green Chemistry (LAQV-REQUIMTE), University NOVA of Lisboa, Lisboa, Caparica, Portugal
| | - Patrícia Poeta
- Microbiology and Antibiotic Resistance Team (MicroART), Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.,Associated Laboratory for Green Chemistry (LAQV-REQUIMTE), University NOVA of Lisboa, Lisboa, Caparica, Portugal.,Animal and Veterinary Research Center (CECAV), Universidade de Trás-os-Montes e Alto Douro, Vila Real, Portugal
| |
Collapse
|
30
|
Queiroz HA, da Silva CR, de Andrade Neto JB, do Av Sá LG, do Nascimento FB, Moreno LS, Barroso FD, da Silva LJ, Cândido TM, de Oliveira LC, de Mesquita JR, de Moraes MO, Cavalcanti BC, Nobre Júnior HV. Synergistic activity of diclofenac sodium with oxacillin against planktonic cells and biofilm of methicillin-resistant Staphylococcus aureus strains. Future Microbiol 2021; 16:375-387. [PMID: 33870731 DOI: 10.2217/fmb-2020-0095] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To evaluate the activity of diclofenac sodium and synergism with oxacillin against clinical strains of SARM in plactonic cells, antibiofilm and biofilm. Materials & methods: Synergism activity was assessed using the fractional inhibitory concentration index and its possible mechanism of action by flow cytometry. Results: The synergistic activity of diclofenac sodium with oxacillin was observed against plactonic cells, antibiofilm and in biofilm formed from clinical methicillin-resistant Staphylococcus aureus strains. Conclusion: This combination caused damage to the integrity of the membrane and ruptures in the DNA of the cells, leading to apoptosis.
Collapse
Affiliation(s)
- Helaine A Queiroz
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Cecília R da Silva
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - João B de Andrade Neto
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil.,University Center Christus, Fortaleza, CE, Brazil
| | - Lívia G do Av Sá
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil.,University Center Christus, Fortaleza, CE, Brazil
| | - Francisca Bsa do Nascimento
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Leticia Ss Moreno
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Fátima Dd Barroso
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Lisandra J da Silva
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Thiago M Cândido
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Leilson C de Oliveira
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - Manoel O de Moraes
- Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Bruno C Cavalcanti
- Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Hélio V Nobre Júnior
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
31
|
Abstract
There is an urgent need for new antibiotics and alternative strategies to combat bacterial pathogens. Molecular docking, antibacterial evaluation in vitro and in vivo, cytotoxicity assessment and enzyme inhibition analyses were performed. Compound 12 exhibited antimicrobial activity against Staphylococcus aureus (MIC: 4 μg/ml), various clinically isolated strains of MRSA (MIC: 4-16 μg/ml) and Acinetobacter baumannii (MIC: 4 μg/ml) when combined with subinhibitory concentrations of colistin B. Compound 12 (20 mg/kg) yielded mild improvement in survival of methicillin-resistant Staphylococcus aureus (MRSA)-infected mice. Additionally, enzyme inhibition tests showed that compound 12 exhibited inhibitory effects against S. aureus dihydrofolate reductase (105.1 μg/ml) and DNA gyrase (122.8 μg/ml). Compound 12 is a promising antibacterial candidate for further development.
Collapse
|
32
|
Brunaugh AD, Sharma S, Smyth H. Inhaled fixed-dose combination powders for the treatment of respiratory infections. Expert Opin Drug Deliv 2021; 18:1101-1115. [PMID: 33632051 DOI: 10.1080/17425247.2021.1886074] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Respiratory infections are a major cause of morbidity and mortality. As an alternative to systemic drug administration, inhaled drug delivery can produce high drug concentrations in the lung tissue to overcome resistant bacteria. The development of inhaled fixed-dose combination powders (I-FDCs) is promising next step in this field, as it would enable simultaneous drug-drug or drug-adjuvant delivery at the site of infection, thereby promoting synergistic activity and improving patient compliance. AREAS COVERED This review covers the clinical and pharmaceutical rationales for the development of I-FDCs for the treatment of respiratory infections, relevant technologies for particle and powder generation, and obstacles which must be addressed to achieve regulatory approval. EXPERT OPINION I-FDCs have been widely successful in the treatment of asthma and chronic obstructive pulmonary disease; however, application of I-FDCs towards the treatment of respiratory infections carries additional challenges related to the high dose requirements and physicochemical characteristics of anti-infective drugs. At present, co-spray drying is an especially promising approach for the development of composite fixed-dose anti-infective particles for inhalation. Though the majority of fixed-dose research has thus far focused on the combination of multiple antibiotics, future work may shift to the additional inclusion of immunomodulatory agents or repurposed non-antibiotics.
Collapse
Affiliation(s)
| | - Shivam Sharma
- Department of Pharmacy & Pharmacology, University of Bath, Bath, UK
| | - Hugh Smyth
- College of Pharmacy, University of Texas at Austin, Austin, USA
| |
Collapse
|
33
|
McClean P, Hunter WR. 17α-ethynylestradiol (EE2) limits the impact of ibuprofen upon respiration by streambed biofilms in a sub-urban stream. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:37149-37154. [PMID: 32681335 PMCID: PMC7456402 DOI: 10.1007/s11356-020-10096-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 07/10/2020] [Indexed: 06/11/2023]
Abstract
Pharmaceutical compounds such as the non-steroidal anti-inflammatory drug ibuprofen and the artificial estrogen 17α-ethynylestradiol (EE2) are contaminants of emerging concern in freshwater systems. Globally, human pharmaceutical use is growing by around ~ 3% per year; yet, we know little about how interactions between different pharmaceuticals may affect aquatic ecosystems. Here, we test how interactions between ibuprofen and EE2 affect the growth and respiration of streambed biofilms. We used contaminant exposure experiments to quantify how these compounds affected biofilm growth (biomass), respiration, net primary production (NPP) and gross primary production (GPP), both individually and in combination. We found no effects of either ibuprofen or EE2 on biofilm biomass (using ash-free dry mass as a proxy) or gross primary production. Ibuprofen significantly reduced biofilm respiration and altered NPP. Concomitant exposure to EE2, however, counteracted the inhibitory effects of ibuprofen upon biofilm respiration. Our study, thus, demonstrates that interactions between pharmaceuticals in the environment may have complex effects upon microbial contributions to aquatic ecosystem functioning.
Collapse
Affiliation(s)
- Peter McClean
- School of Geography and Environmental Science, University of Ulster, Coleraine, BT52 1SA, UK
| | - William Ross Hunter
- School of Geography and Environmental Science, University of Ulster, Coleraine, BT52 1SA, UK.
- Fisheries and Aquatic Ecosystems Branch, Northern Ireland Agri-Food and Bioscience Institute, Belfast, BT9 5PX, UK.
| |
Collapse
|
34
|
Leão C, Borges A, Simões M. NSAIDs as a Drug Repurposing Strategy for Biofilm Control. Antibiotics (Basel) 2020; 9:antibiotics9090591. [PMID: 32927675 PMCID: PMC7558876 DOI: 10.3390/antibiotics9090591] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/01/2020] [Accepted: 09/01/2020] [Indexed: 12/30/2022] Open
Abstract
Persistent infections, usually associated with biofilm-producing bacteria, are challenging for both medical and scientific communities. The potential interest in drug repurposing for biofilm control is growing due to both disinvestment in antibiotic R&D and reduced efficacy of the available panel of antibiotics. In the present study, the antibacterial and antibiofilm activities of four non-steroidal anti-inflammatory drugs (NSAIDs), piroxicam (PXC), diclofenac sodium (DCF), acetylsalicylic acid (ASA) and naproxen sodium (NPX) were evaluated against Escherichia coli and Staphylococcus aureus. The minimum inhibitory/bactericidal concentrations (MICs and MBCs) and the dose–response curves from exposure to the selected NSAIDs were determined. MICs were found for PXC (800 μg/mL) and ASA (1750 μg/mL) against E. coli, and for DCF (2000 μg/mL) and ASA (2000 μg/mL) against S. aureus. No MBCs were found (>2000 μg/mL). The potential of NSAIDs to eradicate preformed biofilms was characterized in terms of biofilm mass, metabolic activity and cell culturability. Additionally, the NSAIDs were tested in combination with kanamycin (KAN) and tetracycline (TET). ASA, DCF and PXC promoted significant reductions in metabolic activity and culturability. However, only PXC promoted biofilm mass removal. Additive interactions were obtained for most of the combinations between NSAIDs and KAN or TET. In general, NSAIDs appear to be a promising strategy to control biofilms as they demonstrated to be more effective than conventional antibiotics.
Collapse
Affiliation(s)
- Cláudia Leão
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal; (C.L.); (A.B.)
| | - Anabela Borges
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal; (C.L.); (A.B.)
- DEQ—Department of Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
| | - Manuel Simões
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal; (C.L.); (A.B.)
- DEQ—Department of Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
- Correspondence: ; Tel.: +351-225-081-654; Fax: +351-225-081-449
| |
Collapse
|
35
|
Mohammed MA, Ahmed MT, Anwer BE, Aboshanab KM, Aboulwafa MM. Propranolol, chlorpromazine and diclofenac restore susceptibility of extensively drug-resistant (XDR)-Acinetobacter baumannii to fluoroquinolones. PLoS One 2020; 15:e0238195. [PMID: 32845920 PMCID: PMC7449414 DOI: 10.1371/journal.pone.0238195] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 08/11/2020] [Indexed: 12/16/2022] Open
Abstract
Nosocomial infections caused by extensively drug-resistant (XDR) or Pan-Drug resistant (PDR) Acinetobacter (A.) baumannii have recently increased dramatically creating a medical challenge as therapeutic options became very limited. The aim of our study was to investigate the antibiotic-resistance profiles and evaluate the various combinations of ciprofloxacin (CIP) or levofloxacin (LEV) with antimicrobial agents and non-antimicrobial agents to combat antimicrobial resistance of XDR A. baumannii. A total of 100 (6.25%) A. baumannii clinical isolates were recovered from 1600 clinical specimens collected from hospitalized patients of two major university hospitals in Upper Egypt. Antimicrobial susceptibility tests were carried out according to CLSI guidelines. Antimicrobial susceptibility testing of the respective isolates showed a high percentage of bacterial resistance to 19 antimicrobial agents ranging from 76 to99%. However, a lower percentage of resistance was observed for only colistin (5%) and doxycycline (57%). The isolates were categorized as PDR (2; 2%), XDR (68; 68%), and multi-drug resistant (MDR) (30; 30%). Genotypic analysis using ERIC-PCR on 2 PDR and 32 selected XDR isolates showed that they were not clonal. Combinations of CIP or LEV with antibiotics (including, ampicillin, ceftriaxone, amikacin, or doxycycline) were tested on these A. baumannii non-clonal isolates using standard protocols where fractional inhibitory concentrations (-FICs) were calculated. Results of the respective combinations showed synergism in 23.5%, 17.65%, 32.35%, 17.65% and 26.47%, 8.28%, 14.71%, 26.47%, of the tested isolates, respectively. CIP or LEV combinations with either chlorpromazine (CPZ) 200 μg/ml, propranolol (PR) in two concentrations, 0.5 mg/ml and 1.0 mg/ml or diclofenac (DIC) 4 mg/ml were carried out and the MIC decrease factor (MDF) of each isolate was calculated and results showed synergism in 44%, 50%, 100%, 100% and 94%, 85%, 100%, 100%, of the tested isolates, respectively. In conclusion, combinations of CIP or LEV with CPZ, PR, or DIC showed synergism in most of the selected PDR and XDR A. baumannii clinical isolates. However, these combinations have to be re-evaluated in vivo using appropriate animal models infected by XDR- or PDR- A. baumannii.
Collapse
Affiliation(s)
- Mostafa A. Mohammed
- Department of Microbiology and Immunology, Faculty of Pharmacy, Al Azhar University, Assiut, Egypt
| | - Mohammed T. Ahmed
- Department of Microbiology and Immunology, Faculty of Pharmacy, Al Azhar University, Assiut, Egypt
| | - Bahaa E. Anwer
- Department of Microbiology and Immunology, Faculty of Pharmacy, Al Azhar University, Assiut, Egypt
| | - Khaled M. Aboshanab
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mohammad M. Aboulwafa
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
36
|
Dubey KK, Indu, Sharma M. Reprogramming of antibiotics to combat antimicrobial resistance. Arch Pharm (Weinheim) 2020; 353:e2000168. [DOI: 10.1002/ardp.202000168] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/04/2020] [Accepted: 07/11/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Kashyap K. Dubey
- Bioprocess Engineering Laboratory, Department of Biotechnology Central University of Haryana Mahendergarh Haryana India
- School of Biotechnology Jawaharlal Nehru University New Delhi India
| | - Indu
- Bioprocess Engineering Laboratory, Department of Biotechnology Central University of Haryana Mahendergarh Haryana India
| | - Manisha Sharma
- Bioprocess Engineering Laboratory, Department of Biotechnology Central University of Haryana Mahendergarh Haryana India
| |
Collapse
|
37
|
Caldara M, Marmiroli N. Known Antimicrobials Versus Nortriptyline in Candida albicans: Repositioning an Old Drug for New Targets. Microorganisms 2020; 8:microorganisms8050742. [PMID: 32429222 PMCID: PMC7284794 DOI: 10.3390/microorganisms8050742] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/05/2020] [Accepted: 05/12/2020] [Indexed: 02/07/2023] Open
Abstract
Candida albicans has the capacity to develop resistance to commonly used antimicrobials, and to solve this problem, drug repositioning and new drug combinations are being studied. Nortriptyline, a tricyclic antidepressant, was shown to have the capacity to inhibit biofilm and hyphae formation, along with the ability to efficiently kill cells in a mature biofilm. To use nortriptyline as a new antimicrobial, or in combination with known drugs to increase their actions, it is important to characterize in more detail the effects of this drug on the target species. In this study, the Candida albicans GRACE™ collection and a Haplo insufficiency profiling were employed to identify the potential targets of nortriptyline, and to classify, in a parallel screening with amphotericin B, caspofungin, and fluconazole, general multi-drug resistance genes. The results identified mutants that, during biofilm formation and upon treatment of a mature biofilm, are sensitive or tolerant to nortriptyline, or to general drug treatments. Gene ontology analysis recognized the categories of ribosome biogenesis and spliceosome as enriched upon treatment with the tricyclic antidepressant, while mutants in oxidative stress response and general stress response were commonly retrieved upon treatment with any other drug. The data presented suggest that nortriptyline can be considered a “new” antimicrobial drug with large potential for application to in vivo infection models.
Collapse
Affiliation(s)
- Marina Caldara
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy;
- Correspondence: ; Tel.: +39-0521-905658
| | - Nelson Marmiroli
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy;
- Interdepartmental Center SITEIA.PARMA, University of Parma, Parco Area delle Scienze 181/A, 43124 Parma, Italy
| |
Collapse
|
38
|
Antibacterial activity of griseofulvin analogues as an example of drug repurposing. Int J Antimicrob Agents 2020; 55:105884. [PMID: 31931149 DOI: 10.1016/j.ijantimicag.2020.105884] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/19/2019] [Accepted: 12/28/2019] [Indexed: 01/30/2023]
Abstract
Griseofulvin is a well-known antifungal drug that was launched in 1962 by Merck & Co. for the treatment of dermatophyte infections. However, according to predictions using the Way2Drug computational drug repurposing platform, it may also have antibacterial activity. As no confirmation of this prediction was found in the published literature, this study estimated in-silico antibacterial activity for 42 griseofulvin derivatives. Antibacterial activity was predicted for 33 of the 42 compounds, which led to the conclusion that this activity might be considered as typical for this chemical series. Therefore, experimental testing of antibacterial activity was performed on a panel of Gram-positive and Gram-negative micro-organisms. Antibacterial activity was evaluated using the microdilution method detecting the minimal inhibitory concentration (MIC) and the minimal bactericidal concentration (MBC). The tested compounds exhibited potent antibacterial activity against all the studied bacteria, with MIC and MBC values ranging from 0.0037 to 0.04 mg/mL and from 0.01 to 0.16 mg/mL, respectively. Activity was 2.5-12 times greater than that of ampicillin and 2-8 times greater than that of streptomycin, which were used as the reference drugs. Similarity analysis for all 42 compounds with the (approximately) 470,000 drug-like compounds indexed in the Clarivate Analytics Integrity database confirmed the significant novelty of the antibacterial activity for the compounds from this chemical class. Therefore, this study demonstrated that by using computer-aided prediction of biological activity spectra for a particular chemical series, it is possible to identify typical biological activities which may be used for discovery of new applications (e.g. drug repurposing).
Collapse
|
39
|
Kumar R, Harilal S, Gupta SV, Jose J, Thomas Parambi DG, Uddin MS, Shah MA, Mathew B. Exploring the new horizons of drug repurposing: A vital tool for turning hard work into smart work. Eur J Med Chem 2019; 182:111602. [PMID: 31421629 PMCID: PMC7127402 DOI: 10.1016/j.ejmech.2019.111602] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/07/2019] [Accepted: 08/07/2019] [Indexed: 02/07/2023]
Abstract
Drug discovery and development are long and financially taxing processes. On an average it takes 12-15 years and costs 1.2 billion USD for successful drug discovery and approval for clinical use. Many lead molecules are not developed further and their potential is not tapped to the fullest due to lack of resources or time constraints. In order for a drug to be approved by FDA for clinical use, it must have excellent therapeutic potential in the desired area of target with minimal toxicities as supported by both pre-clinical and clinical studies. The targeted clinical evaluations fail to explore other potential therapeutic applications of the candidate drug. Drug repurposing or repositioning is a fast and relatively cheap alternative to the lengthy and expensive de novo drug discovery and development. Drug repositioning utilizes the already available clinical trials data for toxicity and adverse effects, at the same time explores the drug's therapeutic potential for a different disease. This review addresses recent developments and future scope of drug repositioning strategy.
Collapse
Affiliation(s)
- Rajesh Kumar
- Department of Pharmacy, Kerala University of Health Sciences, Thrissur, Kerala, India
| | - Seetha Harilal
- Department of Pharmacy, Kerala University of Health Sciences, Thrissur, Kerala, India
| | - Sheeba Varghese Gupta
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA
| | - Jobin Jose
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Science, NITTE Deemed to be University, Manglore, 575018, India
| | - Della Grace Thomas Parambi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Al Jouf, 2014, Saudi Arabia
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Muhammad Ajmal Shah
- Department of Pharmacogonosy, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, 678557, Kerala, India.
| |
Collapse
|
40
|
Nitrofurantoin-Microbial Degradation and Interactions with Environmental Bacterial Strains. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16091526. [PMID: 31052168 PMCID: PMC6539117 DOI: 10.3390/ijerph16091526] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/16/2019] [Accepted: 04/26/2019] [Indexed: 02/07/2023]
Abstract
The continuous exposure of living organisms and microorganisms to antibiotics that have increasingly been found in various environmental compartments may be perilous. One group of antibacterial agents that have an environmental impact that has been very scarcely studied is nitrofuran derivatives. Their representative is nitrofurantoin (NFT)-a synthetic, broad-spectrum antibiotic that is often overdosed. The main aims of the study were to: (a) isolate and characterize new microbial strains that are able to grow in the presence of NFT, (b) investigate the ability of isolates to decompose NFT, and (c) study the impact of NFT on microbial cell properties. As a result, five microbial species were isolated. A 24-h contact of bacteria with NFT provoked modifications in microbial cell properties. The greatest differences were observed in Sphingobacterium thalpophilum P3d, in which a decrease in both total and inner membrane permeability (from 86.7% to 48.3% and from 0.49 to 0.42 µM min-1) as well as an increase in cell surface hydrophobicity (from 28.3% to 39.7%) were observed. Nitrofurantoin removal by selected microbial cultures ranged from 50% to 90% in 28 days, depending on the bacterial strain. Although the isolates were able to decompose the pharmaceutical, its presence significantly affected the bacterial cells. Hence, the environmental impact of NFT should be investigated to a greater extent.
Collapse
|
41
|
Yang X, Huang WT, Wu HY, He RQ, Ma J, Liu AG, Chen G. Novel drug candidate for the treatment of several soft‑tissue sarcoma histologic subtypes: A computational method using survival‑associated gene signatures for drug repurposing. Oncol Rep 2019; 41:2241-2253. [PMID: 30816547 PMCID: PMC6412453 DOI: 10.3892/or.2019.7033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 02/22/2019] [Indexed: 12/11/2022] Open
Abstract
Systemic treatment options for soft tissue sarcomas (STSs) have remained unchanged despite the need for novel drug candidates to improve STS outcomes. Drug repurposing involves the application of clinical drugs to different diseases, reducing development time, and cost. It has also become a fast and effective way to identify drug candidates. The present study used a computational method to screen three drug-gene interaction databases for novel drug candidates for the treatment of several common STS histologic subtypes through drug repurposing. STS survival-associated genes were generated by conducting a univariate cox regression analysis using The Cancer Genome Atlas survival data. These genes were then applied to three databases (the Connectivity Map, the Drug Gene Interaction Database and the L1000 Fireworks Display) to identify drug candidates for STS treatment. Additionally, pathway analysis and molecular docking were conducted to evaluate the molecular mechanisms of the candidate drug. Bepridil was identified as a potential candidate for several STS histologic subtype treatments by overlapping the screening results from three drug-gene interaction databases. The pathway analysis with the Kyoto Encyclopedia of Genes and Genomes predicted that Bepridil may target CRK, fibroblast growth factor receptor 4 (FGFR4), laminin subunit β1 (LAMB1), phosphoinositide-3-kinase regulatory subunit 2 (PIK3R2), WNT5A, cluster of differentiation 47 (CD47), elastase, neutrophil expressed (ELANE), 15-hydroxyprostaglandin dehydrogenase (HPGD) and protein kinase cβ (PRKCB) to suppress STS development. Further molecular docking simulation suggested a relatively stable binding selectivity between Bepridil and eight proteins (CRK, FGFR4, LAMB1, PIK3R2, CD47, ELANE, HPGD, and PRKCB). In conclusion, a computational method was used to identify Bepridil as a potential candidate for the treatment of several common STS histologic subtypes. Experimental validation of these in silico results is necessary before clinical translation can occur.
Collapse
Affiliation(s)
- Xia Yang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Wen-Ting Huang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Hua-Yu Wu
- Department of Cell Biology and Genetics, School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Rong-Quan He
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jie Ma
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - An-Gui Liu
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Gang Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|