1
|
Chung CH, Chang DC, Rhoads NM, Shay MR, Srinivasan K, Okezue MA, Brunaugh AD, Chandrasekaran S. Transfer learning predicts species-specific drug interactions in emerging pathogens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.597386. [PMID: 38895385 PMCID: PMC11185605 DOI: 10.1101/2024.06.04.597386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Machine learning (ML) algorithms are necessary to efficiently identify potent drug combinations within a large candidate space to combat drug resistance. However, existing ML approaches cannot be applied to emerging and under-studied pathogens with limited training data. To address this, we developed a transfer learning and crowdsourcing framework (TACTIC) to train ML models on data from multiple bacteria. TACTIC was built using 2,965 drug interactions from 12 bacterial strains and outperformed traditional ML models in predicting drug interaction outcomes for species that lack training data. Top TACTIC model features revealed genetic and metabolic factors that influence cross-species and species-specific drug interaction outcomes. Upon analyzing ~600,000 predicted drug interactions across 9 metabolic environments and 18 bacterial strains, we identified a small set of drug interactions that are selectively synergistic against Gram-negative (e.g., A. baumannii) and non-tuberculous mycobacteria (NTM) pathogens. We experimentally validated synergistic drug combinations containing clarithromycin, ampicillin, and mecillinam against M. abscessus, an emerging pathogen with growing levels of antibiotic resistance. Lastly, we leveraged TACTIC to propose selectively synergistic drug combinations to treat bacterial eye infections (endophthalmitis).
Collapse
Affiliation(s)
- Carolina H. Chung
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - David C. Chang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Nicole M. Rhoads
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Madeline R. Shay
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Karthik Srinivasan
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Mercy A. Okezue
- Department of Pharmaceutical Sciences, University of Michigan College of Pharmacy, Ann Arbor, MI, 48109, USA
| | - Ashlee D. Brunaugh
- Department of Pharmaceutical Sciences, University of Michigan College of Pharmacy, Ann Arbor, MI, 48109, USA
| | - Sriram Chandrasekaran
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Program in Chemical Biology, University of Michigan, Ann Arbor, MI, 48109, USA
- Center for Bioinformatics and Computational Medicine, Ann Arbor, MI, 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| |
Collapse
|
2
|
Kwankaew P, Sangkanu S, Mitsuwan W, Boonhok R, Lao-On U, Tabo HL, Mahboob T, de Lourdes Pereira M, Tangpong J, Sundar SS, Wiart C, Nissapatorn V. Inhibitory and anti-adherent effects of Piper betle L. leaf extract against Acanthamoeba triangularis in co-infection with Staphylococcus aureus and Pseudomonas aeruginosa: A sustainable one-health approach. Vet World 2024; 17:848-862. [PMID: 38798284 PMCID: PMC11111711 DOI: 10.14202/vetworld.2024.848-862] [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: 01/15/2024] [Accepted: 03/26/2024] [Indexed: 05/29/2024] Open
Abstract
Background and Aim Keratitis is a serious ocular infection often caused by pathogenic microorganisms such as Acanthamoeba spp. Among other harmful microbes, Acanthamoeba keratitis presents a particular challenge due to its resistance to conventional antimicrobial agents. Piper betle Linn., commonly known as betel leaf, has been traditionally used for its medicinal properties. This study aimed to assess the potential of the leaf ethanol extract of P. betle Linn. in the treatment of Acanthamoeba triangularis in monoculture and co-culture with two prevalent pathogenic bacteria, Staphylococcus aureus and Pseudomonas aeruginosa, associated with keratitis. Materials and Methods Minimum inhibitory concentrations (MICs) of A. triangularis, S. aureus, and P. aeruginosa extracts in monoculture and coinfected conditions were examined. In addition, this study explored the potential of the extract in preventing Acanthamoeba adherence in both monoculture and co-culture environments. Scanning electron microscopy (SEM) analysis confirmed the impact of the extract on Acanthamoeba cell membranes, including acanthopodia. Furthermore, a time-kill kinetic assay was used to validate the amoebicidal activity of the extract against A. triangularis and the tested bacteria. Results MICs for trophozoites, cysts, P. aeruginosa, and S. aureus in the monoculture were 0.25, 0.25, 0.51, and 0.128 mg/mL, respectively, whereas the MICs for Acanthamoeba coinfected with bacteria were higher than those in the monoculture. This extract inhibited the growth of A. triangularis trophozoites and cysts for up to 72 h. Moreover, P. betle extract effectively prevented the adherence of Acanthamoeba to contact lenses under monoculture conditions. SEM analysis confirmed that P. betle extract affects the cell membrane of Acanthamoeba, including Acanthopodia. In addition, the time-kill kinetic assay confirmed that the extract contained amoebicidal activity against A. triangularis, including the tested bacteria. Notably, S. aureus was more susceptible than A. triangularis and P. aeruginosa to P. betle extract treatment. Unexpectedly, our study revealed that S. aureus negatively affected A. triangularis in the co-culture after 3 days of incubation, whereas P. aeruginosa facilitated the growth of A. triangularis in the presence of the extract. Conclusion This study provides compelling evidence of the anti-adhesive and anti-Acanthamoeba properties of P. betle leaf extract against A. triangularis under monoculture and co-culture conditions. The observed impact on Acanthamoeba cell membranes, coupled with the time-kill kinetic assay results, underscores the potential of P. betle leaf extract as a promising agent for combating Acanthamoeba-related infections in humans and animals.
Collapse
Affiliation(s)
- Pattamaporn Kwankaew
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, Thailand
| | - Suthinee Sangkanu
- School of Allied Health Sciences, Southeast Asia Water Team (SEA Water Team), World Union for Herbal Drug Discovery, and Research Excellence Center for Innovation and Health Products, Walailak University, Nakhon Si Thammarat, Thailand
| | - Watcharapong Mitsuwan
- Akkhraratchakumari Veterinary College and Research Center of Excellence in Innovation of Essential Oil, Walailak University, Nakhon Si Thammarat, Thailand
| | - Rachasak Boonhok
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, Thailand
| | - Udom Lao-On
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, Thailand
| | - Hazel L. Tabo
- Department of Biological Sciences, College of Science and Computer Studies, De La Salle University-Dasmariñas, Cavite, Philippines
| | - Tooba Mahboob
- Department of Pharmaceutical Biology, Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Maria de Lourdes Pereira
- CICECO-Aveiro Institute of Materials and Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Jitbanjong Tangpong
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, Thailand
| | - Shanmuga S. Sundar
- Department of Biotechnology, Aarupadai Veedu Institute of Technology, Vinayaka Mission’s Research Foundation Chennai Campus, Paiyanoor, Chennai, India
| | - Christophe Wiart
- Institute of Tropical Biology and Conservation, Universiti Malaysia Sabah, Malaysia
| | - Veeranoot Nissapatorn
- School of Allied Health Sciences, Southeast Asia Water Team (SEA Water Team), World Union for Herbal Drug Discovery, and Research Excellence Center for Innovation and Health Products, Walailak University, Nakhon Si Thammarat, Thailand
| |
Collapse
|
3
|
Shah S, Wozniak RAF. Staphylococcus aureus and P seudomonas aeruginosa infectious keratitis: key bacterial mechanisms that mediate pathogenesis and emerging therapeutics. Front Cell Infect Microbiol 2023; 13:1250257. [PMID: 37671149 PMCID: PMC10475732 DOI: 10.3389/fcimb.2023.1250257] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/04/2023] [Indexed: 09/07/2023] Open
Abstract
Bacterial keratitis (bacterial infection of the cornea) is a major cause of vision loss worldwide. Given the rapid and aggressive nature of the disease, immediate broad-spectrum antibiotics are essential to adequately treat this disease. However, rising antibiotic resistance continues to accelerate, rendering many commonly used therapeutics increasingly ineffective. As such, there is a significant effort to understand the basic pathogenesis of common causative organisms implicated in keratitis in part, to fuel the development of novel therapies to treat this blinding disease. This review explores two common causes of bacterial keratitis, Staphylococcus aureus and Pseudomonas aeruginosa, with regards to the bacterial mediators of virulence as well as novel therapies on the horizon.
Collapse
Affiliation(s)
| | - Rachel A. F. Wozniak
- Department of Ophthalmology, The University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| |
Collapse
|
4
|
Al Hagbani T, Rizvi SMD, Shakil S, Lila ASA. Nano-Formulating Besifloxacin and Employing Quercetin as a Synergizer to Enhance the Potency of Besifloxacin against Pathogenic Bacterial Strains: A Nano-Synergistic Approach. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2083. [PMID: 37513094 PMCID: PMC10384196 DOI: 10.3390/nano13142083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/01/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023]
Abstract
The present study applied a nano-synergistic approach to enhance besifloxacin's potency via nano-formulating besifloxacin on gold nanoparticles (Besi-AuNPs) and adding quercetin as a natural synergistic compound. In fact, a one-pot AuNP synthesis approach was applied for the generation of Besi-AuNPs, where besifloxacin itself acted as a reducing and capping agent. Characterization of Besi-AuNPs was performed by spectrophotometry, DLS, FTIR, and electron microscopy techniques. Moreover, antibacterial assessment of pure besifloxacin, Besi-AuNPs, and their combinations with quercetin were performed on Staphylococcus aureus, Pseudomonas aeruginosa, and Escherichia coli. UV-spectra showed a peak of AuNPs at 526 nm, and the electron microscopy-based size was estimated to be 15 ± 3 nm. The effective MIC50 concentrations of besifloxacin after loading on AuNPs were reduced by approximately 50% against the tested bacterial strains. Interestingly, adding quercetin to Besi-AuNPs further enhanced their antibacterial potency, and isobologram analysis showed synergistic potential (combination index below 1) for different quercetin and Besi-AuNP combinations. However, Besi-AuNPs and quercetin combinations were most effective against Gram-positive S. aureus in comparison to Gram-negative P. aeruginosa and E. coli. Their potent activity against S. aureus has its own clinical significance, as it is one the main causative agents of ocular infection, and besifloxacin is primarily used for treating infectious eye diseases. Thus, the outcomes of the present study could be explored further to provide better medication for eye infections caused by resistant pathogens.
Collapse
Affiliation(s)
- Turki Al Hagbani
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Ha'il 81442, Saudi Arabia
| | - Syed Mohd Danish Rizvi
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Ha'il 81442, Saudi Arabia
| | - Shazi Shakil
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Amr Selim Abu Lila
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Ha'il 81442, Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
5
|
Waseem M, Naveed M, Rehman SU, Makhdoom SI, Aziz T, Alharbi M, Alsahammari A, Alasmari AF. Molecular Characterization of spa, hld, fmhA, and l ukD Genes and Computational Modeling the Multidrug Resistance of Staphylococcus Species through Callindra harrisii Silver Nanoparticles. ACS OMEGA 2023; 8:20920-20936. [PMID: 37323409 PMCID: PMC10268295 DOI: 10.1021/acsomega.3c01597] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/24/2023] [Indexed: 06/17/2023]
Abstract
The problem of multidrug resistance in bacterial pathogens is significant and is related to the high morbidity and death rates of living things due to increased levels of beta-lactamases. Plant-derived nanoparticles have gained a great significance in the field of science and technology to combat bacterial diseases, especially multidrug-resistant bacteria. This study examines the multidrug resistance and virulent genes of identified pathogenic Staphylococcus species obtained from Molecular Biotechnology and Bioinformatics Laboratory (MBBL), culture collection. The polymerase chain reaction-based characterization of Staphylococcus aureus and Staphylococcus argenteus having ON875315.1 and ON876003.1 accession IDs revealed the presence of the spa, LukD, fmhA, and hld genes. The green synthesis of silver nanoparticles (AgNPs) was carried out by utilizing the leaf extract of Calliandra harrisii, of which metabolites act as capping and reducing agents for the precursor of nano-synthesis, i.e., AgNO3 of 0.25 M. The synthesized AgNPs were characterized via UV-vis spectroscopy, Fourier transform infrared spectroscopy, scanning electron microscopy, and energy-dispersive X-ray analysis which inferred the bead-like shape of our nanoparticles with the size of 2.21 nm with the existence of aromatic and hydroxyl functional groups at surface plasmon resonance of 477 nm. The antimicrobial activity by AgNPs showed 20 mm inhibition of Staphylococcus species as compared to the vancomycin and cefoxitin antibiotics along with crude plant extract, which showed a minimum zone of inhibition. The synthesized AgNPs were also analyzed for various biological activities like anti-inflammatory with 99.15% inhibition in protein denaturation, antioxidant with 99.8% inhibition in free radical scavenging, antidiabetic with 90.56% inhibition of alpha amylase assay, and anti-haemolytic with 89.9% inhibition in cell lysis which shows good bioavailability and biocompatibility of the nanoparticles with the biological system of the living being. The amplified genes (spa, LukD, fmhA, and hld) were also analyzed for their interaction with AgNPs computationally at the molecular level. The 3-D structure of AgNP and amplified genes was retrieved from ChemSpider (ID: 22394) and Phyre2 online server, respectively. The binding affinities of AgNP with spa, LukD, fmhA, and hld were -7.16, -6.5, -6.45, and -3.3 kJ/mol, respectively, which infers a good docking score except of hld which is -3.3 kJ/mol due to its small size. The salient features of biosynthesized AgNPs proved to be an effective approach in combating the multidrug-resistant Staphylococcus species in the future.
Collapse
Affiliation(s)
- Muhammad Waseem
- Department of Biotechnology,
Faculty of Science and Technology, University
of Central Punjab, Lahore 54590, Pakistan
| | - Muhammad Naveed
- Department of Biotechnology,
Faculty of Science and Technology, University
of Central Punjab, Lahore 54590, Pakistan
| | - Shafiq ur Rehman
- Department of Basic and Applied Chemistry, Faculty of Science and
Technology, University of Central Punjab, Lahore 54000, Pakistan
| | - Syeda Izma Makhdoom
- Department of Biotechnology,
Faculty of Science and Technology, University
of Central Punjab, Lahore 54590, Pakistan
| | - Tariq Aziz
- Department of Agriculture, University of Ioannina, Arta 47100, Greece
| | - Metab Alharbi
- Department of Pharmacology and Toxicology,
College of Pharmacy, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Abdulrahman Alsahammari
- Department of Pharmacology and Toxicology,
College of Pharmacy, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Abdullah F. Alasmari
- Department of Pharmacology and Toxicology,
College of Pharmacy, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
6
|
Nwabor LC, Chukamnerd A, Nwabor OF, Pomwised R, Voravuthikunchai SP, Chusri S. Rifampicin Enhanced Carbapenem Activity with Improved Antibacterial Effects and Eradicates Established Acinetobacter baumannii Biofilms. Pharmaceuticals (Basel) 2023; 16:ph16040477. [PMID: 37111234 PMCID: PMC10141143 DOI: 10.3390/ph16040477] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 04/29/2023] Open
Abstract
Biofilm-mediated infections are critical to public health and a leading cause of resistance among pathogens, amounting to a prolonged hospital stay and increased mortality rate in the intensive care unit. In this study, the antibacterial and antibiofilm activities of rifampicin or carbapenem monotherapies were compared with rifampicin and carbapenem combination therapies against rifampicin-resistant and carbapenem-resistant Acinetobacter baumannii isolates. Among 29 CRAB isolates, 24/29 (83%) were resistant to rifampicin, with MIC values between 2-256 µg/mL. Checkerboard assays disclosed that combination therapies at FICIs between 1/8 and 1/4 improved the activity of carbapenems at subinhibitory concentrations. Time-kill kinetics indicated a 2- to 4-log reduction at 1/2 MIC rifampicin + 1/4 MIC carbapenem and 1/4 MIC rifampicin + 1/4 MIC carbapenem against the isolates, with the MIC values ranging from 2-8 µg/mL. The MTT assay revealed a dose-dependent decrease of the cell viability of established bacterial biofilm at 4 MIC rifampicin + 2 MIC carbapenems, with a percentage reduction of 44-75%, compared with monotherapies at 16 MIC. Scanning electron microscopy further confirmed bacterial cell membrane disruption, suggesting a synergism between carbapenem and rifampicin against a representative isolate. The findings demonstrated that the combination of rifampicin with carbapenems could improve antibacterial activities and eradicate established Acinetobacter baumannii biofilm.
Collapse
Affiliation(s)
- Lois Chinwe Nwabor
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Arnon Chukamnerd
- Division of Infectious Diseases, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Ozioma Forstinus Nwabor
- Division of Infectious Diseases, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Rattanaruji Pomwised
- Division of Biological Science, Faculty of Science, Prince of Songkla University, Songkhla 90110, Thailand
| | - Supayang P Voravuthikunchai
- Division of Biological Science, Faculty of Science, Prince of Songkla University, Songkhla 90110, Thailand
- Natural Product Research Center of Excellence, Faculty of Science, Prince of Songkla University, Songkhla 90110, Thailand
- Center of Antimicrobial Biomaterial Innovation-Southeast Asia, Prince of Songkla University, Songkhla 90110, Thailand
| | - Sarunyou Chusri
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
- Division of Infectious Diseases, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| |
Collapse
|
7
|
Li M, Xin D, Gao J, Yi Q, Yuan J, Bao Y, Gong Y. The protective effect of URP20 on ocular Staphylococcus aureus and Escherichia coli infection in rats. BMC Ophthalmol 2022; 22:517. [PMID: 36585631 PMCID: PMC9801630 DOI: 10.1186/s12886-022-02752-w] [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: 02/28/2022] [Accepted: 12/21/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Infectious keratitis, a medical emergency with acute and rapid disease progression may lead to severe visual impairment and even blindness. Herein, an antimicrobial polypeptide from Crassostrea hongkongensis, named URP20, was evaluated for its therapeutic efficacy against keratitis caused by Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli) infection in rats, respectively. METHODS A needle was used to scratch the surface of the eyeballs of rats and infect them with S. aureus and E.coli to construct a keratitis model. The two models were treated by giving 100 μL 100 μM URP20 drops. Positive drugs for S. aureus and E. coli infection were cefazolin eye drops and tobramycin eye drops, respectively. For the curative effect, the formation of blood vessels in the fundus was observed by a slit lamp (the third day). At the end of the experiment, the condition of the injured eye was photographed by cobalt blue light using 5 μL of 1% sodium fluorescein. The pathological damage to corneal tissues was assessed using hematoxylin-eosin staining, and the expression level of vascular endothelial growth factor (VEGF) was detected by immunohistochemistry. RESULTS URP20 alleviated the symptoms of corneal neovascularization as observed by slit lamp and cobalt blue lamp. The activity of S. aureus and E.coli is inhibited by URP20 to protect corneal epithelial cells and reduce corneal stromal bacterial invasion. It also prevented corneal thickening and inhibited neovascularization by reducing VEGF expression at the cornea. CONCLUSION URP20 can effectively inhibit keratitis caused by E.coli as well as S. aureus in rats, as reflected by the inhibition of corneal neovascularization and the reduction in bacterial damage to the cornea.
Collapse
Affiliation(s)
- Meng Li
- grid.203507.30000 0000 8950 5267School of Medicine, Ningbo University, Ningbo, 315042 China ,Department of Ophtalmology, Ningbo Eye Hospital, Ningbo, 315042 China
| | - Danli Xin
- Department of Ophtalmology, Ningbo Eye Hospital, Ningbo, 315042 China
| | - Jian Gao
- Department of Ophtalmology, Ningbo Eye Hospital, Ningbo, 315042 China
| | - Quanyong Yi
- Department of Ophtalmology, Ningbo Eye Hospital, Ningbo, 315042 China
| | - Jianshu Yuan
- Department of Ophtalmology, Ningbo Eye Hospital, Ningbo, 315042 China
| | - Yongbo Bao
- grid.413076.70000 0004 1760 3510College of Biological & Environmental Sciences, Zhejiang Wanli University, Ningbo, 315100 China
| | - Yan Gong
- Department of Ophtalmology, Ningbo Eye Hospital, Ningbo, 315042 China ,grid.203507.30000 0000 8950 5267Department of Ophtalmology, Medical College of Ningbo University, Ningbo Eye Hospital, No. 599, Beiming Cheng Road, Yinzhou District, Ningbo, 315042 China
| |
Collapse
|
8
|
Hardie KR, Fenn SJ. JMM profile: rifampicin: a broad-spectrum antibiotic. J Med Microbiol 2022; 71. [PMID: 35930318 DOI: 10.1099/jmm.0.001566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Rifampicin (also known as rifampin) inhibits RNA synthesis, and is used to treat tuberculosis, leprosy, staphylococcal infections and legionnaires’ disease. It can also protect at-risk populations from
Haemophilus influenzae
type b and
Neisseria meningitidis
. It is a polyketide antibiotic and is on the World Health Organization (WHO) list of essential medicines due to its critical importance to human medicine. The adverse effect of liver toxicity is controlled by testing during prolonged treatment regimes. Rifampicin’s red–orange colour can result in the colouration of sweat, tears and urine. Resistance to rifampicin arises from mutation of the target RNA polymerase or ADP ribosylation of the antibiotic or efflux. Mycobacteria may become singularly resistant to rifampicin or as part of multidrug or extensive drug resistance.
Collapse
Affiliation(s)
- Kim R Hardie
- School of Life Sciences, Biodiscovery Institute, University Park, Nottingham University, Nottingham, NG7 2RD, UK
| | - Samuel Jacob Fenn
- School of Life Sciences, Biodiscovery Institute, University Park, Nottingham University, Nottingham, NG7 2RD, UK
| |
Collapse
|
9
|
Wang Z, Zhang P, Huang C, Guo Y, Dong X, Li X. Conjunctival sac bacterial culture of patients using levofloxacin eye drops before cataract surgery: a real-world, retrospective study. BMC Ophthalmol 2022; 22:328. [PMID: 35907940 PMCID: PMC9338605 DOI: 10.1186/s12886-022-02544-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/20/2022] [Indexed: 11/21/2022] Open
Abstract
Background The use of antibiotics preoperatively is effective to decrease the incidence of ocular bacterial infections but may lead to high resistance rate, especially on patients with multi-risk clinical factors. This study systematically analyzed real-world data (RWD) of patients to reveal the association between clinical factors and conjunctival sac bacterial load and offer prophylaxis suggestions. Methods We retrieved RWD of patients using levofloxacin eye drops (5 mL: 24.4 mg, 4 times a day for 3 days) preoperatively. Retrieved data included information on the conjunctival sac bacterial culture, sex, presence of hypertension and diabetes mellitus (DM), and history of hospital-based surgeries. Data was analyzed using SPSS 24.0. Results RWD of 15,415 cases (patients) were retrieved. Among these patients, 5,866 (38.1%) were males and 9,549 (61.9%) females. 5,960 (38.7%) patients had a history of hypertension, and 3,493 (22.7%) patients had a history of DM. 7,555 (49.0%) patients had a history of hospital-based operations. There were 274 (1.8%) positive bacterial cultures. Male patients with hypertension and DM may be at increased risk of having positive bacterial cultures (P < 0.05). Staphylococcus epidermidis (n = 56, 20.4%), Kocuria rosea (n = 37, 13.5%), and Micrococcus luteus (n = 32, 11.7%) were the top 3 isolated strains. Most bacterial strains were resistant to various antibiotics except rifampin, and 82.5% (33 of 40 isolates) of Staphylococcus epidermidis isolates had multidrug antibiotic resistance. Numbers of culture-positive Staphylococcus epidermidis isolates in the male group and non-DM group were greater than those in the female and DM groups, respectively. Micrococcus luteus (n = 11, 8.8%) was found less frequently in non-hypertension group than in hypertension group. Conclusion Sex (Male) and the presence of hypertension and DM are risk factors for greater conjunctival sac bacterial loads. We offer a prophylactic suggestion based on the combined use of levofloxacin and rifampin. However, this approach may aggravate risk of multidrug resistance.
Collapse
Affiliation(s)
- Zhenyu Wang
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Pei Zhang
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Chen Huang
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China.,Medical Research Center, Peking University Third Hospital, Beijing, China
| | - Yining Guo
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Xuhe Dong
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Xuemin Li
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China. .,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
10
|
OUP accepted manuscript. J Antimicrob Chemother 2022; 77:1635-1644. [DOI: 10.1093/jac/dkac094] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/28/2022] [Indexed: 11/14/2022] Open
|
11
|
Jadi PK, Sharma P, Bhogapurapu B, Roy S. Alternative Therapeutic Interventions: Antimicrobial Peptides and Small Molecules to Treat Microbial Keratitis. Front Chem 2021; 9:694998. [PMID: 34458234 PMCID: PMC8386189 DOI: 10.3389/fchem.2021.694998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/02/2021] [Indexed: 01/10/2023] Open
Abstract
Microbial keratitis is a leading cause of blindness worldwide and results in unilateral vision loss in an estimated 2 million people per year. Bacteria and fungus are two main etiological agents that cause corneal ulcers. Although antibiotics and antifungals are commonly used to treat corneal infections, a clear trend with increasing resistance to these antimicrobials is emerging at rapid pace. Extensive research has been carried out to determine alternative therapeutic interventions, and antimicrobial peptides (AMPs) are increasingly recognized for their clinical potential in treating infections. Small molecules targeted against virulence factors of the pathogens and natural compounds are also explored to meet the challenges and growing demand for therapeutic agents. Here we review the potential of AMPs, small molecules, and natural compounds as alternative therapeutic interventions for the treatment of corneal infections to combat antimicrobial resistance. Additionally, we have also discussed about the different formats of drug delivery systems for optimal administration of drugs to treat microbial keratitis.
Collapse
Affiliation(s)
- Praveen Kumar Jadi
- Prof, Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, India
| | - Prerana Sharma
- Prof, Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, India
- Department of Animal Sciences, University of Hyderabad, Hyderabad, India
| | - Bharathi Bhogapurapu
- Prof, Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, India
| | - Sanhita Roy
- Prof, Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, India
| |
Collapse
|
12
|
Albash R, M Abdellatif M, Hassan M, M Badawi N. Tailoring Terpesomes and Leciplex for the Effective Ocular Conveyance of Moxifloxacin Hydrochloride (Comparative Assessment): In-vitro, Ex-vivo, and In-vivo Evaluation. Int J Nanomedicine 2021; 16:5247-5263. [PMID: 34376978 PMCID: PMC8349216 DOI: 10.2147/ijn.s316326] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 07/10/2021] [Indexed: 12/17/2022] Open
Abstract
Aim To compare the ability of both terpesomes (TPs) and leciplex (LPs) loaded moxifloxacin hydrochloride (MOX) for enhancing ocular drug conveyance. Methods Two separate 21.31 full-factorial trials were established to determine the influence of multiple variables upon nanovesicles properties and select the optimized formulae using Design Expert® software. The thin-film hydration method was used to formulate TPs, while the single-step procedure was used for LPs. All formulae were characterized for their entrapment efficiency percent (EE%), particle size distribution (PS), polydispersity index (PDI), and zeta potential (ZP). Then, the optimized formulae were selected, evaluated, and compared for additional assessments. Results The optimized formulae TP4 and LP1 showed EE% of 84.14±0.21 and 78.47±0.17%, PS of 578.65±5.65 and 102.41±3.39 nm, PDI of 0.56±0.04 and 0.28±0.01, ZP of -12.50±0.30 and 32.50±0.50 mV, respectively. Further, LP1 showed enhanced corneal permeation across cow cornea compared to MOX solution and TP4. Besides, confocal laser scanning microscopy assessment viewed valuable infiltration from the fluoro-labeled LP through corneal layers compared to TP. LP1 showed spherical morphology and, its ability to adhere to mucus membranes was justified. Further, LP1 showed superiority over MOX solution in biofilm inhibition and eradication in addition to the treatment of infected mice with methicillin-resistant Staphylococcus aureus without any inflammatory response. Finally, the histopathological study verified the harmlessness and biocompatibility of the assembled LPs. Conclusion The gained outcomes confirmed the capability of utilizing LPs as a successful nanovesicle for the ocular conveyance of MOX over TPs and MOX solution.
Collapse
Affiliation(s)
- Rofida Albash
- Department of Pharmaceutics, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, Giza, Egypt
| | - Menna M Abdellatif
- Department of Industrial Pharmacy, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, Giza, Egypt
| | - Mariam Hassan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Noha M Badawi
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| |
Collapse
|
13
|
Optimization of 2-Acylaminocycloalkylthiophene Derivatives for Activity against Staphylococcus aureus RnpA. Antibiotics (Basel) 2021; 10:antibiotics10040369. [PMID: 33807357 PMCID: PMC8066339 DOI: 10.3390/antibiotics10040369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/24/2021] [Accepted: 03/27/2021] [Indexed: 12/15/2022] Open
Abstract
Staphylococcus aureus is well-recognized to cause debilitating bacterial infections that are difficult to treat due to the emergence of antibiotic resistance. As such, there is a need to develop new antimicrobials for the therapeutic intervention of S. aureus disease. To that end, S. aureus RnpA is an essential enzyme that is hypothesized to participate in two required cellular processes, precursor tRNA (ptRNA) maturation and mRNA degradation. Corresponding high throughput screening campaigns have identified the phenylcarbamoyl cyclic thiopenes as a chemical class of RnpA inhibitors that display promising antibacterial effects by reducing RnpA ptRNA and mRNA degradation activities and low human cell toxicity. Herein, we perform a structure activity relationship study of the chemical scaffold. Results revealed that the cycloalkane ring size and trifluoroacetamide moiety are required for antibacterial activity, whereas modifications of the para and/or meta positions of the pharmacophore’s phenyl group allowed tuning of the scaffold’s antimicrobial performance and RnpA inhibitory activity. The top performing compounds with respect to antimicrobial activity also did not exhibit cytotoxicity to human cell lines at concentrations up to 100 µM, greater than 100-fold the minimum inhibitory concentration (MIC). Focused studies of one analog, RNP0012, which exhibited the most potent antimicrobial and inhibition of cellular RnpA activities revealed that the compound reduced bacterial burden in a murine model of S. aureus disease. Taken together, the results presented are expected to provide an early framework for optimization of next-generation of RnpA inhibitor analogues that may represent progenitors of a new class of antimicrobials.
Collapse
|
14
|
Youssef AAA, Cai C, Dudhipala N, Majumdar S. Design of Topical Ocular Ciprofloxacin Nanoemulsion for the Management of Bacterial Keratitis. Pharmaceuticals (Basel) 2021; 14:210. [PMID: 33802394 PMCID: PMC7998883 DOI: 10.3390/ph14030210] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/23/2021] [Accepted: 02/26/2021] [Indexed: 12/13/2022] Open
Abstract
Bacterial keratitis (BK) is a critical ocular infection that can lead to serious visual disability. Ciprofloxacin (CIP), moxifloxacin (MOX), and levofloxacin (LFX) have been accepted as monotherapies by the US Food and Drug Administration for BK treatment. CIP is available commercially at 0.3% w/v concentration as an ophthalmic solution and as an ointment for ocular delivery. Because of solubility issues at physiological pH, CIP precipitation can occur at the corneal surface post instillation of the solution dosage form. Consequently, the ocular bioavailability of CIP is reduced. The ointment dosage form is associated with side effects such as blurred vision, itching, redness, eye discomfort, and eye dryness. This study aimed to design a CIP loaded nanoemulsion (NE; CIP-NE) to facilitate drug penetration into the corneal layers for improved therapeutic outcomes as well as to overcome the drawbacks of the current commercial ophthalmic formulations. CIP-NE formulations were prepared by hot homogenization and ultrasonication, using oleic acid (CIP-O-NE) and Labrafac® Lipophile WL 1349 (CIP-L-NE) as the oily phase, and Tween® 80 and Poloxamer 188 as surfactants. Optimized CIP-NE was further evaluated with respect to in vitro release, ex vivo transcorneal permeation, and moist heat sterilization process, using commercial CIP ophthalmic solution as a control. Optimized CIP-O-NE formulation showed a globule size, polydispersity index, and zeta potential of 121.6 ± 1.5 nm, 0.13 ± 0.01, and -35.1 ± 2.1 mV, respectively, with 100.1 ± 2.0% drug content and was spherical in shape. In vitro release and ex vivo transcorneal permeation studies exhibited sustained release and a 2.1-fold permeation enhancement, respectively, compared with commercial CIP ophthalmic solution. Autoclaved CIP-O-NE formulation was found to be stable for one month (last time-point tested) at refrigerated and room temperature. Therefore, CIP-NE formulation could serve as an effective delivery system for CIP and could improve treatment outcomes in BK.
Collapse
Affiliation(s)
- Ahmed Adel Ali Youssef
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| | - Chuntian Cai
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| | - Narendar Dudhipala
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| | - Soumyajit Majumdar
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
- Research Institute of Pharmaceutical Sciences, University of Mississippi, Oxford, MS 38677, USA
| |
Collapse
|
15
|
Lee JW, Somerville T, Kaye SB, Romano V. Staphylococcus aureus Keratitis: Incidence, Pathophysiology, Risk Factors and Novel Strategies for Treatment. J Clin Med 2021; 10:jcm10040758. [PMID: 33668633 PMCID: PMC7918096 DOI: 10.3390/jcm10040758] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/09/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023] Open
Abstract
Bacterial keratitis is a devastating condition that can rapidly progress to serious complications if not treated promptly. Certain causative microorganisms such as Staphylococcus aureus and Pseudomonas aeruginosa are notorious for their resistance to antibiotics. Resistant bacterial keratitis results in poorer outcomes such as scarring and the need for surgical intervention. Thorough understanding of the causative pathogen and its virulence factors is vital for the discovery of novel treatments to avoid further antibiotic resistance. While much has been previously reported on P. aeruginosa, S. aureus has been less extensively studied. This review aims to give a brief overview of S. aureus epidemiology, pathophysiology and clinical characteristics as well as summarise the current evidence for potential novel therapies.
Collapse
Affiliation(s)
- Jason W. Lee
- School of Medicine, University of Liverpool, Liverpool L69 3GE, UK;
| | - Tobi Somerville
- Department of Eye and Vision Science, University of Liverpool, Liverpool L7 8TX, UK; (T.S.); (S.B.K.)
- St Paul’s Eye Unit, Royal Liverpool University Hospital, Liverpool L7 8XP, UK
| | - Stephen B. Kaye
- Department of Eye and Vision Science, University of Liverpool, Liverpool L7 8TX, UK; (T.S.); (S.B.K.)
- St Paul’s Eye Unit, Royal Liverpool University Hospital, Liverpool L7 8XP, UK
| | - Vito Romano
- Department of Eye and Vision Science, University of Liverpool, Liverpool L7 8TX, UK; (T.S.); (S.B.K.)
- St Paul’s Eye Unit, Royal Liverpool University Hospital, Liverpool L7 8XP, UK
- Correspondence:
| |
Collapse
|
16
|
Multifunctional Monoclonal Antibody Targeting Pseudomonas aeruginosa Keratitis in Mice. Vaccines (Basel) 2020; 8:vaccines8040638. [PMID: 33147726 PMCID: PMC7712430 DOI: 10.3390/vaccines8040638] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/24/2020] [Accepted: 10/29/2020] [Indexed: 01/13/2023] Open
Abstract
A worrisome trend in the study and treatment of infectious disease noted in recent years is the increase in multidrug resistant strains of bacteria concurrent with a scarcity of new antimicrobial agents to counteract this rise. This is particularly true amongst bacteria within the Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species (ESKAPE) designation. P. aeruginosa is one of the most common causes of bacterial keratitis. Therefore, it is of vital importance to characterize new antimicrobial agents with anti-Pseudomonal activity for use with the ocular surface. MEDI3902 is a multifunctional antibody that targets the P. aeruginosa persistence factor Psl exopolysaccharide, and the type 3 secretion protein PcrV. We initially assessed this antibody for ocular surface toxicity. The antimicrobial activity of the antibody was then tested by treating mice with established P. aeruginosa keratitis with both topical and intravenous treatment modalities. MEDI3902, was shown to be non-toxic to the ocular surface of mice when given topically. It was also effective compared to the control antibody at preventing P. aeruginosa keratitis with a one-time treatment at the time of infection. Both topical and intravenous administration of MEDI3902 has been proved significant in treating established keratitis infections as well, speeding the resolution of infection significantly more than that of the control IgG. We report the first use of a topical immunotherapeutic multifunctional agent targeting Psl and type 3 secretion on the ocular surface as an antimicrobial agent. While MEDI3902 has been shown to prevent Pseudomonas biofilm formation in keratitis models when given prophylactically intravitally, we show that MEDI3902 has the capability to also treat an active infection when given intravenously to mice with Pseudomonas keratitis. Our data indicate antibodies are well tolerated and nontoxic on the ocular surface. They reduce infection in mice treated concurrently at inoculation and reduced the signs of cornea pathology in mice with established infection. Taken together, these data indicate treatment with monoclonal antibodies directed against Psl and PcrV may be clinically effective in the treatment of P. aeruginosa keratitis and suggest that the design of further antibodies to be an additional tool in the treatment of bacterial keratitis.
Collapse
|
17
|
Garimella N, Zere T, Hartman N, Gandhi A, Bekele A, Li X, Stone H, Sacks L, Weaver JL. Effect of drug combinations on the kinetics of antibiotic resistance emergence in Escherichia coli CFT073 using an in vitro hollow-fibre infection model. Int J Antimicrob Agents 2019; 55:105861. [PMID: 31838036 DOI: 10.1016/j.ijantimicag.2019.105861] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/04/2019] [Accepted: 12/08/2019] [Indexed: 11/30/2022]
Abstract
Antibiotic resistance is one of the major threats to public health today. To address this problem requires an urgent comprehensive approach. Strategic and multitargeted combination therapy has been increasingly used clinically to treat bacterial infections. The hollow-fibre infection model (HFIM) is a well-controlled in vitro bioreactor system that is increasingly being used in the assessment of resistance emergence with monotherapies and combination antibiotic therapies. In this study, the HFIM was evaluated as a reliable in vitro method to quantitatively and reproducibly analyse the emergence of antibiotic resistance using ampicillin, fosfomycin and ciprofloxacin and their simultaneous combinations against Escherichia coli CFT073, a clinical uropathogenic strain. Bacteria were exposed to clinically relevant pharmacokinetic (PK) concentrations of the drugs for 10 days. Drug and bacterial samples were collected at different time points for PK analysis and to enumerate total and resistant bacterial populations, respectively. The results demonstrated that double or triple combinations significantly delayed the emergence of resistant E. coli CFT073 subpopulations. These findings suggest that strategic combinations of antimicrobials may play a role in controlling the emergence of resistance during treatment. Further animal and human trials will be needed to confirm this and to ensure that there is no adverse impact on the host microbiome or unexpected toxicity. The HFIM system could potentially be used to identify clinically relevant combination dosing regimens for use in a clinical trial evaluating the appearance of resistance to antibacterial drugs.
Collapse
Affiliation(s)
- Narayana Garimella
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, USA
| | - Tesfalem Zere
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, USA
| | - Neil Hartman
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, USA
| | - Adarsh Gandhi
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, USA
| | - Aschalew Bekele
- Division of Microbiology Assessment, Office of Product Quality, Center for Drug Evaluation and Research, US Food and Drug Administration, USA
| | - Xianbin Li
- Division of Biometrics IV, Office of Biostatistics, Center for Drug Evaluation and Research, US Food and Drug Administration, USA
| | - Heather Stone
- Office of Medical Policy, Center for Drug Evaluation and Research, US Food and Drug Administration, USA
| | - Leonard Sacks
- Office of Medical Policy, Center for Drug Evaluation and Research, US Food and Drug Administration, USA
| | - James L Weaver
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, USA.
| |
Collapse
|
18
|
Galdino ACM, de Oliveira MP, Ramalho TC, de Castro AA, Branquinha MH, Santos ALS. Anti-Virulence Strategy against the Multidrug-Resistant Bacterial Pathogen Pseudomonas aeruginosa: Pseudolysin (Elastase B) as a Potential Druggable Target. Curr Protein Pept Sci 2019; 20:471-487. [PMID: 30727891 DOI: 10.2174/1389203720666190207100415] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 01/26/2019] [Accepted: 01/31/2019] [Indexed: 11/22/2022]
Abstract
Pseudomonas aeruginosa is a non-fermentative, gram-negative bacterium that is one of the most common pathogens responsible for hospital-acquired infections worldwide. The management of the infections caused by P. aeruginosa represents a huge challenge in the healthcare settings due to the increased emergence of resistant isolates, some of them resistant to all the currently available antimicrobials, which results in elevated morbimortality rates. Consequently, the development of new therapeutic strategies against multidrug-resistant P. aeruginosa is urgent and needful. P. aeruginosa is wellrecognized for its extreme genetic versatility and its ability to produce a lush variety of virulence factors. In this context, pseudolysin (or elastase B) outstands as a pivotal virulence attribute during the infectious process, playing multifunctional roles in different aspects of the pathogen-host interaction. This protein is a 33-kDa neutral zinc-dependent metallopeptidase that is the most abundant peptidase found in pseudomonal secretions, which contributes to the invasiveness of P. aeruginosa due to its ability to cleave several extracellular matrix proteins and to disrupt the basolateral intercellular junctions present in the host tissues. Moreover, pseudolysin makes P. aeruginosa able to overcome host defenses by the hydrolysis of many immunologically relevant molecules, including antibodies and complement components. The attenuation of this striking peptidase therefore emerges as an alternative and promising antivirulence strategy to combat antibiotic-refractory infections caused by P. aeruginosa. The anti-virulence approach aims to disarm the P. aeruginosa infective arsenal by inhibiting the expression/activity of bacterial virulence factors in order to reduce the invasiveness of P. aeruginosa, avoiding the emergence of resistance since the proliferation is not affected. This review summarizes the most relevant features of pseudolysin and highlights this enzyme as a promising target for the development of new anti-virulence compounds.
Collapse
Affiliation(s)
- Anna Clara M Galdino
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Matheus P de Oliveira
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, United States
| | - Teodorico C Ramalho
- Departamento de Quimica, Universidade Federal de Lavras, Minas Gerais, Brazil
| | | | - Marta H Branquinha
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - André L S Santos
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
19
|
Mohammed I, Said DG, Nubile M, Mastropasqua L, Dua HS. Cathelicidin-Derived Synthetic Peptide Improves Therapeutic Potential of Vancomycin Against Pseudomonas aeruginosa. Front Microbiol 2019; 10:2190. [PMID: 31608030 PMCID: PMC6761703 DOI: 10.3389/fmicb.2019.02190] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 09/06/2019] [Indexed: 12/11/2022] Open
Abstract
Pseudomonas aeruginosa (PA) is the leading cause of corneal blindness worldwide. A constant increase in multi-drug resistant PA strains have heightened the challenge of effectively managing corneal infections with conventional antibiotics. Antimicrobial peptides are promising antibiotic analogs with a unique mode of action. Cathelicidin-derived shorter peptides (FK13 and FK16) have previously been shown to kill a range of pathogens in both in vitro and in vivo systems. Here, our aim was to exploit the potential of FK13 or FK16 to enhance the anti-Pseudomonas activity of vancomycin, which normally has low clinical efficacy against PA. Our results have demonstrated that FK16 is more potent than FK13 against different PA strains including a clinical isolate from a patient's ocular surface. FK16 was shown to enhance the membrane permeability of PAO1 at sub-inhibitory concentrations. Moreover, FK16 at lower concentrations was shown to increase the antibacterial susceptibility of vancomycin against PA strains up to eightfold. The bactericidal synergism between FK16 and vancomycin was shown to be stable in the presence of physiological tear salt concentration and did not cause toxic effects on the human corneal epithelial cells and human red blood cells. Our results have revealed that sub-inhibitory concentration of FK16 could augment the antimicrobial effects of vancomycin against PA. It is anticipated that the future exploitation of the peptide design approach may enhance the effectiveness of FK16 and its application as an adjuvant to antibiotic therapy for the treatment of multi-drug resistant infections.
Collapse
Affiliation(s)
- Imran Mohammed
- Academic Ophthalmology, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Dalia G Said
- Academic Ophthalmology, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Mario Nubile
- Ophthalmology Clinic, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | | | - Harminder S Dua
- Academic Ophthalmology, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
20
|
A Novel, Broad-Spectrum Antimicrobial Combination for the Treatment of Pseudomonas aeruginosa Corneal Infections. Antimicrob Agents Chemother 2019; 63:AAC.00777-19. [PMID: 31332071 DOI: 10.1128/aac.00777-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 07/16/2019] [Indexed: 11/20/2022] Open
Abstract
Bacterial keratitis causes significant blindness, yet antimicrobial resistance has rendered current treatments ineffective. Polymyxin B-trimethoprim (PT) plus rifampin has potent in vitro activity against Staphylococcus aureus and Pseudomonas aeruginosa, two important causes of keratitis. Here we further characterize this combination against P. aeruginosa in a murine keratitis model. PT plus rifampin performed comparably to or better than moxifloxacin, the gold standard, suggesting that the combination may be a promising therapy for bacterial keratitis.
Collapse
|