1
|
Arabestani MR, Saadat M, Taherkhani A. Antibiotic resistance challenge: evaluating anthraquinones as rifampicin monooxygenase inhibitors through integrated bioinformatics analysis. Genomics Inform 2024; 22:13. [PMID: 39232833 PMCID: PMC11375879 DOI: 10.1186/s44342-024-00015-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 07/23/2024] [Indexed: 09/06/2024] Open
Abstract
OBJECTIVE Antibiotic resistance poses a pressing and crucial global public health challenge, leading to significant clinical and health-related consequences. Substantial evidence highlights the pivotal involvement of rifampicin monooxygenase (RIFMO) in the context of antibiotic resistance. Hence, inhibiting RIFMO could offer potential in the treatment of various infections. Anthraquinones, a group of organic compounds, have shown promise in addressing tuberculosis. This study employed integrated bioinformatics approaches to evaluate the potential inhibitory effects of a selection of anthraquinones on RIFMO. The findings were subsequently compared with those of rifampicin (RIF), serving as a positive control inhibitor. METHODS The AutoDock 4.0 tool assessed the binding free energy between 21 anthraquinones and the RIFMO catalytic cleft. The ligands were ranked based on the most favorable scores derived from ΔGbinding. The docking analyses for the highest-ranked anthraquinone and RIF underwent a cross-validation process. This validation procedure utilized the SwissDock server and the Schrödinger Maestro docking software. Molecular dynamics simulations were conducted to scrutinize the stability of the backbone atoms in free RIFMO, RIFMO-RIF, and RIFMO complexed with the top-ranked anthraquinone throughout a 100-ns computer simulation. The Discovery Studio Visualizer tool visualized interactions between RIFMO residues and ligands. An evaluation of the pharmacokinetics and toxicity profiles of the tested compounds was also conducted. RESULTS Five anthraquinones were indicated with ΔGbinding scores less than - 10 kcal/mol. Hypericin emerged as the most potent RIFMO inhibitor, boasting a ΔGbinding score and inhibition constant value of - 12.11 kcal/mol and 798.99 pM, respectively. The agreement across AutoDock 4.0, SwissDock, and Schrödinger Maestro results highlighted hypericin's notable binding affinity to the RIFMO catalytic cleft. The RIFMO-hypericin complex achieved stability after a 70-ns computer simulation, exhibiting a root-mean-square deviation of 0.55 nm. Oral bioavailability analysis revealed that all anthraquinones except hypericin, sennidin A, and sennidin B may be suitable for oral administration. Furthermore, the carcinogenicity prediction analysis indicated a favorable safety profile for all examined anthraquinones. CONCLUSION Inhibiting RIFMO, particularly with anthraquinones such as hypericin, holds promise as a potential therapeutic strategy for infectious diseases.
Collapse
Affiliation(s)
- Mohammad Reza Arabestani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Masoumeh Saadat
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Amir Taherkhani
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
2
|
Li H, Zhu X, Zhang X, Dong C. Caspofungin enhances the potency of rifampin against Gram-negative bacteria. Front Microbiol 2024; 15:1447485. [PMID: 39211315 PMCID: PMC11358092 DOI: 10.3389/fmicb.2024.1447485] [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: 06/11/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Developing antibiotic adjuvants is an effective strategy to combat antimicrobial resistance (AMR). The envelope of Gram-negative bacteria (GNB) is a barrier to prevent the entry of antibiotics, making it an attractive target for novel antibiotic and adjuvant development. Methods and Results In this study, we identified Caspofungin acetate (CAS) as an antibiotic adjuvant against GNB in the repurposing screen of 3,158 FDA-approved drugs. Checkerboard assays suggested that CAS could enhance the antimicrobial activity of rifampin or colistin against various GNB strains in vitro, Moreover, Galleria mellonella larvae infection model also indicated that CAS significantly potentiated the efficacy of rifampin against multidrug-resistant Escherichia coli 72 strain in vivo. Most importantly, resistance development assay showed that CAS was less susceptible to accelerating the resistance development of drug-sensitive strain E. coli MG1655. Functional studies and RNA-seq analysis confirmed that the mechanisms by which CAS enhanced the antimicrobial activities of antibiotics were involved in permeabilizing the bacterial cell envelope, disrupting proton motive force and inhibiting bacterial biofilm formation. Additionally, it has been found that PgaC is the CAS target and enzymatic assay has confirmed the inhibition activity. Discussion Our results illustrate the feasibility of CAS as an antibiotic adjuvant against GNB, which is an alternative strategy of anti-infection.
Collapse
Affiliation(s)
- Haotian Li
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Xiaojing Zhu
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Xing Zhang
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Changjiang Dong
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
3
|
Reinecke P, Morovic P, Niemann M, Renz N, Perka C, Trampuz A, Meller S. Adverse Events Associated with Prolonged Antibiotic Therapy for Periprosthetic Joint Infections-A Prospective Study with a Special Focus on Rifampin. Antibiotics (Basel) 2023; 12:1560. [PMID: 37998762 PMCID: PMC10668752 DOI: 10.3390/antibiotics12111560] [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: 10/03/2023] [Revised: 10/17/2023] [Accepted: 10/21/2023] [Indexed: 11/25/2023] Open
Abstract
Periprosthetic Joint Infection (PJI) is a significant contributor to patient morbidity and mortality, and it can be addressed through a range of surgical interventions coupled with antibiotic therapies. Following surgical intervention(s), prolonged administration of oral antibiotics is recommended to cure PJI. There is a lack of reports on the adverse events (AEs) associated with oral antibiotics, particularly rifampin. This investigation sought to elucidate the occurrence of antibiotic-related AEs after an initial regimen of intravenous antibiotic administration, supplemented by an extended course of oral antibiotics. A prospective study of patients diagnosed with PJI of the hip, knee, or shoulder who underwent single-stage exchange arthroplasty (SSE) (10%), two-stage exchange arthroplasty (TSE) (81%), or debridement, antibiotics, and implant retention (DAIR) (6%) was performed. The primary outcome of interest was the detection of AEs, the secondary outcome the detection of a correlation between rifampin use and the incidence of AEs, and the tertiary outcome was whether oral antibiotic treatment needed to be adjusted or discontinued due to AEs. In addition, subjective tolerability was monitored throughout the study. A total of 336 events were identified for 73 out of 80 patients. The most frequently used antibiotics were rifampin and co-trimoxazole. Most AEs occurred in the gastrointestinal tract (46%). The most frequent AEs were nausea, inappetence, diarrhea, and skin rash. In 6% of cases, the AEs led to antibiotic discontinuation, and in 29% of cases, a dose adjustment of the oral therapy occurred, mainly with amoxicillin or co-trimoxazole. The majority of patients (55%) rated the subjective tolerability as good. In conclusion, AEs during antibiotic treatment for PJI are common. They mainly affect the gastrointestinal tract. Rifampin use might be a reason for the higher incidence of AEs compared to non-rifampin antibiotic treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sebastian Meller
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Center for Musculoskeletal Surgery (CMSC), Augustenburger Platz 1, 13353 Berlin, Germany; (P.R.); (P.M.); (M.N.); (N.R.); (C.P.); (A.T.)
| |
Collapse
|
4
|
Elitas M, Kalayci Demir G, Vural Kaymaz S. Mathematical Model for Growth and Rifampicin-Dependent Killing Kinetics of Escherichia coli Cells. ACS OMEGA 2023; 8:38452-38458. [PMID: 37867679 PMCID: PMC10586251 DOI: 10.1021/acsomega.3c05233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/21/2023] [Indexed: 10/24/2023]
Abstract
Antibiotic resistance is a global health threat. We urgently need better strategies to improve antibiotic use to combat antibiotic resistance. Currently, there are a limited number of antibiotics in the treatment repertoire of existing bacterial infections. Among them, rifampicin is a broad-spectrum antibiotic against various bacterial pathogens. However, during rifampicin exposure, the appearance of persisters or resisters decreases its efficacy. Hence, to benefit more from rifampicin, its current standard dosage might be reconsidered and explored using both computational tools and experimental or clinical studies. In this study, we present the mathematical relationship between the concentration of rifampicin and the growth and killing kinetics of Escherichia coli cells. We generated time-killing curves of E. coli cells in the presence of 4, 16, and 32 μg/mL rifampicin exposures. We specifically focused on the oscillations with decreasing amplitude over time in the growth and killing kinetics of rifampicin-exposed E. coli cells. We propose the solution form of a second-order linear differential equation for a damped oscillator to represent the mathematical relationship. We applied a nonlinear curve fitting solver to time-killing curve data to obtain the model parameters. The results show a high fitting accuracy.
Collapse
Affiliation(s)
- Meltem Elitas
- Faculty
of Engineering and Natural Sciences, Sabanci
University, Istanbul 34956, Turkiye
| | - Guleser Kalayci Demir
- Faculty
of Engineering, Department of Electrical and Electronics Engineering, Dokuz Eylul University, Izmir 35397, Turkey
| | - Sumeyra Vural Kaymaz
- Faculty
of Engineering and Natural Sciences, Sabanci
University, Istanbul 34956, Turkiye
| |
Collapse
|
5
|
Minerdi D, Loqui D, Sabbatini P. Monooxygenases and Antibiotic Resistance: A Focus on Carbapenems. BIOLOGY 2023; 12:1316. [PMID: 37887026 PMCID: PMC10604202 DOI: 10.3390/biology12101316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/08/2023] [Accepted: 09/15/2023] [Indexed: 10/28/2023]
Abstract
Carbapenems are a group of broad-spectrum beta-lactam antibiotics that in many cases are the last effective defense against infections caused by multidrug-resistant bacteria, such as some strains of Klebsiella pneumoniae, Escherichia coli, Pseudomonas aeruginosa, and Acinetobacter baumannii. Resistance to carbapenems has emerged and is beginning to spread, becoming an ongoing public-health problem of global dimensions, causing serious outbreaks, and dramatically limiting treatment options. This paper reviews the role of flavin monooxygenases in antibiotic resistance, with a specific focus on carbapenem resistance and the recently discovered mechanism mediated by Baeyer-Villiger monooxygenases. Flavin monooxygenases are enzymes involved in the metabolism and detoxification of compounds, including antibiotics. Understanding their role in antibiotic resistance is crucial. Carbapenems are powerful antibiotics used to treat severe infections caused by multidrug-resistant bacteria. However, the rise of carbapenem-resistant strains poses a significant challenge. This paper explores the mechanisms by which flavin monooxygenases confer resistance to carbapenems, examining molecular pathways and genetic factors. Additionally, this paper highlights the discovery of Baeyer-Villiger monooxygenases' involvement in antibiotic resistance. These enzymes catalyze the insertion of oxygen atoms into specific chemical bonds. Recent studies have revealed their unexpected role in promoting carbapenem resistance. Through a comprehensive analysis of the literature, this paper contributes to the understanding of the interplay between flavin monooxygenases, carbapenem resistance, and Baeyer-Villiger monooxygenases. By exploring these mechanisms, it aims to inform the development of strategies to combat antibiotic resistance, a critical global health concern.
Collapse
Affiliation(s)
- Daniela Minerdi
- Department of Agricultural, Forestry and Food Sciences, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco, TO, Italy;
| | - Davide Loqui
- Emergency Department, Città della Salute e della Scienza of Turin, 10100 Turin, TO, Italy;
| | - Paolo Sabbatini
- Department of Agricultural, Forestry and Food Sciences, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco, TO, Italy;
| |
Collapse
|
6
|
Hasbun R. Review of Progress and Challenges in Bacterial Meningitis-Reply. JAMA 2023; 329:1407. [PMID: 37097358 DOI: 10.1001/jama.2023.2551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Affiliation(s)
- Rodrigo Hasbun
- Section of Infectious Diseases, UT Health McGovern Medical School, Houston, Texas
| |
Collapse
|
7
|
Drug-in-cyclodextrin-in-polymeric nanoparticles: A promising strategy for rifampicin administration. Eur J Pharm Biopharm 2022; 180:190-200. [DOI: 10.1016/j.ejpb.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/19/2022] [Accepted: 10/01/2022] [Indexed: 11/24/2022]
|
8
|
Sekino N, Selim M, Shehadah A. Sepsis-associated brain injury: underlying mechanisms and potential therapeutic strategies for acute and long-term cognitive impairments. J Neuroinflammation 2022; 19:101. [PMID: 35488237 PMCID: PMC9051822 DOI: 10.1186/s12974-022-02464-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 04/14/2022] [Indexed: 12/29/2022] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. Sepsis causes cerebral dysfunction in the short and long term and induces disruption of the blood–brain barrier (BBB), neuroinflammation, hypoperfusion, and accumulation of amyloid β (Aβ) and tau protein in the brain. White matter changes and brain atrophy can be detected using brain imaging, but unfortunately, there is no specific treatment that directly addresses the underlying mechanisms of cognitive impairments in sepsis. Here, we review the underlying mechanisms of sepsis-associated brain injury, with a focus on BBB dysfunction and Aβ and tau protein accumulation in the brain. We also describe the neurological manifestations and imaging findings of sepsis-associated brain injury, and finally, we propose potential therapeutic strategies for acute and long-term cognitive impairments associated with sepsis. In the acute phase of sepsis, we suggest using antibiotics (such as rifampicin), targeting proinflammatory cytokines, and preventing ischemic injuries and hypoperfusion. In the late phase of sepsis, we suggest targeting neuroinflammation, BBB dysfunction, Aβ and tau protein phosphorylation, glycogen synthase kinase-3 beta (GSK3β), and the receptor for advanced glycation end products (RAGE). These proposed strategies are meant to bring new mechanism-based directions for future basic and clinical research aimed at preventing or ameliorating acute and long-term cognitive impairments in patients with sepsis.
Collapse
Affiliation(s)
- Nobufumi Sekino
- Department of Medicine, Translational Therapeutics Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Magdy Selim
- Department of Neurology, Stroke and Cerebrovascular Diseases Division, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS-641, Boston, MA, 02215, USA
| | - Amjad Shehadah
- Department of Neurology, Stroke and Cerebrovascular Diseases Division, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CLS-641, Boston, MA, 02215, USA.
| |
Collapse
|
9
|
Rizwan M, Selvanathan V, Rasool A, Qureshi MAUR, Iqbal DN, Kanwal Q, Shafqat SS, Rasheed T, Bilal M. Metal-Organic Framework-Based Composites for the Detection and Monitoring of Pharmaceutical Compounds in Biological and Environmental Matrices. WATER, AIR, AND SOIL POLLUTION 2022; 233:493. [PMID: 36466935 PMCID: PMC9685123 DOI: 10.1007/s11270-022-05904-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 10/13/2022] [Indexed: 05/10/2023]
Abstract
The production of synthetic drugs is considered a huge milestone in the healthcare sector, transforming the overall health, aging, and lifestyle of the general population. Due to the surge in production and consumption, pharmaceutical drugs have emerged as potential environmental pollutants that are toxic with low biodegradability. Traditional chromatographic techniques in practice are time-consuming and expensive, despite good precision. Alternatively, electroanalytical techniques are recently identified to be selective, rapid, sensitive, and easier for drug detection. Metal-organic frameworks (MOFs) are known for their intrinsic porous nature, high surface area, and diversity in structural design that provides credible drug-sensing capacities. Long-term reusability and maintaining chemo-structural integrity are major challenges that are countered by ligand-metal combinations, optimization of synthetic conditions, functionalization, and direct MOFs growth over the electrode surface. Moreover, chemical instability and lower conductivities limited the mass commercialization of MOF-based materials in the fields of biosensing, imaging, drug release, therapeutics, and clinical diagnostics. This review is dedicated to analyzing the various combinations of MOFs used for electrochemical detection of pharmaceutical drugs, comprising antibiotics, analgesics, anticancer, antituberculosis, and veterinary drugs. Furthermore, the relationship between the composition, morphology and structural properties of MOFs with their detection capabilities for each drug species is elucidated.
Collapse
Affiliation(s)
- Muhammad Rizwan
- Department of Chemistry, University of Lahore, Lahore, 54000 Punjab Pakistan
| | - Vidhya Selvanathan
- Solar Energy Research Institute (SERI), Universiti Kebangsaan Malaysia (UKM), 43600 Bangi, Selangor Darul Ehsan Malaysia
| | - Atta Rasool
- School of Chemistry, University of the Punjab, Lahore, Pakistan
| | | | - Dure Najaf Iqbal
- Department of Chemistry, University of Lahore, Lahore, 54000 Punjab Pakistan
| | - Qudsia Kanwal
- Department of Chemistry, University of Lahore, Lahore, 54000 Punjab Pakistan
| | - Syed Salman Shafqat
- Department of Chemistry, Division of Science and Technology, University of Education, Lahore, 54000 Pakistan
| | - Tahir Rasheed
- Interdisciplinary Research Centre for Advanced Materials, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, 31261 Saudi Arabia
| | - Muhammad Bilal
- Institute of Chemical Technology and Engineering, Faculty of Chemical Technology, Poznan University of Technology, Berdychowo 4, 60695 Poznan, PL Poland
| |
Collapse
|
10
|
Pietsch M, Hochegger M, Djahani O, Hofmann S, Mlaker G, Eder-Halbedl M, Hofstätter T. A two-stage approach to primary TKA using articulating antibiotic-loaded spacers improve function and eradicate infection in septic arthritic knees. Knee Surg Sports Traumatol Arthrosc 2021; 29:3186-3194. [PMID: 32556435 DOI: 10.1007/s00167-020-06106-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 06/11/2020] [Indexed: 12/01/2022]
Abstract
PURPOSE The treatment of an infected arthritic knee might be challenging. The failure rate has been reported to be high for open or arthroscopic debridement. A subsequently high rate of infection has been noted in these patients undergoing primary total knee arthroplasty (TKA). In the present study, a two-stage approach using an articulating spacer was used. The hypothesis was that the procedure would eradicate the infection and improve pain and function in these patients. METHODS A total of 16 consecutive patients were enrolled in this retrospective study. The mean follow-up time was 6.1 years (range 2.0-9.9 years). Patients with advanced osteoarthritis and infection of the knee were included. All patients had previously undergone one or more failed arthroscopic or open procedures for the eradication of infection. All patients received the same homemade metal-on-plastic articulating antibiotic spacer. Double antibiotic therapy was given for 2 weeks intravenously and orally for 4 weeks. TKA implantation was performed 6 weeks after the first stage. RESULTS The infection was eradicated without recurrence in all patients. The functional results were significantly improved, and pain was significantly reduced after spacer and TKA implantation. The mean amount of knee flexion was 95 ± 30° preoperatively, and it increased to 109 ± 14° (p = 0.012) after spacer implantation and to 119 ± 10° (p = 0.002) after TKA implantation. The mean KSS objective was 58 ± 12 preoperatively, and it increased to 75 ± 14 (p < 0.0001) after spacer implantation and to 96 ± 3 (p < 0.0001) after TKA implantation. The mean KSS function was 17 ± 11 preoperatively, and it increased to 46 ± 10 (p < 0.0001) after spacer implantation and to 86 ± 6 (p < 0.0001) after TKA implantation. The mean VAS score was 65 ± 11 preoperatively, and it decreased to 2 ± 4 (p < 0.0001) after spacer implantation and to 1 ± 2 (p < 0.0001) after TKA implantation. CONCLUSION The two-stage procedure for the treatment of infected arthritic knees after failed eradication surgery was effective in all patients. Using an antibiotic articulating metal-on-plastic cement spacer showed improved functional results between the stages and at the final follow-up. No intra- or postoperative complications occurred.
Collapse
Affiliation(s)
- M Pietsch
- Department of Orthopaedic Surgery, Orthopaedic Hospital Stolzalpe (LKH Murtal), Stolzalpe 38, 8852, Stolzalpe, Austria.
| | - M Hochegger
- Department of Orthopaedic Surgery, Orthopaedic Hospital Stolzalpe (LKH Murtal), Stolzalpe 38, 8852, Stolzalpe, Austria
| | - O Djahani
- Department of Orthopaedic Surgery, Orthopaedic Hospital Stolzalpe (LKH Murtal), Stolzalpe 38, 8852, Stolzalpe, Austria
| | - S Hofmann
- Department of Orthopaedic Surgery, Orthopaedic Hospital Stolzalpe (LKH Murtal), Stolzalpe 38, 8852, Stolzalpe, Austria
| | - G Mlaker
- Department of Orthopaedic Surgery, Orthopaedic Hospital Stolzalpe (LKH Murtal), Stolzalpe 38, 8852, Stolzalpe, Austria
| | - M Eder-Halbedl
- Department of Orthopaedic Surgery, Orthopaedic Hospital Stolzalpe (LKH Murtal), Stolzalpe 38, 8852, Stolzalpe, Austria
| | - Th Hofstätter
- Department of Orthopaedic and Trauma Surgery, Paracelsus Medical University Salzburg, Müllner Hauptstraße 48, 5020, Salzburg, Austria
| |
Collapse
|
11
|
Varghese R, Veeraraghavan B. Decoding the Penicillin Resistance of Streptococcus pneumoniae for Invasive and Noninvasive Infections. Microb Drug Resist 2021; 27:942-950. [DOI: 10.1089/mdr.2020.0233] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Rosemol Varghese
- Department of Clinical Microbiology, Christian Medical College, Vellore, India
| | | |
Collapse
|
12
|
Motiei M, Pleno de Gouveia L, Šopík T, Vícha R, Škoda D, Císař J, Khalili R, Domincová Bergerová E, Münster L, Fei H, Sedlařík V, Sáha P. Nanoparticle-Based Rifampicin Delivery System Development. Molecules 2021; 26:molecules26072067. [PMID: 33916814 PMCID: PMC8038351 DOI: 10.3390/molecules26072067] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/25/2021] [Accepted: 03/29/2021] [Indexed: 12/28/2022] Open
Abstract
The alkaline milieu of chronic wounds severely impairs the therapeutic effect of antibiotics, such as rifampicin; as such, the development of new drugs, or the smart delivery of existing drugs, is required. Herein, two innovative polyelectrolyte nanoparticles (PENs), composed of an amphiphilic chitosan core and a polycationic shell, were synthesized at alkaline pH, and in vitro performances were assessed by 1H NMR, elemental analysis, FT-IR, XRD, DSC, DLS, SEM, TEM, UV/Vis spectrophotometry, and HPLC. According to the results, the nanostructures exhibited different morphologies but similar physicochemical properties and release profiles. It was also hypothesized that the simultaneous use of the nanosystem and an antioxidant could be therapeutically beneficial. Therefore, the simultaneous effects of ascorbic acid and PENs were evaluated on the release profile and degradation of rifampicin, in which the results confirmed their synergistic protective effect at pH 8.5, as opposed to pH 7.4. Overall, this study highlighted the benefits of nanoparticulate development in the presence of antioxidants, at alkaline pH, as an efficient approach for decreasing rifampicin degradation.
Collapse
Affiliation(s)
- Marjan Motiei
- Centre of Polymer Systems, University Institute, TBU, tr. Tomase Bati 5678, 76001 Zlin, Czech Republic; (T.Š.); (D.Š.); (J.C.); (E.D.B.); (L.M.); (H.F.); (V.S.); (P.S.)
- Correspondence:
| | - Luis Pleno de Gouveia
- iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, 169-003 Lisbon, Portugal;
| | - Tomáš Šopík
- Centre of Polymer Systems, University Institute, TBU, tr. Tomase Bati 5678, 76001 Zlin, Czech Republic; (T.Š.); (D.Š.); (J.C.); (E.D.B.); (L.M.); (H.F.); (V.S.); (P.S.)
| | - Robert Vícha
- Department of Chemistry, Faculty of Technology, TBU, Vavrečkova 275, 76001 Zlín, Czech Republic;
| | - David Škoda
- Centre of Polymer Systems, University Institute, TBU, tr. Tomase Bati 5678, 76001 Zlin, Czech Republic; (T.Š.); (D.Š.); (J.C.); (E.D.B.); (L.M.); (H.F.); (V.S.); (P.S.)
| | - Jaroslav Císař
- Centre of Polymer Systems, University Institute, TBU, tr. Tomase Bati 5678, 76001 Zlin, Czech Republic; (T.Š.); (D.Š.); (J.C.); (E.D.B.); (L.M.); (H.F.); (V.S.); (P.S.)
| | - Reza Khalili
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 455/2, 12808 Prague 2, Czech Republic;
| | - Eva Domincová Bergerová
- Centre of Polymer Systems, University Institute, TBU, tr. Tomase Bati 5678, 76001 Zlin, Czech Republic; (T.Š.); (D.Š.); (J.C.); (E.D.B.); (L.M.); (H.F.); (V.S.); (P.S.)
| | - Lukáš Münster
- Centre of Polymer Systems, University Institute, TBU, tr. Tomase Bati 5678, 76001 Zlin, Czech Republic; (T.Š.); (D.Š.); (J.C.); (E.D.B.); (L.M.); (H.F.); (V.S.); (P.S.)
| | - Haojie Fei
- Centre of Polymer Systems, University Institute, TBU, tr. Tomase Bati 5678, 76001 Zlin, Czech Republic; (T.Š.); (D.Š.); (J.C.); (E.D.B.); (L.M.); (H.F.); (V.S.); (P.S.)
| | - Vladimír Sedlařík
- Centre of Polymer Systems, University Institute, TBU, tr. Tomase Bati 5678, 76001 Zlin, Czech Republic; (T.Š.); (D.Š.); (J.C.); (E.D.B.); (L.M.); (H.F.); (V.S.); (P.S.)
| | - Petr Sáha
- Centre of Polymer Systems, University Institute, TBU, tr. Tomase Bati 5678, 76001 Zlin, Czech Republic; (T.Š.); (D.Š.); (J.C.); (E.D.B.); (L.M.); (H.F.); (V.S.); (P.S.)
| |
Collapse
|
13
|
Liu Y, Tong Z, Shi J, Li R, Upton M, Wang Z. Drug repurposing for next-generation combination therapies against multidrug-resistant bacteria. Theranostics 2021; 11:4910-4928. [PMID: 33754035 PMCID: PMC7978324 DOI: 10.7150/thno.56205] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/29/2021] [Indexed: 12/12/2022] Open
Abstract
Antimicrobial resistance has been a global health challenge that threatens our ability to control and treat life-threatening bacterial infections. Despite ongoing efforts to identify new drugs or alternatives to antibiotics, no new classes of antibiotic or their alternatives have been clinically approved in the last three decades. A combination of antibiotics and non-antibiotic compounds that could inhibit bacterial resistance determinants or enhance antibiotic activity offers a sustainable and effective strategy to confront multidrug-resistant bacteria. In this review, we provide a brief overview of the co-evolution of antibiotic discovery and the development of bacterial resistance. We summarize drug-drug interactions and uncover the art of repurposing non-antibiotic drugs as potential antibiotic adjuvants, including discussing classification and mechanisms of action, as well as reporting novel screening platforms. A pathogen-by-pathogen approach is then proposed to highlight the critical value of drug repurposing and its therapeutic potential. Finally, general advantages, challenges and development trends of drug combination strategy are discussed.
Collapse
Affiliation(s)
- Yuan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Ziwen Tong
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jingru Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Ruichao Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Mathew Upton
- School of Biomedical Sciences, University of Plymouth, Drake Circus, Plymouth, UK
| | - Zhiqiang Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
14
|
Yus C, Irusta S, Sebastian V, Arruebo M. Controlling Particle Size and Release Kinetics in the Sustained Delivery of Oral Antibiotics Using pH-Independent Mucoadhesive Polymers. Mol Pharm 2020; 17:3314-3327. [PMID: 32687366 DOI: 10.1021/acs.molpharmaceut.0c00408] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Copolymers synthesized from acrylic acid and methacrylic acid used as gastroprotective and mucoadhesive enteric coatings have been used to prepare micro- (∼2 μm), submicro- (∼200 nm), and nanoparticles (∼20 nm) containing rifampicin (Rif) to obtain time-controlled drug release kinetics. Different particle sizes and drug release kinetics have been obtained using different synthesis conditions and fabrication techniques including the use of an electrosprayer and an interdigital microfabricated micromixer. The antimicrobial action of the encapsulated Rif has been demonstrated against Staphylococcus aureus ATCC 25923 and compared with the effect of the equivalent dose of the free macrolide antibiotic. At low concentrations, the encapsulated antibiotic showed superior antimicrobial activity than the free drug. The stability of the developed particles has been evaluated in vitro under simulated gastric and intestinal conditions. At the concentrations tested, a reduced cytotoxicity against different human cell lines was observed after analyzing their subcytotoxic doses and the influence on their cell cycle by flow cytometry. Drug release kinetics can be tuned by adjusting particle sizes, and it would be possible to reach the minimum inhibitory concentration or the minimum bactericidal concentration at different time points depending on the medical needs.
Collapse
Affiliation(s)
- Cristina Yus
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Aragón Materials Science Institute, ICMA, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain.,Aragon Health Research Institute (IIS Aragon), 50009 Zaragoza, Spain
| | - Silvia Irusta
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Aragón Materials Science Institute, ICMA, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain.,Aragon Health Research Institute (IIS Aragon), 50009 Zaragoza, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, 28029 Madrid, Spain
| | - Victor Sebastian
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Aragón Materials Science Institute, ICMA, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain.,Aragon Health Research Institute (IIS Aragon), 50009 Zaragoza, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, 28029 Madrid, Spain
| | - Manuel Arruebo
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Aragón Materials Science Institute, ICMA, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain.,Aragon Health Research Institute (IIS Aragon), 50009 Zaragoza, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, 28029 Madrid, Spain
| |
Collapse
|
15
|
Pokhrel R, Chapagain P, Siltberg-Liberles J. Potential RNA-dependent RNA polymerase inhibitors as prospective therapeutics against SARS-CoV-2. J Med Microbiol 2020; 69:864-873. [PMID: 32469301 PMCID: PMC7451031 DOI: 10.1099/jmm.0.001203] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 04/27/2020] [Indexed: 01/13/2023] Open
Abstract
Introduction. The emergence of SARS-CoV-2 has taken humanity off guard. Following an outbreak of SARS-CoV in 2002, and MERS-CoV about 10 years later, SARS-CoV-2 is the third coronavirus in less than 20 years to cross the species barrier and start spreading by human-to-human transmission. It is the most infectious of the three, currently causing the COVID-19 pandemic. No treatment has been approved for COVID-19. We previously proposed targets that can serve as binding sites for antiviral drugs for multiple coronaviruses, and here we set out to find current drugs that can be repurposed as COVID-19 therapeutics.Aim. To identify drugs against COVID-19, we performed an in silico virtual screen with the US Food and Drug Administration (FDA)-approved drugs targeting the RNA-dependent RNA polymerase (RdRP), a critical enzyme for coronavirus replication.Methodology. Initially, no RdRP structure of SARS-CoV-2 was available. We performed basic sequence and structural analysis to determine if RdRP from SARS-CoV was a suitable replacement. We performed molecular dynamics simulations to generate multiple starting conformations that were used for the in silico virtual screen. During this work, a structure of RdRP from SARS-CoV-2 became available and was also included in the in silico virtual screen.Results. The virtual screen identified several drugs predicted to bind in the conserved RNA tunnel of RdRP, where many of the proposed targets were located. Among these candidates, quinupristin is particularly interesting because it is expected to bind across the RNA tunnel, blocking access from both sides and suggesting that it has the potential to arrest viral replication by preventing viral RNA synthesis. Quinupristin is an antibiotic that has been in clinical use for two decades and is known to cause relatively minor side effects.Conclusion. Quinupristin represents a potential anti-SARS-CoV-2 therapeutic. At present, we have no evidence that this drug is effective against SARS-CoV-2 but expect that the biomedical community will expeditiously follow up on our in silico findings.
Collapse
Affiliation(s)
- Rudramani Pokhrel
- Department of Physics, Florida International University, 11200 SW 8th St, Miami, FL 33199, USA
| | - Prem Chapagain
- Department of Physics, Florida International University, 11200 SW 8th St, Miami, FL 33199, USA
- Biomolecular Sciences Institute, Florida International University, 11200 SW 8th St, Miami, FL 33199, USA
| | - Jessica Siltberg-Liberles
- Biomolecular Sciences Institute, Florida International University, 11200 SW 8th St, Miami, FL 33199, USA
- Department of Biological Sciences, Florida International University, 11200 SW 8th St, Miami, FL 33199, USA
| |
Collapse
|
16
|
Ali AE, Mahdy HM, Elsherbiny DM, Azab SS. Rifampicin ameliorates lithium-pilocarpine-induced seizures, consequent hippocampal damage and memory deficit in rats: Impact on oxidative, inflammatory and apoptotic machineries. Biochem Pharmacol 2018; 156:431-443. [PMID: 30195730 DOI: 10.1016/j.bcp.2018.09.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 09/05/2018] [Indexed: 01/28/2023]
Abstract
Epilepsy is one of the serious neurological sequelae of bacterial meningitis. Rifampicin, the well-known broad spectrum antibiotic, is clinically used for chemoprophylaxis of meningitis. Besides its antibiotic effects, rifampicin has been proven to be an effective neuroprotective candidate in various experimental models of neurological diseases. In addition, rifampicin was found to have promising antioxidant, anti-inflammatory and anti-apoptotic effects. Herein, we investigated the anticonvulsant effect of rifampicin at experimental meningitis dose (20 mg/kg, i.p.) using lithium-pilocarpine model of status epilepticus (SE) in rats. Additionally, we studied the effect of rifampicin on seizure induced histopathological, neurochemical and behavioral abnormalities. Our study showed that rifampicin pretreatment attenuated seizure activity and the resulting hippocampal insults marked by hematoxylin and eosin. Markers of oxidative stress, neuroinflammation and apoptosis were evaluated, in the hippocampus, 24 h after SE induction. We found that rifampicin pretreatment suppressed oxidative stress as indicated by normalized malondialdehyde and glutathione levels. Rifampicin pretreatment attenuated SE-induced neuroinflammation and decreased the hippocampal expression of interleukin-1β, tumor necrosis factor-α, nuclear factor kappa-B, and cyclooxygenase-2. Moreover, rifampicin mitigated SE-induced neuronal apoptosis as indicated by fewer positive cytochrome c immunostained cells and lower caspase-3 activity in the hippocampus. Furthermore, Morris water maze testing at 7 days after SE induction showed that rifampicin pretreatment can improve cognitive dysfunction. Therefore, rifampicin, currently used in the management of meningitis, has a potential additional advantage of ameliorating its epileptic sequelae.
Collapse
Affiliation(s)
- Alaa E Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Heba M Mahdy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Doaa M Elsherbiny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Samar S Azab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
17
|
Ho KK, Conley AC, Kennis RA, Hathcock TL, Boothe DM, White AG. Minimum inhibitory concentration and killing properties of rifampicin against canine Staphylococcus pseudintermedius isolates from dogs in the southeast USA. Vet Dermatol 2018; 29:302. [PMID: 29845664 DOI: 10.1111/vde.12653] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2018] [Indexed: 02/28/2024]
Abstract
BACKGROUND Meticillin-resistant (MR) staphylococcal pyoderma in dogs has led to increased use of alternate antibiotics such as rifampicin (RFP). However, little information exists regarding its pharmacodynamics in MR Staphylococcus pseudintermedius. HYPOTHESIS/OBJECTIVES To determine the minimum inhibitory concentration (MIC) and killing properties of RFP for canine Staphylococcus pseudintermedius isolates. METHODS The MIC of RFP was determined using the ETEST® for 50 meticillin-susceptible (MS) and 50 MR S. pseudintermedius isolates collected from dogs. From these isolates, two MS isolates (RFP MIC of 0.003 and 0.008 μg/mL, respectively) and two MR isolates (RFP MIC of 0.003 and 0.012 μg/mL, respectively) were subjected to time-kill studies. Mueller-Hinton broth was supplemented with RFP at 0, 0.5, 1, 2, 4, 8, 16 and 32 times the MIC for 0, 2, 4, 10, 16 and 24 h. The number of viable colony forming units in each sample was determined using a commercial luciferase assay kit. RESULTS The MIC50 and MIC90 were the same for MS and MR isolates, at 0.004 μg/mL and 0.008 μg/mL, respectively. Rifampicin kill curves were not indicative of concentration-dependency, suggesting time-dependent activity. Two isolates (MS 0.003 and 0.008 μg/mL) exhibited bacteriostatic activity, whereas two others (MR 0.003 and 0.012 μg/mL) exhibited bactericidal activity. CONCLUSIONS AND CLINICAL IMPORTANCE This study demonstrated that MS and MR S. pseudintermedius isolates were equally susceptible to rifampicin and that dosing intervals should be designed for time-dependent efficacy. These data can support pharmacokinetic studies of RFP in dogs with susceptible infections caused by S. pseudintermedius.
Collapse
Affiliation(s)
- Karen K Ho
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, 1220 Wire Road, Auburn, AL, 36849, USA
| | - Austin C Conley
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, 109 Greene Hall, Auburn, AL, 36849, USA
| | - Robert A Kennis
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, 1220 Wire Road, Auburn, AL, 36849, USA
| | - Terri L Hathcock
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, 264 Greene Hall, Auburn, AL, 36849, USA
| | - Dawn M Boothe
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, 109 Greene Hall, Auburn, AL, 36849, USA
| | - Amelia G White
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, 1220 Wire Road, Auburn, AL, 36849, USA
| |
Collapse
|
18
|
Liu LK, Dai Y, Abdelwahab H, Sobrado P, Tanner JJ. Structural Evidence for Rifampicin Monooxygenase Inactivating Rifampicin by Cleaving Its Ansa-Bridge. Biochemistry 2018; 57:2065-2068. [PMID: 29578336 DOI: 10.1021/acs.biochem.8b00190] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Rifampicin monooxygenase (RIFMO) decreases the potency of rifampicin (RIF) by converting it to oxidative products. Further decomposition of RIF has been observed in bacteria producing RIFMO and contributes to RIFMO-mediated drug resistance. Here we report the first crystal structure of RIFMO in complex with the hydroxylated RIF product. The 2.10 Å resolution structure reveals a breach of the ansa aliphatic chain of RIF between naphthoquinone C2 and amide N1. Our data suggest that RIFMO catalyzes the hydroxylation of RIF at the C2 atom followed by cleavage of the ansa linkage, which leads to inactivation of the antibiotic by preventing key contacts with the RNA polymerase target.
Collapse
Affiliation(s)
| | - Yumin Dai
- Department of Biochemistry , Virginia Tech , Blacksburg , Virginia 24061 , United States
| | - Heba Abdelwahab
- Department of Biochemistry , Virginia Tech , Blacksburg , Virginia 24061 , United States
| | - Pablo Sobrado
- Department of Biochemistry , Virginia Tech , Blacksburg , Virginia 24061 , United States
| | | |
Collapse
|