1
|
Wang X, Shao Y, Yao C, Huang L, Song W, Yang X, Zhang Z. 2D/2D Co 3O 4/BiOCl nanocomposite with enhanced antibacterial activity under full spectrum: Synergism of mesoporous structure, photothermal effect and photocatalytic reactive oxygen species. J Colloid Interface Sci 2025; 678:30-41. [PMID: 39180846 DOI: 10.1016/j.jcis.2024.08.145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/25/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024]
Abstract
The overuse of antibiotics has caused the emergence of drug-resistant bacteria and even superbugs, which makes it imperative to develop promising antibiotic-free alternatives. Herein, a multimodal antibacterial nanoplatform of two dimensional/two dimensional (2D/2D) mesoporous Co3O4/BiOCl nanocomposite is constructed, which possesses the effect of "kill three birds with one stone": (1) the use of mesoporous Co3O4 can enlarge the surface area of the nanocomposite and promote the adsorption of bacteria; (2) Co3O4 displays remarkable full-spectrum absorption and photo-induced self-heating effect, which can raise the temperature of Co3O4/BiOCl and help to kill bacteria; (3) the p-type Co3O4 and n-type BiOCl form a p-n heterojunction, which promotes the separation of photoelectrons and holes, thus producing more reactive oxygen species (ROS) for killing bacteria. The synergism of mesoporous structure, photothermal effect and photocatalytic ROS makes the developed Co3O4/BiOCl a promising antibacterial material, which shows outstanding antibacterial activity with an inhibition rate of nearly 100 % against Escherichia coli (E. coli) within 8 min. This work provides inspiration for designing multimodal synergistic nanoplatform for antibacterial applications.
Collapse
Affiliation(s)
- Xuesheng Wang
- School of Materials Science and Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, PR China
| | - Yi Shao
- Institute for Agro-food Standards and Testing Technology, Shanghai Academy of Agricultural Sciences, 1000 Jinqi Road, Shanghai 201403, PR China
| | - Chunxia Yao
- Institute for Agro-food Standards and Testing Technology, Shanghai Academy of Agricultural Sciences, 1000 Jinqi Road, Shanghai 201403, PR China.
| | - Liujuan Huang
- Institute for Agro-food Standards and Testing Technology, Shanghai Academy of Agricultural Sciences, 1000 Jinqi Road, Shanghai 201403, PR China
| | - Wei Song
- Institute for Agro-food Standards and Testing Technology, Shanghai Academy of Agricultural Sciences, 1000 Jinqi Road, Shanghai 201403, PR China
| | - Xianli Yang
- Institute for Agro-food Standards and Testing Technology, Shanghai Academy of Agricultural Sciences, 1000 Jinqi Road, Shanghai 201403, PR China
| | - Zhijie Zhang
- School of Materials Science and Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, PR China.
| |
Collapse
|
2
|
Pacios O, Herrera-Espejo S, Armán L, Ibarguren-Quiles C, Blasco L, Bleriot I, Fernández-García L, Ortiz-Cartagena C, Paniagua M, Barrio-Pujante A, Aracil B, Cisneros JM, Pachón-Ibáñez ME, Tomás M. Mitomycin C as an Anti-Persister Strategy against Klebsiella pneumoniae: Toxicity and Synergy Studies. Antibiotics (Basel) 2024; 13:815. [PMID: 39334989 PMCID: PMC11428439 DOI: 10.3390/antibiotics13090815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
The combination of several therapeutic strategies is often seen as a good way to decrease resistance rates, since bacteria can more easily overcome single-drug treatments than multi-drug ones. This strategy is especially attractive when several targets and subpopulations are affected, as it is the case of Klebsiella pneumoniae persister cells, a subpopulation of bacteria able to transiently survive antibiotic exposures. This work aims to evaluate the potential of a repurposed anticancer drug, mitomycin C, combined with the K. pneumoniae lytic phage vB_KpnM-VAC13 in vitro and its safety in an in vivo murine model against two clinical isolates of this pathogen, one of them exhibiting an imipenem-persister phenotype. At the same time, we verified the absence of toxicity of mitomycin C at the concentration using the human chondrocyte cell line T/C28a2. The viability of these human cells was checked using both cytotoxicity assays and flow cytometry.
Collapse
Affiliation(s)
- Olga Pacios
- Translational and Multidisciplinary Microbiology Research Group (MicroTM)-Microbiology Department, Biomedical Research Institute of A Coruña (INIBIC), A Coruña Hospital (CHUAC), University of A Coruña (UDC), 15006 A Coruña, Spain; (O.P.); (L.A.); (C.I.-Q.); (L.B.); (I.B.); (L.F.-G.); (C.O.-C.); (A.B.-P.)
- Mechanisms of Antimicrobial Resistance Study Group (GEMARA) on Behalf of the Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC), 28003 Madrid, Spain
| | - Soraya Herrera-Espejo
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, 41013 Seville, Spain; (S.H.-E.); (M.P.); (J.M.C.); (M.E.P.-I.)
| | - Lucía Armán
- Translational and Multidisciplinary Microbiology Research Group (MicroTM)-Microbiology Department, Biomedical Research Institute of A Coruña (INIBIC), A Coruña Hospital (CHUAC), University of A Coruña (UDC), 15006 A Coruña, Spain; (O.P.); (L.A.); (C.I.-Q.); (L.B.); (I.B.); (L.F.-G.); (C.O.-C.); (A.B.-P.)
- Mechanisms of Antimicrobial Resistance Study Group (GEMARA) on Behalf of the Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC), 28003 Madrid, Spain
| | - Clara Ibarguren-Quiles
- Translational and Multidisciplinary Microbiology Research Group (MicroTM)-Microbiology Department, Biomedical Research Institute of A Coruña (INIBIC), A Coruña Hospital (CHUAC), University of A Coruña (UDC), 15006 A Coruña, Spain; (O.P.); (L.A.); (C.I.-Q.); (L.B.); (I.B.); (L.F.-G.); (C.O.-C.); (A.B.-P.)
- Mechanisms of Antimicrobial Resistance Study Group (GEMARA) on Behalf of the Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC), 28003 Madrid, Spain
| | - Lucía Blasco
- Translational and Multidisciplinary Microbiology Research Group (MicroTM)-Microbiology Department, Biomedical Research Institute of A Coruña (INIBIC), A Coruña Hospital (CHUAC), University of A Coruña (UDC), 15006 A Coruña, Spain; (O.P.); (L.A.); (C.I.-Q.); (L.B.); (I.B.); (L.F.-G.); (C.O.-C.); (A.B.-P.)
- Mechanisms of Antimicrobial Resistance Study Group (GEMARA) on Behalf of the Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC), 28003 Madrid, Spain
- MEPRAM, Project of Personalized Medicine against Antimicrobial Resistance, 28029 Madrid, Spain;
| | - Inés Bleriot
- Translational and Multidisciplinary Microbiology Research Group (MicroTM)-Microbiology Department, Biomedical Research Institute of A Coruña (INIBIC), A Coruña Hospital (CHUAC), University of A Coruña (UDC), 15006 A Coruña, Spain; (O.P.); (L.A.); (C.I.-Q.); (L.B.); (I.B.); (L.F.-G.); (C.O.-C.); (A.B.-P.)
- Mechanisms of Antimicrobial Resistance Study Group (GEMARA) on Behalf of the Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC), 28003 Madrid, Spain
| | - Laura Fernández-García
- Translational and Multidisciplinary Microbiology Research Group (MicroTM)-Microbiology Department, Biomedical Research Institute of A Coruña (INIBIC), A Coruña Hospital (CHUAC), University of A Coruña (UDC), 15006 A Coruña, Spain; (O.P.); (L.A.); (C.I.-Q.); (L.B.); (I.B.); (L.F.-G.); (C.O.-C.); (A.B.-P.)
- Mechanisms of Antimicrobial Resistance Study Group (GEMARA) on Behalf of the Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC), 28003 Madrid, Spain
| | - Concha Ortiz-Cartagena
- Translational and Multidisciplinary Microbiology Research Group (MicroTM)-Microbiology Department, Biomedical Research Institute of A Coruña (INIBIC), A Coruña Hospital (CHUAC), University of A Coruña (UDC), 15006 A Coruña, Spain; (O.P.); (L.A.); (C.I.-Q.); (L.B.); (I.B.); (L.F.-G.); (C.O.-C.); (A.B.-P.)
- Mechanisms of Antimicrobial Resistance Study Group (GEMARA) on Behalf of the Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC), 28003 Madrid, Spain
| | - María Paniagua
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, 41013 Seville, Spain; (S.H.-E.); (M.P.); (J.M.C.); (M.E.P.-I.)
- CIBER of Infectious Diseases (CIBERINFEC), Health Institute Carlos III, 28029 Madrid, Spain
| | - Antonio Barrio-Pujante
- Translational and Multidisciplinary Microbiology Research Group (MicroTM)-Microbiology Department, Biomedical Research Institute of A Coruña (INIBIC), A Coruña Hospital (CHUAC), University of A Coruña (UDC), 15006 A Coruña, Spain; (O.P.); (L.A.); (C.I.-Q.); (L.B.); (I.B.); (L.F.-G.); (C.O.-C.); (A.B.-P.)
- Mechanisms of Antimicrobial Resistance Study Group (GEMARA) on Behalf of the Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC), 28003 Madrid, Spain
| | - Belén Aracil
- MEPRAM, Project of Personalized Medicine against Antimicrobial Resistance, 28029 Madrid, Spain;
- CIBER of Infectious Diseases (CIBERINFEC), Health Institute Carlos III, 28029 Madrid, Spain
- Reference Laboratory of Antimicrobial Resistance, National Center of Microbiology, Health Institute Carlos III, Majadahonda, 28222 Madrid, Spain
| | - José Miguel Cisneros
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, 41013 Seville, Spain; (S.H.-E.); (M.P.); (J.M.C.); (M.E.P.-I.)
- MEPRAM, Project of Personalized Medicine against Antimicrobial Resistance, 28029 Madrid, Spain;
- CIBER of Infectious Diseases (CIBERINFEC), Health Institute Carlos III, 28029 Madrid, Spain
| | - María Eugenia Pachón-Ibáñez
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, 41013 Seville, Spain; (S.H.-E.); (M.P.); (J.M.C.); (M.E.P.-I.)
- MEPRAM, Project of Personalized Medicine against Antimicrobial Resistance, 28029 Madrid, Spain;
- CIBER of Infectious Diseases (CIBERINFEC), Health Institute Carlos III, 28029 Madrid, Spain
| | - María Tomás
- Translational and Multidisciplinary Microbiology Research Group (MicroTM)-Microbiology Department, Biomedical Research Institute of A Coruña (INIBIC), A Coruña Hospital (CHUAC), University of A Coruña (UDC), 15006 A Coruña, Spain; (O.P.); (L.A.); (C.I.-Q.); (L.B.); (I.B.); (L.F.-G.); (C.O.-C.); (A.B.-P.)
- Mechanisms of Antimicrobial Resistance Study Group (GEMARA) on Behalf of the Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC), 28003 Madrid, Spain
- MEPRAM, Project of Personalized Medicine against Antimicrobial Resistance, 28029 Madrid, Spain;
| |
Collapse
|
3
|
Al-Rabia MW, Asfour HZ, Alhakamy NA, Bazuhair MA, Ibrahim TS, Abbas HA, Mansour B, Hegazy WAH, Seleem NM. Cilostazol is a promising anti-pseudomonal virulence drug by disruption of quorum sensing. AMB Express 2024; 14:87. [PMID: 39090255 PMCID: PMC11294311 DOI: 10.1186/s13568-024-01740-1] [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: 04/01/2024] [Accepted: 07/03/2024] [Indexed: 08/04/2024] Open
Abstract
Resistance to antibiotics is a critical growing public health problem that desires urgent action to combat. To avoid the stress on bacterial growth that evokes the resistance development, anti-virulence agents can be an attractive strategy as they do not target bacterial growth. Quorum sensing (QS) systems play main roles in controlling the production of diverse virulence factors and biofilm formation in bacteria. Thus, interfering with QS systems could result in mitigation of the bacterial virulence. Cilostazol is an antiplatelet and a vasodilator FDA approved drug. This study aimed to evaluate the anti-virulence activities of cilostazol in the light of its possible interference with QS systems in Pseudomonas aeruginosa. Additionally, the study examines cilostazol's impact on the bacterium's ability to induce infection in vivo, using sub-inhibitory concentrations to minimize the risk of resistance development. In this context, the biofilm formation, the production of virulence factors and influence on the in vivo ability to induce infection were assessed in the presence of cilostazol at sub-inhibitory concentration. Furthermore, the outcome of combination with antibiotics was evaluated. Cilostazol interfered with biofilm formation in P. aeruginosa. Moreover, swarming motility, biofilm formation and production of virulence factors were significantly diminished. Histopathological investigation revealed that liver, spleen and kidney tissues damage was abolished in mice injected with cilostazol-treated bacteria. Cilostazol exhibited a synergistic outcome when used in combination with antibiotics. At the molecular level, cilostazol downregulated the QS genes and showed considerable affinity to QS receptors. In conclusion, Cilostazol could be used as adjunct therapy with antibiotics for treating Pseudomonal infections. This research highlights cilostazol's potential to combat bacterial infections by targeting virulence mechanisms, reducing the risk of antibiotic resistance, and enhancing treatment efficacy against P. aeruginosa. These findings open avenues for repurposing existing drugs, offering new, safer, and more effective infection control strategies.
Collapse
Affiliation(s)
- Mohammed W Al-Rabia
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Hani Z Asfour
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Nabil A Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Mohammed A Bazuhair
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Tarek S Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Hisham A Abbas
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Basem Mansour
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt
- Department of Pharmaceutical Chemistry, Kut University College, Al Kut, Wasit, 52001, Iraq
| | - Wael A H Hegazy
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
- Department of Pharmaceutical Sciences, Pharmacy Program, College of Health Sciences, 113, Muscat, Oman.
| | - Noura M Seleem
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| |
Collapse
|
4
|
Wu W, Wang Y, Yang H, Chen H, Wang C, Liang J, Song Y, Xu S, Sun Y, Wang L. Antibacterial and Biofilm Removal Strategies Based on Micro/Nanomotors in the Biomedical Field. ChemMedChem 2024:e202400349. [PMID: 38965060 DOI: 10.1002/cmdc.202400349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/30/2024] [Accepted: 07/04/2024] [Indexed: 07/06/2024]
Abstract
Bacterial infection, which can trigger varieties of diseases and tens of thousands of deaths each year, poses serious threats to human health. Particularly, the new dilemma caused by biofilms is gradually becoming a severe and tough problem in the biomedical field. Thus, the strategies to address these problems are considered an urgent task at present. Micro/nanomotors (MNMs), also named micro/nanoscale robots, are mostly driven by chemical energy or external field, exhibiting strong diffusion and self-propulsion in the liquid media, which has the potential for antibacterial applications. In particular, when MNMs are assembled in swarms, they become robust and efficient for biofilm removal. However, there is a lack of comprehensive review discussing the progress in this aspect. Bearing it in mind and based on our own research experience in this regard, the studies on MNMs driven by different mechanisms orchestrated for antibacterial activity and biofilm removal are timely and concisely summarized and discussed in this work, aiming to show the advantages of MNMs brought to this field. In addition, an outlook was proposed, hoping to provide the fundamental guidance for future development in this area.
Collapse
Affiliation(s)
- Wenlu Wu
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Yuxin Wang
- Emergency Department, Harbin First Hospital, Harbin, 150010, China
| | - Haiyue Yang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Haixu Chen
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Cong Wang
- Department of Microwave Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Junge Liang
- Department of Electronic Engineering, Jiangnan University, Wuxi, 214122, China
| | - Yiran Song
- Department of Electronic Engineering, Jiangnan University, Wuxi, 214122, China
| | - Shanshan Xu
- Emergency Department, Harbin First Hospital, Harbin, 150010, China
| | - Yuan Sun
- Center of Pharmaceutical Engineering and Technology, Harbin University of Commerce, Harbin, 150076, China
| | - Lei Wang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| |
Collapse
|
5
|
Al-Fadhli AH, Jamal WY. Recent advances in gene-editing approaches for tackling antibiotic resistance threats: a review. Front Cell Infect Microbiol 2024; 14:1410115. [PMID: 38994001 PMCID: PMC11238145 DOI: 10.3389/fcimb.2024.1410115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 06/11/2024] [Indexed: 07/13/2024] Open
Abstract
Antibiotic resistance, a known global health challenge, involves the flow of bacteria and their genes among animals, humans, and their surrounding environment. It occurs when bacteria evolve and become less responsive to the drugs designated to kill them, making infections harder to treat. Despite several obstacles preventing the spread of genes and bacteria, pathogens regularly acquire novel resistance factors from other species, which reduces their ability to prevent and treat such bacterial infections. This issue requires coordinated efforts in healthcare, research, and public awareness to address its impact on human health worldwide. This review outlines how recent advances in gene editing technology, especially CRISPR/Cas9, unveil a breakthrough in combating antibiotic resistance. Our focus will remain on the relationship between CRISPR/cas9 and its impact on antibiotic resistance and its related infections. Moreover, the prospects of this new advanced research and the challenges of adopting these technologies against infections will be outlined by exploring its different derivatives and discussing their advantages and limitations over others, thereby providing a corresponding reference for the control and prevention of the spread of antibiotic resistance.
Collapse
Affiliation(s)
- Amani H Al-Fadhli
- Laboratory Sciences, Department of Medical, Faculty of Allied Health Sciences, Health Sciences Center (HSC), Kuwait University, Jabriya, Kuwait
| | - Wafaa Yousef Jamal
- Department of Microbiology, College of Medicine, Kuwait University, Jabriya, Kuwait
| |
Collapse
|
6
|
Lu Y, Zhang L, Wang J, Bian L, Ding Z, Yang C. Hyperspectral upgrade solution for biomicroscope combined with Transformer network to classify infectious bacteria. JOURNAL OF BIOPHOTONICS 2024; 17:e202300484. [PMID: 38297446 DOI: 10.1002/jbio.202300484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 02/02/2024]
Abstract
Infectious diseases caused by bacterial pathogens pose a significant public health threat, emphasizing the need for swift and accurate bacterial species detection methods. Hyperspectral microscopic imaging (HMI) offers nondestructive, rapid, and data-rich advantages, making it a promising tool for microbial detection. In this research, we present a highly compatible and cost-effective approach to extend a standard biomicroscope system into a hyperspectral biomicroscope using a prism-grating-prism configuration. Using this prototype, we generate 600 hyperspectral data cubes for Listeria, Bacillus typhi, Bacillus pestis, and Bacillus anthracis. Additionally, we propose a Transformer-based classification network that achieves a 99.44% accuracy in classifying these infectious pathogens, outperforming traditional methods. Our results suggest that the successful combination of HMI and the optimized Transformer-based classification network highlights the potential for rapid and precise detection of infectious disease pathogens .
Collapse
Affiliation(s)
- You Lu
- Engineering Research Center of Semiconductor Power Device Reliability Ministry of Education, Guizhou University, Guiyang, China
| | - Lan Zhang
- Engineering Research Center of Semiconductor Power Device Reliability Ministry of Education, Guizhou University, Guiyang, China
| | - Jihong Wang
- Engineering Research Center of Semiconductor Power Device Reliability Ministry of Education, Guizhou University, Guiyang, China
| | - Lifeng Bian
- Frontier Institute of Chip and System, Fudan University, Shanghai, China
| | - Zhao Ding
- Engineering Research Center of Semiconductor Power Device Reliability Ministry of Education, Guizhou University, Guiyang, China
| | - Chen Yang
- Engineering Research Center of Semiconductor Power Device Reliability Ministry of Education, Guizhou University, Guiyang, China
| |
Collapse
|
7
|
Allemailem KS. Recent Advances in Understanding the Molecular Mechanisms of Multidrug Resistance and Novel Approaches of CRISPR/Cas9-Based Genome-Editing to Combat This Health Emergency. Int J Nanomedicine 2024; 19:1125-1143. [PMID: 38344439 PMCID: PMC10859101 DOI: 10.2147/ijn.s453566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 01/26/2024] [Indexed: 02/15/2024] Open
Abstract
The rapid spread of multidrug resistance (MDR), due to abusive use of antibiotics has led to global health emergency, causing substantial morbidity and mortality. Bacteria attain MDR by different means such as antibiotic modification/degradation, target protection/modification/bypass, and enhanced efflux mechanisms. The classical approaches of counteracting MDR bacteria are expensive and time-consuming, thus, it is highly significant to understand the molecular mechanisms of this resistance to curb the problem from core level. The revolutionary approach of clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated sequence 9 (CRISPR/Cas9), considered as a next-generation genome-editing tool presents an innovative opportunity to precisely target and edit bacterial genome to alter their MDR strategy. Different bacteria possessing antibiotic resistance genes such as mecA, ermB, ramR, tetA, mqrB and blaKPC that have been targeted by CRISPR/Cas9 to re-sensitize these pathogens against antibiotics, such as methicillin, erythromycin, tigecycline, colistin and carbapenem, respectively. The CRISPR/Cas9 from S. pyogenes is the most widely studied genome-editing tool, consisting of a Cas9 DNA endonuclease associated with tracrRNA and crRNA, which can be systematically coupled as sgRNA. The targeting strategies of CRISPR/Cas9 to bacterial cells is mediated through phage, plasmids, vesicles and nanoparticles. However, the targeting approaches of this genome-editing tool to specific bacteria is a challenging task and still remains at a very preliminary stage due to numerous obstacles awaiting to be solved. This review elaborates some recent updates about the molecular mechanisms of antibiotic resistance and the innovative role of CRISPR/Cas9 system in modulating these resistance mechanisms. Furthermore, the delivery approaches of this genome-editing system in bacterial cells are discussed. In addition, some challenges and future prospects are also described.
Collapse
Affiliation(s)
- Khaled S Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah51452, Saudi Arabia
| |
Collapse
|
8
|
Jothi R, Hong ST, Enkhtsatsral M, Pandian SK, Gowrishankar S. ROS mediated anticandidal efficacy of 3-Bromopyruvate prevents vulvovaginal candidiasis in mice model. PLoS One 2023; 18:e0295922. [PMID: 38153954 PMCID: PMC10754460 DOI: 10.1371/journal.pone.0295922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 12/03/2023] [Indexed: 12/30/2023] Open
Abstract
Candidal infections, particularly vulvovaginal candidiasis (VVC), necessitate effective therapeutic interventions in clinical settings owing to their intricate clinical nature and elusive understanding of their etiological mechanisms. Given the challenges in developing effective antifungal therapies, the strategy of repurposing existing pharmaceuticals has emerged as a promising approach to combat drug-resistant fungi. In this regard, the current study investigates molecular insights on the anti-candidal efficacy of a well-proven anticancer small molecule -3-bromopyruvate (3BP) against three clinically significant VVC causing Candida species viz., C. albicans, C. tropicalis and C. glabrata. Furthermore, the study validates 3BP's therapeutic application by developing it as a vaginal cream for the treatment of VVC. 3BP exhibited phenomenal antifungal efficacy (killing >99%) with minimum inhibitory concentrations (MIC) and minimum fungicidal concentrations (MFC) of 256 μg/mL against all tested Candida spp. Time killing kinetics experiment revealed 20 min as the minimum time required for 3BP at 2XMIC to achieve complete-killing (99.9%) in all Candida strains. Moreover, the ergosterol or sorbitol experiment explicated that the antifungal activity of 3BP does not stem from targeting the cell wall or the membrane component ergosterol. Instead, 3BP was observed to instigate a sequence of pre-apoptotic cascade events, such as phosphatidylserine (PS) externalization, nuclear condensation and ROS accumulations, as evidenced by PI, DAPI and DCFH-DA staining methods. Furthermore, 3BP demonstrated a remarkable efficacy in eradicating mature biofilms of Candida spp., achieving a maximum eradication level of 90%. Toxicity/safety profiling in both in vitro erythrocyte lysis and in vivo Galleria mellonella survival assay authenticated the non-toxic nature of 3BP up to 512 μg/mL. Finally, a vaginal cream formulated with 3BP was found to be effective in VVC-induced female mice model, as it significantly decreasing fungal load and protecting vaginal mucosa. Concomitantly, the present study serves as a clear demonstration of antifungal mechanistic action of anticancer drug -3BP, against Candida species. This finding holds significant potential for mitigating candidal infections, particularly VVC, within healthcare environments.
Collapse
Affiliation(s)
- Ravi Jothi
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Seong-Tshool Hong
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, South Korea
| | - Munkhtur Enkhtsatsral
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, South Korea
| | | | | |
Collapse
|
9
|
Uddin MJ, Overkleeft HS, Lentz CS. Activity-Based Protein Profiling in Methicillin-Resistant Staphylococcus aureus Reveals the Broad Reactivity of a Carmofur-Derived Probe. Chembiochem 2023; 24:e202300473. [PMID: 37552008 DOI: 10.1002/cbic.202300473] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/07/2023] [Accepted: 08/07/2023] [Indexed: 08/09/2023]
Abstract
Activity-based protein profiling is a powerful chemoproteomic technique to detect active enzymes and identify targets and off-targets of drugs. Here, we report the use of carmofur- and activity-based probes to identify biologically relevant enzymes in the bacterial pathogen Staphylococcus aureus. Carmofur is an anti-neoplastic prodrug of 5-fluorouracil and also has antimicrobial and anti-biofilm activity. Carmofur probes were originally designed to target human acid ceramidase, a member of the NTN hydrolase family with an active-site cysteine nucleophile. Here, we first profiled the targets of a fluorescent carmofur probe in live S. aureus under biofilm-promoting conditions and in liquid culture, before proceeding to target identification by liquid chromatography/mass spectrometry. Treatment with a carmofur-biotin probe led to enrichment of 20 enzymes from diverse families awaiting further characterization, including the NTN hydrolase-related IMP cyclohydrolase PurH. However, the probe preferentially labeled serine hydrolases, thus displaying a reactivity profile similar to that of carbamates. Our results suggest that the electrophilic N-carbamoyl-5-fluorouracil scaffold could potentially be optimized to achieve selectivity towards diverse enzyme families. The observed promiscuous reactivity profile suggests that the clinical use of carmofur presumably leads to inactivation of a number human and microbial enzymes, which could lead to side effects and/or contribute to therapeutic efficacy.
Collapse
Affiliation(s)
- Md Jalal Uddin
- Department of Medical Biology, UiT- The Arctic University of Norway, 9019, Tromsø, Norway
| | - Hermen S Overkleeft
- Department of Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Christian S Lentz
- Department of Medical Biology, UiT- The Arctic University of Norway, 9019, Tromsø, Norway
| |
Collapse
|
10
|
Khattak AA, Qian J, Xu L, Tomah AA, Ibrahim E, Khan MZI, Ahmed T, Hatamleh AA, Al-Dosary MA, Ali HM, Li B. Precision drug design against Acidovorax oryzae: leveraging bioinformatics to combat rice brown stripe disease. Front Cell Infect Microbiol 2023; 13:1225285. [PMID: 37886665 PMCID: PMC10598866 DOI: 10.3389/fcimb.2023.1225285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 09/15/2023] [Indexed: 10/28/2023] Open
Abstract
Bacterial brown stripe disease caused by Acidovorax oryzae is a major threat to crop yields, and the current reliance on pesticides for control is unsustainable due to environmental pollution and resistance. To address this, bacterial-based ligands have been explored as a potential treatment solution. In this study, we developed a protein-protein interaction (PPI) network for A. oryzae by utilizing shared differentially expressed genes (DEGs) and the STRING database. Using a maximal clique centrality (MCC) approach through CytoHubba and Network Analyzer, we identified hub genes within the PPI network. We then analyzed the genomic data of the top 10 proteins, and further narrowed them down to 2 proteins by utilizing betweenness, closeness, degree, and eigenvector studies. Finally, we used molecular docking to screen 100 compounds against the final two proteins (guaA and metG), and Enfumafungin was selected as a potential treatment for bacterial resistance caused by A. oryzae based on their binding affinity and interaction energy. Our approach demonstrates the potential of utilizing bioinformatics and molecular docking to identify novel drug candidates for precision treatment of bacterial brown stripe disease caused by A. oryzae, paving the way for more targeted and sustainable control strategies. The efficacy of Enfumafungin in inhibiting the growth of A. oryzae strain RS-1 was investigated through both computational and wet lab methods. The models of the protein were built using the Swiss model, and their accuracy was confirmed via a Ramachandran plot. Additionally, Enfumafungin demonstrated potent inhibitory action against the bacterial strain, with an MIC of 100 µg/mL, reducing OD600 values by up to 91%. The effectiveness of Enfumafungin was further evidenced through agar well diffusion assays, which exhibited the highest zone of inhibition at 1.42 cm when the concentration of Enfumafungin was at 100 µg/mL. Moreover, Enfumafungin was also able to effectively reduce the biofilm of A. oryzae RS-1 in a concentration-dependent manner. The swarming motility of A. oryzae RS-1 was also found to be significantly inhibited by Enfumafungin. Further validation through TEM observation revealed that bacterial cells exposed to Enfumafungin displayed mostly red fluorescence, indicating destruction of the bacterial cell membrane.
Collapse
Affiliation(s)
- Arif Ali Khattak
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
| | - Jiahui Qian
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
| | - Lihui Xu
- Institute of Eco-Environmental Protection, Shanghai Academy of Agricultural Sciences, Shanghai, China
| | - Ali Athafah Tomah
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
- Plant Protection, College of Agriculture, University of Misan, AL-Amarah, Iraq
| | - Ezzeldin Ibrahim
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
- Department of Vegetable Diseases Research, Plant Pathology Research Institute, Agriculture Research Centre, Giza, Egypt
| | | | - Temoor Ahmed
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
- Xianghu Laboratory, Hangzhou, China
| | - Ashraf Atef Hatamleh
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | | | - Hayssam M. Ali
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Bin Li
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Ferrando R, Mitchell SG, Atrián-Blasco E, Cerrada E. Antibacterial properties of phosphine gold(I) complexes with 5-fluorouracil. Dalton Trans 2023. [PMID: 37448318 DOI: 10.1039/d3dt01159c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/15/2023]
Abstract
New gold(I) complexes with coordination to 5-fluorouracil (5-FU), an anticancer drug with antibacterial properties, have been synthesised and characterised, and are the first reported examples of 5-FU-Au compounds. These new complexes show high solution stability, even in the presence of a cysteine derivative, and so were evaluated as antibacterial compounds against model Gram-positive and Gram-negative bacteria. All the complexes show excellent antibacterial activity against Gram-positive B. subtilis, most of them improving the activity of 5-FU alone. Furthermore, these new complexes are also active against Gram-negative E. coli, where [Au(5-FU)(PTA)], the complex with the smallest phosphane, is the most bactericidal, 32 times more active than 5-FU on its own.
Collapse
Affiliation(s)
- Ricardo Ferrando
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-CSIC, 50009 Zaragoza, Spain.
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
| | - Scott G Mitchell
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Elena Atrián-Blasco
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza 50009, Spain.
| | - Elena Cerrada
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-CSIC, 50009 Zaragoza, Spain.
| |
Collapse
|
12
|
Wang B, Chen S, Sun X, Shan X, Zhu X, Yuan B, Wang H, Zhou G, Liu J. A Photothermally Enhanced Vancomycin-Coated Liquid Metal Antimicrobial Agent with Targeting Capability. Bioengineering (Basel) 2023; 10:748. [PMID: 37508775 PMCID: PMC10376194 DOI: 10.3390/bioengineering10070748] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/14/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
The targeted antimicrobial efficacy of Vancomycin decreases significantly over time due to bacterial resistance, whereas Ga-based liquid metals, which are less prone to inducing bacterial resistance, face challenges in achieving targeted antimicrobial effects. To tackle these issues, a highly efficient antimicrobial agent with targeting properties has been developed by combining Ga-based liquid metals and Vancomycin. Moreover, the performance of this antimicrobial agent can be greatly enhanced through the use of near-infrared light. Microscopic observations reveal that Vancomycin can be effectively encapsulated on the surface of liquid metal, facilitated by the presence of the oxide layer. The resulting core–shell structured antimicrobial agent demonstrates notable targeted antimicrobial effects against S. aureus. Antibacterial tests indicate that Vancomycin effectively improves the antibacterial properties of pure liquid metal. Additionally, this study unveils the excellent photothermal conversion capabilities of liquid metal, enabling the antimicrobial agent exposed to 808nm near-infrared light to exhibit significantly strengthened bactericidal performance. In this scenario, the antimicrobial agent can achieve nearly 100% effectiveness. This work enriches the investigation of integrating Ga-based antimicrobial agents with traditional antibiotics, showcasing promising antibacterial effects and establishing the groundwork for subsequent clinical applications.
Collapse
Affiliation(s)
- Bo Wang
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China; (B.W.)
| | - Sen Chen
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xuyang Sun
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China; (B.W.)
| | - Xiaohui Shan
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xiyu Zhu
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Bo Yuan
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Hongzhang Wang
- Center of Double Helix, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Beijing Key Lab of Cryo-Biomedical Engineering, Technical Institute of Physics and Chemistry Chinese Academy of Sciences, Beijing 100190, China
| | - Gang Zhou
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China; (B.W.)
| | - Jing Liu
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China
- Beijing Key Lab of Cryo-Biomedical Engineering, Technical Institute of Physics and Chemistry Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
13
|
Kantiwal U, Pandey J. Efficient Inhibition of Bacterial Biofilm Through Interference of Protein-Protein Interaction of Master Regulator Proteins: a Proof of Concept Study with SinR- SinI Complex of Bacillus subtilis. Appl Biochem Biotechnol 2023; 195:1947-1967. [PMID: 36401726 DOI: 10.1007/s12010-022-04231-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2022] [Indexed: 11/21/2022]
Abstract
Biofilm-associated microbial growth is a major cause of environmental, industrial, and public health concern. Therefore, there is a pressing need to discover and develop efficient antibiofilm strategies. Regulatory proteins vital for biofilm formation might be ideal targets for developing novel antibiofilm therapeutics. Their activities often depend on protein-protein interactions. Therefore, such targets present unique opportunities and challenges to drug discovery. In Bacillus subtilis, a model organism for studying biofilms, SinR acts as the master regulator of the biofilm formation cascade. Under favourable growth conditions, it represses the epsA-O and tapA-sipW-tasA operons, which encode for essential structural components of biofilms. Under unfavourable growth conditions, SinI, an agonist protein, inactivates SinR by forming a heterotrimeric complex. This results in derepression of epsA-O and tapA-sipW-tasA operons and leads to the phenotypic switch from planktonic to biofilm-associated form. We hypothesized that inhibiting SinR-SinI interaction might warrant repression of epsA-O and tapA-sipW-tasA operons and inhibit biofilm formation. To evaluate this hypothesis, we carried out a drug repurposing study for identifying potential inhibitors of SinI. Cefoperazone and itraconazole were identified as potential inhibitors with virtual screening. The stability of their interaction with SinI was assessed in extended MD performed over 100 ns. Both cefoperazone and itraconazole showed stable interaction. In in vitro studies, cefoperazone hindered the interaction of purified recombinant SinI and SinR. In the whole cell-based biofilm inhibition assays also cefoperazone was found to efficiently inhibited biofilm formation. These results provide proof of concept for targeting protein-protein interaction of master regulators as potential target for discovery and development of antibiofilm therapeutics. We propose that similar drug repurposing studies targeting key regulators of biofilm formation cascade could be an efficient approach for discovering novel anti-biofilm therapeutics against priority pathogens.
Collapse
Affiliation(s)
- Usha Kantiwal
- Laboratory of Molecular Microbiology, Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, Bandarsindri, NH-8, Kishangarh, Ajmer, 305817, Rajasthan, India
| | - Janmejay Pandey
- Laboratory of Molecular Microbiology, Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, Bandarsindri, NH-8, Kishangarh, Ajmer, 305817, Rajasthan, India.
| |
Collapse
|
14
|
Tao C, Du J, Tang Y, Wang J, Dong K, Yang M, Hu B, Zhang Z. A Deep-Learning Based System for Rapid Genus Identification of Pathogens under Hyperspectral Microscopic Images. Cells 2022; 11:cells11142237. [PMID: 35883680 PMCID: PMC9315805 DOI: 10.3390/cells11142237] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/06/2022] [Accepted: 07/15/2022] [Indexed: 11/21/2022] Open
Abstract
Infectious diseases have always been a major threat to the survival of humanity. Additionally, they bring an enormous economic burden to society. The conventional methods for bacteria identification are expensive, time-consuming and laborious. Therefore, it is of great importance to automatically rapidly identify pathogenic bacteria in a short time. Here, we constructed an AI-assisted system for automating rapid bacteria genus identification, combining the hyperspectral microscopic technology and a deep-learning-based algorithm Buffer Net. After being trained and validated in the self-built dataset, which consists of 11 genera with over 130,000 hyperspectral images, the accuracy of the algorithm could achieve 94.9%, which outperformed 1D-CNN, 2D-CNN and 3D-ResNet. The AI-assisted system we developed has great potential in assisting clinicians in identifying pathogenic bacteria at the single-cell level with high accuracy in a cheap, rapid and automatic way. Since the AI-assisted system can identify the pathogenic genus rapidly (about 30 s per hyperspectral microscopic image) at the single-cell level, it can shorten the time or even eliminate the demand for cultivating. Additionally, the system is user-friendly for novices.
Collapse
Affiliation(s)
- Chenglong Tao
- Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Sciences, Xi’an 710119, China; (C.T.); (J.D.); (J.W.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Biomedical Spectroscopy of Xi’an, Xi’an 710119, China
| | - Jian Du
- Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Sciences, Xi’an 710119, China; (C.T.); (J.D.); (J.W.)
- Key Laboratory of Biomedical Spectroscopy of Xi’an, Xi’an 710119, China
| | | | - Junjie Wang
- Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Sciences, Xi’an 710119, China; (C.T.); (J.D.); (J.W.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ke Dong
- The Second Affiliated Hospital of Air Force Military Medical University, Xi’an 710119, China; (K.D.); (M.Y.)
| | - Ming Yang
- The Second Affiliated Hospital of Air Force Military Medical University, Xi’an 710119, China; (K.D.); (M.Y.)
| | - Bingliang Hu
- Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Sciences, Xi’an 710119, China; (C.T.); (J.D.); (J.W.)
- Key Laboratory of Biomedical Spectroscopy of Xi’an, Xi’an 710119, China
- Correspondence: (B.H.); (Z.Z.)
| | - Zhoufeng Zhang
- Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Sciences, Xi’an 710119, China; (C.T.); (J.D.); (J.W.)
- Key Laboratory of Biomedical Spectroscopy of Xi’an, Xi’an 710119, China
- Correspondence: (B.H.); (Z.Z.)
| |
Collapse
|
15
|
Abbas HA, Shaker GH, Mosallam FM, Gomaa SE. Novel silver metformin nano-structure to impede virulence of Staphylococcus aureus. AMB Express 2022; 12:84. [PMID: 35771288 PMCID: PMC9247137 DOI: 10.1186/s13568-022-01426-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 06/21/2022] [Indexed: 11/25/2022] Open
Abstract
Staphylococcus aureus is a prevalent etiological agent of health care associated and community acquired infections. Antibiotic abuse resulted in developing multidrug resistance in S. aureus that complicates treatment of infections. Targeting bacterial virulence using FDA approved medication offers an alternative to the antibiotics with no stress on bacterial viability. Using nanomaterials as anti-virulence agent against S. aureus virulence factors is a valuable approach. This study aims to investigate the impact of metformin (MET), metformin nano (MET-Nano), silver metformin nano structure (Ag-MET-Ns) and silver nanoparticles (AgNPs) on S. aureus virulence and pathogenicity. The in vitro results showed a higher inhibitory activity against S. aureus virulence factors with both MET-Nano and Ag-MET-Ns treatment. However, genotypically, it was found that except for agrA and icaR genes that are upregulated, the tested agents significantly downregulated the expression of crtM, sigB, sarA and fnbA genes, with Ag-MET-Ns being the most efficient one. MET-Nano exhibited the highest protection against S. aureus infection in mice. These data indicate the promising anti-virulence activity of nanoformulations especially Ag-MET-Ns against multidrug resistant S. aureus by inhibiting quorum sensing signaling system. A new formation of silver metformin nanostructure. The in vitro inhibition of S. aureus virulence factors. Nano structure form improves the activity of anti-virulence agents.
Collapse
Affiliation(s)
- Hisham A Abbas
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University,, Zagazig, Egypt
| | - Ghada H Shaker
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University,, Zagazig, Egypt
| | - Farag M Mosallam
- Drug Microbiology Lab., Drug Radiation Research Department, Biotechnology Division, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt.
| | - Salwa E Gomaa
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University,, Zagazig, Egypt
| |
Collapse
|
16
|
Knocking down Pseudomonas aeruginosa virulence by oral hypoglycemic metformin nano emulsion. World J Microbiol Biotechnol 2022; 38:119. [PMID: 35644864 PMCID: PMC9148876 DOI: 10.1007/s11274-022-03302-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/05/2022] [Indexed: 11/18/2022]
Abstract
Long-term antibiotic treatment results in the spread of multi-drug resistance in Pseudomonas aeruginosa that complicates treatment. Anti-virulence agents can be viewed as alternative options that cripple virulence factors of the bacteria to facilitate their elimination by the host immunity. The use of nanoparticles in the inhibition of P. aeruginosa virulence factors is a promising strategy. This study aims to study the effect of metformin (MET), metformin nano emulsions (MET-NEs), silver metformin nano emulsions (Ag-MET-NEs) and silver nanoparticles (AgNPs) on P. aeruginosa virulence factors’ expression. The phenotypic results showed that MET-NEs had the highest virulence inhibitory activity. However, concerning RT-PCR results, all tested agents significantly decreased the expression of quorum sensing regulatory genes of P. aeruginosa; lasR, lasI, pqsA, fliC, exoS and pslA, with Ag-MET-NEs being the most potent one, however, it failed to protect mice from P. aeruginosa pathogenesis. MET-NEs showed the highest protective activity against pseudomonal infection in vivo. Our findings support the promising use of nano formulations particularly Ag-MET-NEs as an alternative against multidrug resistant pseudomonal infections via inhibition of virulence factors and quorum sensing gene expression.
Collapse
|
17
|
Kim HR, Eom YB. Auranofin promotes antibacterial effect of doripenem against carbapenem-resistant Acinetobacter baumannii. J Appl Microbiol 2022; 133:1422-1433. [PMID: 35633297 DOI: 10.1111/jam.15644] [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: 08/04/2021] [Revised: 03/10/2022] [Accepted: 05/25/2022] [Indexed: 11/29/2022]
Abstract
AIMS This study was performed to identify the potential for repurposing auranofin as an antibiotic adjuvant against carbapenemase-producing A. baumannii. METHODS AND RESULTS The clinically isolated A. baumannii strains used in this study were all resistant to carbapenems and harbored the blaOXA-23 gene. The synergistic effect of auranofin and doripenem against carbapenemase-producing A. baumannii was confirmed through checkerboard and growth kinetic analyses. This study also demonstrated the inhibitory effects of auranofin against A. baumannii biofilms. The anti-biofilm effects of auranofin were visualized by confocal laser scanning microscopy (CLSM). Furthermore, auranofin inhibited motility, one of the virulence factors. Additionally, the changes in the expression of carbapenemase-, biofilm- and efflux pump-related genes induced by auranofin were confirmed via quantitative polymerase chain reaction (qPCR). CONCLUSIONS Our results demonstrated that auranofin has an antibacterial effect with doripenem and an inhibitory effect on several factors related to carbapenem resistance. SIGNIFICANCE AND IMPACT OF THE STUDY This study suggests that auranofin is a promising antibiotic adjuvant that can be used to prevent antibiotic resistance in carbapenem-resistant A. baumannii.
Collapse
Affiliation(s)
- H-R Kim
- Department of Medical Sciences, Graduate School, Soonchunhyang University, Asan, Chungnam 31538, Republic of Korea
| | - Y-B Eom
- Department of Medical Sciences, Graduate School, Soonchunhyang University, Asan, Chungnam 31538, Republic of Korea.,Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan, Chungnam 31538, Republic of Korea
| |
Collapse
|
18
|
Folliero V, Dell’Annunziata F, Roscetto E, Cammarota M, De Filippis A, Schiraldi C, Catania MR, Casolaro V, Perrella A, Galdiero M, Franci G. Niclosamide as a Repurposing Drug against Corynebacterium striatum Multidrug-Resistant Infections. Antibiotics (Basel) 2022; 11:antibiotics11050651. [PMID: 35625295 PMCID: PMC9137567 DOI: 10.3390/antibiotics11050651] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 02/06/2023] Open
Abstract
Corynebacterium striatum (C. striatum) is an emerging multidrug-resistant (MDR) pathogen associated with nosocomial infections. In this scenario, we screened the antimicrobial activity of the anthelmintic drugs doramectin, moxidectin, selamectin and niclosamide against 20 C. striatum MDR clinical isolates. Among these, niclosamide was the best performing drug against C. striatum. Niclosamide cytotoxicity was evaluated by a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay on immortalized human keratinocyte cells (HaCaT). After 20 h of treatment, the recorded 50% cytotoxic concentration (CC50) was 2.56 μg/mL. The antibacterial efficacy was determined via disc diffusion, broth microdilution method and time-killing. Against C. striatum, niclosamide induced a growth inhibitory area of 22 mm and the minimum inhibitory concentration that inhibits 90% of bacteria (MIC90) was 0.39 μg/mL, exhibiting bactericidal action. The biofilm biomass eradicating action was investigated through crystal violet (CV), MTT and confocal laser scanning microscopy (CLSM). Niclosamide affected the biofilm viability in a dose-dependent manner and degraded biomass by 55 and 49% at 0.39 μg/mL and 0.19 μg/mL. CLSM images confirmed the biofilm biomass degradation, showing a drastic reduction in cell viability. This study could promote the drug-repurposing of the anthelmintic FDA-approved niclosamide as a therapeutic agent to counteract the C. striatum MDR infections.
Collapse
Affiliation(s)
- Veronica Folliero
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (V.F.); (F.D.); (M.C.); (A.D.F.); (C.S.)
| | - Federica Dell’Annunziata
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (V.F.); (F.D.); (M.C.); (A.D.F.); (C.S.)
| | - Emanuela Roscetto
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (E.R.); (M.R.C.)
| | - Marcella Cammarota
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (V.F.); (F.D.); (M.C.); (A.D.F.); (C.S.)
| | - Anna De Filippis
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (V.F.); (F.D.); (M.C.); (A.D.F.); (C.S.)
| | - Chiara Schiraldi
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (V.F.); (F.D.); (M.C.); (A.D.F.); (C.S.)
| | - Maria Rosaria Catania
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (E.R.); (M.R.C.)
| | - Vincenzo Casolaro
- Department of Medicine Surgery and Dentistry, University of Salerno, Baronissi, 84081 Salerno, Italy;
| | - Alessandro Perrella
- Division Emerging Infectious Disease and High Contagiousness, Hospital D Cotugno, 80131 Naples, Italy;
| | - Massimiliano Galdiero
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (V.F.); (F.D.); (M.C.); (A.D.F.); (C.S.)
- Correspondence: (M.G.); (G.F.)
| | - Gianluigi Franci
- Department of Medicine Surgery and Dentistry, University of Salerno, Baronissi, 84081 Salerno, Italy;
- Clinical Pathology and Microbiology Unit, San Giovanni di Dio e Ruggi D’Aragona University Hospital, 84126 Salerno, Italy
- Correspondence: (M.G.); (G.F.)
| |
Collapse
|
19
|
Di Bonaventura G, Lupetti V, De Fabritiis S, Piccirilli A, Porreca A, Di Nicola M, Pompilio A. Giving Drugs a Second Chance: Antibacterial and Antibiofilm Effects of Ciclopirox and Ribavirin against Cystic Fibrosis Pseudomonas aeruginosa Strains. Int J Mol Sci 2022; 23:ijms23095029. [PMID: 35563420 PMCID: PMC9102761 DOI: 10.3390/ijms23095029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
Drug repurposing is an attractive strategy for developing new antibacterial molecules. Herein, we evaluated the in vitro antibacterial, antibiofilm, and antivirulence activities of eight FDA-approved “non-antibiotic” drugs, comparatively to tobramycin, against selected Pseudomonas aeruginosa strains from cystic fibrosis patients. MIC and MBC values were measured by broth microdilution method. Time–kill kinetics was studied by the macro dilution method, and synergy studies were performed by checkerboard microdilution assay. The activity against preformed biofilms was measured by crystal violet and viable cell count assays. The effects on gene expression were studied by real-time quantitative PCR, while the cytotoxic potential was evaluated against IB3-1 bronchial CF cells. Ciclopirox, 5-fluorouracil, and actinomycin D showed the best activity against P. aeruginosa planktonic cells and therefore underwent further evaluation. Time–kill assays indicated actinomycin D and ciclopirox, contrarily to 5-fluorouracil and tobramycin, have the potential for bacterial eradication, although with strain-dependent efficacy. Ciclopirox was the most effective against the viability of the preformed biofilm. A similar activity was observed for other drugs, although they stimulate extracellular polymeric substance production. Ribavirin showed a specific antibiofilm effect, not dependent on bacterial killing. Exposure to drugs and tobramycin generally caused hyperexpression of the virulence traits tested, except for actinomycin D, which downregulated the expression of alkaline protease and alginate polymerization. Ciclopirox and actinomycin D revealed high cytotoxic potential. Ciclopirox and ribavirin might provide chemical scaffolds for anti-P. aeruginosa drugs. Further studies are warranted to decrease ciclopirox cytotoxicity and evaluate the in vivo protective effects.
Collapse
Affiliation(s)
- Giovanni Di Bonaventura
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (V.L.); (A.P.); (M.D.N.); (A.P.)
- Center of Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
- Correspondence:
| | - Veronica Lupetti
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (V.L.); (A.P.); (M.D.N.); (A.P.)
- Center of Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Simone De Fabritiis
- Center of Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
- Department of Medicine and Aging Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Alessandra Piccirilli
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Annamaria Porreca
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (V.L.); (A.P.); (M.D.N.); (A.P.)
| | - Marta Di Nicola
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (V.L.); (A.P.); (M.D.N.); (A.P.)
| | - Arianna Pompilio
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (V.L.); (A.P.); (M.D.N.); (A.P.)
- Center of Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
| |
Collapse
|
20
|
Tena-Garitaonaindia M, Ceacero-Heras D, Montoro MDMM, de Medina FS, Martínez-Augustin O, Daddaoua A. A Standardized Extract of Lentinula edodes Cultured Mycelium Inhibits Pseudomonas aeruginosa Infectivity Mechanisms. Front Microbiol 2022; 13:814448. [PMID: 35369436 PMCID: PMC8966770 DOI: 10.3389/fmicb.2022.814448] [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: 11/13/2021] [Accepted: 02/14/2022] [Indexed: 12/03/2022] Open
Abstract
The priority pathogen list of the World Health Organization classified Pseudomonas aeruginosa as the second top critical pathogen. Hence, the development of novel antibacterial strategies to tackle this bacterium is highly necessary. Herein we explore the potential antibacterial effect of a standardized extract of cultured mycelium of Lentinula edodes (AHCC®) on P. aeruginosa. AHCC® was found to inhibit the growth rate and biofilm formation of strain PAO1. No change in swarming was observed, but AHCC® hampered swimming and twitching motility. In accordance, a decreased expression of metabolism, growth, and biofilm formation genes was shown. AHCC® also diminished the levels of exotoxin A and bacteria inside IEC18 cells and the secretion of IL-6, IL-10 and TNF by infected macrophages. This effect was related to a reduced phosphorylation of MAPKs and to bacteria internalization. Taken together, our data suggest that AHCC® has a potential role to prevent P. aeruginosa infections and may lead to the development of new therapies.
Collapse
Affiliation(s)
- Mireia Tena-Garitaonaindia
- Department of Biochemistry and Molecular Biology II, Pharmacy School, University of Granada, Granada, Spain
| | - Diego Ceacero-Heras
- Department of Biochemistry and Molecular Biology II, Pharmacy School, University of Granada, Granada, Spain
| | - María Del Mar Maldonado Montoro
- Clinical Analysis Service, Hospital Campus de la Salud, Granada, Spain.,Instituto de Investigación Biosanitaria (IBS), Granada, Spain
| | - Fermín Sánchez de Medina
- Department of Pharmacology, School of Pharmacy, University of Granada, Granada, Spain.,Department of Pharmacology, Pharmacy School, University of Granada, Granada, Spain
| | - Olga Martínez-Augustin
- Department of Biochemistry and Molecular Biology II, Pharmacy School, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria (IBS), Granada, Spain.,Department of Pharmacology, Pharmacy School, University of Granada, Granada, Spain.,Institute of Nutrition and Food Technology "José Mataix," Center of Biomedical Research, University of Granada, Granada, Spain
| | - Abdelali Daddaoua
- Department of Biochemistry and Molecular Biology II, Pharmacy School, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria (IBS), Granada, Spain.,Institute of Nutrition and Food Technology "José Mataix," Center of Biomedical Research, University of Granada, Granada, Spain
| |
Collapse
|
21
|
Repurposing of Ciclopirox to Overcome the Limitations of Zidovudine (Azidothymidine) against Multidrug-Resistant Gram-Negative Bacteria. Pharmaceutics 2022; 14:pharmaceutics14030552. [PMID: 35335928 PMCID: PMC8950944 DOI: 10.3390/pharmaceutics14030552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/17/2022] [Accepted: 02/28/2022] [Indexed: 11/17/2022] Open
Abstract
Multidrug-resistant (MDR) Gram-negative bacteria are the top-priority pathogens to be eradicated. Drug repurposing (e.g., the use of non-antibiotics to treat bacterial infections) may be helpful to overcome the limitations of current antibiotics. Zidovudine (azidothymidine, AZT), a licensed oral antiviral agent, is a leading repurposed drug against MDR Gram-negative bacterial infections. However, the rapid emergence of bacterial resistance due to long-term exposure, overuse, or misuse limits its application, making it necessary to develop new alternatives. In this study, we investigated the efficacy of ciclopirox (CPX) as an alternative to AZT. The minimum inhibitory concentrations of AZT and CPX against MDR Gram-negative bacteria were determined; CPX appeared more active against β-lactamase-producing Escherichia coli, whereas AZT displayed no selectivity for any antibiotic-resistant strain. Motility assays revealed that β-lactamase-producing Escherichia coli strains were less motile in nature and more strongly affected by CPX than a parental strain. Resistance against CPX was not observed in E. coli even after 25 days of growth, whereas AZT resistance was observed in less than 2 days. Moreover, CPX effectively killed AZT-resistant strains with different resistance mechanisms. Our findings indicate that CPX may be utilized as an alternative or supplement to AZT-based medications to treat opportunistic Gram-negative bacterial infections.
Collapse
|
22
|
Olaifa K, Ajunwa O, Marsili E. Electroanalytic evaluation of antagonistic effect of azole fungicides on Acinetobacter baumannii biofilms. Electrochim Acta 2022. [DOI: 10.1016/j.electacta.2022.139837] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
|
23
|
Das R, Kotra K, Singh P, Loh B, Leptihn S, Bajpai U. Alternative Treatment Strategies for Secondary Bacterial and Fungal Infections Associated with COVID-19. Infect Dis Ther 2022; 11:53-78. [PMID: 34807451 PMCID: PMC8607056 DOI: 10.1007/s40121-021-00559-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/21/2021] [Indexed: 01/08/2023] Open
Abstract
Antimicrobials are essential for combating infectious diseases. However, an increase in resistance to them is a major cause of concern. The empirical use of drugs in managing COVID-19 and the associated secondary infections have further exacerbated the problem of antimicrobial resistance. Hence, the situation mandates exploring and developing efficient alternatives for the treatment of bacterial and fungal infections in patients suffering from COVID-19 or other viral infections. In this review, we have described the alternatives to conventional antimicrobials that have shown promising results and are at various stages of development. An acceleration of efforts to investigate their potential as therapeutics can provide more treatment options for clinical management of drug-resistant secondary bacterial and fungal infections in the current pandemic and similar potential outbreaks in the future. The alternatives include bacteriophages and their lytic enzymes, anti-fungal enzymes, antimicrobial peptides, nanoparticles and small molecule inhibitors among others. What is required at this stage is to critically examine the challenges in developing the listed compounds and biomolecules as therapeutics and to establish guidelines for their safe and effective application within a suitable time frame. In this review, we have attempted to highlight the importance of rational use of antimicrobials in patients suffering from COVID-19 and boost the deployment of alternative therapeutics.
Collapse
Affiliation(s)
- Ritam Das
- Department of Life Science, Acharya Narendra Dev College, University of Delhi, New Delhi, 110019 India
| | - Komal Kotra
- Department of Zoology, Acharya Narendra Dev College, University of Delhi, New Delhi, 110019 India
| | - Pulkit Singh
- Department of Zoology, Acharya Narendra Dev College, University of Delhi, New Delhi, 110019 India
| | - Belinda Loh
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 314400 People’s Republic of China
| | - Sebastian Leptihn
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 314400 People’s Republic of China
| | - Urmi Bajpai
- Department of Biomedical Science, Acharya Narendra Dev College, University of Delhi, Govindpuri, Kalkaji, New Delhi, 110019 India
| |
Collapse
|
24
|
Yang L, Li M, Wang Y, Zhang Y, Liu Z, Ruan S, Wang Z, Wang S. An isocamphanyl-based fluorescent "turn-on" probe for highly sensitive and selective detection of Ga 3+ and application in vivo and in vitro. Analyst 2021; 146:7294-7305. [PMID: 34749386 DOI: 10.1039/d1an01368h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A novel fluorescent probe 2-(4-(diethylamino)-2-hydroxybenzylidene)-N-(2,3,3-trimethylbicyclo[2.2.1]heptan-2-yl)hydrazinecarbothioamide (HT) was prepared in this study by a condensation reaction. HT has been confirmed to possess high specificity toward Ga3+ over other metal ions (including Al3+ and In3+) via a distinct fluorescence light-up response. Moreover, HT exhibited good detection performances for Ga3+ including high selectivity, excellent anti-interference ability, a wide working pH range, and good reversibility. The association constant and limit of detection (LOD) were calculated to be 5.34 × 103 M-1 and 1.18 × 10-6 M, respectively. The detection mechanism of HT toward Ga3+ was proposed and confirmed by 1H NMR analysis, HRMS analysis, and DFT calculations. A simple test strip-based portable detecting device and a molecular INHIBIT logic circuit were established for improving its practical applicability. Furthermore, the desirable sensing performance of HT for Ga3+ was successfully reconfirmed in MCF-7 cells and zebrafish.
Collapse
Affiliation(s)
- Lijuan Yang
- Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China.
| | - Mingxin Li
- Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China.
| | - Yunyun Wang
- Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China.
| | - Yan Zhang
- Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China.
| | - Zhipeng Liu
- College of Materials Science and Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Shutang Ruan
- Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China.
| | - Zhonglong Wang
- Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China.
| | - Shifa Wang
- Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China.
| |
Collapse
|
25
|
Yang P, Luo Y, Kurnaz LB, Bam M, Yang X, Decho AW, Nagarkatti M, Tang C. Biodegradable polycaprolactone metallopolymer-antibiotic bioconjugates containing phenylboronic acid and cobaltocenium for antimicrobial application. Biomater Sci 2021; 9:7237-7246. [PMID: 34596174 PMCID: PMC8551062 DOI: 10.1039/d1bm00970b] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
This paper reports antimicrobial metallopolymers containing biodegradable polycaprolactone as the backbone with boronic acid and cobaltocenium as the side chain. While boronic acid promotes interactions with bacterial cells via boronolectin with lipopolysaccharides, cationic cobaltocenium facilitates the unique complexation with anionic β-lactam antibiotics. The synergistic interactions in these metallopolymer-antibiotic bioconjugates were evidenced by re-sensitized efficacy of penicillin-G against four different Gram-negative bacteria (E. coli, P. vulgaris, P. aeruginosa and K. pneumoniae). The degradability of the polyester backbone was validated through tests under physiological pH (7.4) and acidic pH (5.5) or under enzymatic conditions. These metallopolymers exhibited time-dependent uptake and reduction of cobalt metals in different organs of mice via in vivo absorption, distribution, metabolism, and excretion (ADME) tests.
Collapse
Affiliation(s)
- Peng Yang
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, USA.
| | - Yuanyuan Luo
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, USA.
| | - Leman B Kurnaz
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, USA.
| | - Marpe Bam
- Department of Pathology, Microbiology and Immunology, University of South Carolina, School of Medicine, Columbia, South Carolina 29209, USA
| | - Xiaoming Yang
- Department of Pathology, Microbiology and Immunology, University of South Carolina, School of Medicine, Columbia, South Carolina 29209, USA
| | - Alan W Decho
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina 29208, USA
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina, School of Medicine, Columbia, South Carolina 29209, USA
| | - Chuanbing Tang
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, USA.
| |
Collapse
|
26
|
Rossato L, Arantes JP, Ribeiro SM, Simionatto S. Antibacterial activity of gallium nitrate against polymyxin-resistant Klebsiella pneumoniae strains. Diagn Microbiol Infect Dis 2021; 102:115569. [PMID: 34775292 DOI: 10.1016/j.diagmicrobio.2021.115569] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 07/19/2021] [Accepted: 10/02/2021] [Indexed: 11/03/2022]
Abstract
Iron uptake and metabolism have become attractive targets for the development of new antibacterial drugs. In this scenario, the FDA-approved iron mimetic metal gallium [Ga (III)] has been successfully researched as an antimicrobial drug. Ga (III) inhibits microbial growth by disrupting ferric iron-dependent metabolic pathways. In this study, we revealed that gallium nitrate III (GaN) inhibits the growth of a collection of twenty polymyxin-resistant Klebsiella pneumoniae strains at concentrations ranging from 2 to 16µg/mL, using a medium, on which the low iron content and the presence of human serum better mimic the in vivo environment. GaN was also successful in protecting Caenorhabditis elegans from polymyxin-resistant K. pneumoniae strains lethal infection, with survival rates of >75%. GaN also exhibited synergism with polymyxin B, suggesting that a polymyxin B-GaN combination holds promise like as one alternative therapy for infections caused by resistant polymyxin B K. pneumoniae strains.
Collapse
Affiliation(s)
- Luana Rossato
- Laboratório de Pesquisa em Ciências da Saúde, Universidade Federal da Grande Dourados - UFGD, Dourados, Mato Grosso do Sul, Brazil
| | - Julia Pimentel Arantes
- Laboratório de Pesquisa em Ciências da Saúde, Universidade Federal da Grande Dourados - UFGD, Dourados, Mato Grosso do Sul, Brazil
| | - Suzana Meira Ribeiro
- Laboratório de Pesquisa em Ciências da Saúde, Universidade Federal da Grande Dourados - UFGD, Dourados, Mato Grosso do Sul, Brazil
| | - Simone Simionatto
- Laboratório de Pesquisa em Ciências da Saúde, Universidade Federal da Grande Dourados - UFGD, Dourados, Mato Grosso do Sul, Brazil.
| |
Collapse
|
27
|
Pi H, Ogunniyi AD, Savaliya B, Nguyen HT, Page SW, Lacey E, Venter H, Trott DJ. Repurposing of the Fasciolicide Triclabendazole to Treat Infections Caused by Staphylococcus spp. and Vancomycin-Resistant Enterococci. Microorganisms 2021; 9:microorganisms9081697. [PMID: 34442776 PMCID: PMC8398527 DOI: 10.3390/microorganisms9081697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 12/25/2022] Open
Abstract
One approach to combat the increasing incidence of multidrug-resistant (MDR) bacterial pathogens involves repurposing existing compounds with known safety and development pathways as new antibacterial classes with potentially novel mechanisms of action. Here, triclabendazole (TCBZ), a drug originally developed to treat Fasciola hepatica (liver fluke) in sheep and cattle, and later in humans, was evaluated as an antibacterial alone or in combination with sub-inhibitory concentrations of polymyxin B (PMB) against clinical isolates and reference strains of key Gram-positive and Gram-negative bacteria. We show for the first time that in vitro, TCBZ selectively kills methicillin-sensitive and methicillin-resistant Staphylococcus aureus and Staphylococcus pseudintermedius at a minimum inhibitory concentration (MIC) range of 2–4 µg/mL, and vancomycin-resistant enterococci at a MIC range of 4–8 µg/mL. TCBZ also inhibited key Gram-negative bacteria in the presence of sub-inhibitory concentrations of PMB, returning MIC90 values of 1 µg/mL for Escherichia coli, 8 µg/mL for Klebsiella pneumoniae, 2 µg/mL for Acinetobacter baumannii and 4 µg/mL for Pseudomonasaeruginosa. Interestingly, TCBZ was found to be bacteriostatic against intracellular S. aureus but bactericidal against intracellular S. pseudintermedius. Additionally, TCBZ’s favourable pharmacokinetic (PK) and pharmacodynamic (PD) profile was further explored by in vivo safety and efficacy studies using a bioluminescent mouse model of S. aureus sepsis. We show that repeated four-hourly oral treatment of mice with 50 mg/kg TCBZ after systemic S. aureus challenge resulted in a significant reduction in S. aureus populations in the blood to 18 h post-infection (compared to untreated mice) but did not clear the bacterial infection from the bloodstream, consistent with in vivo bacteriostatic activity. These results indicate that additional pharmaceutical development of TCBZ may enhance its PK/PD, allowing it to be an appropriate candidate for the treatment of serious MDR bacterial pathogens.
Collapse
Affiliation(s)
- Hongfei Pi
- Australian Centre for Antimicrobial Resistance Ecology, Roseworthy Campus, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA 5371, Australia; (H.P.); (A.D.O.); (B.S.); (H.T.N.)
| | - Abiodun D. Ogunniyi
- Australian Centre for Antimicrobial Resistance Ecology, Roseworthy Campus, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA 5371, Australia; (H.P.); (A.D.O.); (B.S.); (H.T.N.)
| | - Bhumi Savaliya
- Australian Centre for Antimicrobial Resistance Ecology, Roseworthy Campus, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA 5371, Australia; (H.P.); (A.D.O.); (B.S.); (H.T.N.)
| | - Hang Thi Nguyen
- Australian Centre for Antimicrobial Resistance Ecology, Roseworthy Campus, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA 5371, Australia; (H.P.); (A.D.O.); (B.S.); (H.T.N.)
- Department of Pharmacology, Toxicology, Internal Medicine and Diagnostics, Faculty of Veterinary Medicine, Vietnam National University of Agriculture, Hanoi 100000, Vietnam
| | | | - Ernest Lacey
- Microbial Screening Technologies Pty Ltd., Smithfield, NSW 2164, Australia;
| | - Henrietta Venter
- Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia;
| | - Darren J. Trott
- Australian Centre for Antimicrobial Resistance Ecology, Roseworthy Campus, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA 5371, Australia; (H.P.); (A.D.O.); (B.S.); (H.T.N.)
- Correspondence:
| |
Collapse
|
28
|
Mitidieri E, Visaggio D, Frangipani E, Turnaturi C, Vanacore D, Provenzano R, Costabile G, Sorrentino R, Ungaro F, Visca P, d'Emmanuele di Villa Bianca R. Intra-tracheal administration increases gallium availability in lung: implications for antibacterial chemotherapy. Pharmacol Res 2021; 170:105698. [PMID: 34058327 DOI: 10.1016/j.phrs.2021.105698] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/11/2021] [Accepted: 05/26/2021] [Indexed: 12/01/2022]
Abstract
The emergence of pan-resistant strains in nosocomial settings underscores the urgent need of novel therapies targeting vital bacterial functions. Bacterial iron metabolism is a fascinating target for new antimicrobials. Iron mimetic metal Ga(III) has been repurposed as an antimicrobial drug, in pre-clinical studies and recent clinical studies have raised the possibility of using Ga(III) for the treatment of P. aeruginosa pulmonary infection. Ga(III) has been approved by FDA for the treatment of cancer, autoimmune and bone resorption disorders. However, some critical issues affect the therapeutic schedule of Ga(III), principally the intra-venous (i.v.) administration, and the nephrotoxicity caused by prolonged administration. Ga(III) aerosolization could represent a viable alternative for treatment of lung infections, since delivery of antimicrobial agents to the airways maximizes drug concentration at the site of infection, improves the therapeutic efficacy, and alleviates systemic toxic effects. We demonstrate the advantage of inhaled vs i.v. administered Ga(III), in terms of bio-distribution and lung acute toxicity, by using a rat model. In vivo results support the use of Ga(III) for inhalation since intra-tracheal Ga(III) delivery improved its persistence in the lung, while the i.v. administration caused rapid clearance and did not allow to attain a significant Ga(III) concentration in this organ. Moreover, local and systemic acute toxicity following intra-tracheal administration was not observed, since no significant signs of inflammation were found. At this stage of evidence, the direct administration of Ga(III) to the lung appears feasible and safe, boosting the development of Ga(III)-based drugs for inhalation therapy.
Collapse
Affiliation(s)
- Emma Mitidieri
- Department of Pharmacy, School of Medicine and Surgery, University of Napoli Federico II, Napoli, Italy
| | | | - Emanuela Frangipani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Carlotta Turnaturi
- Department of Pharmacy, School of Medicine and Surgery, University of Napoli Federico II, Napoli, Italy
| | - Domenico Vanacore
- Department of Pharmacy, School of Medicine and Surgery, University of Napoli Federico II, Napoli, Italy
| | - Romina Provenzano
- Department of Pharmacy, School of Medicine and Surgery, University of Napoli Federico II, Napoli, Italy
| | - Gabriella Costabile
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Raffaella Sorrentino
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Italy.
| | - Francesca Ungaro
- Department of Pharmacy, School of Medicine and Surgery, University of Napoli Federico II, Napoli, Italy
| | - Paolo Visca
- Department of Science, Roma Tre University, Rome, Italy
| | | |
Collapse
|
29
|
Sedlmayer F, Woischnig AK, Unterreiner V, Fuchs F, Baeschlin D, Khanna N, Fussenegger M. 5-Fluorouracil blocks quorum-sensing of biofilm-embedded methicillin-resistant Staphylococcus aureus in mice. Nucleic Acids Res 2021; 49:e73. [PMID: 33856484 PMCID: PMC8287944 DOI: 10.1093/nar/gkab251] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 03/02/2021] [Accepted: 03/29/2021] [Indexed: 02/05/2023] Open
Abstract
Antibiotic-resistant pathogens often escape antimicrobial treatment by forming protective biofilms in response to quorum-sensing communication via diffusible autoinducers. Biofilm formation by the nosocomial pathogen methicillin-resistant Staphylococcus aureus (MRSA) is triggered by the quorum-sensor autoinducer-2 (AI-2), whose biosynthesis is mediated by methylthioadenosine/S-adenosylhomocysteine nucleosidase (MTAN) and S-ribosylhomocysteine lyase (LuxS). Here, we present a high-throughput screening platform for small-molecular inhibitors of either enzyme. This platform employs a cell-based assay to report non-toxic, bioavailable and cell-penetrating inhibitors of AI-2 production, utilizing engineered human cells programmed to constitutively secrete AI-2 by tapping into the endogenous methylation cycle via ectopic expression of codon-optimized MTAN and LuxS. Screening of a library of over 5000 commercial compounds yielded 66 hits, including the FDA-licensed cytostatic anti-cancer drug 5-fluorouracil (5-FU). Secondary screening and validation studies showed that 5-FU is a potent quorum-quencher, inhibiting AI-2 production and release by MRSA, Staphylococcus epidermidis, Escherichia coli and Vibrio harveyi. 5-FU efficiently reduced adherence and blocked biofilm formation of MRSA in vitro at an order-of-magnitude-lower concentration than that clinically relevant for anti-cancer therapy. Furthermore, 5-FU reestablished antibiotic susceptibility and enabled daptomycin-mediated prevention and clearance of MRSA infection in a mouse model of human implant-associated infection.
Collapse
Affiliation(s)
- Ferdinand Sedlmayer
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Anne-Kathrin Woischnig
- Laboratory of Infection Biology, Department of Biomedicine, University and University Hospital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
| | - Vincent Unterreiner
- Novartis Institutes for BioMedical Research (NIBR), Chemical Biology and Therapeutics (CBT), CH-4033, Basel, Switzerland
| | - Florian Fuchs
- Novartis Institutes for BioMedical Research (NIBR), Chemical Biology and Therapeutics (CBT), CH-4033, Basel, Switzerland
| | - Daniel Baeschlin
- Novartis Institutes for BioMedical Research (NIBR), Chemical Biology and Therapeutics (CBT), CH-4033, Basel, Switzerland
| | - Nina Khanna
- Laboratory of Infection Biology, Department of Biomedicine, University and University Hospital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Basel, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, CH-4058 Basel, Switzerland
- Faculty of Science, University of Basel, Mattenstrasse 26, CH-4058 Basel, Switzerland
| |
Collapse
|
30
|
Nazir S, Umar Aslam Khan M, Shamsan Al-Arjan W, Izwan Abd Razak S, Javed A, Rafiq Abdul Kadir M. Nanocomposite hydrogels for melanoma skin cancer care and treatment: In-vitro drug delivery, drug release kinetics and anti-cancer activities. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2021.103120] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
31
|
Aguilar-Vega L, López-Jácome LE, Franco B, Muñoz-Carranza S, Vargas-Maya N, Franco-Cendejas R, Hernández-Durán M, Otero-Zúñiga M, Campo-Beleño C, Jiménez-Cortés JG, Martínez-Vázquez M, Rodríguez-Zavala JS, Maeda T, Zurabian R, García-Contreras R. Antibacterial properties of phenothiazine derivatives against multidrug-resistant Acinetobacter baumannii strains. J Appl Microbiol 2021; 131:2235-2243. [PMID: 33884726 DOI: 10.1111/jam.15109] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 11/29/2022]
Abstract
AIM As options to treat recalcitrant bacterial infections which are increasingly limited due to multidrug-resistant strains, searching for new, effective antibacterial compounds is necessary. One strategy is to generate treatment alternatives by drug repurposing. METHODS AND RESULTS In this work, phenotypic microarrays were used for the screening of miscellaneous compounds against the growth and biofilm formation of Acinetobacter baumannii, an important emergent multidrug-resistant opportunistic pathogen. The results showed that the phenothiazine derivatives, such as promethazine, trifluoperazine, thioridazine, and chlorpromazine, inhibited the growth of antibiotic-sensitive and multidrug-resistant strains (showing minimal inhibitory concentrations ranging from 0·05 to 0·6 g l-1 and minimal bactericidal concentrations ranging from 0·1 to 2·5 g l-1 ). All phenothiazine derivatives were active against biofilm cells (with minimal biofilm eradication concentrations ranging from 0·5 to >3 g l-1 ). Chlorpromazine promoted reactive oxigen species (ROS) production, and cell membrane and DNA damage. Chlorpromazine showed synergy with antibiotics such as ceftazidime, meropenem, and colistin and was an effective treatment for experimentally infected Galleria mellonella when combined with ceftazidime. CONCLUSIONS It was demonstrated that phenothiazine derivatives, especially chlorpromazine, are drugs with attractive antibacterial properties against nosocomial MDR strains of A. baumannii, by generating ROS and cell membrane and DNA damage. SIGNIFICANCE AND IMPACT OF THE STUDY The present study indicates that repurposing phenothiazine derivatives for treating recalcitrant infections by A. baumannii could be promising.
Collapse
Affiliation(s)
- L Aguilar-Vega
- Departamento de Microbiología y Parasitología, Facultad de Medicina, UNAM, Ciudad de México, México
| | - L E López-Jácome
- Departamento de Microbiología y Parasitología, Facultad de Medicina, UNAM, Ciudad de México, México.,División de Enfermedades Infecciosas, Instituto Nacional de Rehabilitación "Luis G. Ibarra Ibarra", Ciudad de México, México
| | - B Franco
- División de Ciencias Naturales y Exactas, Departamento de Biología, Universidad de Guanajuato, Guanajuato, México
| | - S Muñoz-Carranza
- División de Ciencias Naturales y Exactas, Departamento de Biología, Universidad de Guanajuato, Guanajuato, México
| | - N Vargas-Maya
- División de Ciencias Naturales y Exactas, Departamento de Biología, Universidad de Guanajuato, Guanajuato, México
| | - R Franco-Cendejas
- División de Enfermedades Infecciosas, Instituto Nacional de Rehabilitación "Luis G. Ibarra Ibarra", Ciudad de México, México
| | - M Hernández-Durán
- División de Enfermedades Infecciosas, Instituto Nacional de Rehabilitación "Luis G. Ibarra Ibarra", Ciudad de México, México
| | - M Otero-Zúñiga
- Departamento de Psiquiatría y Salud Mental, Facultad de Medicina, UNAM, Ciudad de México, México
| | - C Campo-Beleño
- Departamento de Microbiología y Parasitología, Facultad de Medicina, UNAM, Ciudad de México, México
| | - J G Jiménez-Cortés
- Departamento de Microbiología y Parasitología, Facultad de Medicina, UNAM, Ciudad de México, México
| | - M Martínez-Vázquez
- Departamento de Química de Productos Naturales, Instituto de Química, UNAM, Ciudad de México, México
| | - J S Rodríguez-Zavala
- Departamento de Bioquímica, Instituto Nacional de Cardiología "Ignacio Chávez", Ciudad de México, México
| | - T Maeda
- Department of Biological Functions Engineering, Kyushu Institute of Technology, Kitakyushu, Japan
| | - R Zurabian
- Departamento de Microbiología y Parasitología, Facultad de Medicina, UNAM, Ciudad de México, México
| | - R García-Contreras
- Departamento de Microbiología y Parasitología, Facultad de Medicina, UNAM, Ciudad de México, México
| |
Collapse
|
32
|
Lin Z, Gao C, Wang D, He Q. Bubble‐Propelled Janus Gallium/Zinc Micromotors for the Active Treatment of Bacterial Infections. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202016260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Zhihua Lin
- Key Laboratory of Microsystems and Microstructures Manufacturing Harbin Institute of Technology 92 West Dazhi Street Harbin 150080 China
| | - Changyong Gao
- Key Laboratory of Microsystems and Microstructures Manufacturing Harbin Institute of Technology 92 West Dazhi Street Harbin 150080 China
| | - Daolin Wang
- Key Laboratory of Microsystems and Microstructures Manufacturing Harbin Institute of Technology 92 West Dazhi Street Harbin 150080 China
| | - Qiang He
- Key Laboratory of Microsystems and Microstructures Manufacturing Harbin Institute of Technology 92 West Dazhi Street Harbin 150080 China
| |
Collapse
|
33
|
Abstract
There is an urgent need for new antibiotics and alternative strategies to combat bacterial pathogens. Molecular docking, antibacterial evaluation in vitro and in vivo, cytotoxicity assessment and enzyme inhibition analyses were performed. Compound 12 exhibited antimicrobial activity against Staphylococcus aureus (MIC: 4 μg/ml), various clinically isolated strains of MRSA (MIC: 4-16 μg/ml) and Acinetobacter baumannii (MIC: 4 μg/ml) when combined with subinhibitory concentrations of colistin B. Compound 12 (20 mg/kg) yielded mild improvement in survival of methicillin-resistant Staphylococcus aureus (MRSA)-infected mice. Additionally, enzyme inhibition tests showed that compound 12 exhibited inhibitory effects against S. aureus dihydrofolate reductase (105.1 μg/ml) and DNA gyrase (122.8 μg/ml). Compound 12 is a promising antibacterial candidate for further development.
Collapse
|
34
|
Lin Z, Gao C, Wang D, He Q. Bubble‐Propelled Janus Gallium/Zinc Micromotors for the Active Treatment of Bacterial Infections. Angew Chem Int Ed Engl 2021; 60:8750-8754. [PMID: 33481280 DOI: 10.1002/anie.202016260] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/11/2021] [Indexed: 12/13/2022]
Affiliation(s)
- Zhihua Lin
- Key Laboratory of Microsystems and Microstructures Manufacturing Harbin Institute of Technology 92 West Dazhi Street Harbin 150080 China
| | - Changyong Gao
- Key Laboratory of Microsystems and Microstructures Manufacturing Harbin Institute of Technology 92 West Dazhi Street Harbin 150080 China
| | - Daolin Wang
- Key Laboratory of Microsystems and Microstructures Manufacturing Harbin Institute of Technology 92 West Dazhi Street Harbin 150080 China
| | - Qiang He
- Key Laboratory of Microsystems and Microstructures Manufacturing Harbin Institute of Technology 92 West Dazhi Street Harbin 150080 China
| |
Collapse
|
35
|
Rosa TFD, Machado CDS, Serafin MB, Bottega A, Coelho SS, Foletto VS, Rampelotto RF, Lorenzoni VV, Mainardi A, Hörner R. Repurposing of escitalopram oxalate and clonazepam in combination with ciprofloxacin and sulfamethoxazole-trimethoprim for treatment of multidrug-resistant microorganisms and evaluation of the cleavage capacity of plasmid DNA. Can J Microbiol 2021; 67:599-612. [PMID: 33481681 DOI: 10.1139/cjm-2020-0546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Bacterial resistance has become one of the most serious public health problems, globally, and drug repurposing is being investigated to speed up the identification of effective drugs. The aim of this study was to investigate the repurposing of escitalopram oxalate and clonazepam drugs individually, and in combination with the antibiotics ciprofloxacin and sulfamethoxazole-trimethoprim, to treat multidrug-resistant (MDR) microorganisms and to evaluate the potential chemical nuclease activity. The minimum inhibitory concentration, minimum bactericidal concentration, fractional inhibitory concentration index, and tolerance level were determined for each microorganism tested. In vitro antibacterial activity was evaluated against 47 multidrug-resistant clinical isolates and 11 standard bacterial strains from the American Type Culture Collection. Escitalopram oxalate was mainly active against Gram-positive bacteria, and clonazepam was active against both Gram-positive and Gram-negative bacteria. When associated with the two antibiotics mentioned, they had a significant synergistic effect. Clonazepam cleaved plasmid DNA, and the mechanisms involved were oxidative and hydrolytic. These results indicate the potential for repurposing these non-antibiotic drugs to treat bacterial infections. However, further studies on the mechanism of action of these drugs should be performed to ensure their safe use.
Collapse
Affiliation(s)
- Taciéli F da Rosa
- Laboratory of Bacteriology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil.,Department of Clinical and Toxicological Analysis, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Catrine de S Machado
- Laboratory of Bacteriology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil.,Department of Clinical and Toxicological Analysis, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Marissa B Serafin
- Laboratory of Bacteriology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil.,Department of Clinical and Toxicological Analysis, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Angelita Bottega
- Laboratory of Bacteriology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil.,Department of Clinical and Toxicological Analysis, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Silvana S Coelho
- Laboratory of Bacteriology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil.,Department of Clinical and Toxicological Analysis, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Vitória S Foletto
- Laboratory of Bacteriology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil.,Department of Clinical and Toxicological Analysis, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Roberta F Rampelotto
- Laboratory of Bacteriology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil.,Department of Clinical and Toxicological Analysis, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Vinícius V Lorenzoni
- Laboratory of Bacteriology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil.,Department of Clinical and Toxicological Analysis, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Amanda Mainardi
- Laboratory of Bacteriology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil.,Department of Clinical and Toxicological Analysis, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Rosmari Hörner
- Laboratory of Bacteriology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil.,Department of Clinical and Toxicological Analysis, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| |
Collapse
|
36
|
Holbein BE, Ang MTC, Allan DS, Chen W, Lehmann C. Iron-withdrawing anti-infectives for new host-directed therapies based on iron dependence, the Achilles' heel of antibiotic-resistant microbes. ENVIRONMENTAL CHEMISTRY LETTERS 2021; 19:2789-2808. [PMID: 33907538 PMCID: PMC8062846 DOI: 10.1007/s10311-021-01242-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/13/2021] [Indexed: 05/02/2023]
Abstract
The iron dependence of antibiotic-resistant microbes represents an Achilles' heel that can be exploited broadly. The growing global problem of antibiotic resistance of microbial pathogens wherein microbes become resistant to the very antibiotics used against them during infection is linked not only to our health uses but also to agribusiness practices and the changing environment. Here we review mechanisms of microbial iron acquisition and host iron withdrawal defense, and the influence of iron withdrawal on the antimicrobial activity of antibiotics. Antibiotic-resistant microbes are unaltered in their iron requirements, but iron withdrawal from microbes enhances the activities of various antibiotics and importantly suppresses outgrowth of antibiotic-exposed resistant microbial survivors. Of the three therapeutic approaches available to exploit microbial iron susceptibility, including (1) use of gallium as a non-functional iron analogue, (2) Trojan horse conjugates of microbial siderophores carrying antibiotics, and (3) new generation iron chelators, purposely designed as anti-microbials, the latter offers various advantages. For instance, these novel anti-microbial chelators overcome the limitations of conventional clinically-used hematological chelators which display host toxicity and are not useful antimicrobials. 3-Hydroxypyridin-4-one-containing polymeric chelators appear to have the highest potential. DIBI (developmental code name) is a well-developed lead candidate, being a low molecular weight, water-soluble copolymer with enhanced iron binding characteristics, strong anti-microbial and anti-inflammatory activities, low toxicity for animals and demonstrated freedom from microbial resistance development. DIBI has been shown to enhance antibiotic efficacy for antibiotic-resistant microbes during infection, and it also prevents recovery growth and resistance development during microbe exposure to various antibiotics. Because DIBI bolsters innate iron withdrawal defenses of the infected host, it has potential to provide a host-directed anti-infective therapy.
Collapse
Affiliation(s)
- Bruce E. Holbein
- Chelation Partners Inc., #58, The Labs at Innovacorp, Life Sciences Research Institute, 1344 Summer Street, Halifax, NS B3H OA8 Canada
- Department of Microbiology and Immunology, Dalhousie University, 5859 College St., Halifax, NS B3H 1X5 Canada
| | - M. Trisha C. Ang
- Chelation Partners Inc., #58, The Labs at Innovacorp, Life Sciences Research Institute, 1344 Summer Street, Halifax, NS B3H OA8 Canada
| | - David S. Allan
- Chelation Partners Inc., #58, The Labs at Innovacorp, Life Sciences Research Institute, 1344 Summer Street, Halifax, NS B3H OA8 Canada
| | - Wangxue Chen
- Human Health Therapeutics Research Center, National Research Council Canada, 100 Sussex Drive, Ottawa, ON K1A 0R6 Canada
| | - Christian Lehmann
- Department of Microbiology and Immunology, Dalhousie University, 5859 College St., Halifax, NS B3H 1X5 Canada
- Department of Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS Canada
| |
Collapse
|
37
|
Baldelli V, D’Angelo F, Pavoncello V, Fiscarelli EV, Visca P, Rampioni G, Leoni L. Identification of FDA-approved antivirulence drugs targeting the Pseudomonas aeruginosa quorum sensing effector protein PqsE. Virulence 2020; 11:652-668. [PMID: 32423284 PMCID: PMC7549961 DOI: 10.1080/21505594.2020.1770508] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/03/2020] [Accepted: 05/05/2020] [Indexed: 12/13/2022] Open
Abstract
The ability of the bacterial pathogen Pseudomonas aeruginosa to cause both chronic and acute infections mainly relies on its capacity to finely modulate the expression of virulence factors through a complex network of regulatory circuits, including the pqs quorum sensing (QS) system. While in most QS systems the signal molecule/receptor complexes act as global regulators that modulate the expression of QS-controlled genes, the main effector protein of the pqs system is PqsE. This protein is involved in the synthesis of the QS signal molecules 2-alkyl-4(1H)-quinolones (AQs), but it also modulates the expression of genes involved in virulence factors production and biofilm formation via AQ-independent pathway(s). P. aeruginosa pqsE mutants disclose attenuated virulence in plant and animal infection models, hence PqsE is considered a good target for the development of antivirulence drugs against P. aeruginosa. In this study, the negative regulation exerted by PqsE on its own transcription has been exploited to develop a screening system for the identification of PqsE inhibitors in a library of FDA-approved drugs. This led to the identification of nitrofurazone and erythromycin estolate, two antibiotic compounds that reduce the expression of PqsE-dependent virulence traits and biofilm formation in the model strain P. aeruginosa PAO1 at concentrations far below those affecting the bacterial growth rate. Notably, both drugs reduce the production of the PqsE-controlled virulence factor pyocyanin also in P. aeruginosa strains isolated from cystic fibrosis patients, and do not antagonize the activity of antibiotics commonly used to treat P. aeruginosa infection.
Collapse
Affiliation(s)
| | | | | | | | - Paolo Visca
- Department of Science, University Roma Tre, Rome, Italy
| | | | - Livia Leoni
- Department of Science, University Roma Tre, Rome, Italy
| |
Collapse
|
38
|
Gilbert-Girard S, Savijoki K, Yli-Kauhaluoma J, Fallarero A. Screening of FDA-Approved Drugs Using a 384-Well Plate-Based Biofilm Platform: The Case of Fingolimod. Microorganisms 2020; 8:microorganisms8111834. [PMID: 33233348 PMCID: PMC7700524 DOI: 10.3390/microorganisms8111834] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/12/2020] [Accepted: 11/20/2020] [Indexed: 12/19/2022] Open
Abstract
In an effort to find new repurposed antibacterial compounds, we performed the screening of an FDA-approved compounds library against Staphylococcus aureus American Type Culture Collection (ATCC) 25923. Compounds were evaluated for their capacity to prevent both planktonic growth and biofilm formation as well as to disrupt pre-formed biofilms. One of the identified initial hits was fingolimod (FTY720), an immunomodulator approved for the treatment of multiple sclerosis, which was then selected for follow-up studies. Fingolimod displayed a potent activity against S. aureus and S. epidermidis with a minimum inhibitory concentration (MIC) within the range of 12–15 µM at which concentration killing of all the bacteria was confirmed. A time–kill kinetic study revealed that fingolimod started to drastically reduce the viable bacterial count within two hours and we showed that no resistance developed against this compound for up to 20 days. Fingolimod also displayed a high activity against Acinetobacter baumannii (MIC 25 µM) as well as a modest activity against Escherichia coli and Pseudomonas aeruginosa. In addition, fingolimod inhibited quorum sensing in Chromobacterium violaceum and might therefore target this signaling pathway in certain Gram-negative bacteria. In conclusion, we present the identification of fingolimod from a compound library and its evaluation as a potential repurposed antibacterial compound.
Collapse
Affiliation(s)
- Shella Gilbert-Girard
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland; (K.S.); (A.F.)
- Correspondence:
| | - Kirsi Savijoki
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland; (K.S.); (A.F.)
| | - Jari Yli-Kauhaluoma
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland;
| | - Adyary Fallarero
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland; (K.S.); (A.F.)
| |
Collapse
|
39
|
Centola G, Deredge DJ, Hom K, Ai Y, Dent AT, Xue F, Wilks A. Gallium(III)-Salophen as a Dual Inhibitor of Pseudomonas aeruginosa Heme Sensing and Iron Acquisition. ACS Infect Dis 2020; 6:2073-2085. [PMID: 32551497 DOI: 10.1021/acsinfecdis.0c00138] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pseudomonas aeruginosa is an opportunistic bacterium that causes life-threatening infections in immunocompromised patients. In infection, it uses heme as a primary iron source and senses the availability of exogenous heme through the heme assimilation system (Has), an extra cytoplasmic function σ-factor system. A secreted hemophore HasAp scavenges heme and, upon interaction with the outer-membrane receptor HasR, activates a signaling cascade, which in turn creates a positive feedback loop critical for sensing and adaptation within the host. The ability to sense and respond to heme as an iron source contributes to virulence. Consequently, the inhibition of this system will lead to a disruption in iron homeostasis, decreasing virulence. We have identified a salophen scaffold that successfully inhibits the activation of the Has signaling system while simultaneously targeting iron uptake via xenosiderophore receptors. We propose this dual mechanism wherein free Ga3+-salophen reduces growth through uptake and iron mimicry. A dual mechanism targeting extracellular heme signaling and uptake together with Ga3+-induced toxicity following active Ga3+salophen uptake provides a significant therapeutic advantage while reducing the propensity to develop resistance.
Collapse
Affiliation(s)
- Garrick Centola
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Daniel J. Deredge
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Kellie Hom
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Yong Ai
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Alecia T. Dent
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Fengtian Xue
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Angela Wilks
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| |
Collapse
|
40
|
Weber BS, De Jong AM, Guo AB, Dharavath S, French S, Fiebig-Comyn AA, Coombes BK, Magolan J, Brown ED. Genetic and Chemical Screening in Human Blood Serum Reveals Unique Antibacterial Targets and Compounds against Klebsiella pneumoniae. Cell Rep 2020; 32:107927. [DOI: 10.1016/j.celrep.2020.107927] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 04/30/2020] [Accepted: 06/26/2020] [Indexed: 12/15/2022] Open
|
41
|
Marchetti M, De Bei O, Bettati S, Campanini B, Kovachka S, Gianquinto E, Spyrakis F, Ronda L. Iron Metabolism at the Interface between Host and Pathogen: From Nutritional Immunity to Antibacterial Development. Int J Mol Sci 2020; 21:E2145. [PMID: 32245010 PMCID: PMC7139808 DOI: 10.3390/ijms21062145] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 02/08/2023] Open
Abstract
Nutritional immunity is a form of innate immunity widespread in both vertebrates and invertebrates. The term refers to a rich repertoire of mechanisms set up by the host to inhibit bacterial proliferation by sequestering trace minerals (mainly iron, but also zinc and manganese). This strategy, selected by evolution, represents an effective front-line defense against pathogens and has thus inspired the exploitation of iron restriction in the development of innovative antimicrobials or enhancers of antimicrobial therapy. This review focuses on the mechanisms of nutritional immunity, the strategies adopted by opportunistic human pathogen Staphylococcus aureus to circumvent it, and the impact of deletion mutants on the fitness, infectivity, and persistence inside the host. This information finally converges in an overview of the current development of inhibitors targeting the different stages of iron uptake, an as-yet unexploited target in the field of antistaphylococcal drug discovery.
Collapse
Affiliation(s)
- Marialaura Marchetti
- Interdepartmental Center Biopharmanet-TEC, University of Parma, 43124 Parma, Italy; (M.M.); (S.B.)
| | - Omar De Bei
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (O.D.B.); (B.C.)
| | - Stefano Bettati
- Interdepartmental Center Biopharmanet-TEC, University of Parma, 43124 Parma, Italy; (M.M.); (S.B.)
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Institute of Biophysics, National Research Council, 56124 Pisa, Italy
- National Institute of Biostructures and Biosystems, 00136 Rome, Italy
| | - Barbara Campanini
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (O.D.B.); (B.C.)
| | - Sandra Kovachka
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (S.K.); (E.G.); (F.S.)
| | - Eleonora Gianquinto
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (S.K.); (E.G.); (F.S.)
| | - Francesca Spyrakis
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy; (S.K.); (E.G.); (F.S.)
| | - Luca Ronda
- Interdepartmental Center Biopharmanet-TEC, University of Parma, 43124 Parma, Italy; (M.M.); (S.B.)
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Institute of Biophysics, National Research Council, 56124 Pisa, Italy
| |
Collapse
|
42
|
Howard KC, Dennis EK, Watt DS, Garneau-Tsodikova S. A comprehensive overview of the medicinal chemistry of antifungal drugs: perspectives and promise. Chem Soc Rev 2020; 49:2426-2480. [PMID: 32140691 DOI: 10.1039/c9cs00556k] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The emergence of new fungal pathogens makes the development of new antifungal drugs a medical imperative that in recent years motivates the talents of numerous investigators across the world. Understanding not only the structural families of these drugs but also their biological targets provides a rational means for evaluating the merits and selectivity of new agents for fungal pathogens and normal cells. An equally important aspect of modern antifungal drug development takes a balanced look at the problems of drug potency and drug resistance. The future development of new antifungal agents will rest with those who employ synthetic and semisynthetic methodology as well as natural product isolation to tackle these problems and with those who possess a clear understanding of fungal cell architecture and drug resistance mechanisms. This review endeavors to provide an introduction to a growing and increasingly important literature, including coverage of the new developments in medicinal chemistry since 2015, and also endeavors to spark the curiosity of investigators who might enter this fascinatingly complex fungal landscape.
Collapse
Affiliation(s)
- Kaitlind C Howard
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596, USA.
| | | | | | | |
Collapse
|
43
|
Strategies to Combat Multidrug-Resistant and Persistent Infectious Diseases. Antibiotics (Basel) 2020; 9:antibiotics9020065. [PMID: 32041137 PMCID: PMC7168131 DOI: 10.3390/antibiotics9020065] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/16/2020] [Accepted: 02/01/2020] [Indexed: 12/21/2022] Open
Abstract
Antibiotic failure is one of the most worrying health problems worldwide. We are currently facing an international crisis with several problematic facets: new antibiotics are no longer being discovered, resistance mechanisms are occurring in almost all clinical isolates of bacteria, and recurrent infections caused by persistent bacteria are hampering the successful treatment of infections. In this context, new anti-infectious strategies against multidrug-resistant (MDR) and persistent bacteria, as well as the rescue of Food and Drug Administration (FDA)-approved compounds (drug repurposing), are being explored. Among the highlighted new anti-infectious strategies, in this review, we focus on antimicrobial peptides, anti-virulence compounds, phage therapy, and new molecules. As drugs that are being repurposed, we highlight anti-inflammatory compounds, anti-psychotics, anti-helminthics, anti-cancerous drugs, and statins.
Collapse
|
44
|
Foerster S, Gustafsson TN, Brochado AR, Desilvestro V, Typas A, Unemo M. The first wide-scale drug repurposing screen using the Prestwick Chemical Library (1200 bioactive molecules) against Neisseria gonorrhoeae identifies high in vitro activity of auranofin and many additional drugs. APMIS 2020; 128:242-250. [PMID: 31811739 DOI: 10.1111/apm.13014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/18/2019] [Indexed: 12/22/2022]
Abstract
Treatment options for gonorrhoea are scarce. Drug repurposing of bioactive molecules approved for other conditions might therefore be of value. We developed a method for wide-scale, systematic drug repurposing screen to identify molecules with activity against Neisseria gonorrhoeae and screened the Prestwick Chemical Library (1200 FDA-approved drugs). As a proof-of-concept, we further examined one promising and interesting screening hit (auranofin; antirheumatic agent). Three WHO gonococcal reference strains (WHO F, O, P) were used for the Library screening. The strains were grown in presence of a fixed concentration of the library drugs in 384-well plates for 12 h, and the remaining bacterial respiration, to reflect growth, was then quantitatively measured using optical density (OD) 450 nm and a resazurin assay. The activity of auranofin was further examined using in vitro susceptibility testing (minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC)) against genetically diverse antimicrobial-resistant N. gonorrhoeae strains and time-kill assays. Sixty-eight molecules significantly inhibited bacterial growth of WHO F, O and P. Auranofin showed potent in vitro bactericidal activity (in MIC-, MBC- and time-kill assays) against four WHO reference strains. No cross-resistance between auranofin and any antimicrobial currently or previously used for gonorrhoea treatment was found when examining 51 selected antimicrobial-resistant gonococcal strains. In conclusion, this is the first wide-scale systematic screening effort for repurposing drugs for future treatment of gonorrhoea. Additional studies examining mechanism(s) of action, resistance development, in vivo anti-gonococcal activity and pharmacokinetics/pharmacodynamics for gonococcal infections of auranofin and several other significant screening hits would be valuable.
Collapse
Affiliation(s)
- Sunniva Foerster
- WHO Collaborating Centre for Gonorrhoea and other STIs, Department of Laboratory Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden.,European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Tomas N Gustafsson
- Department of Clinical Microbiology, Sunderby Research Unit, Umeå University, Umeå, Sweden
| | - Anna Rita Brochado
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | | | - Athanasios Typas
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Magnus Unemo
- WHO Collaborating Centre for Gonorrhoea and other STIs, Department of Laboratory Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
45
|
Vlad IM, Nuta DC, Chirita C, Caproiu MT, Draghici C, Dumitrascu F, Bleotu C, Avram S, Udrea AM, Missir AV, Marutescu LG, Limban C. In Silico and In Vitro Experimental Studies of New Dibenz[ b, e]oxepin-11(6 H)one O-(arylcarbamoyl)-oximes Designed as Potential Antimicrobial Agents. Molecules 2020; 25:molecules25020321. [PMID: 31941125 PMCID: PMC7024208 DOI: 10.3390/molecules25020321] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 12/24/2019] [Accepted: 01/06/2020] [Indexed: 01/28/2023] Open
Abstract
In a drug-repurposing-driven approach for speeding up the development of novel antimicrobial agents, this paper presents for the first time in the scientific literature the synthesis, physico-chemical characterization, in silico analysis, antimicrobial activity against bacterial and fungal strains in planktonic and biofilm growth state, as well as the in vitro cytotoxicity of some new 6,11-dihydrodibenz[b,e]oxepin-11(6H)one O-(arylcarbamoyl)oximes. The structures of intermediary and final substances (compounds 7a–j) were confirmed by 1H-NMR, 13C-NMR and IR spectra, as well as by elemental analysis. The in silico bioinformatic and cheminformatic studies evidenced an optimal pharmacokinetic profile for the synthesized compounds 7a–j, characterized by an average lipophilic character predicting good cell membrane permeability and intestinal absorption; low maximum tolerated dose for humans; potassium channels encoded by the hERG I and II genes as potential targets and no carcinogenic effects. The obtained compounds exhibited a higher antimicrobial activity against the planktonic Gram-positive Staphylococcus aureus and Bacillus subtilis strains and the Candida albicans fungal strain. The obtained compounds also inhibited the ability of S. aureus, B. subtilis, Escherichia coli and C. albicans strains to colonize the inert substratum, accounting for their possible use as antibiofilm agents. All the active compounds exhibited low or acceptable cytotoxicity levels on the HCT8 cells, ensuring the potential use of these compounds for the development of new antimicrobial drugs with minimal side effects on the human cells and tissues.
Collapse
Affiliation(s)
- Ilinca Margareta Vlad
- Department of Pharmaceutical Chemistry, University of Medicine and Pharmacy “Carol Davila”, Traian Vuia 6, sect. 2, 020956 Bucharest, Romania; (I.M.V.); (D.C.N.); (A.V.M.); (C.L.)
| | - Diana Camelia Nuta
- Department of Pharmaceutical Chemistry, University of Medicine and Pharmacy “Carol Davila”, Traian Vuia 6, sect. 2, 020956 Bucharest, Romania; (I.M.V.); (D.C.N.); (A.V.M.); (C.L.)
| | - Cornel Chirita
- Department of Pharmacology and Clinical Pharmacy, University of Medicine and Pharmacy “Carol Davila”, Traian Vuia 6, sect. 2, 020956 Bucharest, Romania;
| | - Miron Teodor Caproiu
- The Organic Chemistry Center of Romanian Academy “Costin D. Nenitescu”, Splaiul Independenței 202B, 77208 Bucharest, Romania; (M.T.C.); (C.D.); (F.D.)
| | - Constantin Draghici
- The Organic Chemistry Center of Romanian Academy “Costin D. Nenitescu”, Splaiul Independenței 202B, 77208 Bucharest, Romania; (M.T.C.); (C.D.); (F.D.)
| | - Florea Dumitrascu
- The Organic Chemistry Center of Romanian Academy “Costin D. Nenitescu”, Splaiul Independenței 202B, 77208 Bucharest, Romania; (M.T.C.); (C.D.); (F.D.)
| | - Coralia Bleotu
- Ștefan S Nicolau Institute of Virology, Romanian Academy, 285 Mihai Bravu Avenue, 030304 Bucharest, Romania
- Research Institute of the University of Bucharest (ICUB) and Microbiology Immunology Department, Faculty of Biology, University of Bucharest, 1-3 Ale. Portocalelor, 060101 Bucharest, Romania;
- Correspondence: ; Tel.: +40-213242590
| | - Speranța Avram
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, 1-3 Ale. Portocalelor, 060101 Bucharest, Romania; (S.A.); (A.M.U.)
| | - Ana Maria Udrea
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, 1-3 Ale. Portocalelor, 060101 Bucharest, Romania; (S.A.); (A.M.U.)
- Laser Department, National Institute for Laser, Plasma and Radiation Physics, Atomistilor Street 409, 077125 Magurele, Romania
| | - Alexandru Vasile Missir
- Department of Pharmaceutical Chemistry, University of Medicine and Pharmacy “Carol Davila”, Traian Vuia 6, sect. 2, 020956 Bucharest, Romania; (I.M.V.); (D.C.N.); (A.V.M.); (C.L.)
| | - Luminita Gabriela Marutescu
- Research Institute of the University of Bucharest (ICUB) and Microbiology Immunology Department, Faculty of Biology, University of Bucharest, 1-3 Ale. Portocalelor, 060101 Bucharest, Romania;
| | - Carmen Limban
- Department of Pharmaceutical Chemistry, University of Medicine and Pharmacy “Carol Davila”, Traian Vuia 6, sect. 2, 020956 Bucharest, Romania; (I.M.V.); (D.C.N.); (A.V.M.); (C.L.)
| |
Collapse
|
46
|
In-Silico Repurposing of Anticancer Drug (5-FU) as an Antimicrobial Agent Against Methicillin-Resistant Staphylococcus aureus (MRSA). Int J Pept Res Ther 2020. [DOI: 10.1007/s10989-019-10010-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
47
|
Konreddy AK, Rani GU, Lee K, Choi Y. Recent Drug-Repurposing-Driven Advances in the Discovery of Novel Antibiotics. Curr Med Chem 2019; 26:5363-5388. [PMID: 29984648 DOI: 10.2174/0929867325666180706101404] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/26/2018] [Accepted: 05/03/2018] [Indexed: 12/18/2022]
Abstract
Drug repurposing is a safe and successful pathway to speed up the novel drug discovery and development processes compared with de novo drug discovery approaches. Drug repurposing uses FDA-approved drugs and drugs that failed in clinical trials, which have detailed information on potential toxicity, formulation, and pharmacology. Technical advancements in the informatics, genomics, and biological sciences account for the major success of drug repurposing in identifying secondary indications of existing drugs. Drug repurposing is playing a vital role in filling the gap in the discovery of potential antibiotics. Bacterial infections emerged as an ever-increasing global public health threat by dint of multidrug resistance to existing drugs. This raises the urgent need of development of new antibiotics that can effectively fight multidrug-resistant bacterial infections (MDRBIs). The present review describes the key role of drug repurposing in the development of antibiotics during 2016-2017 and of the details of recently FDA-approved antibiotics, pipeline antibiotics, and antibacterial properties of various FDA-approved drugs of anti-cancer, anti-fungal, anti-hyperlipidemia, antiinflammatory, anti-malarial, anti-parasitic, anti-viral, genetic disorder, immune modulator, etc. Further, in view of combination therapies with the existing antibiotics, their potential for new implications for MDRBIs is discussed. The current review may provide essential data for the development of quick, safe, effective, and novel antibiotics for current needs and suggest acuity in its effective implications for inhibiting MDRBIs by repurposing existing drugs.
Collapse
Affiliation(s)
- Ananda Kumar Konreddy
- College of Life Sciences and Biotechnology, Korea University, Seoul 136- 713, South Korea
| | - Grandhe Usha Rani
- College of Pharmacy, Dongguk University-Seoul, Goyang 410-820, South Korea
| | - Kyeong Lee
- College of Pharmacy, Dongguk University-Seoul, Goyang 410-820, South Korea
| | - Yongseok Choi
- College of Life Sciences and Biotechnology, Korea University, Seoul 136- 713, South Korea
| |
Collapse
|
48
|
Maddila S, Gorle S, Jonnalagadda SB. Drug screening of rhodanine derivatives for antibacterial activity. Expert Opin Drug Discov 2019; 15:203-229. [PMID: 31777321 DOI: 10.1080/17460441.2020.1696768] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Bacteriological infections are a major risk to human health. These include all hospital and public-acquired infections. In drug discovery, rhodanines are privileged heterocyclic frameworks. Their derivatives possess strong anti-bacterial activity and some of them have shown potent activity against multidrug-resistant pathogens, both under in vitro and in vivo conditions. To treat multi-drug resistant pathogens, the development of novel potent drugs, with superior anti-bacterial efficacy, is paramount. One avenue which shows promise is the design and development of novel rhodanines.Areas covered: This review summarizes the status on rhodanine-based derivatives and their anti-bacterial activity, based on published research over the past six years. Furthermore, to facilitate the design of novel derivatives with improved functions, their structure-activity relationships are assessed with reference to their efficacy as anti-bacterial agents and their toxicity.Expert opinion: The pharmacological activity of molecules bearing a rhodanine scaffold needs to be very critically assessed in spite of considerable information available from various biological evaluations. Although, some data on structure-activity relationship frameworks is available, information is not adequate to optimize the efficacy of rhodanine derivatives for different applications.
Collapse
Affiliation(s)
- Suresh Maddila
- School of Chemistry & Physics, University of KwaZulu-Natal, Westville Campus, Durban, South Africa.,Department of Chemistry, GITAM Institute of Sciences, GITAM University, Visakhapatnam, India
| | - Sridevi Gorle
- Department of Microbiology and Food Science & Technology, GITAM Institute of Sciences, GITAM University, Visakhapatnam, India
| | | |
Collapse
|
49
|
Pogodin PV, Lagunin AA, Rudik AV, Druzhilovskiy DS, Filimonov DA, Poroikov VV. AntiBac-Pred: A Web Application for Predicting Antibacterial Activity of Chemical Compounds. J Chem Inf Model 2019; 59:4513-4518. [PMID: 31661960 DOI: 10.1021/acs.jcim.9b00436] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Discovery of new antibacterial agents is a never-ending task of medicinal chemistry. Every new drug brings significant improvement to patients with bacterial infections, but prolonged usage of antibacterials leads to the emergence of resistant strains. Therefore, novel active structures with new modes of action are required. We describe a web application called AntiBac-Pred aimed to help users in the rational selection of the chemical compounds for experimental studies of antibacterial activity. This application is developed using antibacterial activity data available in ChEMBL and PASS software. It allows users to classify chemical structures of interest into growth inhibitors or noninhibitors of 353 different bacteria strains, including both resistant and nonresistant ones.
Collapse
Affiliation(s)
- Pavel V Pogodin
- Department for Bioinformatics , Institute of Biomedical Chemistry (IBMC) , Moscow 119121 , Russia
| | - Alexey A Lagunin
- Department for Bioinformatics , Institute of Biomedical Chemistry (IBMC) , Moscow 119121 , Russia.,Department of Bioinformatics , Pirogov Russian National Research Medical University , Moscow 117997 , Russia
| | - Anastasia V Rudik
- Department for Bioinformatics , Institute of Biomedical Chemistry (IBMC) , Moscow 119121 , Russia
| | - Dmitry S Druzhilovskiy
- Department for Bioinformatics , Institute of Biomedical Chemistry (IBMC) , Moscow 119121 , Russia
| | - Dmitry A Filimonov
- Department for Bioinformatics , Institute of Biomedical Chemistry (IBMC) , Moscow 119121 , Russia
| | - Vladimir V Poroikov
- Department for Bioinformatics , Institute of Biomedical Chemistry (IBMC) , Moscow 119121 , Russia
| |
Collapse
|
50
|
Mellini M, Di Muzio E, D’Angelo F, Baldelli V, Ferrillo S, Visca P, Leoni L, Polticelli F, Rampioni G. In silico Selection and Experimental Validation of FDA-Approved Drugs as Anti-quorum Sensing Agents. Front Microbiol 2019; 10:2355. [PMID: 31649658 PMCID: PMC6796623 DOI: 10.3389/fmicb.2019.02355] [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: 08/01/2019] [Accepted: 09/27/2019] [Indexed: 12/13/2022] Open
Abstract
The emergence of antibiotic resistant bacterial pathogens is increasing at an unprecedented pace, calling for the development of new therapeutic options. Small molecules interfering with virulence processes rather than growth hold promise as an alternative to conventional antibiotics. Anti-virulence agents are expected to decrease bacterial virulence and to pose reduced selective pressure for the emergence of resistance. In the opportunistic pathogen Pseudomonas aeruginosa the expression of key virulence traits is controlled by quorum sensing (QS), an intercellular communication process that coordinates gene expression at the population level. Hence, QS inhibitors represent promising anti-virulence agents against P. aeruginosa. Virtual screenings allow fast and cost-effective selection of target ligands among vast libraries of molecules, thus accelerating the time and limiting the cost of conventional drug-discovery processes, while the drug-repurposing approach is based on the identification of off-target activity of FDA-approved drugs, likely endowed with low cytotoxicity and favorable pharmacological properties. This study aims at combining the advantages of virtual screening and drug-repurposing approaches to identify new QS inhibitors targeting the pqs QS system of P. aeruginosa. An in silico library of 1,467 FDA-approved drugs has been screened by molecular docking, and 5 hits showing the highest predicted binding affinity for the pqs QS receptor PqsR (also known as MvfR) have been selected. In vitro experiments have been performed by engineering ad hoc biosensor strains, which were used to verify the ability of hit compounds to decrease PqsR activity in P. aeruginosa. Phenotypic analyses confirmed the impact of the most promising hit, the antipsychotic drug pimozide, on the expression of P. aeruginosa PqsR-controlled virulence traits. Overall, this study highlights the potential of virtual screening campaigns of FDA-approved drugs to rapidly select new inhibitors of important bacterial functions.
Collapse
Affiliation(s)
- Marta Mellini
- Department of Science, University Roma Tre, Rome, Italy
| | | | | | | | | | - Paolo Visca
- Department of Science, University Roma Tre, Rome, Italy
| | - Livia Leoni
- Department of Science, University Roma Tre, Rome, Italy
| | - Fabio Polticelli
- Department of Science, University Roma Tre, Rome, Italy
- National Institute of Nuclear Physics, Roma Tre Section, Rome, Italy
| | | |
Collapse
|