1
|
Xie Y, Liu J, Ma J, Shi N, Zhang X. Excavation of resources of Streptomyces species in frozen soils of the Qinghai-Tibet Plateau based on RpfA protein of Streptomyces coelicolor. Front Microbiol 2025; 16:1557511. [PMID: 40264977 PMCID: PMC12011840 DOI: 10.3389/fmicb.2025.1557511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/21/2025] [Indexed: 04/24/2025] Open
Abstract
This study is aimed at the actual demand for exploring new species resources of Streptomyces, and aims to solve the technical bottleneck of Streptomyces isolation and culture. A new method was established based on the resuscitation function of the RpfA protein from Streptomyces coelicolor CGMCC 4.1658T to isolate unculturable or difficult-to-culture Streptomyces species, and it was applied to explore Streptomyces species resources in special habitats in the frozen soils of the Qinghai-Tibet Plateau. The RpfA protein of S. coelicolor was heterologously expressed and validated for its in vitro activity. The purified RpfA protein was then used to isolate Streptomyces from soil samples in the frozen soils of the Qinghai-Tibet Plateau, followed by an investigation into the impact of the RpfA protein on the cultivability of Streptomyces species. The results showed that the RpfA protein had a significant promoting effect on the germination of spores of both S. coelicolor itself and other species of the Streptomyces genus, and when a suitable concentration of RpfA protein was added to the culture medium, it could significantly improve the culturability of members of phylum Actinomycetota, especially Streptomyces species. In addition, many new species of the genus Streptomyces and other genera of phylum Actinomycetota were discovered. This study provides a new approach for further exploring Streptomyces species resources in special environments such as the Qinghai-Tibet Plateau and developing new biologically active substances produced by Streptomyces.
Collapse
Affiliation(s)
- Yuxiao Xie
- College of Life Sciences, Hebei University, Baoding, China
| | - Jingjing Liu
- College of Life Sciences, Hebei University, Baoding, China
| | - Jun Ma
- College of Life Sciences, Hebei University, Baoding, China
| | - Nan Shi
- College of Life Sciences, Hebei University, Baoding, China
- Key Laboratory of Microbial Diversity Research and Application of Hebei Province, Baoding, China
- Engineering Research Center of Microbial Breeding and Conservation, Baoding, China
| | - Xiumin Zhang
- College of Life Sciences, Hebei University, Baoding, China
- Key Laboratory of Microbial Diversity Research and Application of Hebei Province, Baoding, China
- Engineering Research Center of Microbial Breeding and Conservation, Baoding, China
| |
Collapse
|
2
|
Avci FG. Unraveling bacterial stress responses: implications for next-generation antimicrobial solutions. World J Microbiol Biotechnol 2024; 40:285. [PMID: 39073503 PMCID: PMC11286680 DOI: 10.1007/s11274-024-04090-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024]
Abstract
The accelerated spread of antimicrobial-resistant bacteria has caused a serious health problem and rendered antimicrobial treatments ineffective. Innovative approaches are crucial to overcome the health threat posed by resistant pathogens and prevent the emergence of untreatable infections. Triggering stress responses in bacteria can diminish susceptibility to various antimicrobials by inducing resistance mechanisms. Therefore, a thorough understanding of stress response control, especially in relation to antimicrobial resistance, offers valuable perspectives for innovative and efficient therapeutic approaches to combat antimicrobial resistance. The aim of this study was to evaluate the stress responses of 8 different bacteria by analyzing reporter metabolites, around which significant alterations were observed, using a pathway-driven computational approach. For this purpose, the transcriptomic data that the bacterial pathogens were grown under 11 different stress conditions mimicking the human host environments were integrated with the genome-scale metabolic models of 8 pathogenic species (Enterococcus faecalis OG1R, Escherichia coli EPEC O127:H6 E2348/69, Escherichia coli ETEC H10407, Escherichia coli UPEC 536, Klebsiella pneumoniae MGH 78578, Pseudomonas aeruginosa PAO1, Staphylococcus aureus MRSA252, and Staphylococcus aureus MSSA476). The resulting reporter metabolites were enriched in multiple metabolic pathways, with cofactor biosynthesis being the most important. The results of this study will serve as a guide for the development of antimicrobial agents as they provide a first insight into potential drug targets.
Collapse
Affiliation(s)
- Fatma Gizem Avci
- Department of Bioengineering, Faculty of Engineering and Natural Sciences, Üsküdar University, Istanbul, Türkiye.
- Genetics of Prokaryotes, Faculty of Biology and Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany.
| |
Collapse
|
3
|
Glajzner P, Bernat A, Jasińska-Stroschein M. Improving the treatment of bacterial infections caused by multidrug-resistant bacteria through drug repositioning. Front Pharmacol 2024; 15:1397602. [PMID: 38910882 PMCID: PMC11193365 DOI: 10.3389/fphar.2024.1397602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
Drug repurposing (repositioning) is a dynamically-developing area in the search for effective therapy of infectious diseases. Repositioning existing drugs with a well-known pharmacological and toxicological profile is an attractive method for quickly discovering new therapeutic indications. The off-label use of drugs for infectious diseases requires much less capital and time, and can hasten progress in the development of new antimicrobial drugs, including antibiotics. The use of drug repositioning in searching for new therapeutic options has brought promising results for many viral infectious diseases, such as Ebola, ZIKA, Dengue, and HCV. This review describes the most favorable results for repositioned drugs for the treatment of bacterial infections. It comprises publications from various databases including PubMed and Web of Science published from 2015 to 2023. The following search keywords/strings were used: drug repositioning and/or repurposing and/or antibacterial activity and/or infectious diseases. Treatment options for infections caused by multidrug-resistant bacteria were taken into account, including methicillin-resistant staphylococci, multidrug-resistant Mycobacterium tuberculosis, or carbapenem-resistant bacteria from the Enterobacteriaceae family. It analyses the safety profiles of the included drugs and their synergistic combinations with antibiotics and discusses the potential of antibacterial drugs with antiparasitic, anticancer, antipsychotic effects, and those used in metabolic diseases. Drug repositioning may be an effective response to public health threats related to the spread of multidrug-resistant bacterial strains and the growing antibiotic resistance of microorganisms.
Collapse
Affiliation(s)
- Paulina Glajzner
- Department of Biopharmacy, Faculty of Pharmacy, Medical University of Lodz, Łódź, Poland
| | | | | |
Collapse
|
4
|
Fatemi N, Karimpour M, Bahrami H, Zali MR, Chaleshi V, Riccio A, Nazemalhosseini-Mojarad E, Totonchi M. Current trends and future prospects of drug repositioning in gastrointestinal oncology. Front Pharmacol 2024; 14:1329244. [PMID: 38239190 PMCID: PMC10794567 DOI: 10.3389/fphar.2023.1329244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 12/11/2023] [Indexed: 01/22/2024] Open
Abstract
Gastrointestinal (GI) cancers comprise a significant number of cancer cases worldwide and contribute to a high percentage of cancer-related deaths. To improve survival rates of GI cancer patients, it is important to find and implement more effective therapeutic strategies with better prognoses and fewer side effects. The development of new drugs can be a lengthy and expensive process, often involving clinical trials that may fail in the early stages. One strategy to address these challenges is drug repurposing (DR). Drug repurposing is a developmental strategy that involves using existing drugs approved for other diseases and leveraging their safety and pharmacological data to explore their potential use in treating different diseases. In this paper, we outline the existing therapeutic strategies and challenges associated with GI cancers and explore DR as a promising alternative approach. We have presented an extensive review of different DR methodologies, research efforts and examples of repurposed drugs within various GI cancer types, such as colorectal, pancreatic and liver cancers. Our aim is to provide a comprehensive overview of employing the DR approach in GI cancers to inform future research endeavors and clinical trials in this field.
Collapse
Affiliation(s)
- Nayeralsadat Fatemi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mina Karimpour
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hoda Bahrami
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Chaleshi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Andrea Riccio
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università degli Studi della Campania “Luigi Vanvitelli”, Caserta, Italy
- Institute of Genetics and Biophysics (IGB) “Adriano Buzzati-Traverso”, Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | - Ehsan Nazemalhosseini-Mojarad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Totonchi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università degli Studi della Campania “Luigi Vanvitelli”, Caserta, Italy
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
5
|
Stromberg ZR, Phillips SMB, Omberg KM, Hess BM. High-throughput functional trait testing for bacterial pathogens. mSphere 2023; 8:e0031523. [PMID: 37702517 PMCID: PMC10597404 DOI: 10.1128/msphere.00315-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023] Open
Abstract
Functional traits are characteristics that affect the fitness and metabolic function of a microorganism. There is growing interest in using high-throughput methods to characterize bacterial pathogens based on functional virulence traits. Traditional methods that phenotype a single organism for a single virulence trait can be time consuming and labor intensive. Alternatively, machine learning of whole-genome sequences (WGS) has shown some success in predicting virulence. However, relying solely on WGS can miss functional traits, particularly for organisms lacking classical virulence factors. We propose that high-throughput assays for functional virulence trait identification should become a prominent method of characterizing bacterial pathogens on a population scale. This work is critical as we move from compiling lists of bacterial species associated with disease to pathogen-agnostic approaches capable of detecting novel microbes. We discuss six key areas of functional trait testing and how advancing high-throughput methods could provide a greater understanding of pathogens.
Collapse
Affiliation(s)
- Zachary R. Stromberg
- Chemical and Biological Signatures Group, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Shelby M. B. Phillips
- Chemical and Biological Signatures Group, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Kristin M. Omberg
- Chemical and Biological Signatures Group, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Becky M. Hess
- Chemical and Biological Signatures Group, Pacific Northwest National Laboratory, Richland, Washington, USA
| |
Collapse
|
6
|
Risk Factors of Clonally Related, Multi, and Extensively Drug-Resistant Acinetobacter baumannii in Severely Ill COVID-19 Patients. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2023; 2023:3139270. [PMID: 36814503 PMCID: PMC9940951 DOI: 10.1155/2023/3139270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/30/2022] [Accepted: 02/06/2023] [Indexed: 02/15/2023]
Abstract
Background The secondary infection of multi and extensively drug-resistant "Acinetobacter baumannii" in severely ill COVID-19 individuals is usually associated with extended hospitalisation and a high mortality rate. The current study aimed to assess the exact incidence rate of A. baumannii coinfection in severely ill COVID-19 patients admitted to intensive care unit (ICUs), to identify the possible mechanism of A. baumannii transfer to COVID-19 patients and to find out their resistance rate against different antibiotics. Methods Fifty severely ill "COVID-19" individuals on respiratory support were selected with samples being collected from the pharynx. In addition, another 60 samples were collected from the surrounding environment. Bacterial isolates were diagnosed by microbiological cultures and confirmed by "Vitek 2 system" and real-time PCR. The "Vitek 2 Compact system" was used to evaluate these isolates for antimicrobial susceptibility. The recovered isolates' DNA fingerprints and genetic similarities were performed using ERIC-PCR. Results Twenty-six samples were tested positive for A. baumannii (20 out of 50 samples taken from patients, 40%; 6 out of 60 swabs from a nosocomial setting, 10%). All A. baumannii strains isolated from the nosocomial sites were clonally related (have the same genetic lineage) to some strains isolated from patients. However, the majority of the patients' strains were categorised as belonging to the same genetic lineage. Furthermore, "the multi and extensively drug" resistance patterns were seen in all isolates. In addition, total isolates showed resistance to the most commonly tested antibiotics, while none of them was found to be resistant to tigecycline. Conclusion Secondary "A. baumannii" infection in severely ill "COVID-19" patients is a serious matter, especially when it has one spot of transmission in the ICU as well as when it is extensively drug-resistant, necessitating an immediate and tactical response to secure the issue.
Collapse
|
7
|
She P, Li Z, Li Y, Liu S, Li L, Yang Y, Zhou L, Wu Y. Pixantrone Sensitizes Gram-Negative Pathogens to Rifampin. Microbiol Spectr 2022; 10:e0211422. [PMID: 36318018 PMCID: PMC9769682 DOI: 10.1128/spectrum.02114-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 10/17/2022] [Indexed: 12/24/2022] Open
Abstract
The emergence of bacterial drug resistance poses a severe threat to global public health. In particular, antimicrobial-resistant pathogens lead to a high rate of treatment failure and significantly increase mortality. Repurposing FDA-approved compounds to sensitize superbugs to conventional antibiotics provides a promising strategy to alleviate such crises. Pixantrone (PIX) has been approved for treating aggressive B-cell non-Hodgkin's lymphoma. By high-throughput drug screening, we profiled the synergistic activity between PIX and rifampin (RFP) against Gram-negative extensively drug-resistant isolates by checkerboard assay. Mechanistic studies demonstrated that PIX impacted the flagellum assembly, induced irreversible intracellular reactive oxygen species accumulation and disrupted proton motive force. In addition, the combination of PIX with RFP possesses effective antimicrobial activity against multidrug-resistant strains in vivo without detected toxicity. Collectively, these results reveal the potential of PIX in combination with RFP as a therapy option for refractory infections caused by Gram-negative pathogens. IMPORTANCE Bacterial resistance has become increasingly serious because of the widespread use and abuse of antibiotics. In particular, the emergence of multidrug-resistant bacteria has posed a serious threat to human public health. Drug repurposing, the process of finding new uses for existing drugs, provide a promising pathway to solve antimicrobial resistance. Compared to the development of novel antibiotics, this strategy leverages well-characterized pharmacology and toxicology of known drugs and is more cost-effective.
Collapse
Affiliation(s)
- Pengfei She
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zehao Li
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yimin Li
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Shasha Liu
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Linhui Li
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yifan Yang
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Linying Zhou
- Department of Laboratory Medicine, The First Hospital of Changsha, Changsha, Hunan, China
| | - Yong Wu
- Department of Laboratory Medicine, The First Hospital of Changsha, Changsha, Hunan, China
| |
Collapse
|
8
|
Akkermansia muciniphila and Faecalibacterium prausnitzii in Immune-Related Diseases. Microorganisms 2022; 10:microorganisms10122382. [PMID: 36557635 PMCID: PMC9782003 DOI: 10.3390/microorganisms10122382] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/27/2022] [Accepted: 11/28/2022] [Indexed: 12/04/2022] Open
Abstract
Probiotics and synbiotics are used to treat chronic illnesses due to their roles in immune system modulation and anti-inflammatory response. They have been shown to reduce inflammation in a number of immune-related disorders, including systemic lupus erythematosus (SLE), human immunodeficiency virus (HIV), and chronic inflammatory skin conditions such as psoriasis and atopic dermatitis (AD). Akkermansia muciniphila (A. muciniphila) and Faecalibacterium prausnitzii (F. prausnitzii) are two different types of bacteria that play a significant part in this function. It has been established that Akkermansia and Faecalibacterium are abundant in normal populations and have protective benefits on digestive health while also enhancing the immune system, metabolism, and gut barrier of the host. They have the potential to be a therapeutic target in diseases connected to the microbiota, such as immunological disorders and cancer immunotherapy. There has not been a review of the anti-inflammatory effects of Akkermansia and Faecalibacterium, particularly in immunological diseases. In this review, we highlight the most recent scientific findings regarding A. muciniphila and F. prausnitzii as two significant gut microbiota for microbiome alterations and seek to provide cutting-edge insight in terms of microbiome-targeted therapies as promising preventive and therapeutic tools in immune-related diseases and cancer immunotherapy.
Collapse
|
9
|
Jameel S, Farooq S, Gani I, Riyaz-Ul-Hassan S, Bhat KA. Ultrasound assisted facile synthesis of Boron-Heck coupled sclareol analogs as potential antibacterial agents against Staphylococcus aureus. J Appl Microbiol 2022; 133:3678-3689. [PMID: 36064938 DOI: 10.1111/jam.15805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/19/2022] [Accepted: 08/23/2022] [Indexed: 10/14/2022]
Abstract
AIM To evaluate the antimicrobial capability of sclareol and its derivatives against Staphylococcus aureus and its Methicillin-resistant strain (MRSA). METHODS AND RESULTS A new series of Boron-Heck-coupled sclareol analogs were prepared by structural modifications at C-15 terminal double bond of sclareol using ultrasonication. The structural modifications were designed to keep the stereochemistry of all the five chiral centres of sclareol intact. A two-step reaction scheme consisting of Boron-Heck coupling of sclareol followed by Wittig reaction was carried out to produce novel sclareol congeners for antintimicrobial evaluation. Three compounds SAJ-1, SAJ-2 and SB-11 exhibited strong antibacterial activity against Staphylococcus aureus and Methicillin-resistant strain (MRSA) with MIC values between 3 to 11 μM. Among all the screened compounds, SAJ-1 and SAJ-2 showed the best anti-biofilm profiles against both the strains. Moreover SAJ-1 and SAJ-2 acted synergistically with streptomycin against S. aureus while creating varying outcomes in combination with ciprofloxacin, penicillin, and ampicillin. SAJ-1 also acted synergistically with ampicillin against S. aureus, while SB-11 showed synergism with ciprofloxacin against both pathogens. Moreover, SAJ-1 and SAJ-2 also inhibited staphyloxanthin production in S. aureus and MRSA and induced post-antibiotic effects against both pathogens. CONCLUSIONS It can be inferred that SAJ-1, SAJ-2 and SB-11 may act as potential chemical entities for the development of antibacterial substances. The study revealed that SAJ-1 and SAJ-2 are most suitable sclareol analogs for further studies towards the development of antibacterial substances. SIGNIFICANCE AND IMPACT OF THE STUDY SAJ-1, SAJ-2 and SB-11 show promising antibacterial properties against Staphylococcus aureus. Efforts should be made and more research should be done, utilising in vivo models to determine their efficacy as antibiotics.
Collapse
Affiliation(s)
- Salman Jameel
- Bioorganic Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Sanatnagar, 190005, Srinagar, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad- 201002, India
| | - Sadaqat Farooq
- Fermentation and Microbial Biotechnology Division, CSIR-Indian Institute of Integrative Medicine, Sanatnagar, 190005, Srinagar, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad- 201002, India
| | - Ifshana Gani
- Bioorganic Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Sanatnagar, 190005, Srinagar, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad- 201002, India
| | - Syed Riyaz-Ul-Hassan
- Fermentation and Microbial Biotechnology Division, CSIR-Indian Institute of Integrative Medicine, Sanatnagar, 190005, Srinagar, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad- 201002, India
| | - Khursheed Ahmad Bhat
- Bioorganic Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Sanatnagar, 190005, Srinagar, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad- 201002, India
| |
Collapse
|
10
|
Piludiya RI, Dholaria PV, Jivani AJ, Kapadiya KM. Bis-triazole Heterocycles as Antitubercular and Antimicrobial Agents: Synthesis Using Copper-Catalyzed Click Chemistry Approach. RUSSIAN JOURNAL OF ORGANIC CHEMISTRY 2022. [DOI: 10.1134/s1070428022090135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
11
|
Cressey P, Bronstein LG, Benmahmoudi R, Rosilio V, Regeard C, Makky A. Novel liposome-like assemblies composed of phospholipid-porphyrin conjugates with photothermal and photodynamic activities against bacterial biofilms. Int J Pharm 2022; 623:121915. [PMID: 35716977 DOI: 10.1016/j.ijpharm.2022.121915] [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: 04/22/2022] [Revised: 06/10/2022] [Accepted: 06/11/2022] [Indexed: 10/18/2022]
Abstract
Phospholipid-Porphyrin (PL-Por) conjugates are unique building blocks that can self assemble into liposome-like structures with improved photophysical properties compared to their monomeric counterparts. The high packing density of porphyrin moieties enables these assemblies to exhibit high photothermal conversion efficiency as well as photodynamic activity. Thus, PL-Por conjugates assemblies can be used for photodynamic therapy (PDT) and photothermal therapy (PTT) applications against resistant bacteria and biofilms. In order to tune the PD/PT properties of such nanosystems, we developed six different supramolecular assemblies composed of newly synthesized PL-Por conjugates bearing either pheophorbide-a (PhxLPC) or pyropheophorbide-a (PyrxLPC) photosensitizers (PSs) for combined PDT/PTT against planktonic bacteria and their biofilms. In this study, the influence of the chemical structure of the phospholipid backbone as well as that of the PS on the photothermal conversion efficiency, the photodynamic activity and the stability of these assemblies in biological medium were determined. Then their antimicrobial efficiency was assessed on S. aureus and P. aeruginosa planktonic cultures and biofilms. The two studied systems show almost the same photothermal effect against planktonic cultures and biofilms of S. aureus and P. aeruginosa. However, PhxLPC vesicles exhibit superior photodynamic activity, making them the best combination for PTT/PDT. Such results highlight the higher potential of the photodynamic activity of PL-Por nanoassemblies compared to their photothermal conversion in combating bacterial infections.
Collapse
Affiliation(s)
- Paul Cressey
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 92296 Châtenay-Malabry cedex, France
| | - Louis-Gabriel Bronstein
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 92296 Châtenay-Malabry cedex, France
| | - Rayene Benmahmoudi
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 92296 Châtenay-Malabry cedex, France; Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Véronique Rosilio
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 92296 Châtenay-Malabry cedex, France
| | - Christophe Regeard
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France..
| | - Ali Makky
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 92296 Châtenay-Malabry cedex, France.
| |
Collapse
|
12
|
A systematic review of disulfiram as an antibacterial agent: What is the evidence? Int J Antimicrob Agents 2022; 59:106578. [DOI: 10.1016/j.ijantimicag.2022.106578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 02/24/2022] [Accepted: 03/20/2022] [Indexed: 11/18/2022]
|
13
|
Peyclit L, Baron SA, Hadjadj L, Rolain JM. In Vitro Screening of a 1280 FDA-Approved Drugs Library against Multidrug-Resistant and Extensively Drug-Resistant Bacteria. Antibiotics (Basel) 2022; 11:antibiotics11030291. [PMID: 35326755 PMCID: PMC8944690 DOI: 10.3390/antibiotics11030291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/18/2022] [Accepted: 02/20/2022] [Indexed: 12/29/2022] Open
Abstract
Alternative strategies against multidrug-resistant (MDR) bacterial infections are suggested to clinicians, such as drug repurposing, which uses rapidly available and marketed drugs. We gathered a collection of MDR bacteria from our hospital and performed a phenotypic high-throughput screening with a 1280 FDA-approved drug library. We used two Gram positive (Enterococcus faecium P5014 and Staphylococcus aureus P1943) and six Gram negative (Acinetobacter baumannii P1887, Klebsiella pneumoniae P9495, Pseudomonas aeruginosa P6540, Burkholderia multivorans P6539, Pandoraea nosoerga P8103, and Escherichia coli DSM105182 as the reference and control strain). The selected MDR strain panel carried resistance genes or displayed phenotypic resistance to last-line therapies such as carbapenems, vancomycin, or colistin. A total of 107 compounds from nine therapeutic classes inhibited >90% of the growth of the selected Gram negative and Gram positive bacteria at a drug concentration set at 10 µmol/L, and 7.5% were anticancer drugs. The common hit was the antiseptic chlorhexidine. The activity of niclosamide, carmofur, and auranofin was found against the selected methicillin-resistant S. aureus. Zidovudine was effective against colistin-resistant E. coli and carbapenem-resistant K. pneumoniae. Trifluridine, an antiviral, was effective against E. faecium. Deferoxamine mesylate inhibited the growth of XDR P. nosoerga. Drug repurposing by an in vitro screening of a drug library is a promising approach to identify effective drugs for specific bacteria.
Collapse
Affiliation(s)
- Lucie Peyclit
- Aix Marseille University, IRD, APHM, MEPHI, 19-21 Boulevard Jean Moulin, CEDEX 05, 13385 Marseille, France; (L.P.); (S.A.B.); (L.H.)
- IHU-Méditerranée Infection, 19-21 Boulevard Jean Moulin, CEDEX 05, 13385 Marseille, France
| | - Sophie Alexandra Baron
- Aix Marseille University, IRD, APHM, MEPHI, 19-21 Boulevard Jean Moulin, CEDEX 05, 13385 Marseille, France; (L.P.); (S.A.B.); (L.H.)
- IHU-Méditerranée Infection, 19-21 Boulevard Jean Moulin, CEDEX 05, 13385 Marseille, France
| | - Linda Hadjadj
- Aix Marseille University, IRD, APHM, MEPHI, 19-21 Boulevard Jean Moulin, CEDEX 05, 13385 Marseille, France; (L.P.); (S.A.B.); (L.H.)
- IHU-Méditerranée Infection, 19-21 Boulevard Jean Moulin, CEDEX 05, 13385 Marseille, France
| | - Jean-Marc Rolain
- Aix Marseille University, IRD, APHM, MEPHI, 19-21 Boulevard Jean Moulin, CEDEX 05, 13385 Marseille, France; (L.P.); (S.A.B.); (L.H.)
- IHU-Méditerranée Infection, 19-21 Boulevard Jean Moulin, CEDEX 05, 13385 Marseille, France
- Correspondence: ; Tel.: +33-4-13-73-24-01
| |
Collapse
|
14
|
Liu Y, Lu Y, Xu Z, Ma X, Chen X, Liu W. Repurposing of the gold drug auranofin and a review of its derivatives as antibacterial therapeutics. Drug Discov Today 2022; 27:1961-1973. [DOI: 10.1016/j.drudis.2022.02.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 01/22/2022] [Accepted: 02/16/2022] [Indexed: 12/18/2022]
|
15
|
Targeting Staphylococcus aureus and its biofilms with novel antibacterial compounds produced by Lactiplantibacillus plantarum SJ33. Arch Microbiol 2021; 204:20. [DOI: 10.1007/s00203-021-02630-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 11/26/2022]
|
16
|
Payne JAE, Tailhades J, Ellett F, Kostoulias X, Fulcher AJ, Fu T, Leung R, Louch S, Tran A, Weber SA, Schittenhelm RB, Lieschke GJ, Qin CH, Irima D, Peleg AY, Cryle MJ. Antibiotic-chemoattractants enhance neutrophil clearance of Staphylococcus aureus. Nat Commun 2021; 12:6157. [PMID: 34697316 PMCID: PMC8546149 DOI: 10.1038/s41467-021-26244-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 09/22/2021] [Indexed: 01/28/2023] Open
Abstract
The pathogen Staphylococcus aureus can readily develop antibiotic resistance and evade the human immune system, which is associated with reduced levels of neutrophil recruitment. Here, we present a class of antibacterial peptides with potential to act both as antibiotics and as neutrophil chemoattractants. The compounds, which we term 'antibiotic-chemoattractants', consist of a formylated peptide (known to act as chemoattractant for neutrophil recruitment) that is covalently linked to the antibiotic vancomycin (known to bind to the bacterial cell wall). We use a combination of in vitro assays, cellular assays, infection-on-a-chip and in vivo mouse models to show that the compounds improve the recruitment, engulfment and killing of S. aureus by neutrophils. Furthermore, optimizing the formyl peptide sequence can enhance neutrophil activity through differential activation of formyl peptide receptors. Thus, we propose antibiotic-chemoattractants as an alternate approach for antibiotic development.
Collapse
Affiliation(s)
- Jennifer A E Payne
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia.
- ARC Centre of Excellence for Innovations in Peptide and Protein Science, Monash University, Clayton, Victoria, 3800, Australia.
- EMBL Australia, Monash University, Clayton, Victoria, 3800, Australia.
| | - Julien Tailhades
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
- ARC Centre of Excellence for Innovations in Peptide and Protein Science, Monash University, Clayton, Victoria, 3800, Australia
- EMBL Australia, Monash University, Clayton, Victoria, 3800, Australia
| | - Felix Ellett
- BioMEMS Resource Center, Center for Engineering in Medicine and Surgical Services, Massachusetts General Hospital, Shriners Hospital for Children, and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Xenia Kostoulias
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Alex J Fulcher
- Monash Micro Imaging, Monash University, Clayton, Victoria, 3800, Australia
| | - Ting Fu
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Ryan Leung
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Stephanie Louch
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Amy Tran
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
| | - Severin A Weber
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia
- EMBL Australia, Monash University, Clayton, Victoria, 3800, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics and Metabolomics Facility, Monash University, Clayton, Victoria, 3800, Australia
| | - Graham J Lieschke
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, 3800, Australia
| | - Chengxue Helena Qin
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia
| | - Daniel Irima
- BioMEMS Resource Center, Center for Engineering in Medicine and Surgical Services, Massachusetts General Hospital, Shriners Hospital for Children, and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Anton Y Peleg
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, 3800, Australia
- Department of Infectious Diseases, The Alfred Hospital, Melbourne, Victoria, 3004, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, 3004, Australia
| | - Max J Cryle
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3800, Australia.
- ARC Centre of Excellence for Innovations in Peptide and Protein Science, Monash University, Clayton, Victoria, 3800, Australia.
- EMBL Australia, Monash University, Clayton, Victoria, 3800, Australia.
| |
Collapse
|
17
|
Asadpoor M, Ithakisiou GN, van Putten JPM, Pieters RJ, Folkerts G, Braber S. Antimicrobial Activities of Alginate and Chitosan Oligosaccharides Against Staphylococcus aureus and Group B Streptococcus. Front Microbiol 2021; 12:700605. [PMID: 34589067 PMCID: PMC8473942 DOI: 10.3389/fmicb.2021.700605] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 08/17/2021] [Indexed: 12/22/2022] Open
Abstract
The bacterial pathogens Streptococcus agalactiae (GBS) and Staphylococcus aureus (S. aureus) cause serious infections in humans and animals. The emergence of antibiotic-resistant isolates and bacterial biofilm formation entails the urge of novel treatment strategies. Recently, there is a profound scientific interest in the capabilities of non-digestible oligosaccharides as antimicrobial and anti-biofilm agents as well as adjuvants in antibiotic combination therapies. In this study, we investigated the potential of alginate oligosaccharides (AOS) and chitosan oligosaccharides (COS) as alternative for, or in combination with antibiotic treatment. AOS (2-16%) significantly decreased GBS V growth by determining the minimum inhibitory concentration. Both AOS (8 and 16%) and COS (2-16%) were able to prevent biofilm formation by S. aureus wood 46. A checkerboard biofilm formation assay demonstrated a synergistic effect of COS and clindamycin on the S. aureus biofilm formation, while AOS (2 and 4%) were found to sensitize GBS V to trimethoprim. In conclusion, AOS and COS affect the growth of GBS V and S. aureus wood 46 and can function as anti-biofilm agents. The promising effects of AOS and COS in combination with different antibiotics may offer new opportunities to combat antimicrobial resistance.
Collapse
Affiliation(s)
- Mostafa Asadpoor
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Georgia-Nefeli Ithakisiou
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Jos P. M. van Putten
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Roland J. Pieters
- Division of Medicinal Chemistry and Chemical Biology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Saskia Braber
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
18
|
Caldara M, Marmiroli N. Antimicrobial Properties of Antidepressants and Antipsychotics-Possibilities and Implications. Pharmaceuticals (Basel) 2021; 14:ph14090915. [PMID: 34577614 PMCID: PMC8470654 DOI: 10.3390/ph14090915] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/03/2021] [Accepted: 09/08/2021] [Indexed: 12/13/2022] Open
Abstract
The spreading of antibiotic resistance is responsible annually for over 700,000 deaths worldwide, and the prevision is that this number will increase exponentially. The identification of new antimicrobial treatments is a challenge that requires scientists all over the world to collaborate. Developing new drugs is an extremely long and costly process, but it could be paralleled by drug repositioning. The latter aims at identifying new clinical targets of an “old” drug that has already been tested, approved, and even marketed. This approach is very intriguing as it could reduce costs and speed up approval timelines, since data from preclinical studies and on pharmacokinetics, pharmacodynamics, and toxicity are already available. Antidepressants and antipsychotics have been described to inhibit planktonic and sessile growth of different yeasts and bacteria. The main findings in the field are discussed in this critical review, along with the description of the possible microbial targets of these molecules. Considering their antimicrobial activity, the manuscript highlights important implications that the administration of antidepressants and antipsychotics may have on the gut microbiome.
Collapse
Affiliation(s)
- Marina Caldara
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy;
- Interdepartmental Center SITEIA.PARMA, University of Parma, Parco Area delle Scienze 181/A, 43124 Parma, Italy
- Correspondence:
| | - Nelson Marmiroli
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy;
- Interdepartmental Center SITEIA.PARMA, University of Parma, Parco Area delle Scienze 181/A, 43124 Parma, Italy
- Italian National Interuniversity Consortium for Environmental Sciences (CINSA), University of Parma, 43124 Parma, Italy
| |
Collapse
|
19
|
Yang TY, Kao HY, Lu PL, Chen PY, Wang SC, Wang LC, Hsieh YJ, Tseng SP. Evaluation of the Organotellurium Compound AS101 for Treating Colistin- and Carbapenem-Resistant Klebsiella pneumoniae. Pharmaceuticals (Basel) 2021; 14:ph14080795. [PMID: 34451891 PMCID: PMC8400984 DOI: 10.3390/ph14080795] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/01/2021] [Accepted: 08/09/2021] [Indexed: 01/21/2023] Open
Abstract
Colistin- and carbapenem-resistant Enterobacteriaceae cases are increasing at alarming rates worldwide. Drug repurposing is receiving greater attention as an alternative approach in light of economic and technical barriers in antibiotics research. The immunomodulation agent ammonium trichloro(dioxoethylene-O,O’-)tellurate (AS101) was repurposed as an antimicrobial agent against colistin- and carbapenem-resistant Klebsiella pneumoniae (CRKP). 134 CRKP isolates were collected between 2012 and 2015 in Taiwan. The in vitro antibacterial activities of AS101 was observed through broth microdilution, time-kill assay, and electron microscopy. Pharmaceutical manipulation and RNA microarray were applied to investigate these antimicrobial mechanisms. Caenorhabditis elegans, a nematode animal model, and the Institute for Cancer Research (ICR) mouse model was employed for the evaluation of in vivo efficacy. The in vitro antibacterial results were found for AS101 against colistin- and CRKP isolates, with minimum inhibitory concentration (MIC) values ranging from <0.5 to 32 μg/mL. ROS-mediated antibacterial activity eliminated 99.9% of bacteria within 2–4 h. AS101 also extended the median survival time in a C. elegans animal model infected with a colistin-resistant CRKP isolate and rescued lethally infected animals in a separate mouse model of mono-bacterial sepsis by eliminating bacterial organ loads. These findings support the use of AS101 as an antimicrobial agent for addressing the colistin and carbapenem resistance crisis.
Collapse
Affiliation(s)
- Tsung-Ying Yang
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-Y.Y.); (P.-Y.C.); (S.-C.W.)
| | - Hao-Yun Kao
- Department of Healthcare Administration and Medical Informatics, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Po-Liang Lu
- Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- School of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Pei-Yu Chen
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-Y.Y.); (P.-Y.C.); (S.-C.W.)
| | - Shu-Chi Wang
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-Y.Y.); (P.-Y.C.); (S.-C.W.)
- Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Liang-Chun Wang
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan;
| | - Ya-Ju Hsieh
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Correspondence: (Y.-J.H.); (S.-P.T.); Tel.: +886-7-312-1101 (ext. 2350) (Y.-J.H.); +886-7-312-1101 (ext. 2356-22) (S.-P.T.)
| | - Sung-Pin Tseng
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-Y.Y.); (P.-Y.C.); (S.-C.W.)
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan;
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 900391, Taiwan
- Correspondence: (Y.-J.H.); (S.-P.T.); Tel.: +886-7-312-1101 (ext. 2350) (Y.-J.H.); +886-7-312-1101 (ext. 2356-22) (S.-P.T.)
| |
Collapse
|
20
|
Vogel AL, Knebel AR, Faupel-Badger JM, Portilla LM, Simeonov A. A systems approach to enable effective team science from the internal research program of the National Center for Advancing Translational Sciences. J Clin Transl Sci 2021; 5:e163. [PMID: 34527302 PMCID: PMC8427549 DOI: 10.1017/cts.2021.811] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 12/17/2022] Open
Abstract
The internal research program of the National Center for Advancing Translational Sciences (NCATS) at the National Institutes of Health aims to fundamentally transform the preclinical translational research process to get more treatments to more people more quickly. The program develops and implements innovative scientific and operational approaches that accelerate and enhance translation across many diverse projects. Cross-disciplinary team science is a defining feature of our organization, with scientists at all levels engaged in multiple research teams. Here, we share our systems approach to nurturing cross-disciplinary team science, which leverages organizational policies, structures, and processes. Policies including the organizational mission statement, principles for ethical conduct of research, performance review criteria, and training program objectives and approaches reinforce the value of team science to achieve the program's scientific goals. Structures including an organizational structure designed around solving translational problems, co-location of employees in a single state-of-the-art scientific facility, and shared-use laboratories, expertise and instrumentation facilitate collaboration. Processes including fluid team assembly, specialized project management, cross-agency partnerships, and decision making based on clear screening criteria and milestones enable effective team assembly and functioning. We share evidence of the impact of these approaches on the science and commercialization of findings and discuss pathways to broad adoption of similar approaches.
Collapse
Affiliation(s)
- Amanda L. Vogel
- National Institutes of Health (NIH); National Center for Advancing Translational Sciences (NCATS); Office of Policy, Communications and Education; Education Branch; Bethesda, MD, USA
| | - Ann R. Knebel
- National Institutes of Health (NIH), National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation (DPI), Office of the Scientific Director, Rockville, MD, USA
| | - Jessica M. Faupel-Badger
- National Institutes of Health (NIH); National Center for Advancing Translational Sciences (NCATS); Office of Policy, Communications and Education; Education Branch; Bethesda, MD, USA
| | - Lili M. Portilla
- National Institutes of Health (NIH), National Center for Advancing Translational Sciences (NCATS), Office of Strategic Alliances (OSA), Rockville, MD, USA
| | - Anton Simeonov
- National Institutes of Health (NIH), National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation (DPI), Office of the Scientific Director, Rockville, MD, USA
| |
Collapse
|
21
|
Feng X, Liu S, Wang Y, Zhang Y, Sun L, Li H, Wang C, Liu Y, Cao B. Synergistic Activity of Colistin Combined With Auranofin Against Colistin-Resistant Gram-Negative Bacteria. Front Microbiol 2021; 12:676414. [PMID: 34248888 PMCID: PMC8267823 DOI: 10.3389/fmicb.2021.676414] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 06/03/2021] [Indexed: 11/13/2022] Open
Abstract
Colistin-resistant (Col-R) bacteria are steadily increasing, and are extremely difficult to treat. New drugs or therapies are urgently needed to treat infections caused by these pathogens. Combination therapy with colistin and other old drugs, is an important way to restore the activity of colistin. This study aimed to investigate the activity of colistin in combination with the anti-rheumatic drug auranofin against Col-R Gram-negative bacteria. The results of checkerboard analysis demonstrated that auranofin synergized with colistin against Col-R Gram-negative bacteria. Time-kill assays showed significant synergistic antimicrobial activity of colistin combined with auranofin. Electron microscopy revealed that the combination resulted in more cellular structural alterations compared to each drug alone. Auranofin enhanced the therapeutic effectiveness of colistin in mouse peritoneal infection models. These results suggested that the combination of colistin and auranofin might be a potential alternative for the treatment of Col-R Gram-negative bacterial infections.
Collapse
Affiliation(s)
- Xiaoxuan Feng
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,Department of Pulmonary and Critical Care Medicine, Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China
| | - Shuai Liu
- Department of Pulmonary and Critical Care Medicine, Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China.,China-Japan Friendship Hospital, National Clinical Research Center for Respiratory Diseases, Clinical Center for Pulmonary Infections, Capital Medical University, Beijing, China
| | - Yang Wang
- Department of Pulmonary and Critical Care Medicine, Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China.,China-Japan Friendship Hospital, National Clinical Research Center for Respiratory Diseases, Clinical Center for Pulmonary Infections, Capital Medical University, Beijing, China
| | - Yulin Zhang
- Department of Pulmonary and Critical Care Medicine, Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China.,China-Japan Friendship Hospital, National Clinical Research Center for Respiratory Diseases, Clinical Center for Pulmonary Infections, Capital Medical University, Beijing, China.,Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Lingxiao Sun
- Department of Pulmonary and Critical Care Medicine, Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China.,China-Japan Friendship Hospital, National Clinical Research Center for Respiratory Diseases, Clinical Center for Pulmonary Infections, Capital Medical University, Beijing, China
| | - Haibo Li
- Department of Pulmonary and Critical Care Medicine, Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China.,China-Japan Friendship Hospital, National Clinical Research Center for Respiratory Diseases, Clinical Center for Pulmonary Infections, Capital Medical University, Beijing, China.,Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Chunlei Wang
- Department of Pulmonary and Critical Care Medicine, Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China.,China-Japan Friendship Hospital, National Clinical Research Center for Respiratory Diseases, Clinical Center for Pulmonary Infections, Capital Medical University, Beijing, China.,Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Yingmei Liu
- Department of Pulmonary and Critical Care Medicine, Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China.,China-Japan Friendship Hospital, National Clinical Research Center for Respiratory Diseases, Clinical Center for Pulmonary Infections, Capital Medical University, Beijing, China.,Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Bin Cao
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,Department of Pulmonary and Critical Care Medicine, Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China.,China-Japan Friendship Hospital, National Clinical Research Center for Respiratory Diseases, Clinical Center for Pulmonary Infections, Capital Medical University, Beijing, China.,Laboratory of Clinical Microbiology and Infectious Diseases, Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, China.,Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
22
|
Pharmacodynamic evaluation of suppression of in vitro resistance in Acinetobacter baumannii strains using polymyxin B-based combination therapy. Sci Rep 2021; 11:11339. [PMID: 34059725 PMCID: PMC8167102 DOI: 10.1038/s41598-021-90709-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/13/2021] [Indexed: 02/04/2023] Open
Abstract
The emergence of polymyxin resistance in Gram-negative bacteria infections has motivated the use of combination therapy. This study determined the mutant selection window (MSW) of polymyxin B alone and in combination with meropenem and fosfomycin against A. baumannii strains belonging to clonal lineages I and III. To evaluate the inhibition of in vitro drug resistance, we investigate the MSW-derived pharmacodynamic indices associated with resistance to polymyxin B administrated regimens as monotherapy and combination therapy, such as the percentage of each dosage interval that free plasma concentration was within the MSW (%TMSW) and the percentage of each dosage interval that free plasma concentration exceeded the mutant prevention concentration (%T>MPC). The MSW of polymyxin B varied between 1 and 16 µg/mL for polymyxin B-susceptible strains. The triple combination of polymyxin B with meropenem and fosfomycin inhibited the polymyxin B-resistant subpopulation in meropenem-resistant isolates and polymyxin B plus meropenem as a double combination sufficiently inhibited meropenem-intermediate, and susceptible strains. T>MPC 90% was reached for polymyxin B in these combinations, while %TMSW was 0 against all strains. TMSW for meropenem and fosfomycin were also reduced. Effective antimicrobial combinations significantly reduced MSW. The MSW-derived pharmacodynamic indices can be used for the selection of effective combination regimen to combat the polymyxin B-resistant strain.
Collapse
|
23
|
Jain P, Jain SK, Jain M. Harnessing Drug Repurposing for Exploration of New Diseases: An Insight to Strategies and Case Studies. Curr Mol Med 2021; 21:111-132. [PMID: 32560606 DOI: 10.2174/1566524020666200619125404] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Traditional drug discovery is time consuming, costly, and risky process. Owing to the large investment, excessive attrition, and declined output, drug repurposing has become a blooming approach for the identification and development of new therapeutics. The method has gained momentum in the past few years and has resulted in many excellent discoveries. Industries are resurrecting the failed and shelved drugs to save time and cost. The process accounts for approximately 30% of the new US Food and Drug Administration approved drugs and vaccines in recent years. METHODS A systematic literature search using appropriate keywords were made to identify articles discussing the different strategies being adopted for repurposing and various drugs that have been/are being repurposed. RESULTS This review aims to describe the comprehensive data about the various strategies (Blinded search, computational approaches, and experimental approaches) used for the repurposing along with success case studies (treatment for orphan diseases, neglected tropical disease, neurodegenerative diseases, and drugs for pediatric population). It also inculcates an elaborated list of more than 100 drugs that have been repositioned, approaches adopted, and their present clinical status. We have also attempted to incorporate the different databases used for computational repurposing. CONCLUSION The data presented is proof that drug repurposing is a prolific approach circumventing the issues poised by conventional drug discovery approaches. It is a highly promising approach and when combined with sophisticated computational tools, it also carries high precision. The review would help researches in prioritizing the drugrepositioning method much needed to flourish the drug discovery research.
Collapse
Affiliation(s)
- Priti Jain
- Department of Pharmaceutical Chemistry and Computational Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dhule (425405) Maharashtra, India
| | - Shreyans K Jain
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Munendra Jain
- SVKM's Department of Sciences, Narsee Monjee Institute of Management Studies, Indore, Madhya Pradesh, India
| |
Collapse
|
24
|
Cheng YS, Roma JS, Shen M, Mota Fernandes C, Tsang PS, Forbes HE, Boshoff H, Lazzarini C, Del Poeta M, Zheng W, Williamson PR. Identification of Antifungal Compounds against Multidrug-Resistant Candida auris Utilizing a High-Throughput Drug-Repurposing Screen. Antimicrob Agents Chemother 2021; 65:e01305-20. [PMID: 33468482 PMCID: PMC8097445 DOI: 10.1128/aac.01305-20] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/31/2020] [Indexed: 12/30/2022] Open
Abstract
Candida auris is an emerging fatal fungal infection that has resulted in several outbreaks in hospitals and care facilities. Current treatment options are limited by the development of drug resistance. Identification of new pharmaceuticals to combat these drug-resistant infections will thus be required to overcome this unmet medical need. We have established a bioluminescent ATP-based assay to identify new compounds and potential drug combinations showing effective growth inhibition against multiple strains of multidrug-resistant Candida auris The assay is robust and suitable for assessing large compound collections by high-throughput screening (HTS). Utilizing this assay, we conducted a screen of 4,314 approved drugs and pharmacologically active compounds that yielded 25 compounds, including 6 novel anti-Candida auris compounds and 13 sets of potential two-drug combinations. Among the drug combinations, the serine palmitoyltransferase inhibitor myriocin demonstrated a combinational effect with flucytosine against all tested isolates during screening. This combinational effect was confirmed in 13 clinical isolates of Candida auris.
Collapse
Affiliation(s)
- Yu-Shan Cheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Jose Santinni Roma
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Min Shen
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Caroline Mota Fernandes
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Patricia S Tsang
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - He Eun Forbes
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Helena Boshoff
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Cristina Lazzarini
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Maurizio Del Poeta
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
- Division of Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
- Veterans Affairs Medical Center, Northport, New York, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Peter R Williamson
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
25
|
Gur D, Chitlaru T, Mamroud E, Zauberman A. Screening of an FDA-Approved Library for Novel Drugs against Y. pestis. Antibiotics (Basel) 2021; 10:antibiotics10010040. [PMID: 33401634 PMCID: PMC7823876 DOI: 10.3390/antibiotics10010040] [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: 12/15/2020] [Revised: 12/29/2020] [Accepted: 12/30/2020] [Indexed: 11/16/2022] Open
Abstract
Yersinia pestis is a Gram-negative pathogen that causes plague, a devastating disease that kills millions worldwide. Although plague is efficiently treatable by recommended antibiotics, the time of antibiotic therapy initiation is critical, as high mortality rates have been observed if treatment is delayed for longer than 24 h after symptom onset. To overcome the emergence of antibiotic resistant strains, we attempted a systematic screening of Food and Drug Administration (FDA)-approved drugs to identify alternative compounds which may possess antibacterial activity against Y. pestis. Here, we describe a drug-repurposing approach, which led to the identification of two antibiotic-like activities of the anticancer drugs bleomycin sulfate and streptozocin that have the potential for designing novel antiplague therapy approaches. The inhibitory characteristics of these two drugs were further addressed as well as their efficiency in affecting the growth of Y. pestis strains resistant to doxycycline and ciprofloxacin, antibiotics recommended for plague treatment.
Collapse
|
26
|
Sysel AM, Dunphy MJ, Bauer JA. Antimicrobial properties of diethylamine NONOate, a nitric oxide donor, against Escherichia coli: a pilot study. J Antibiot (Tokyo) 2021; 74:260-265. [PMID: 33361779 PMCID: PMC7767638 DOI: 10.1038/s41429-020-00397-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 01/29/2023]
Abstract
The emergence of SARS-CoV-2, the causative agent of COVID-19, highlights the increasing need for new and effective antiviral and antimicrobial agents. The FDA has recently banned several active ingredients used in hand sanitizers, including triclosan and benzethonium chloride. Nitric oxide (NO) is involved in the innate immune response and is a major component of macrophage-mediated attack on foreign viruses and bacteria. The specific aim of this study was to assess the antibacterial effects of 2-(N,N-diethylamino)-diazenolate-2-oxide (DEA-NONOate) against Escherichia coli (E. coli). A bacterial growth assay was compared to an adenosine triphosphate (ATP) activity assay at various time points to assess effects of DEA-NONOate on E. coli growth. A UV/Vis spectrophotometer was used to determine concentration of E. coli by measuring optical density (OD) at 630 nm. A luminescent assay was used to measure ATP activity correlating to viable cells. DEA-NONOate at a concentration of 65 mM was able to inhibit the growth of E. coli with the same efficacy as 1 μg ml-1 concentration of ciprofloxacin. Both the OD and ATP assays demonstrated a 99.9% reduction in E. coli. Both a 1 μg ml-1 concentration of ciprofloxacin and a 65 mM concentration of DEA-NONOate achieved 99.9% inhibition of E. coli, verified using both optical density measurement of bacterial cultures in 96 well plates and a luminescent ATP activity assay. The bactericidal effects of DEA-NONOate against E. coli is proof-of-concept to pursue evaluation of nitric oxide-based formulations as antimicrobial and antiviral agents as hand sanitizers.
Collapse
Affiliation(s)
- Annette M. Sysel
- grid.427858.4Bauer Research Foundation, Inc, North Canton, OH USA ,Nitric Oxide Services, LLC, North Canton, OH USA
| | - Michael J. Dunphy
- Nitric Oxide Services, LLC, North Canton, OH USA ,grid.412869.0Walsh University, North Canton, OH USA
| | - Joseph A. Bauer
- grid.427858.4Bauer Research Foundation, Inc, North Canton, OH USA ,Nitric Oxide Services, LLC, North Canton, OH USA
| |
Collapse
|
27
|
Li S, She P, Zhou L, Zeng X, Xu L, Liu Y, Chen L, Wu Y. High-Throughput Identification of Antibacterials Against Pseudomonas aeruginosa. Front Microbiol 2020; 11:591426. [PMID: 33362739 PMCID: PMC7755642 DOI: 10.3389/fmicb.2020.591426] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/16/2020] [Indexed: 12/16/2022] Open
Abstract
Antibiotic resistance is a growing public health concern, though the constant development of new antibiotics. The combination of high-throughput screening and drug repurposing is an effective way to develop new therapeutic uses of drugs. In this study, we screened a drug library consisting of 1,573 drugs already approved by the Food and Drug Administration and 903 drugs from the natural product library, to identify antimicrobials against Pseudomonas aeruginosa. A high-throughput screening assay based on microtiter plate was used to screen 39 drugs that inhibit the planktonic or biofilm formation of P. aeruginosa while most of them are antibiotics. The antimicrobial activities of these drugs were evaluated by phenotypic analysis. Further studies showed the combined therapy of tetracycline antibiotics demeclocycline hydrochloride (DMCT) and the novel antimicrobial peptide SAAP-148 has an effective synergistic antibacterial effect on P. aeruginosa PAO1 and P. aeruginosa ATCC27853. Moreover, the time-kill curve assay and murine model of cutaneous abscesses further confirmed the synergistic effect. In addition, the combination of DMCT and SAAP-148 has the potential to combat clinically isolated multidrug-resistant (MDR) P. aeruginosa strains. Our results clearly indicate that DMCT and SAAP-148 combined therapy could be an effective method to combat MDR P. aeruginosa-related infections.
Collapse
Affiliation(s)
- Shijia Li
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Pengfei She
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Linying Zhou
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xianghai Zeng
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Lanlan Xu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yaqian Liu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Lihua Chen
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yong Wu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
28
|
Croppi G, Zhou Y, Yang R, Bian Y, Zhao M, Hu Y, Ruan BH, Yu J, Wu F. Discovery of an Inhibitor for Bacterial 3-Mercaptopyruvate Sulfurtransferase that Synergistically Controls Bacterial Survival. Cell Chem Biol 2020; 27:1483-1499.e9. [DOI: 10.1016/j.chembiol.2020.10.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 08/12/2020] [Accepted: 10/22/2020] [Indexed: 12/19/2022]
|
29
|
Xu XL, Kang XQ, Qi J, Jin FY, Liu D, Du YZ. Novel Antibacterial Strategies for Combating Bacterial Multidrug Resistance. Curr Pharm Des 2020; 25:4717-4724. [PMID: 31642769 DOI: 10.2174/1381612825666191022163237] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/14/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Antibacterial multidrug resistance has emerged as one of the foremost global problems affecting human health. The emergence of resistant infections with the increasing number of multidrug-resistant pathogens has posed a serious problem, which required innovative collaborations across multiple disciplines to address this issue. METHODS In this review, we will explain the mechanisms of bacterial multidrug resistance and discuss different strategies for combating it, including combination therapy, the use of novel natural antibiotics, and the use of nanotechnology in the development of efflux pump inhibitors. RESULTS While combination therapy will remain the mainstay of bacterial multi-drug resistance treatment, nanotechnology will play critical roles in the development of novel treatments in the coming years. CONCLUSION Nanotechnology provides an encouraging platform for the development of clinically relevant and practical strategies to overcome drug resistance in the future.
Collapse
Affiliation(s)
- Xiao-Ling Xu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Xu-Qi Kang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Jing Qi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Fei-Yang Jin
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Di Liu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Yong-Zhong Du
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| |
Collapse
|
30
|
Zeng P, Xu C, Liu C, Liu J, Cheng Q, Gao W, Yang X, Chen S, Chan KF, Wong KY. De Novo Designed Hexadecapeptides Synergize Glycopeptide Antibiotics Vancomycin and Teicoplanin against Pathogenic Klebsiella pneumoniae via Disruption of Cell Permeability and Potential. ACS APPLIED BIO MATERIALS 2020; 3:1738-1752. [DOI: 10.1021/acsabm.0c00044] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Ping Zeng
- State Key Laboratory of Chemical Biology and Drug Discovery and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon 852, Hong Kong
| | - Chen Xu
- State Key Laboratory of Chemical Biology and Drug Discovery and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon 852, Hong Kong
| | - Chenyu Liu
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon +852, Hong Kong
| | - Jun Liu
- State Key Laboratory of Chemical Biology and Drug Discovery and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon 852, Hong Kong
| | - Qipeng Cheng
- State Key Laboratory of Chemical Biology and Drug Discovery and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon 852, Hong Kong
| | - Wei Gao
- State Key Laboratory of Chemical Biology and Drug Discovery and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon 852, Hong Kong
| | - Xuemei Yang
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon +852, Hong Kong
| | - Sheng Chen
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon +852, Hong Kong
| | - Kin-Fai Chan
- State Key Laboratory of Chemical Biology and Drug Discovery and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon 852, Hong Kong
| | - Kwok-Yin Wong
- State Key Laboratory of Chemical Biology and Drug Discovery and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon 852, Hong Kong
| |
Collapse
|
31
|
Cesur MF, Siraj B, Uddin R, Durmuş S, Çakır T. Network-Based Metabolism-Centered Screening of Potential Drug Targets in Klebsiella pneumoniae at Genome Scale. Front Cell Infect Microbiol 2020; 9:447. [PMID: 31993376 PMCID: PMC6970976 DOI: 10.3389/fcimb.2019.00447] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 12/12/2019] [Indexed: 01/28/2023] Open
Abstract
Klebsiella pneumoniae is an opportunistic bacterial pathogen leading to life-threatening nosocomial infections. Emergence of highly resistant strains poses a major challenge in the management of the infections by healthcare-associated K. pneumoniae isolates. Thus, despite intensive efforts, the current treatment strategies remain insufficient to eradicate such infections. Failure of the conventional infection-prevention and treatment efforts explicitly indicates the requirement of new therapeutic approaches. This prompted us to systematically analyze the K. pneumoniae metabolism to investigate drug targets. Genome-scale metabolic networks (GMNs) facilitating the systematic analysis of the metabolism are promising platforms. Thus, we used a GMN of K. pneumoniae MGH 78578 to determine putative targets through gene- and metabolite-centric approaches. To develop more realistic infection models, we performed the bacterial growth simulations within different host-mimicking media, using an improved biomass formation reaction. We selected more suitable targets based on several property-based prioritization procedures. KdsA was identified as the high-ranked putative target satisfying most of the target prioritization criteria specified under the gene-centric approach. Through a structure-based virtual screening protocol, we identified potential KdsA inhibitors. In addition, the metabolite-centric approach extended the drug target list based on synthetic lethality. This revealed the importance of combined metabolic analyses for a better understanding of the metabolism. To our knowledge, this is the first comprehensive effort on the investigation of the K. pneumoniae metabolism for drug target prediction through the constraint-based analysis of its GMN in conjunction with several bioinformatic approaches. This study can guide the researchers for the future drug designs by providing initial findings regarding crucial components of the Klebsiella metabolism.
Collapse
Affiliation(s)
- Müberra Fatma Cesur
- Computational Systems Biology Group, Department of Bioengineering, Gebze Technical University, Gebze, Turkey
| | - Bushra Siraj
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Reaz Uddin
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Saliha Durmuş
- Computational Systems Biology Group, Department of Bioengineering, Gebze Technical University, Gebze, Turkey
| | - Tunahan Çakır
- Computational Systems Biology Group, Department of Bioengineering, Gebze Technical University, Gebze, Turkey
| |
Collapse
|
32
|
Ianevski A, Giri AK, Gautam P, Kononov A, Potdar S, Saarela J, Wennerberg K, Aittokallio T. Prediction of drug combination effects with a minimal set of experiments. NAT MACH INTELL 2019; 1:568-577. [PMID: 32368721 DOI: 10.1038/s42256-019-0122-4] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
High-throughput drug combination screening provides a systematic strategy to discover unexpected combinatorial synergies in pre-clinical cell models. However, phenotypic combinatorial screening with multi-dose matrix assays is experimentally expensive, especially when the aim is to identify selective combination synergies across a large panel of cell lines or patient samples. Here we implemented DECREASE, an efficient machine learning model that requires only a limited set of pairwise dose-response measurements for accurate prediction of drug combination synergy and antagonism. Using a compendium of 23,595 drug combination matrices tested in various cancer cell lines, and malaria and Ebola infection models, we demonstrate how cost-effective experimental designs with DECREASE capture almost the same degree of information for synergy and antagonism detection as the fully-measured dose-response matrices. Measuring only the diagonal of the matrix provides an accurate and practical option for combinatorial screening. The open-source web-implementation enables applications of DECREASE to both pre-clinical and translational studies.
Collapse
Affiliation(s)
- Aleksandr Ianevski
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, FI-00290 Helsinki, Finland.,Helsinki Institute for Information Technology (HIIT), Department of Computer Science, Aalto University, FI-02150 Espoo, Finland
| | - Anil K Giri
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, FI-00290 Helsinki, Finland
| | - Prson Gautam
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, FI-00290 Helsinki, Finland
| | - Alexander Kononov
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, FI-00290 Helsinki, Finland
| | - Swapnil Potdar
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, FI-00290 Helsinki, Finland
| | - Jani Saarela
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, FI-00290 Helsinki, Finland
| | - Krister Wennerberg
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, FI-00290 Helsinki, Finland.,Biotech Research & Innovation Centre (BRIC) and the Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, FI-00290 Helsinki, Finland.,Helsinki Institute for Information Technology (HIIT), Department of Computer Science, Aalto University, FI-02150 Espoo, Finland.,Department of Mathematics and Statistics, University of Turku, Quantum, FI-20014 Turku, Finland
| |
Collapse
|
33
|
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: 5.5] [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
|
34
|
Ren J, Li X, Wang L, Liu M, Zheng K, Wang Y. Risk Factors and Drug Resistance of the MDR Acinetobacter Baumannii in Pneumonia Patients in ICU. Open Med (Wars) 2019; 14:772-777. [PMID: 31667355 PMCID: PMC6814959 DOI: 10.1515/med-2019-0090] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 05/20/2019] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To investigate the risk factors and drug resistance of MDR Acinetobacter baumannii in pneumonia patients. METHODS From January 2013 to February 2016, 98 pneumonia patients with MDR Acinetobacter baumannii in our hospital ICU were selected as the observation group, and 49 pneumonia patients with not-MDR Acinetobacter baumannii in our hospital ICU were selected as the control group in accordance with the proportion of 2:1. Sputum samples were collected from the two groups for drug resistance, and the risk factors and prognosis of MDR Acinetobacter baumannii in pneumonia patients were given survey analysis. RESULTS The observation group was highly resistant to cefotaxime, piperacillin, imipenem, levofloxacin, gentamicin, tetracycline and ceftazidime, and was only sensitive to polymyxin. In addition to piperacillin, polymyxin B, the other antimicrobial drug resistance rates in the control group was significantly lower than in the observation group (P<0.05). Univariate analysis showed that diabetes, infection before hospitalization, admission 24h score of APACHE II and GCS scores, deep venous catheterization, and mechanical ventilation were related to the MDR Acinetobacter baumannii in pneumonia patients(P<0.05). Non conditional logistic regression analysis showed that diabetes mellitus, infection before hospitalization, admission 24h score of APACHE II and GCS scores were the independent risk factors for the MDR Acinetobacter baumannii in pneumonia patients(P<0.05). CONCLUSION MDR Acinetobacter baumannii in pneumonia patients in ICU is common, where diabetes infection before hospitalization, admission 24h score of APACHE II and GCS scores are the main risk factors, and the vast majority of the antibiotics are resistant to the MDR Acinetobacter baumannii that can lead to poorer prognosis and followed-up of patients with increased mortality.
Collapse
Affiliation(s)
- Jichen Ren
- Jilin Tumor Hospital,Changchun130012, China
| | - Xiaomeng Li
- Endoscopy Center of China Japan Union Hospital to Jilin University, Changchun130033, P.R.C, China
| | - Libo Wang
- Jilin Tumor Hospital,Changchun130012, China
| | | | - Ke Zheng
- Jilin Tumor Hospital,Changchun130012, China
| | | |
Collapse
|
35
|
Huang R, Zhu H, Shinn P, Ngan D, Ye L, Thakur A, Grewal G, Zhao T, Southall N, Hall MD, Simeonov A, Austin CP. The NCATS Pharmaceutical Collection: a 10-year update. Drug Discov Today 2019; 24:2341-2349. [PMID: 31585169 DOI: 10.1016/j.drudis.2019.09.019] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 09/16/2019] [Accepted: 09/24/2019] [Indexed: 12/25/2022]
Abstract
The National Center for Advancing Translational Sciences (NCATS) Pharmaceutical Collection (NPC), a comprehensive collection of clinically approved drugs, was made a public resource in 2011. Over the past decade, the NPC has been systematically profiled for activity across an array of pathways and disease models, generating an unparalleled amount of data. These data have not only enabled the identification of new repurposing candidates with several in clinical trials, but also uncovered new biological insights into drug targets and disease mechanisms. This retrospective provides an update on the NPC in terms of both successes and lessons learned. We also report our efforts in bringing the NPC up-to-date with drugs approved in recent years.
Collapse
Affiliation(s)
- Ruili Huang
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD 20850, USA.
| | - Hu Zhu
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Paul Shinn
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Deborah Ngan
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Lin Ye
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Ashish Thakur
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Gurmit Grewal
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Tongan Zhao
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Noel Southall
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Mathew D Hall
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Anton Simeonov
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Christopher P Austin
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD 20850, USA
| |
Collapse
|
36
|
Wu B, Yang X, Yan M. Synthesis and Structure-Activity Relationship Study of Antimicrobial Auranofin against ESKAPE Pathogens. J Med Chem 2019; 62:7751-7768. [PMID: 31386365 PMCID: PMC7941214 DOI: 10.1021/acs.jmedchem.9b00550] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Auranofin, an FDA-approved arthritis drug, has recently been repurposed as a potential antimicrobial agent; it performed well against many Gram-positive bacteria, including multidrug resistant strains. It is, however, inactive toward Gram-negative bacteria, for which we are in dire need of new therapies. In this work, 40 auranofin analogues were synthesized by varying the structures of the thiol and phosphine ligands, and their activities were tested against ESKAPE pathogens. The study identified compounds that exhibited bacterial inhibition (MIC) and killing (MBC) activities up to 65 folds higher than that of auranofin, making them effective against Gram-negative pathogens. Both thiol and the phosphine structures influence the activities of the analogues. The trimethylphosphine and triethylphosphine ligands gave the highest activities against Gram-negative and Gram-positive bacteria, respectively. Our SAR study revealed that the thiol ligand is also very important, the structure of which can modulate the activities of the AuI complexes for both Gram-negative and Gram-positive bacteria. Moreover, these analogues had mammalian cell toxicities either similar to or lower than that of auranofin.
Collapse
Affiliation(s)
- Bin Wu
- Department of Chemistry, The University of Massachusetts, Lowell, Massachusetts 01854, United States
| | - Xiaojian Yang
- Department of Chemistry, The University of Massachusetts, Lowell, Massachusetts 01854, United States
| | - Mingdi Yan
- Department of Chemistry, The University of Massachusetts, Lowell, Massachusetts 01854, United States
| |
Collapse
|
37
|
Cheng YS, Williamson PR, Zheng W. Improving therapy of severe infections through drug repurposing of synergistic combinations. Curr Opin Pharmacol 2019; 48:92-98. [PMID: 31454708 DOI: 10.1016/j.coph.2019.07.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/19/2019] [Accepted: 07/20/2019] [Indexed: 01/17/2023]
Abstract
Infections from multidrug resistant (MDR) pathogens and emerging viruses present challenges for effective clinical treatments. Drug repurposing and combination screens may provide therapies at a fraction of the time and cost of traditional methods of drug development. Synergistic combinations of two or three known compounds can increase therapeutic efficacy and reduce concentrations required for individual drugs, in turn, reducing the risk of drug toxicity. Using libraries of approved drugs, traditionally non-antibiotic compounds identified in repurposing screens can quickly move into clinical trials, since safety profiles have been previously established. Herein we summarize recent advances in identifying synergistic drug combinations and the use of drug screens for personalized medicine treatments of infections caused by MDR pathogens and emerging viruses.
Collapse
Affiliation(s)
- Yu-Shan Cheng
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892-3375, USA
| | - Peter R Williamson
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States.
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD 20892-3375, USA.
| |
Collapse
|
38
|
Auranofin Activity Exposes Thioredoxin Reductase as a Viable Drug Target in Mycobacterium abscessus. Antimicrob Agents Chemother 2019; 63:AAC.00449-19. [PMID: 31262763 DOI: 10.1128/aac.00449-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/23/2019] [Indexed: 02/01/2023] Open
Abstract
Nontuberculous mycobacteria (NTM) are highly drug-resistant, opportunistic pathogens that can cause pulmonary disease. The outcomes of the currently recommended treatment regimens are poor, especially for Mycobacterium abscessus New or repurposed drugs are direly needed. Auranofin, a gold-based antirheumatic agent, was investigated for Mycobacterium tuberculosis Here, we test auranofin against NTM in vitro and ex vivo We tested the susceptibility of 63 NTM isolates to auranofin using broth microdilution. Next, we assessed synergy between auranofin and antimycobacterial drugs using the checkerboard method and calculated the fractional inhibition concentration index (FICI). Using time-kill kinetics assays (TK), we assessed pharmacodynamics of auranofin alone and in combination with drug combinations showing the lowest FICIs for M. abscessus CIP 104536. A response surface analysis was used to assess synergistic interactions over time in TKs. Primary isolated macrophages were infected with M. abscessus and treated with auranofin. Finally, using KEGG Orthology, we looked for orthologues to auranofins drug target in M. tuberculosis M. abscessus had the lowest auranofin MIC50 (2 μg/ml) among the tested NTM. The lowest average FICIs were observed between auranofin and amikacin (0.45) and linezolid (0.50). Auranofin exhibited concentration-dependent killing of M. abscessus, with >1-log killing at concentrations of >2× MIC. Only amikacin was synergistic with auranofin according to Bliss independence. Auranofin could not lower the intracellular bacterial load in macrophages. Auranofin itself may not be feasible for M. abscessus treatment, but these data point toward a promising, unutilized drug target.
Collapse
|
39
|
Peyclit L, Baron SA, Rolain JM. Drug Repurposing to Fight Colistin and Carbapenem-Resistant Bacteria. Front Cell Infect Microbiol 2019; 9:193. [PMID: 31245302 PMCID: PMC6579884 DOI: 10.3389/fcimb.2019.00193] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/20/2019] [Indexed: 12/23/2022] Open
Abstract
The emergence of new resistance mechanisms, the failure of classical antibiotics in clinic, the decrease in the development of antibiotics in the industry are all challenges that lead us to consider new strategies for the treatment of infectious diseases. Indeed, in recent years controversy has intensified over strains resistant to carbapenem and/or colistin. Various therapeutic solutions are used to overcome administration of last line antibiotics. In this context, drug repurposing, which consists of using a non-antibiotic compound to treat multi-drug resistant bacteria (MDR), is encouraged. In this review, we first report what may have led to drug repurposing. Main definitions, advantages and drawbacks are summarized. Three major methods are described: phenotypic, computational and serendipity. In a second time we will focus on the current knowledge in drug repurposing for carbapenem and colistin-resistant bacteria with different studies describing repurposed compounds tested on Gram-negative bacteria. Furthermore, we show that drug combination therapies can increase successful by drug repurposing strategy. In conclusion, we discuss the pharmaceutical industries that have little interest in reprofiling drugs due to lack of profits. We also consider what a clinician might think of the indications of these uncommon biologists to treat MDR bacterial infections and avoid therapeutic impasses.
Collapse
Affiliation(s)
- Lucie Peyclit
- Faculté de Médecine et de Pharmacie, IRD, APHM, MEPHI, Aix Marseille Univ, Marseille, France.,IHU Méditerranée Infection, Marseille, France
| | - Sophie Alexandra Baron
- Faculté de Médecine et de Pharmacie, IRD, APHM, MEPHI, Aix Marseille Univ, Marseille, France.,IHU Méditerranée Infection, Marseille, France
| | - Jean-Marc Rolain
- Faculté de Médecine et de Pharmacie, IRD, APHM, MEPHI, Aix Marseille Univ, Marseille, France.,IHU Méditerranée Infection, Marseille, France
| |
Collapse
|
40
|
Cheng YS, Sun W, Xu M, Shen M, Khraiwesh M, Sciotti RJ, Zheng W. Repurposing Screen Identifies Unconventional Drugs With Activity Against Multidrug Resistant Acinetobacter baumannii. Front Cell Infect Microbiol 2019; 8:438. [PMID: 30662875 PMCID: PMC6328479 DOI: 10.3389/fcimb.2018.00438] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 12/10/2018] [Indexed: 12/28/2022] Open
Abstract
Antibiotic-resistant nosocomial infections are an emerging public health issue; carbapenem-resistant gram-negative bacteria such as Acinetobacter baumannii are among the pathogens against which new therapeutic agents are desperately needed. Drug repurposing has recently emerged as an alternative approach to rapidly identifying effective drugs and drug combinations to combat drug resistant bacteria. We performed a drug repurposing screen against a highly virulent, multidrug resistant, Acinetobacter baumannii strain AB5075. This strain, isolated from a patient, is resistant to 25 first-line antibiotics for gram-negative bacteria. A compound screen using a bacterial growth assay led to identification and confirmation of 43 active compounds. Among these confirmed compounds, seven are approved drugs or pharmacologically active compounds for non-antimicrobial indications. Three of these drugs, 5-fluorouracil, fluspirilene, and Bay 11-7082 resensitized strain AB5075 to azithromycin and colistin in a two-drug combination format. The approach using a drug repurposing screen with a pathogen sample isolated from a patient and a high throughput bacterial growth assay led to the successful identification of new drug combinations to overcome a multidrug resistant bacterial infection.
Collapse
Affiliation(s)
- Yu-Shan Cheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| | - Wei Sun
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| | - Miao Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| | - Min Shen
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| | - Mozna Khraiwesh
- Experimental Therapeutics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Richard J Sciotti
- Experimental Therapeutics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
41
|
Ramalingam V, Rajaram R. 2-Ethoxycarbonyl-2-β-hydroxy-a-nor-cholest-5-ene-4one: Extraction, structural characterization, antimicrobial, antioxidant, anticancer and acute toxicity studies. Steroids 2018; 140:11-23. [PMID: 30149072 DOI: 10.1016/j.steroids.2018.08.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 08/20/2018] [Accepted: 08/21/2018] [Indexed: 12/27/2022]
Abstract
Identification and characterization of marine natural products with antimicrobial, antioxidant activity with minimal toxicity has received much interest over the past few years. Among, Acropora formosa is one of the unexplored marine organism for the screening of natural products in marine resources. In this study, a novel steroid 2-ethoxycarbonyl-2-β-hydroxy-A-nor-cholest-5-ene-4one (ECHC) was isolated from butanol extracts of A. formosa using vacuum liquid chromatography and sequentially purified by column chromatography. The chemical structure of the compound was elucidated based on spectroscopic analysis including GC-MS, 1H NMR and 13C NMR and identified as ECHC. Moreover, in vitro antioxidant activity showed that ECHC was highly scavenged the oxidative stress generative molecules. The in vitro cytotoxic activity of ECHC showed excellent activity against human breast cancer cells. Further, in vivo acute toxicity of ECHC on zebrafish Danio rerio was showed no toxicity as well as no morphological damage was observed after 21 days exposure. Histological analysis revealed that there is no apparent difference was observed between ECHC exposure and control group of D. rerio. Together, these results confirmed that ECHC has in vitro antioxidant and anticancer activity and could be developed as a potential drug against most contagious disease like cancer.
Collapse
Affiliation(s)
- Vaikundamoorthy Ramalingam
- DNA Barcoding and Marine Genomics Lab, Department of Marine Science, Bharathidasan University, Tiruchirappalli, India
| | - Rajendran Rajaram
- DNA Barcoding and Marine Genomics Lab, Department of Marine Science, Bharathidasan University, Tiruchirappalli, India.
| |
Collapse
|
42
|
Torres NS, Montelongo-Jauregui D, Abercrombie JJ, Srinivasan A, Lopez-Ribot JL, Ramasubramanian AK, Leung KP. Antimicrobial and Antibiofilm Activity of Synergistic Combinations of a Commercially Available Small Compound Library With Colistin Against Pseudomonas aeruginosa. Front Microbiol 2018; 9:2541. [PMID: 30410476 PMCID: PMC6209680 DOI: 10.3389/fmicb.2018.02541] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 10/04/2018] [Indexed: 01/21/2023] Open
Abstract
Biofilm-associated Pseudomonas aeruginosa infections remain a significant clinical challenge since the conventional antibiotic treatment or combination therapies are largely ineffective; and new approaches are needed. To circumvent the major challenges associated with discovery of new antimicrobials, we have screened a library of compounds that are commercially available and approved by the FDA (Prestwick Chemical Library) against P. aeruginosa for effective antimicrobial and anti-biofilm activity. A preliminary screen of the Prestwick Chemical Library alone did not yield any repositionable candidates, but in a screen of combinations with a fixed sub-inhibitory concentration of the antibiotic colistin we observed 10 drugs whose bacterial inhibiting activity was reproducibly enhanced, seven of which were enhanced by more than 50%. We performed checkerboard assays of these seven drugs in combination with colistin against planktonic cells, and analysis of their interactions over the complete combination matrix using the Zero Interaction Potency (ZIP) model revealed interactions that varied from highly synergistic to completely antagonistic. Of these, five combinations that showed synergism were down-selected and tested against preformed biofilms of P. aeruginosa. Two of the five combinations were active against preformed biofilms of both laboratory and clinical strain of P. aeruginosa, resulting in a 2-log reduction in culturable cells. In summary, we have identified synergistic combinations of five commercially available, FDA-approved drugs and colistin that show antimicrobial activity against planktonic P. aeruginosa (Clomiphene Citrate, Mitoxantrone Dihydrochloride, Methyl Benzethonium Chloride, Benzethonium Chloride, and Auranofin) as well as two combinations (Auranofin and Clomiphene Citrate) with colistin that show antibiofilm activity.
Collapse
Affiliation(s)
- Nelson S Torres
- Dental and Craniofacial Trauma Research and Tissue Regeneration Directorate, Institute of Surgical Research, San Antonio, TX, United States.,Department of Biomedical Engineering, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Daniel Montelongo-Jauregui
- Department of Biology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Johnathan J Abercrombie
- Dental and Craniofacial Trauma Research and Tissue Regeneration Directorate, Institute of Surgical Research, San Antonio, TX, United States
| | - Anand Srinivasan
- Department of Biomedical Engineering, The University of Texas at San Antonio, San Antonio, TX, United States.,Department of Biology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Jose L Lopez-Ribot
- Department of Biology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Anand K Ramasubramanian
- Department of Chemical and Materials Engineering, San José State University, San José, CA, United States
| | - Kai P Leung
- Dental and Craniofacial Trauma Research and Tissue Regeneration Directorate, Institute of Surgical Research, San Antonio, TX, United States
| |
Collapse
|
43
|
Sun W, Hesse S, Xu M, Childs RW, Zheng W, Williamson PR. "Real-Time" High-Throughput Drug and Synergy Testing for Multidrug-Resistant Bacterial Infection: A Case Report. Front Med (Lausanne) 2018; 5:267. [PMID: 30298132 PMCID: PMC6160733 DOI: 10.3389/fmed.2018.00267] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 09/03/2018] [Indexed: 11/17/2022] Open
Abstract
Antibiotic management of infections with multidrug-resistant organisms (MDRO) represents a complex clinical challenge. We report here the first patient with a severe MDRO infection managed with assistance of a novel “real-time” 3-day high-throughput screen (HTS) that allowed screening of 9 drugs in 14 combinations in 2,304 total samplings. Identified synergies were used to modify patient therapy with the goal of reducing drug-induced toxicity. The desired clinical outcome was achieved on the HTS-informed therapeutic regimen, supporting the utility of HTS technology to expand standard antimicrobial susceptibility testing.
Collapse
Affiliation(s)
- Wei Sun
- National Center for Advancing Translational Sciences, Bethesda, MD, United States
| | - Shayla Hesse
- Laboratory of Molecular Biology, National Cancer Institute, Bethesda, MD, United States
| | - Miao Xu
- National Center for Advancing Translational Sciences, Bethesda, MD, United States
| | - Richard W Childs
- Transplantation Immunotherapy, Hematology Branch, National Heart Lung and Blood Institute, Bethesda, MD, United States
| | - Wei Zheng
- National Center for Advancing Translational Sciences, Bethesda, MD, United States
| | - Peter R Williamson
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
44
|
Gorshkov K, Chen CZ, Marshall RE, Mihatov N, Choi Y, Nguyen DT, Southall N, Chen KG, Park JK, Zheng W. Advancing precision medicine with personalized drug screening. Drug Discov Today 2018; 24:272-278. [PMID: 30125678 DOI: 10.1016/j.drudis.2018.08.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/26/2018] [Accepted: 08/13/2018] [Indexed: 01/15/2023]
Abstract
Personalized drug screening (PDS) of approved drug libraries enables rapid development of specific small-molecule therapies for individual patients. With a multidisciplinary team including clinicians, researchers, ethicists, informaticians and regulatory professionals, patient treatment can be optimized with greater efficacy and fewer adverse effects by using PDS as an approach to find remedies. In addition, PDS has the potential to rapidly identify therapeutics for a patient suffering from a disease without an existing therapy. From cancer to bacterial infections, we review specific maladies addressed with PDS campaigns. We predict that PDS combined with personal genomic analyses will contribute to the development of future precision medicine endeavors.
Collapse
Affiliation(s)
- Kirill Gorshkov
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892-3375, USA
| | - Catherine Z Chen
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892-3375, USA
| | - Raisa E Marshall
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892-3375, USA
| | - Nino Mihatov
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892-3375, USA
| | - Yong Choi
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892-3375, USA
| | - Dac-Trung Nguyen
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892-3375, USA
| | - Noel Southall
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892-3375, USA
| | - Kevin G Chen
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892-3375, USA
| | - John K Park
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892-3375, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892-3375, USA.
| |
Collapse
|
45
|
Meng Y, Chen CW, Yung MMH, Sun W, Sun J, Li Z, Li J, Li Z, Zhou W, Liu SS, Cheung ANY, Ngan HYS, Braisted JC, Kai Y, Peng W, Tzatsos A, Li Y, Dai Z, Zheng W, Chan DW, Zhu W. DUOXA1-mediated ROS production promotes cisplatin resistance by activating ATR-Chk1 pathway in ovarian cancer. Cancer Lett 2018; 428:104-116. [PMID: 29704517 PMCID: PMC7474466 DOI: 10.1016/j.canlet.2018.04.029] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 04/18/2018] [Accepted: 04/20/2018] [Indexed: 01/16/2023]
Abstract
The acquisition of resistance is a major obstacle to the clinical use of platinum drugs for ovarian cancer treatment. Increase of DNA damage response is one of major mechanisms contributing to platinum-resistance. However, how DNA damage response is regulated in platinum-resistant ovarian cancer cells remains unclear. Using quantitative high throughput combinational screen (qHTCS) and RNA-sequencing (RNA-seq), we show that dual oxidase maturation factor 1 (DUOXA1) is overexpressed in platinum-resistant ovarian cancer cells, resulting in over production of reactive oxygen species (ROS). Elevated ROS level sustains the activation of ATR-Chk1 pathway, leading to resistance to cisplatin in ovarian cancer cells. Moreover, using qHTCS we identified two Chk1 inhibitors (PF-477736 and AZD7762) that re-sensitize resistant cells to cisplatin. Blocking this novel pathway by inhibiting ROS, DUOXA1, ATR or Chk1 effectively overcomes cisplatin resistance in vitro and in vivo. Significantly, the clinical studies also confirm the activation of ATR and DOUXA1 in ovarian cancer patients, and elevated DOUXA1 or ATR-Chk1 pathway correlates with poor prognosis. Taken together, our findings not only reveal a novel mechanism regulating cisplatin resistance, but also provide multiple combinational strategies to overcome platinum-resistance in ovarian cancer.
Collapse
Affiliation(s)
- Yunxiao Meng
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA; GW Cancer Center, The George Washington University, Washington, DC, 20052, USA
| | - Chi-Wei Chen
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA; GW Cancer Center, The George Washington University, Washington, DC, 20052, USA
| | - Mingo M H Yung
- Department of Obstetrics and Gynecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Wei Sun
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jing Sun
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA; GW Cancer Center, The George Washington University, Washington, DC, 20052, USA
| | - Zhuqing Li
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA; GW Cancer Center, The George Washington University, Washington, DC, 20052, USA
| | - Jing Li
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA; GW Cancer Center, The George Washington University, Washington, DC, 20052, USA
| | - Zongzhu Li
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA; GW Cancer Center, The George Washington University, Washington, DC, 20052, USA
| | - Wei Zhou
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA; GW Cancer Center, The George Washington University, Washington, DC, 20052, USA; Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Stephanie S Liu
- Department of Obstetrics and Gynecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Annie N Y Cheung
- Department of Pathology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Hextan Y S Ngan
- Department of Obstetrics and Gynecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - John C Braisted
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yan Kai
- GW Cancer Center, The George Washington University, Washington, DC, 20052, USA; Department of Physics, The George Washington University Columbian College of Arts & Sciences, Washington, DC, 20052, USA
| | - Weiqun Peng
- Department of Physics, The George Washington University Columbian College of Arts & Sciences, Washington, DC, 20052, USA
| | - Alexandros Tzatsos
- GW Cancer Center, The George Washington University, Washington, DC, 20052, USA; Department of Anatomy and Regenerative Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Yiliang Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, 300192, China
| | - Zhijun Dai
- Department of Oncology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - David W Chan
- Department of Obstetrics and Gynecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| | - Wenge Zhu
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA; GW Cancer Center, The George Washington University, Washington, DC, 20052, USA.
| |
Collapse
|
46
|
Sima N, Sun W, Gorshkov K, Shen M, Huang W, Zhu W, Xie X, Zheng W, Cheng X. Small Molecules Identified from a Quantitative Drug Combinational Screen Resensitize Cisplatin's Response in Drug-Resistant Ovarian Cancer Cells. Transl Oncol 2018; 11:1053-1064. [PMID: 29982103 PMCID: PMC6034569 DOI: 10.1016/j.tranon.2018.06.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 06/10/2018] [Accepted: 06/11/2018] [Indexed: 12/29/2022] Open
Abstract
Drug resistance to chemotherapy occurs in many ovarian cancer patients resulting in failure of treatment. Exploration of drug resistance mechanisms and identification of new therapeutics that overcome the drug resistance can improve patient prognosis. Following a quantitative combination screen of 6060 approved drugs and bioactive compounds in a cisplatin-resistant A2780-cis ovarian cancer cell line, 38 active compounds with IC50s under 1 μM suppressed the growth of cisplatin-resistant ovarian cancer cells. Among these confirmed compounds, CUDC-101, OSU-03012, oligomycin A, VE-821, or Torin2 in a combination with cisplatin restored cisplatin's apoptotic response in the A2780-cis cells, while SR-3306, GSK-923295, SNX-5422, AT-13387, and PF-05212384 directly suppressed the growth of A2780-cis cells. One of the mechanisms for overcoming cisplatin resistance in these cells is mediated by the inhibition of epidermal growth factor receptor (EGFR), though not all the EGFR inhibitors are equally active. The increased levels of total EGFR and phosphorylated-EGFR (p-EGFR) in the A2780-cis cells were reduced after the combined treatment of cisplatin with EGFR inhibitors. In addition, a knockdown of EGFR mRNA reduced cisplatin resistance in the A2780-cis cells. Therefore, the top active compounds identified in this work can be studied further as potential treatments for cisplatin-resistant ovarian cancer. The quantitative combinational screening approach is a useful method for identifying effective compounds and drug combinations against drug-resistant cancer cells.
Collapse
Affiliation(s)
- Ni Sima
- Department of Gynecologic Oncology, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Wei Sun
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Kirill Gorshkov
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Min Shen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Wei Huang
- Department of Gynecologic Oncology, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Wenge Zhu
- Department of Biochemistry and Molecular Biology, The George Washington University Medical School, Washington, DC
| | - Xing Xie
- Department of Gynecologic Oncology, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA.
| | - Xiaodong Cheng
- Department of Gynecologic Oncology, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China.
| |
Collapse
|
47
|
Zhou W, Sun W, Yung MMH, Dai S, Cai Y, Chen CW, Meng Y, Lee JB, Braisted JC, Xu Y, Southall NT, Shinn P, Huang X, Song Z, Chen X, Kai Y, Cai X, Li Z, Hao Q, Cheung ANY, Ngan HYS, Liu SS, Barak S, Hao J, Dai Z, Tzatsos A, Peng W, Pei H, Han Z, Chan DW, Zheng W, Zhu W. Autocrine activation of JAK2 by IL-11 promotes platinum drug resistance. Oncogene 2018; 37:3981-3997. [PMID: 29662190 PMCID: PMC6054535 DOI: 10.1038/s41388-018-0238-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/20/2018] [Accepted: 03/02/2018] [Indexed: 02/08/2023]
Abstract
Antineoplastic platinum agents are used in first-line treatment of ovarian cancer, but treatment failure frequently results from platinum drug resistance. Emerging observations suggest a role of reactive oxygen species (ROS) in the resistance of cancer drugs including platinum drugs. However, the molecular link between ROS and cellular survival pathway is poorly understood. Using quantitative high-throughput combinational screen (qHTCS) and genomic sequencing, we show that in platinum-resistant ovarian cancer elevated ROS levels sustain high level of IL-11 by stimulating FRA1-mediated IL-11 expression and increased IL-11 causes resistance to platinum drugs by constitutively activating JAK2-STAT5 via an autocrine mechanism. Inhibition of JAK2 by LY2784544 or IL-11 by anti-IL-11 antibody overcomes the platinum resistance in vitro or in vivo. Significantly, clinic studies also confirm the activated IL-11-JAK2 pathway in platinum-resistant ovarian cancer patients, which highly correlates with poor prognosis. These findings not only identify a novel ROS-IL-11-JAK2-mediated platinum resistance mechanism but also provide a new strategy for using LY2784544- or IL-11-mediated immunotherapy to treat platinum-resistant ovarian cancer.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA
| | - Wei Sun
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mingo M H Yung
- Department of Obstetrics and Gynecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sheng Dai
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yihua Cai
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA
| | - Chi-Wei Chen
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA
| | - Yunxiao Meng
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA
| | - Jennifer B Lee
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA
| | - John C Braisted
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yinghua Xu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Noel T Southall
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Paul Shinn
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xuefeng Huang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Zhangfa Song
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Xiulei Chen
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Yan Kai
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA
- Department of Physics, The George Washington University Columbian College of Arts & Sciences, Washington, DC, 20052, USA
| | - Xin Cai
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA
| | - Zongzhu Li
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA
| | - Qiang Hao
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA
| | - Annie N Y Cheung
- Department of Pathology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Hextan Y S Ngan
- Department of Pathology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Stephanie S Liu
- Department of Pathology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Stephanie Barak
- Department of Pathology, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Jing Hao
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Zhijun Dai
- Department of Oncology, the Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Alexandros Tzatsos
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA
- Department of Anatomy and Regenerative Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Weiqun Peng
- Department of Physics, The George Washington University Columbian College of Arts & Sciences, Washington, DC, 20052, USA
| | - Huadong Pei
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA
| | - Zhiyong Han
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - David W Chan
- Department of Obstetrics and Gynecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Wenge Zhu
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA.
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA.
| |
Collapse
|
48
|
Synergistic combination of two antimicrobial agents closing each other's mutant selection windows to prevent antimicrobial resistance. Sci Rep 2018; 8:7237. [PMID: 29740150 PMCID: PMC5940791 DOI: 10.1038/s41598-018-25714-z] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 04/27/2018] [Indexed: 12/18/2022] Open
Abstract
Antimicrobial resistance seriously threatened human health. Combination therapy is generally an effective strategy to fight resistance, while some data on its effects are conflicting. To explore the reasons, the fractional inhibitory concentration indexes (FICIs) of three designed combinations against methicillin-resistant Staphylococcus aureus (MRSA) were determined using checkerboard method, and their minimal concentrations inhibiting colony formation by 99% (MIC99%s) and mutant prevention concentrations (MPCs) alone or in combinations including different proportions were first determined using agar plates. The results indicated that different proportions of a combination had presented different MPCs and mutant selection window (MSWs), and also showed that the smaller the FICIs of two agents in combinations were, the more probable their MSWs were to close each other. As two agents of a combination had different pharmacokinetic characters, the ratios of two agents in blood and infectious sites were likely different even though a specific proportion was administrated, which would lead to different effects preventing resistance. Thereby, these experimental results theoretically indicated that synergistic combination closing each other’s MSWs had a great potency to prevent resistance according to the hypotheses of MSW and MPC, and deduced that in vivo synergistic validity of a combination was likely a key to prevent resistance. Moreover, a synergistic combination of roxithromycin/doxycycline with the FICIs of 0.26–0.50 and 0.28–0.38 respectively against MRSA 01 and 02 was obtained, and the MSWs of these two agents could be simultaneously closed each other in a certain range of proportions, but for others. Meanwhile, its effect preventing resistance needs to be further verified.
Collapse
|
49
|
Coussens NP, Molinaro AL, Culbertson KJ, Peryea T, Zahoránszky-Köhalmi G, Hall MD, Daines DA. Better living through chemistry: Addressing emerging antibiotic resistance. Exp Biol Med (Maywood) 2018; 243:538-553. [PMID: 29409348 PMCID: PMC5882019 DOI: 10.1177/1535370218755659] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The increasing emergence of multidrug-resistant bacteria is recognized as a major threat to human health worldwide. While the use of small molecule antibiotics has enabled many modern medical advances, it has also facilitated the development of resistant organisms. This minireview provides an overview of current small molecule drugs approved by the US Food and Drug Administration (FDA) for use in humans, the unintended consequences of antibiotic use, and the mechanisms that underlie the development of drug resistance. Promising new approaches and strategies to counter antibiotic-resistant bacteria with small molecules are highlighted. However, continued public investment in this area is critical to maintain an edge in our evolutionary "arms race" against antibiotic-resistant microorganisms. Impact statement The alarming increase in antibiotic-resistant microorganisms is a rapidly emerging threat to human health throughout the world. Historically, small molecule drugs have played a major role in controlling bacterial infections and they continue to offer tremendous potential in countering resistant organisms. This minireview provides a broad overview of the relevant issues, including the diversity of FDA-approved small molecule drugs and mechanisms of drug resistance, unintended consequences of antibiotic use, the current state of development for small molecule antibacterials and financial challenges that impact progress towards novel therapies. The content will be informative to diverse stakeholders, including clinicians, basic scientists, translational scientists and policy makers, and may be used as a bridge between these key players to advance the development of much-needed therapeutics.
Collapse
Affiliation(s)
- Nathan P Coussens
- National Center for Advancing Translational Sciences, Division of Pre-Clinical Innovation, Rockville, MD 20850, USA
| | - Ashley L Molinaro
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| | - Kayla J Culbertson
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| | - Tyler Peryea
- National Center for Advancing Translational Sciences, Division of Pre-Clinical Innovation, Rockville, MD 20850, USA
| | - Gergely Zahoránszky-Köhalmi
- National Center for Advancing Translational Sciences, Division of Pre-Clinical Innovation, Rockville, MD 20850, USA
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, Division of Pre-Clinical Innovation, Rockville, MD 20850, USA
| | - Dayle A Daines
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| |
Collapse
|
50
|
Wang C, Gu B, Liu Q, Pang Y, Xiao R, Wang S. Combined use of vancomycin-modified Ag-coated magnetic nanoparticles and secondary enhanced nanoparticles for rapid surface-enhanced Raman scattering detection of bacteria. Int J Nanomedicine 2018. [PMID: 29520142 PMCID: PMC5834169 DOI: 10.2147/ijn.s150336] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Pathogenic bacteria have always been a significant threat to human health. The detection of pathogens needs to be rapid, accurate, and convenient. Methods We present a sensitive surface-enhanced Raman scattering (SERS) biosensor based on the combination of vancomycin-modified Ag-coated magnetic nanoparticles (Fe3O4@Ag-Van MNPs) and Au@Ag nanoparticles (NPs) that can effectively capture and discriminate bacterial pathogens from solution. The high-performance Fe3O4@Ag MNPs were modified with vancomycin and used as bacteria capturer for magnetic separation and enrichment. The modified MNPS were found to exhibit strong affinity with a broad range of Gram-positive and Gram-negative bacteria. After separating and rinsing bacteria, Fe3O4@Ag-Van MNPs and Au@Ag NPs were synergistically used to construct a very large number of hot spots on bacteria cells, leading to ultrasensitive SERS detection. Results The dominant merits of our dual enhanced strategy included high bacterial-capture efficiency (>65%) within a wide pH range (pH 3.0–11.0), a short assay time (<30 min), and a low detection limit (5×102 cells/mL). Moreover, the spiked tests show that this method is still valid in milk and blood samples. Owing to these capabilities, the combined system enabled the sensitive and specific discrimination of different pathogens in complex solution, as verified by its detection of Gram-positive bacterium Escherichia coli, Gram-positive bacterium Staphylococcus aureus, and methicillin-resistant S. aureus. Conclusion This method has great potential for field applications in food safety, environmental monitoring, and infectious disease diagnosis.
Collapse
Affiliation(s)
- Chongwen Wang
- Beijing Key Laboratory of New Molecular Diagnosis Technologies for Infectious Diseases, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.,College of Life Sciences and Bio-Engineering, Beijing University of Technology, Beijing, People's Republic of China
| | - Bing Gu
- Medical Technology School, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Qiqi Liu
- Beijing Key Laboratory of New Molecular Diagnosis Technologies for Infectious Diseases, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Yuanfeng Pang
- College of Life Sciences and Bio-Engineering, Beijing University of Technology, Beijing, People's Republic of China.,Department of Toxicology, Capital Medical University, Beijing, People's Republic of China
| | - Rui Xiao
- Beijing Key Laboratory of New Molecular Diagnosis Technologies for Infectious Diseases, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Shengqi Wang
- Beijing Key Laboratory of New Molecular Diagnosis Technologies for Infectious Diseases, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.,College of Life Sciences and Bio-Engineering, Beijing University of Technology, Beijing, People's Republic of China.,Medical Technology School, Xuzhou Medical University, Xuzhou, People's Republic of China
| |
Collapse
|