1
|
Díaz-Guerrero MÁ, Castillo-Juárez I, Zurabian R, Valdez A, Kota K, Hoshiko Y, Ramesh E, Martínez-Vazquez M, Ceapă CD, Hernandez-Garnica M, Cadet F, García-Contreras R. Reviving the past for a healthier future: ancient molecules and remedies as a solution to the antibiotic crisis. Future Microbiol 2025; 20:429-441. [PMID: 40099865 PMCID: PMC11980515 DOI: 10.1080/17460913.2025.2476290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 03/04/2025] [Indexed: 03/20/2025] Open
Abstract
Options to combat bacterial infections are becoming scarce. We require innovative approaches to enhance the discovery of effective antimicrobials capable of combating bacteria resistant to multiple or all antibiotics. These methods should either directly eliminate resistant bacteria or indirectly influence their viability by inhibiting their virulence or reducing their resistance to antibiotics. One interesting approach is to analyze ancient remedies used to treat bacterial infections, formulate them, and test them against modern microbes. This field has recently been named "ancientbiotics." This approach allows us to leverage centuries of empirical knowledge accumulated, from traditional medicines across various ancient cultures worldwide. The strategy has already yielded promising formulations to combat the ESKAPE group of nosocomial pathogens. Additionally, molecular de-extinction, which involves genome analysis of extinct species to search for useful antimicrobials, such as peptides, offers another avenue. In this review, we compile the antimicrobial effects of ancient remedies and de-extinct molecules known to modern science and discuss possible new strategies to further harness the potential of past remedies and molecules to fight the rise of superbugs.
Collapse
Affiliation(s)
| | - Israel Castillo-Juárez
- Conahcyt-Instituto de Ciencias Básicas e Ingeniería, Universidad Autónoma del Estado de Hidalgo, Mineral de la Reforma, Hidalgo, México
| | - Rimma Zurabian
- Departamento de Microbiología y Parasitología, Facultad de Medicina, UNAM, Mexico City, Mexico
| | - Alejandra Valdez
- Laboratorio de Interacciones Microbianas, Planta Piloto de Procesos Industriales Microbiológicos, PROIMI, CONICET, San Miguel de Tucumán, Tucumán, Argentina
- Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, San Miguel de Tucumán, Argentina
| | - Kokila Kota
- Department of Biology, Ramapo College of New Jersey, Mahwah, NJ, USA
| | - Yuki Hoshiko
- Department of Health Science, School of Allied Health Sciences, Kitasato University, Sagamihara, Japan
| | - Ekaprana Ramesh
- Department of Biology, Ramapo College of New Jersey, Mahwah, NJ, USA
| | | | - Corina Diana Ceapă
- Laboratory of Microbiology, Institute of Chemistry, National Autonomous University of Mexico, Mexico City, Mexico
| | | | - Frederic Cadet
- PEACCEL, Artificial Intelligence Department, AI for Biologics, Paris, France
| | | |
Collapse
|
2
|
Lorente-Torres B, Llano-Verdeja J, Castañera P, Ferrero HÁ, Fernández-Martínez S, Javadimarand F, Mateos LM, Letek M, Mourenza Á. Innovative Strategies in Drug Repurposing to Tackle Intracellular Bacterial Pathogens. Antibiotics (Basel) 2024; 13:834. [PMID: 39335008 PMCID: PMC11428606 DOI: 10.3390/antibiotics13090834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
Intracellular bacterial pathogens pose significant public health challenges due to their ability to evade immune defenses and conventional antibiotics. Drug repurposing has recently been explored as a strategy to discover new therapeutic uses for established drugs to combat these infections. Utilizing high-throughput screening, bioinformatics, and systems biology, several existing drugs have been identified with potential efficacy against intracellular bacteria. For instance, neuroleptic agents like thioridazine and antipsychotic drugs such as chlorpromazine have shown effectiveness against Staphylococcus aureus and Listeria monocytogenes. Furthermore, anticancer drugs including tamoxifen and imatinib have been repurposed to induce autophagy and inhibit bacterial growth within host cells. Statins and anti-inflammatory drugs have also demonstrated the ability to enhance host immune responses against Mycobacterium tuberculosis. The review highlights the complex mechanisms these pathogens use to resist conventional treatments, showcases successful examples of drug repurposing, and discusses the methodologies used to identify and validate these drugs. Overall, drug repurposing offers a promising approach for developing new treatments for bacterial infections, addressing the urgent need for effective antimicrobial therapies.
Collapse
Affiliation(s)
- Blanca Lorente-Torres
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain
| | - Jesús Llano-Verdeja
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain
| | - Pablo Castañera
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain
| | - Helena Á Ferrero
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain
| | | | - Farzaneh Javadimarand
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain
| | - Luis M Mateos
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain
- Instituto de Biología Molecular, Genómica y Proteómica (INBIOMIC), Universidad de León, 24071 León, Spain
| | - Michal Letek
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain
- Instituto de Desarrollo Ganadero y Sanidad Animal (INDEGSAL), Universidad de León, 24071 León, Spain
| | - Álvaro Mourenza
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071 León, Spain
| |
Collapse
|
3
|
Mathuria A, Ali N, Kataria N, Mani I. Drug repurposing for fungal infections. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 207:59-78. [PMID: 38942545 DOI: 10.1016/bs.pmbts.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
The rise of multidrug-resistant bacteria is a well-recognized threat to world health, necessitating the implementation of effective treatments. This issue has been identified as a top priority on the global agenda by the World Health Organization. Certain strains, such as Candida glabrata, Candida krusei, Candida lusitaniae, Candida auris, select cryptococcal species, and opportunistic Aspergillus or Fusarium species, have significant intrinsic resistance to numerous antifungal medicines. This inherent resistance and subsequent suboptimal clinical outcomes underscore the critical imperative for enhanced therapeutic alternatives and management protocols. The challenge of effectively treating fungal infections, compounded by the protracted timelines involved in developing novel drugs, underscores the pressing need to explore alternative therapeutic avenues. Among these, drug repurposing emerges as a particularly promising and expeditious solution, providing cost-effective solutions and safety benefits. In the fight against life-threatening resistant fungal infections, the idea of repurposing existing medications has encouraged research into both established and new compounds as a last-resort therapy. This chapter seeks to provide a comprehensive overview of contemporary antifungal drugs, as well as their key resistance mechanisms. Additionally, it seeks to provide insight into the antimicrobial properties of non-traditional drugs, thereby offering a holistic perspective on the evolving landscape of antifungal therapeutics.
Collapse
Affiliation(s)
- Anshu Mathuria
- Department of Biochemistry, Sri Venkateswara College, University of Delhi, New Delhi, India
| | - Namra Ali
- Department of Microbiology, Gargi College, University of Delhi, New Delhi, India
| | - Naina Kataria
- Department of Biochemistry, Sri Venkateswara College, University of Delhi, New Delhi, India
| | - Indra Mani
- Department of Microbiology, Gargi College, University of Delhi, New Delhi, India.
| |
Collapse
|
4
|
Ashok G, Basu S, Priyamvada P, Anbarasu A, Chintala S, Ramaiah S. Coinfections in human papillomavirus associated cancers and prophylactic recommendations. Rev Med Virol 2024; 34:e2524. [PMID: 38375992 DOI: 10.1002/rmv.2524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/01/2023] [Accepted: 02/06/2024] [Indexed: 02/21/2024]
Abstract
The Human Papillomavirus (HPV) infection is responsible for more than 80% of reported cervical cancer and other virus-associated tumours. Although this global threat can be controlled using effective vaccination strategies, a growing perturbation of HPV infection is an emerging coinfection likely to increase the severity of the infection in humans. Moreover, these coinfections prolong the HPV infections, thereby risking the chances for oncogenic progression. The present review consolidated the clinically significant microbial coinfections/co-presence associated with HPV and their underlying molecular mechanisms. We discussed the gaps and concerns associated with demography, present vaccination strategies, and other prophylactic limitations. We concluded our review by highlighting the potential clinical as well as emerging computational intervention measures to kerb down HPV-associated severities.
Collapse
Affiliation(s)
- Gayathri Ashok
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
- Department of Bio-Sciences, SBST, VIT, Vellore, Tamil Nadu, India
| | - Soumya Basu
- Department of Biotechnology, SBST, VIT, Vellore, Tamil Nadu, India
- Department of Biotechnology, NIST University, Berhampur, Odisha, India
| | | | - Anand Anbarasu
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
- Department of Biotechnology, SBST, VIT, Vellore, Tamil Nadu, India
| | - Sreenivasulu Chintala
- Department of Pediatrics, Indiana University, School of Medicine, Indianapolis, Indiana, USA
| | - Sudha Ramaiah
- Medical and Biological Computing Laboratory, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
- Department of Bio-Sciences, SBST, VIT, Vellore, Tamil Nadu, India
| |
Collapse
|
5
|
Svedholm E, Bruce B, Parcell BJ, Coote PJ. Repurposing Mitomycin C in Combination with Pentamidine or Gentamicin to Treat Infections with Multi-Drug-Resistant (MDR) Pseudomonas aeruginosa. Antibiotics (Basel) 2024; 13:177. [PMID: 38391563 PMCID: PMC10886254 DOI: 10.3390/antibiotics13020177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 02/24/2024] Open
Abstract
The aims of this study were (i) to determine if the combination of mitomycin C with pentamidine or existing antibiotics resulted in enhanced efficacy versus infections with MDR P. aeruginosa in vivo; and (ii) to determine if the doses of mitomycin C and pentamidine in combination can be reduced to levels that are non-toxic in humans but still retain antibacterial activity. Resistant clinical isolates of P. aeruginosa, a mutant strain over-expressing the MexAB-OprM resistance nodulation division (RND) efflux pump and a strain with three RND pumps deleted, were used. MIC assays indicated that all strains were sensitive to mitomycin C, but deletion of three RND pumps resulted in hypersensitivity and over-expression of MexAB-OprM caused some resistance. These results imply that mitomycin C is a substrate of the RND efflux pumps. Mitomycin C monotherapy successfully treated infected Galleria mellonella larvae, albeit at doses too high for human administration. Checkerboard and time-kill assays showed that the combination of mitomycin C with pentamidine, or the antibiotic gentamicin, resulted in synergistic inhibition of most P. aeruginosa strains in vitro. In vivo, administration of a combination therapy of mitomycin C with pentamidine, or gentamicin, to G. mellonella larvae infected with P. aeruginosa resulted in enhanced efficacy compared with monotherapies for the majority of MDR clinical isolates. Notably, the therapeutic benefit conferred by the combination therapy occurred with doses of mitomycin C close to those used in human medicine. Thus, repurposing mitomycin C in combination therapies to target MDR P. aeruginosa infections merits further investigation.
Collapse
Affiliation(s)
- Elin Svedholm
- Biomedical Sciences Research Complex, School of Biology, University of St Andrews, The North Haugh, St Andrews, Fife KY16 9ST, UK
| | - Benjamin Bruce
- Biomedical Sciences Research Complex, School of Biology, University of St Andrews, The North Haugh, St Andrews, Fife KY16 9ST, UK
| | - Benjamin J Parcell
- NHS Tayside, Medical Microbiology, Ninewells Hospital and Medical School, Dundee DD1 9SY, UK
| | - Peter J Coote
- Biomedical Sciences Research Complex, School of Biology, University of St Andrews, The North Haugh, St Andrews, Fife KY16 9ST, UK
| |
Collapse
|
6
|
Eldeghidy A, Abdel-Fattah G, El-Sayed ASA, Abdel-Fattah GG. Production, bioprocessing and antiproliferative activity of camptothecin from Aspergillus terreus, endophyte of Cinnamomum camphora: restoring their biosynthesis by indigenous microbiome of C. camphora. Microb Cell Fact 2023; 22:143. [PMID: 37533061 PMCID: PMC10399021 DOI: 10.1186/s12934-023-02158-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/21/2023] [Indexed: 08/04/2023] Open
Abstract
Fungal producing potency of camptothecin (CPT) raise the hope for their usage to be a platform for industrial production of CPT, nevertheless, attenuation of their productivity of CPT with the subculturing and preservation is the challenge. So, screening for novel endophytic fungal isolates with a reliable CPT-biosynthetic stability was the objective. Among the isolated endophytic fungi from the tested medicinal plants, Aspergillus terreus OQ642314.1, endophyte of Cinnamomum camphora, exhibits the highest yield of CPT (89.4 μg/l). From the NMR, FT-IR and LC-MS/MS analyses, the extracted CPT from A. terreus gave the same structure and molecular mass fragmentation pattern of authentic CPT (349 m/z). The putative CPT had a significant activity against MCF7 (0.27 µM) and HEPG-2 (0.8 µM), with a strong affinity to inhibits the human Topoisomerase 1 activity (IC50 0.362 μg/ml) as revealed from the Gel-based DNA relaxation assay. The purified CPT displayed a strong antimicrobial activity for various bacterial (E. coli and B. cereus) and fungal (A. flavus and A. parasiticus) isolates, ensuring the unique tertiary, and stereo-structure of A. terreus for penetrating the microbial cell walls and targeting the topoisomerase I. The higher dual activity of the purified CPT as antimicrobial and antitumor, emphasize their therapeutic efficiency, especially with growth of the opportunistic microorganisms due to the suppression of human immune system with the CPT uses in vivo. The putative CPT had an obvious activity against the tumor cell (MCF7) metastasis, and migration as revealed from the wound healing assay. The overall yield of A. terreus CPT was maximized with the Blackett-Burman design by twofolds increment (164.8 μg/l). The CPT yield by A. terreus was successively diminished with the multiple fungal subculturing, otherwise, the CPT productivity of A. terreus was restored, and increased over the zero culture upon coculturing with C. camphora microbiome (1.5% w/v), ensuring the restoring of CPT biosynthetic potency of A. terreus by the plant microbiome-derived chemical signals "microbial communication". This is the first report exploring the feasibility of A. terreus "endophyte of C. camphora" to be a preliminary platform for commercial production of CPT with a reliable sustainability upon uses of indigenous C. camphora microbiome.
Collapse
Affiliation(s)
- Abeer Eldeghidy
- Botany and Microbiology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Gamal Abdel-Fattah
- Botany and Microbiology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Ashraf S A El-Sayed
- Enzymology and Fungal Biotechnology Lab, Botany and Microbiology, Faculty of Science, Zagazig University, Zagazig, Egypt.
| | - Ghada G Abdel-Fattah
- Botany and Microbiology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| |
Collapse
|
7
|
Sharma K, Ahmed F, Sharma T, Grover A, Agarwal M, Grover S. Potential Repurposed Drug Candidates for Tuberculosis Treatment: Progress and Update of Drugs Identified in Over a Decade. ACS OMEGA 2023; 8:17362-17380. [PMID: 37251185 PMCID: PMC10210030 DOI: 10.1021/acsomega.2c05511] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/23/2022] [Indexed: 05/31/2023]
Abstract
The devastating impact of Tuberculosis (TB) has been a menace to mankind for decades. The World Health Organization (WHO) End TB Strategy aims to reduce TB mortality up to 95% and 90% of overall TB cases worldwide, by 2035. This incessant urge will be achieved with a breakthrough in either a new TB vaccine or novel drugs with higher efficacy. However, the development of novel drugs is a laborious process involving a timeline of almost 20-30 years with huge expenditure; on the other hand, repurposing previously approved drugs is a viable technique for overcoming current bottlenecks in the identification of new anti-TB agents. The present comprehensive review discusses the progress of almost all the repurposed drugs that have been identified to the present day (∼100) and are in the development or clinical testing phase against TB. We have also emphasized the efficacy of repurposed drugs in combination with already available frontline anti-TB medications along with the scope of future investigations. This study would provide the researchers a detailed overview of nearly all identified anti-TB repurposed drugs and may assist them in selecting the lead compounds for further in vivo/clinical research.
Collapse
Affiliation(s)
- Khushbu Sharma
- Department
of Molecular Medicine, Jamia Hamdard, New Delhi, 110062, India
| | - Faraz Ahmed
- Department
of Molecular Medicine, Jamia Hamdard, New Delhi, 110062, India
| | - Tarina Sharma
- New
Jersey Medical School, Rutgers, The State
University of New Jersey, Newark, New Jersey 07103, United States
| | - Abhinav Grover
- School
of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Meetu Agarwal
- Department
of Molecular Medicine, Jamia Hamdard, New Delhi, 110062, India
| | - Sonam Grover
- Department
of Molecular Medicine, Jamia Hamdard, New Delhi, 110062, India
| |
Collapse
|
8
|
Salim SA, Badawi NM, El-Moslamy SH, Kamoun EA, Daihom BA. Novel long-acting brimonidine tartrate loaded-PCL/PVP nanofibers for versatile biomedical applications: fabrication, characterization and antimicrobial evaluation. RSC Adv 2023; 13:14943-14957. [PMID: 37200698 PMCID: PMC10186146 DOI: 10.1039/d3ra02244g] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/08/2023] [Indexed: 05/20/2023] Open
Abstract
The global state of antibiotic resistance highlights the necessity for new drugs that can treat a wide range of microbial infections. Drug repurposing has several advantages, including lower costs and improved safety compared to developing a new compound. The aim of the current study is to evaluate the repurposed antimicrobial activity of Brimonidine tartrate (BT), a well-known antiglaucoma drug, and to potentiate its antimicrobial effect by using electrospun nanofibrous scaffolds. BT-loaded nanofibers were fabricated in different drug concentrations (1.5, 3, 6, and 9%) via the electrospinning technique using two biopolymers (PCL and PVP). Then, the prepared nanofibers were characterized by SEM, XRD, FTIR, swelling ratio, and in vitro drug release. Afterward, the antimicrobial activities of the prepared nanofibers were investigated in vitro using different methods against several human pathogens and compared to the free BT. The results showed that all nanofibers were prepared successfully with a smooth surface. The diameters of nanofibers were reduced after loading of BT compared to the unloaded ones. In addition, scaffolds showed controlled-drug release profiles that were maintained for more than 7 days. The in vitro antimicrobial assessments revealed good activities for all scaffolds against most of the investigated human pathogens, particularly the one prepared with 9% BT which showed superiority in the antimicrobial effect over other scaffolds. To conclude, our findings proved the capability of nanofibers in loading BT and improving its repurposed antimicrobial efficacy. Therefore, it could be a promising carrier for BT to be used in combating numerous human pathogens.
Collapse
Affiliation(s)
- Samar A Salim
- Nanotechnology Research Center (NTRC), The British University in Egypt (BUE) Cairo 11837 Egypt
| | - Noha M Badawi
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, The British University in Egypt (BUE) Cairo 11837 Egypt
| | - Shahira H El-Moslamy
- Bioprocess Development Department (BID), Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City) New Borg El-Arab City Alexandria 21934 Egypt
- Polymeric Materials Research Dep., Advanced Technology and New Materials Research Institute (ATNMRI), City of Scientific Research and Technological Applications (SRTA-City) Alexandria 21934 Egypt
| | - Elbadawy A Kamoun
- Polymeric Materials Research Dep., Advanced Technology and New Materials Research Institute (ATNMRI), City of Scientific Research and Technological Applications (SRTA-City) Alexandria 21934 Egypt
- Biomaterials for Medical and Pharmaceutical Applications Research Group, Nanotechnology Research Center (NTRC), The British University in Egypt (BUE) Cairo 11837 Egypt
| | - Baher A Daihom
- Department of Pharmaceutics and Industrial Pharmacy, Cairo University Cairo Egypt
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, University of Texas at Austin 78712 USA
| |
Collapse
|
9
|
Rabaan AA, Sulaiman T, Al-Ahmed SH, Buhaliqah ZA, Buhaliqah AA, AlYuosof B, Alfaresi M, Al Fares MA, Alwarthan S, Alkathlan MS, Almaghrabi RS, Abuzaid AA, Altowaileb JA, Al Ibrahim M, AlSalman EM, Alsalman F, Alghounaim M, Bueid AS, Al-Omari A, Mohapatra RK. Potential Strategies to Control the Risk of Antifungal Resistance in Humans: A Comprehensive Review. Antibiotics (Basel) 2023; 12:antibiotics12030608. [PMID: 36978475 PMCID: PMC10045400 DOI: 10.3390/antibiotics12030608] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/09/2023] [Accepted: 03/11/2023] [Indexed: 03/30/2023] Open
Abstract
Fungal infections are becoming one of the main causes of morbidity and mortality in people with weakened immune systems. Mycoses are becoming more common, despite greater knowledge and better treatment methods, due to the regular emergence of resistance to the antifungal medications used in clinical settings. Antifungal therapy is the mainstay of patient management for acute and chronic mycoses. However, the limited availability of antifungal drug classes limits the range of available treatments. Additionally, several drawbacks to treating mycoses include unfavourable side effects, a limited activity spectrum, a paucity of targets, and fungal resistance, all of which continue to be significant issues in developing antifungal drugs. The emergence of antifungal drug resistance has eliminated accessible drug classes as treatment choices, which significantly compromises the clinical management of fungal illnesses. In some situations, the emergence of strains resistant to many antifungal medications is a major concern. Although new medications have been developed to address this issue, antifungal drug resistance has grown more pronounced, particularly in patients who need long-term care or are undergoing antifungal prophylaxis. Moreover, the mechanisms that cause resistance must be well understood, including modifications in drug target affinities and abundances, along with biofilms and efflux pumps that diminish intracellular drug levels, to find novel antifungal drugs and drug targets. In this review, different classes of antifungal agents, and their resistance mechanisms, have been discussed. The latter part of the review focuses on the strategies by which we can overcome this serious issue of antifungal resistance in humans.
Collapse
Affiliation(s)
- Ali A Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan
| | - Tarek Sulaiman
- Infectious Diseases Section, Medical Specialties Department, King Fahad Medical City, Riyadh 12231, Saudi Arabia
| | - Shamsah H Al-Ahmed
- Specialty Paediatric Medicine, Qatif Central Hospital, Qatif 32654, Saudi Arabia
| | - Zainab A Buhaliqah
- Department of Family Medicine, Primary Healthcare Center, Dammam 32433, Saudi Arabia
| | - Ali A Buhaliqah
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Buthina AlYuosof
- Directorate of Public Health, Dammam Network, Eastern Health Cluster, Dammam 31444, Saudi Arabia
| | - Mubarak Alfaresi
- Department of Pathology and Laboratory Medicine, Zayed Military Hospital, Abu Dhabi 3740, United Arab Emirates
- Department of Pathology, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai 505055, United Arab Emirates
| | - Mona A Al Fares
- Department of Internal Medicine, King Abdulaziz University Hospital, Jeddah 21589, Saudi Arabia
| | - Sara Alwarthan
- Department of Internal Medicine, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia
| | - Mohammed S Alkathlan
- Infectious Diseases Department, King Fahad Specialist Hospital, Buraydah 52382, Saudi Arabia
| | - Reem S Almaghrabi
- Organ Transplant Center of Excellence, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - Abdulmonem A Abuzaid
- Medical Microbiology Department, Security Forces Hospital Programme, Dammam 32314, Saudi Arabia
| | - Jaffar A Altowaileb
- Microbiology Laboratory, Laboratory Department, Qatif Central Hospital, Qatif 32654, Saudi Arabia
| | - Maha Al Ibrahim
- Microbiology Laboratory, Laboratory Department, Qatif Central Hospital, Qatif 32654, Saudi Arabia
| | - Eman M AlSalman
- Department of Family Medicine, Primary Health Care Centers, Qatif Health Network, Qatif 31911, Saudi Arabia
| | - Fatimah Alsalman
- Department of Emergency Medicine, Oyun City Hospital, Al-Ahsa 36312, Saudi Arabia
| | | | - Ahmed S Bueid
- Microbiology Laboratory, King Faisal General Hospital, Al-Ahsa 31982, Saudi Arabia
| | - Awad Al-Omari
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Research Center, Dr. Sulaiman Al Habib Medical Group, Riyadh 11372, Saudi Arabia
| | - Ranjan K Mohapatra
- Department of Chemistry, Government College of Engineering, Keonjhar 758002, India
| |
Collapse
|
10
|
El-Sayed ASA, George NM, Abou-Elnour A, El-Mekkawy RM, El-Demerdash MM. Production and bioprocessing of camptothecin from Aspergillus terreus, an endophyte of Cestrum parqui, restoring their biosynthetic potency by Citrus limonum peel extracts. Microb Cell Fact 2023; 22:4. [PMID: 36609265 PMCID: PMC9824926 DOI: 10.1186/s12934-022-02012-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 12/27/2022] [Indexed: 01/07/2023] Open
Abstract
The metabolic potency of fungi as camptothecin producer elevates their prospective use as an industrial platform for commercial production, however, the loss of camptothecin productivity by fungi with the storage and subculturing are the major obstacle. Thus, screening for endophytic fungal isolates inhabiting ethnopharmacological plants with an obvious metabolic stability and sustainability for camptothecin biosynthesis could be one of the most feasible paradigms. Aspergillus terreus ON908494.1, an endophyte of Cestrum parqui was morphologically and molecularly verified, displaying the most potent camptothecin biosynthetic potency. The chemical identity of A. terreus camptothecin was confirmed from the HPLC, FTIR and LC-MS/MS analyses, gave the same molecular structure and mass fragmentation patterns of authentic one. The purified putative camptothecin displayed a strong anticancer activity towards HepG-2 and MCF-7 with IC50 values 0.96 and 1.4 µM, respectively, with no toxicity to OEC normal cells. As well as, the purified camptothecin displayed a significant antifungal activity towards fungal human pathogen Candida albicans, Aspergillus flavus, and A. parasiticus, ensuring the unique structural activity relationships of A. terreus camptothecin, as a powerful dually active anticancer and antimicrobial agent. The camptothecin productivity of A. terreus was maximized by bioprocessing with Plackett-Burman design, with an overall 1.5 folds increment (170.5 µg/L), comparing to control culture. So, the optimal medium components for maximum yield of camptothecin by A. terreus was acid why (2.0 mL/L), Diaion HP20 (2.0 g/L), Amberlite XAD (2.0 g/L), dextrin (5.0 g/L), glucose (10.0 g/L), salicylic acid (2.0 g/L), serine (4.0 g/L), cysteine (4.0 g/L) and glutamate (10.0 g/L), at pH 6 for 15 days incubation. By the 5th generation of A. terreus, the camptothecin yield was reduced by 60%, comparing to zero culture. Interestingly, the productivity of camptothecin by A. terreus has been completely restored and over increased (210 µg/L), comparing to the 3rd generation A. terreus (90 µg/L) upon addition of methanolic extracts of Citrus limonum peels, revealing the presence of some chemical signals that triggers the camptothecin biosynthetic machinery. The feasibility of complete restoring of camptothecin biosynthetic-machinery of A. terreus for stable and sustainable production of camptothecin, pave the way for using this fungal isolate as new platform for scaling-up the camptothecin production.
Collapse
Affiliation(s)
- Ashraf S. A. El-Sayed
- grid.31451.320000 0001 2158 2757Enzymology and Fungal Biotechnology Lab, Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Nelly M. George
- grid.31451.320000 0001 2158 2757Enzymology and Fungal Biotechnology Lab, Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Amira Abou-Elnour
- grid.31451.320000 0001 2158 2757Enzymology and Fungal Biotechnology Lab, Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Rasha M. El-Mekkawy
- grid.31451.320000 0001 2158 2757Enzymology and Fungal Biotechnology Lab, Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Marwa M. El-Demerdash
- grid.31451.320000 0001 2158 2757Enzymology and Fungal Biotechnology Lab, Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig, Egypt
| |
Collapse
|
11
|
Zhao S, Wang Z, Lin Z, Wei G, Wen X, Li S, Yang X, Zhang Q, Jing C, Dai Y, Guo J, He Y. Drug Repurposing by Siderophore Conjugation: Synthesis and Biological Evaluation of Siderophore‐Methotrexate Conjugates as Antibiotics. Angew Chem Int Ed Engl 2022; 61:e202204139. [DOI: 10.1002/anie.202204139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Sheng Zhao
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research Innovative Drug Research Center School of Pharmaceutical Sciences Chongqing University Chongqing 401331 P. R. China
| | - Zhi‐Peng Wang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research Innovative Drug Research Center School of Pharmaceutical Sciences Chongqing University Chongqing 401331 P. R. China
- Chongqing Institute of Green and Intelligent Technology Chinese Academy of Sciences Chongqing 400714 P. R. China
| | - Zihua Lin
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research Innovative Drug Research Center School of Pharmaceutical Sciences Chongqing University Chongqing 401331 P. R. China
| | - Guoxing Wei
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research Innovative Drug Research Center School of Pharmaceutical Sciences Chongqing University Chongqing 401331 P. R. China
| | - Xumei Wen
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research Innovative Drug Research Center School of Pharmaceutical Sciences Chongqing University Chongqing 401331 P. R. China
| | - Siyu Li
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research Innovative Drug Research Center School of Pharmaceutical Sciences Chongqing University Chongqing 401331 P. R. China
| | - Xiaohong Yang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research Innovative Drug Research Center School of Pharmaceutical Sciences Chongqing University Chongqing 401331 P. R. China
- Chongqing Institute of Green and Intelligent Technology Chinese Academy of Sciences Chongqing 400714 P. R. China
| | - Qun Zhang
- Medicine Laboratory Children's Hospital of Chongqing Medical University Ministry of Education Key Laboratory of Child Development and Disorders 136 Zhongshan 2nd Rd Yuzhong, Chongqing 400014 P. R. China
| | - Chunmei Jing
- Medicine Laboratory Children's Hospital of Chongqing Medical University Ministry of Education Key Laboratory of Child Development and Disorders 136 Zhongshan 2nd Rd Yuzhong, Chongqing 400014 P. R. China
| | - Yuanwei Dai
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research Innovative Drug Research Center School of Pharmaceutical Sciences Chongqing University Chongqing 401331 P. R. China
| | - Jian Guo
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research Innovative Drug Research Center School of Pharmaceutical Sciences Chongqing University Chongqing 401331 P. R. China
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research Innovative Drug Research Center School of Pharmaceutical Sciences Chongqing University Chongqing 401331 P. R. China
| |
Collapse
|
12
|
Bhattacharjee R, Dey T, Kumar L, Kar S, Sarkar R, Ghorai M, Malik S, Jha NK, Vellingiri B, Kesari KK, Pérez de la Lastra JM, Dey A. Cellular landscaping of cisplatin resistance in cervical cancer. Biomed Pharmacother 2022; 153:113345. [PMID: 35810692 DOI: 10.1016/j.biopha.2022.113345] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 12/11/2022] Open
Abstract
Cervical cancer (CC) caused by human papillomavirus (HPV) is one of the largest causes of malignancies in women worldwide. Cisplatin is one of the widely used drugs for the treatment of CC is rendered ineffective owing to drug resistance. This review highlights the cause of resistance and the mechanism of cisplatin resistance cells in CC to develop therapeutic ventures and strategies that could be utilized to overcome the aforementioned issue. These strategies would include the application of nanocarries, miRNA, CRIPSR/Cas system, and chemotherapeutics in synergy with cisplatin to not only overcome the issues of drug resistance but also enhance its anti-cancer efficiency. Moreover, we have also discussed the signaling network of cisplatin resistance cells in CC that would provide insights to develop therapeutic target sites and inhibitors. Furthermore, we have discussed the role of CC metabolism on cisplatin resistance cells and the physical and biological factors affecting the tumor microenvironments.
Collapse
Affiliation(s)
- Rahul Bhattacharjee
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar 751024, Odisha, India
| | - Tanima Dey
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar 751024, Odisha, India
| | - Lamha Kumar
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, Kerala, India
| | - Sulagna Kar
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar 751024, Odisha, India
| | - Ritayan Sarkar
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar 751024, Odisha, India
| | - Mimosa Ghorai
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India
| | - Sumira Malik
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, Jharkhand 834001, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida, Uttar Pradesh 201310, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, India.
| | - Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641-046, India
| | - Kavindra Kumar Kesari
- Department of Applied Physics, School of Science, Aalto University, Espoo 00076, Finland; Department of Bio-products and Bio-systems, School of Chemical Engineering, Aalto University, Espoo 00076, Finland
| | - José M Pérez de la Lastra
- Biotechnology of Macromolecules, Instituto de Productos Naturales y Agrobiología, IPNA (CSIC), Avda. Astrofísico Francisco Sánchez, 3, 38206 San Cristóbal de la Laguna (Santa Cruz de Tenerife), Spain.
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, West Bengal, India.
| |
Collapse
|
13
|
Zhao S, Wang ZP, Lin Z, Wei G, Wen X, Li S, Yang X, Zhang Q, Jing C, Dai Y, Guo J, He Y. Drug Repurposing by Siderophore Conjugation: Synthesis and Biological Evaluation of Siderophore‐Methotrexate Conjugates as Antibiotics. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202204139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Sheng Zhao
- Chongqing University School of Pharmaceutical Sciences CHINA
| | - Zhi-Peng Wang
- Chongqing University School of Pharmaceutical Sciences CHINA
| | - Zihua Lin
- Chongqing University School of Pharmaceutical Sciences CHINA
| | - Guoxing Wei
- Chongqing University School of Pharmaceutical Sciences CHINA
| | - Xumei Wen
- Chongqing University School of Pharmaceutical Sciences CHINA
| | - Siyu Li
- Chongqing University School of Pharmaceutical Sciences CHINA
| | - Xiaohong Yang
- Chongqing University School of Pharmaceutical Sciences CHINA
| | - Qun Zhang
- Chongqing Medical University Affiliated Children's Hospital Medicine Laboratory CHINA
| | - Chunmei Jing
- Chongqing Medical University Affiliated Children's Hospital Department of Clinical Laboratory CHINA
| | - Yuanwei Dai
- Chongqing University School of Pharmaceutical Sciences CHINA
| | - Jian Guo
- Chongqing University School of Pharmaceutical Sciences CHINA
| | - Yun He
- Chongqing University School of Pharmaceutical Sciences Daxuecheng South Road 401331 Chongqing CHINA
| |
Collapse
|
14
|
Poonacha KNT, Villa TG, Notario V. The Interplay among Radiation Therapy, Antibiotics and the Microbiota: Impact on Cancer Treatment Outcomes. Antibiotics (Basel) 2022; 11:331. [PMID: 35326794 PMCID: PMC8944497 DOI: 10.3390/antibiotics11030331] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/27/2022] [Accepted: 02/28/2022] [Indexed: 12/01/2022] Open
Abstract
Radiation therapy has been used for more than a century, either alone or in combination with other therapeutic modalities, to treat most types of cancer. On average, radiation therapy is included in the treatment plans for over 50% of all cancer patients, and it is estimated to contribute to about 40% of curative protocols, a success rate that may reach 90%, or higher, for certain tumor types, particularly on patients diagnosed at early disease stages. A growing body of research provides solid support for the existence of bidirectional interaction between radiation exposure and the human microbiota. Radiation treatment causes quantitative and qualitative changes in the gut microbiota composition, often leading to an increased abundance of potentially hazardous or pathogenic microbes and a concomitant decrease in commensal bacteria. In turn, the resulting dysbiotic microbiota becomes an important contributor to worsen the adverse events caused in patients by the inflammatory process triggered by the radiation treatment and a significant determinant of the radiation therapy anti-tumor effectiveness. Antibiotics, which are frequently included as prophylactic agents in cancer treatment protocols to prevent patient infections, may affect the radiation/microbiota interaction through mechanisms involving both their antimicrobial activity, as a mediator of microbiota imbalances, and their dual capacity to act as pro- or anti-tumorigenic effectors and, consequently, as critical determinants of radiation therapy outcomes. In this scenario, it becomes important to introduce the use of probiotics and/or other agents that may stabilize the healthy microbiota before patients are exposed to radiation. Ultimately, newly developed methodologies may facilitate performing personalized microbiota screenings on patients before radiation therapy as an accurate way to identify which antibiotics may be used, if needed, and to inform the overall treatment planning. This review examines currently available data on these issues from the perspective of improving radiation therapy outcomes.
Collapse
Affiliation(s)
| | - Tomás G. Villa
- Department of Microbiology, Faculty of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, 15705 La Coruña, Spain;
| | - Vicente Notario
- Department of Radiation Medicine, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
15
|
Allioux FM, Ghasemian MB, Xie W, O'Mullane AP, Daeneke T, Dickey MD, Kalantar-Zadeh K. Applications of liquid metals in nanotechnology. NANOSCALE HORIZONS 2022; 7:141-167. [PMID: 34982812 DOI: 10.1039/d1nh00594d] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Post-transition liquid metals (LMs) offer new opportunities for accessing exciting dynamics for nanomaterials. As entities with free electrons and ions as well as fluidity, LM-based nanomaterials are fundamentally different from their solid counterparts. The low melting points of most post-transition metals (less than 330 °C) allow for the formation of nanodroplets from bulk metal melts under mild mechanical and chemical conditions. At the nanoscale, these liquid state nanodroplets simultaneously offer high electrical and thermal conductivities, tunable reactivities and useful physicochemical properties. They also offer specific alloying and dealloying conditions for the formation of multi-elemental liquid based nanoalloys or the synthesis of engineered solid nanomaterials. To date, while only a few nanosized LM materials have been investigated, extraordinary properties have been observed for such systems. Multi-elemental nanoalloys have shown controllable homogeneous or heterogeneous core and surface compositions with interfacial ordering at the nanoscale. The interactions and synergies of nanosized LMs with polymeric, inorganic and bio-materials have also resulted in new compounds. This review highlights recent progress and future directions for the synthesis and applications of post-transition LMs and their alloys. The review presents the unique properties of these LM nanodroplets for developing functional materials for electronics, sensors, catalysts, energy systems, and nanomedicine and biomedical applications, as well as other functional systems engineered at the nanoscale.
Collapse
Affiliation(s)
- Francois-Marie Allioux
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, NSW 2052, Australia.
| | - Mohammad B Ghasemian
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, NSW 2052, Australia.
| | - Wanjie Xie
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, NSW 2052, Australia.
| | - Anthony P O'Mullane
- School of Chemistry and Physics, Queensland University of Technology (QUT), Brisbane, QLD 4001, Australia
| | - Torben Daeneke
- School of Engineering, RMIT University, Melbourne, Victoria, 3001, Australia
| | - Michael D Dickey
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA
| | - Kourosh Kalantar-Zadeh
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, NSW 2052, Australia.
| |
Collapse
|
16
|
Multidrug resistance crisis during COVID-19 pandemic: Role of anti-microbial peptides as next-generation therapeutics. Colloids Surf B Biointerfaces 2021; 211:112303. [PMID: 34952285 PMCID: PMC8685351 DOI: 10.1016/j.colsurfb.2021.112303] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 12/03/2021] [Accepted: 12/16/2021] [Indexed: 02/07/2023]
Abstract
The decreasing effectiveness of conventional drugs due to multidrug-resistance is a major challenge for the scientific community, necessitating development of novel antimicrobial agents. In the present era of coronavirus 2 (COVID-19) pandemic, patients are being widely exposed to antimicrobial drugs and hence the problem of multidrug-resistance shall be aggravated in the days to come. Consequently, revisiting the phenomena of multidrug resistance leading to formulation of effective antimicrobial agents is the need of the hour. As a result, this review sheds light on the looming crisis of multidrug resistance in wake of the COVID-19 pandemic. It highlights the problem, significance and approaches for tackling microbial resistance with special emphasis on anti-microbial peptides as next-generation therapeutics against multidrug resistance associated diseases. Antimicrobial peptides exhibit exceptional mechanism of action enabling rapid killing of microbes at low concentration, antibiofilm activity, immunomodulatory properties along with a low tendency for resistance development providing them an edge over conventional antibiotics. The review is unique as it discusses the mode of action, pharmacodynamic properties and application of antimicrobial peptides in areas ranging from therapeutics to agriculture.
Collapse
|
17
|
Cattò C, Villa F, Cappitelli F. Understanding the Role of the Antioxidant Drug Erdosteine and Its Active Metabolite on Staphylococcus aureus Methicillin Resistant Biofilm Formation. Antioxidants (Basel) 2021; 10:antiox10121922. [PMID: 34943025 PMCID: PMC8698571 DOI: 10.3390/antiox10121922] [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: 10/27/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 01/02/2023] Open
Abstract
Increasing numbers of researches have suggested that some drugs with reactive oxygen species (ROS)-mediated mechanisms of action modulate biofilm formation of some pathogenic strains. However, the full contribution of ROS to biofilm development is still an open question. In this paper, the correlations between the antioxidant drug Erdosteine (Er) and its active Metabolite I (Met I), ROS and biofilm development of two strains of methicillin resistant Staphylococcus aureus are presented. Experiments revealed that Er and Met I at 2 and 5 mg/L increased up to three orders of magnitude the number of biofilm-dwelling cells, while the content of ROS within the biofilms was reduced above the 87%, with a major effect of Met I in comparison to Er. Comparative proteomics showed that, 5 mg/L Met I modified the expression of 30% and 65% of total proteins in the two strains respectively. Some proteins involved in cell replication were upregulated, and a nitric oxide-based mechanism is assumed to modulate the biofilm development by changing quorum sensitive pathways. Additionally, several proteins involved in virulence were downregulated in the presence of Met I, suggesting that treated cells, despite being greater in number, might have lost part of their virulence.
Collapse
|
18
|
Li H, Li T, Zhang L, Hu Q, Liao X, Jiang Q, Qiu X, Li L, Draheim RR, Huang Q, Zhou R. Antimicrobial compounds from an FDA-approved drug library with activity against Streptococcus suis. J Appl Microbiol 2021; 132:1877-1886. [PMID: 34800069 DOI: 10.1111/jam.15377] [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: 06/15/2021] [Revised: 10/12/2021] [Accepted: 11/17/2021] [Indexed: 02/03/2023]
Abstract
AIM Antimicrobial resistance (AMR) has become a global concern. Developing novel antimicrobials is one of the most effective approaches in tackling AMR. Considering its relatively low cost and risk, drug repurposing has been proposed as a valuable approach for novel antimicrobial discovery. The aim of this study was to screen for antimicrobial compounds against Streptococcus suis, an important zoonotic bacterial pathogen, from an Food and Drug Administration (FDA)-approved drug library. METHODS AND RESULTS In this study, we tested the antimicrobial activity of 1815 FDA-approved drugs against S. suis. Sixty-seven hits were obtained that showed a growth inhibition of more than 98%. After excluding already known antibiotics and antiseptics, 12 compounds were subjected to minimal inhibition concentration (MIC) assessment against S. suis. This showed that pralatrexate, daunorubicin (hydrochloride), teniposide, aclacinomycin A hydrochloride and floxuridine gave a relatively low MIC, ranging from 0.85 to 5.25 μg/ml. Apart from pralatrexate, the remaining four drugs could also inhibit the growth of antimicrobial-resistant S. suis. It was also demonstrated that these four drugs had better efficacy against Gram-positive bacteria than Gram-negative bacteria. Cytotoxicity assays showed that floxuridine and teniposide had a relatively high 50% cytotoxic concentration (CC50 ). The in vivo efficacy of floxuridine was analysed using a Galleria mellonella larvae infection model, and the results showed that floxuridine was effective in treating S. suis infection in vivo. CONCLUSIONS Five compounds from the FDA-approved drug library showed high antimicrobial activity against S. suis, among which floxuridine displayed potent in vivo efficacy that is worth further development. SIGNIFICANCE AND IMPACT OF STUDY Our study identified several antimicrobial compounds that are effective against S. suis, which provides a valuable starting point for further antimicrobial development.
Collapse
Affiliation(s)
- Haotian Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Tingting Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Liangsheng Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qiao Hu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xia Liao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qinggen Jiang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiuxiu Qiu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Lu Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Cooperative Innovation Center of Sustainable Pig Production, Wuhan, China.,International Research Center for Animal Disease (Ministry of Science & Technology of China), Wuhan, China
| | - Roger R Draheim
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Qi Huang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Cooperative Innovation Center of Sustainable Pig Production, Wuhan, China.,International Research Center for Animal Disease (Ministry of Science & Technology of China), Wuhan, China
| | - Rui Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Cooperative Innovation Center of Sustainable Pig Production, Wuhan, China.,International Research Center for Animal Disease (Ministry of Science & Technology of China), Wuhan, China
| |
Collapse
|
19
|
García-Trejo JJ, Ortega R, Zarco-Zavala M. Putative Repurposing of Lamivudine, a Nucleoside/Nucleotide Analogue and Antiretroviral to Improve the Outcome of Cancer and COVID-19 Patients. Front Oncol 2021; 11:664794. [PMID: 34367956 PMCID: PMC8335563 DOI: 10.3389/fonc.2021.664794] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 06/21/2021] [Indexed: 12/24/2022] Open
Abstract
Lamivudine, also widely known as 3TC belongs to a family of nucleotide/nucleoside analogues of cytidine or cytosine that inhibits the Reverse Transcriptase (RT) of retroviruses such as HIV. Lamivudine is currently indicated in combination with other antiretroviral agents for the treatment of HIV-1 infection or for chronic Hepatitis B (HBV) virus infection associated with evidence of hepatitis B viral replication and active liver inflammation. HBV reactivation in patients with HBV infections who receive anticancer chemotherapy can be a life-threatening complication during and after the completion of chemotherapy. Lamivudine is used, as well as other antiretrovirals, to prevent the reactivation of the Hepatitis B virus during and after chemotherapy. In addition, Lamivudine has been shown to sensitize cancer cells to chemotherapy. Lamivudine and other similar analogues also have direct positive effects in the prevention of cancer in hepatitis B or HIV positive patients, independently of chemotherapy or radiotherapy. Recently, it has been proposed that Lamivudine might be also repurposed against SARS-CoV-2 in the context of the COVID-19 pandemic. In this review we first examine recent reports on the re-usage of Lamivudine or 3TC against the SARS-CoV-2, and we present docking evidence carried out in silico suggesting that Lamivudine may bind and possibly work as an inhibitor of the SARS-CoV-2 RdRp RNA polymerase. We also evaluate and propose assessment of repurposing Lamivudine as anti-SARS-CoV-2 and anti-COVID-19 antiviral. Secondly, we summarize the published literature on the use of Lamivudine or (3TC) before or during chemotherapy to prevent reactivation of HBV, and examine reports of enhanced effectiveness of radiotherapy in combination with Lamivudine treatment against the cancerous cells or tissues. We show that the anti-cancer properties of Lamivudine are well established, whereas its putative anti-COVID effect is under investigation. The side effects of lamivudine and the appearance of resistance to 3TC are also discussed.
Collapse
Affiliation(s)
- José J García-Trejo
- Department of Biology, Laboratory of Bioenergetics, Chemistry Faculty and School, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Raquel Ortega
- Department of Biology, Laboratory of Bioenergetics, Chemistry Faculty and School, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Mariel Zarco-Zavala
- Department of Biology, Laboratory of Bioenergetics, Chemistry Faculty and School, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| |
Collapse
|
20
|
Ibrahim MAA, Badr EAA, Abdelrahman AHM, Almansour NM, Shawky AM, Mekhemer GAH, Alrumaihi F, Moustafa MF, Atia MAM. Prospective Drug Candidates as Human Multidrug Transporter ABCG2 Inhibitors: an In Silico Drug Discovery Study. Cell Biochem Biophys 2021; 79:189-200. [PMID: 33954893 DOI: 10.1007/s12013-021-00985-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2021] [Indexed: 12/22/2022]
Abstract
Breast cancer resistance protein (ABCG2) is a human ATP-binding cassette (ABC) that plays a paramount role in multidrug resistance (MDR) in cancer therapy. The discovery of ABCG2 inhibitors could assist in designing unprecedented therapeutic strategies for cancer treatment. There is as yet no approved drug targeting ABCG2, although a large number of drug candidates have been clinically investigated. In this work, binding affinities of 181 drug candidates in clinical-trial or investigational stages as ABCG2 inhibitors were inspected using in silico techniques. Based on available experimental data, the performance of AutoDock4.2.6 software was first validated to predict the inhibitor-ABCG2 binding mode and affinity. Combined molecular docking calculations and molecular dynamics (MD) simulations, followed by molecular mechanics-generalized Born surface area (MM-GBSA) binding energy calculations, were then performed to filter out the studied drug candidates. From the estimated docking scores and MM-GBSA binding energies, six auspicious drug candidates-namely, pibrentasvir, venetoclax, ledipasvir, avatrombopag, cobicistat, and revefenacin-exhibited auspicious binding energies with value < -70.0 kcal/mol. Interestingly, pibrentasvir, venetoclax, and ledipasvir were observed to show even higher binding affinities with the ABCG2 transporter with binding energies of < -80.0 kcal/mol over long MD simulations of 100 ns. The stabilities of these three promising candidates in complex with ABCG2 transporter were demonstrated by their energetics and structural analyses throughout the 100 ns MD simulations. The current study throws new light on pibrentasvir, venetoclax, and ledipasvir as curative options for multidrug resistant cancers by inhibiting ABCG2 transporter.
Collapse
Affiliation(s)
- Mahmoud A A Ibrahim
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, Egypt.
| | - Esraa A A Badr
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, Egypt
| | - Alaa H M Abdelrahman
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, Egypt
| | - Nahlah Makki Almansour
- Department of Biology, College of Science, University of Hafr Al Batin, Hafr Al Batin, Saudi Arabia
| | - Ahmed M Shawky
- Science and Technology Unit (STU), Umm Al-Qura University, Makkah, Saudi Arabia
| | - Gamal A H Mekhemer
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, Egypt
| | - Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Mahmoud F Moustafa
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
- Department of Botany & Microbiology, Faculty of Science, South Valley University, Qena, Egypt
| | - Mohamed A M Atia
- Molecular Genetics and Genome Mapping Laboratory, Genome Mapping Department, Agricultural Genetic Engineering Research Institute (AGERI), Agricultural Research Center (ARC), Giza, Egypt.
| |
Collapse
|
21
|
Santos ALS, Braga-Silva LA, Gonçalves DS, Ramos LS, Oliveira SSC, Souza LOP, Oliveira VS, Lins RD, Pinto MR, Muñoz JE, Taborda CP, Branquinha MH. Repositioning Lopinavir, an HIV Protease Inhibitor, as a Promising Antifungal Drug: Lessons Learned from Candida albicans-In Silico, In Vitro and In Vivo Approaches. J Fungi (Basel) 2021; 7:jof7060424. [PMID: 34071195 PMCID: PMC8229492 DOI: 10.3390/jof7060424] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/25/2021] [Accepted: 05/25/2021] [Indexed: 12/20/2022] Open
Abstract
The repurposing strategy was applied herein to evaluate the effects of lopinavir, an aspartic protease inhibitor currently used in the treatment of HIV-infected individuals, on the globally widespread opportunistic human fungal pathogen Candida albicans by using in silico, in vitro and in vivo approaches in order to decipher its targets on fungal cells and its antifungal mechanisms of action. Secreted aspartic proteases (Saps) are the obviously main target of lopinavir. To confirm this hypothesis, molecular docking assays revealed that lopinavir bound to the Sap2 catalytic site of C. albicans as well as inhibited the Sap hydrolytic activity in a typically dose-dependent manner. The inhibition of Saps culminated in the inability of C. albicans yeasts to assimilate the unique nitrogen source (albumin) available in the culture medium, culminating with fungal growth inhibition (IC50 = 39.8 µM). The antifungal action of lopinavir was corroborated by distinct microscopy analyses, which evidenced drastic and irreversible changes in the morphology that justified the fungal death. Furthermore, our results revealed that lopinavir was able to (i) arrest the yeasts-into-hyphae transformation, (ii) disturb the synthesis of neutral lipids, including ergosterol, (iii) modulate the surface-located molecules, such as Saps and mannose-, sialic acid- and N-acetylglucosamine-containing glycoconjugates, (iv) diminish the secretion of hydrolytic enzymes, such as Saps and esterase, (v) negatively influence the biofilm formation on polystyrene surface, (vi) block the in vitro adhesion to epithelial cells, (vii) contain the in vivo infection in both immunocompetent and immunosuppressed mice and (viii) reduce the Sap production by yeasts recovered from kidneys of infected animals. Conclusively, the exposed results highlight that lopinavir may be used as a promising repurposing drug against C. albicans infection as well as may be used as a lead compound for the development of novel antifungal drugs.
Collapse
Affiliation(s)
- André L. S. Santos
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (L.A.B.-S.); (D.S.G.); (L.S.R.); (S.S.C.O.); (L.O.P.S.)
- Programa de Pós-Graduação em Bioquímica (PPGBq), Instituto de Química (IQ), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-909, Brazil
- Correspondence: (A.L.S.S.); (M.H.B.); Tel.: +55-21-3938-0366 (A.L.S.S.)
| | - Lys A. Braga-Silva
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (L.A.B.-S.); (D.S.G.); (L.S.R.); (S.S.C.O.); (L.O.P.S.)
- Programa de Pós-Graduação em Bioquímica (PPGBq), Instituto de Química (IQ), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-909, Brazil
| | - Diego S. Gonçalves
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (L.A.B.-S.); (D.S.G.); (L.S.R.); (S.S.C.O.); (L.O.P.S.)
- Programa de Pós-Graduação em Bioquímica (PPGBq), Instituto de Química (IQ), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-909, Brazil
| | - Lívia S. Ramos
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (L.A.B.-S.); (D.S.G.); (L.S.R.); (S.S.C.O.); (L.O.P.S.)
| | - Simone S. C. Oliveira
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (L.A.B.-S.); (D.S.G.); (L.S.R.); (S.S.C.O.); (L.O.P.S.)
| | - Lucieri O. P. Souza
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (L.A.B.-S.); (D.S.G.); (L.S.R.); (S.S.C.O.); (L.O.P.S.)
| | - Vanessa S. Oliveira
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Recife 50740-465, Brazil; (V.S.O.); (R.D.L.)
| | - Roberto D. Lins
- Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Recife 50740-465, Brazil; (V.S.O.); (R.D.L.)
| | - Marcia R. Pinto
- Departamento de Microbiologia e Parasitologia, Instituto Biomédico, Universidade Federal Fluminense (UFF), Niterói 24210-130, Brazil;
| | - Julian E. Muñoz
- MICROS Group, Medicine Traslacional Institute, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia;
| | - Carlos P. Taborda
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo 05508-060, Brazil;
- Laboratório de Micologia Médica—LIM53/IMTSP, Universidade de São Paulo (USP), São Paulo 05508-000, Brazil
| | - Marta H. Branquinha
- Laboratório de Estudos Avançados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-901, Brazil; (L.A.B.-S.); (D.S.G.); (L.S.R.); (S.S.C.O.); (L.O.P.S.)
- Correspondence: (A.L.S.S.); (M.H.B.); Tel.: +55-21-3938-0366 (A.L.S.S.)
| |
Collapse
|
22
|
Drug Repurposing in Medical Mycology: Identification of Compounds as Potential Antifungals to Overcome the Emergence of Multidrug-Resistant Fungi. Pharmaceuticals (Basel) 2021; 14:ph14050488. [PMID: 34065420 PMCID: PMC8161392 DOI: 10.3390/ph14050488] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/13/2021] [Accepted: 05/18/2021] [Indexed: 02/07/2023] Open
Abstract
Immunodepression, whether due to HIV infection or organ transplantation, has increased human vulnerability to fungal infections. These conditions have created an optimal environment for the emergence of opportunistic infections, which is concomitant to the increase in antifungal resistance. The use of conventional antifungal drugs as azoles and polyenes can lead to clinical failure, particularly in immunocompromised individuals. Difficulties related to treating fungal infections combined with the time required to develop new drugs, require urgent consideration of other therapeutic alternatives. Drug repurposing is one of the most promising and rapid solutions that the scientific and medical community can turn to, with low costs and safety advantages. To treat life-threatening resistant fungal infections, drug repurposing has led to the consideration of well-known and potential molecules as a last-line therapy. The aim of this review is to provide a summary of current antifungal compounds and their main resistance mechanisms, following by an overview of the antifungal activity of non-traditional antimicrobial drugs. We provide their eventual mechanisms of action and the synergistic combinations that improve the activity of current antifungal treatments. Finally, we discuss drug repurposing for the main emerging multidrug resistant (MDR) fungus, including the Candida auris, Aspergillus or Cryptococcus species.
Collapse
|
23
|
Structure-activity relationships of furanones, dihydropyrrolones and thiophenones as potential quorum sensing inhibitors. Future Med Chem 2020; 12:1925-1943. [PMID: 33094640 DOI: 10.4155/fmc-2020-0244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Since their initial isolation from the marine alga Delisea pulchra, bromofuranones have been investigated as potential inhibitors of quorum sensing (QS) in various bacterial strains. QS is an important mechanism by which bacteria co-ordinate their molecular response to the environment. QS is intrinsically linked to bacterial antibiotic resistance. Inspired by nature, chemists have developed a wide variety of synthetic analogs in an effort to elucidate the structure-activity relationships of these compounds, and to ultimately develop novel antimicrobial agents. In this work, we describe advances in this field while paying particular attention to apparent structure-activity relationships. This review is organized according to the main ring systems under investigation, namely furanones, dihydropyrrolones and thiophenones.
Collapse
|