1
|
Xiong J, Lu H, Jiang Y. Mechanisms of Azole Potentiation: Insights from Drug Repurposing Approaches. ACS Infect Dis 2025. [PMID: 39749640 DOI: 10.1021/acsinfecdis.4c00657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
The emergence of azole resistance and tolerance in pathogenic fungi has emerged as a significant public health concern, emphasizing the urgency for innovative strategies to bolster the efficacy of azole-based treatments. Drug repurposing stands as a promising and practical avenue for advancing antifungal therapy, with the potential for swift clinical translation. This review offers a comprehensive overview of azole synergistic agents uncovered through drug repurposing strategies, alongside an in-depth exploration of the mechanisms by which these agents augment azole potency. Drawing from these mechanisms, we delineate strategies aimed at enhancing azole effectiveness, such as inhibiting efflux pumps to elevate azole concentrations within fungal cells, intensifying ergosterol synthesis inhibition, mitigating fungal cell resistance to azoles, and disrupting biological processes extending beyond ergosterol synthesis. This review is beneficial for the development of these potentiators, as it meticulously examines instances and provides nuanced discussions on the mechanisms underlying the progression of azole potentiators through drug repurposing strategies.
Collapse
Affiliation(s)
- Juan Xiong
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Hui Lu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yuanying Jiang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| |
Collapse
|
2
|
Wang J, Hu T, He X, Zhang Z, Yu N, Liu Z. Omeprazole and its analogs exhibit insecticidal potencies as inhibitors of insect choline acetyltransferase. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 206:106207. [PMID: 39672619 DOI: 10.1016/j.pestbp.2024.106207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 12/15/2024]
Abstract
Choline acetyltransferase (ChAT) is crucial for acetylcholine synthesis and regulates diverse functions in numerous biological processes. Omeprazole, an inhibitor on human ChAT, was evaluated here on insect ChAT as a potential inhibitor, as well as its insecticidal potency on Nilaparvata lugens, a major insect pest on rice. The evaluation also included omeprazole analogs and α-NETA, in order to explore a superior leading compound targeting on insect ChAT. In toxicity test, α-NETA and omeprazole exhibited insecticidal activity, among which omeprazole exhibited activity with a mortality of around 50 % on N. lugens nymphs at 0.4 mg/mL. In vitro crude enzyme assays showed that omeprazole acted as an inhibitor on insect ChAT with a high selectivity and exciting potency compared with α-NETA and control. Three residues (Tyr84, Val95, Tyr589) was critical in N. lugens ChAT for interacting with its substrate choline through molecular docking, and it also revealed that omeprazole exhibited a higher binding affinity toward ChAT catalytic tunnel compared with α-NETA. Based on this, we screened omeprazole analogs for their affinity to N. lugens ChAT, and two compounds stood out. The 5-hydroxy omeprazole had the highest binding affinity by prediction, and 5-O-desmethyl omeprazole was with the lowest binding affinity. The toxicity bioassay and enzyme activity test were then performed on these two compounds. Aligned with the docking results, 5-hydroxy omeprazole showed a strong inhibitory effect and insecticidal activity. In summary, omeprazole and 5-hydroxy omeprazole could serve as lead compounds for insecticides targeting on insect ChAT, a novel target.
Collapse
Affiliation(s)
- Jingting Wang
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China
| | - Tianye Hu
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China
| | - Xu He
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhen Zhang
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China
| | - Na Yu
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China
| | - Zewen Liu
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
3
|
Nobre AFD, Sousa AMSD, Costa ADC, Fernandes MR, Kumar R, Ponne S, Rocha MG, Rodrigues AM, Camargo ZPD, Brilhante RSN. Effect of proton pump inhibitors on susceptibility and melanogenesis of Sporothrix species. J Med Microbiol 2024; 73. [PMID: 39145374 DOI: 10.1099/jmm.0.001870] [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] [Indexed: 08/16/2024] Open
Abstract
Introduction. Sporotrichosis is a subcutaneous infection caused by dimorphic Sporothrix species embedded in the clinical clade. Fungi have virulence factors, such as biofilm and melanin production, which contribute to their survival and are related to the increase in the number of cases of therapeutic failure, making it necessary to search for new options.Gap statement. Proton pump inhibitors (PPIs) have already been shown to inhibit the growth and melanogenesis of other fungi.Aim. Therefore, this study aimed to evaluate the effect of the PPIs omeprazole (OMP), rabeprazole (RBP), esomeprazole, pantoprazole and lansoprazole on the susceptibility and melanogenesis of Sporothrix species, and their interactions with itraconazole, terbinafine and amphotericin B.Methodology. The antifungal activity of PPIs was evaluated using the microdilution method, and the combination of PPIs with itraconazole, terbinafine and amphotericin B was assessed using the checkerboard method. The assessment of melanogenesis inhibition was assessed using grey scale.Results. The OMP and RBP showed significant MIC results ranging from 32 to 256 µg ml-1 and 32 to 128 µg ml-1, respectively. Biofilms were sensitive, with a significant reduction (P<0.05) in metabolic activity of 52% for OMP and 50% for RBP at a concentration of 512 µg ml-1 and of biomass by 53% for OMP and 51% for RBP at concentrations of 512 µg ml-1. As for the inhibition of melanogenesis, only OMP showed inhibition, with a 54% reduction.Conclusion. It concludes that the PPIs OMP and RBP have antifungal activity in vitro against planktonic cells and biofilms of Sporothrix species and that, in addition, OMP can inhibit the melanization process in Sporothrix species.
Collapse
Affiliation(s)
- Augusto Feynman Dias Nobre
- One Health Microbiology Laboratory, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Alanna Mayara Soares de Sousa
- One Health Microbiology Laboratory, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Anderson da Cunha Costa
- One Health Microbiology Laboratory, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Mirele Rodrigues Fernandes
- One Health Microbiology Laboratory, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Rajender Kumar
- Division of Glycoscience, Department of Chemistry, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 10691 Stockholm, Sweden
| | - Saravanaraman Ponne
- Department of Medical Biotechnology, Aarupadai Veedu Medical College and Hospital, Vinayaka Mission's Research Foundation (Deemed to be University), Kirumampakkam, Puducherry 607402, India
| | - Maria Gleiciane Rocha
- One Health Microbiology Laboratory, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| | - Anderson Messias Rodrigues
- Laboratory of Emerging Fungal Pathogens, Department of Microbiology, Immunology, and Parasitology, Discipline of Cellular Biology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
- National Institute of Science and Technology in Human Pathogenic Fungi, São Paulo, Brazil
| | - Zoilo Pires de Camargo
- National Institute of Science and Technology in Human Pathogenic Fungi, São Paulo, Brazil
| | - Raimunda Sâmia Nogueira Brilhante
- One Health Microbiology Laboratory, Postgraduate Program in Medical Microbiology, Department of Pathology and Legal Medicine, Federal University of Ceará, Rua Cel. Nunes de Melo, 1315 - Rodolfo Teófilo - CEP: 60430-275, Fortaleza, Ceará, Brazil
| |
Collapse
|
4
|
Zou P, Liu J, Li P, Luan Q. Antifungal Activity, Synergism with Fluconazole or Amphotericin B and Potential Mechanism of Direct Current against Candida albicans Biofilms and Persisters. Antibiotics (Basel) 2024; 13:521. [PMID: 38927187 PMCID: PMC11200915 DOI: 10.3390/antibiotics13060521] [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: 04/30/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Candida albicans, as a notorious fungal pathogen, is associated with high morbidity and mortality worldwide due to its ability to form biofilms and persisters that can withstand currently available antifungals. Direct current (DC) has demonstrated a promising antimicrobial effect and synergistic effect with antimicrobials against various infections. Here, we first found DC exerted a killing effect on C. albicans planktonic and biofilm cells. Moreover, DC showed a synergistic effect with fluconazole (FLC) and amphotericin B (AMB). Notably, near-to-complete eradication of AMB-tolerant C. albicans biofilm persisters was achieved upon DC treatment. Next, the mechanism of action of DC was explored through mapping the genes and proteomic profiles of DC-treated C. albicans. The multi-omics analysis, quantitative real-time PCR and assay of reactive oxygen species (ROS) demonstrated DC exerted an antifungal effect on C. albicans by increasing cellular oxidative stress. As revealed by multiple analyses (e.g., protein assay based on absorbance at 280 nm and rhodamine 6G assay), DC was able to enhance membrane permeability, inhibit drug efflux and increase cellular FLC/AMB concentration of C. albicans, thereby mediating its synergism with the antifungals. Furthermore, DC inhibited superoxide dismutase 2 (SOD2) expression and manganese-containing SOD (Mn SOD) activity, leading to ROS production and enhanced killing of C. albicans biofilm persisters. The current findings demonstrate that the adjunctive use of DC in combination with antifungals is a promising strategy for effective control of C. albicans infections and management of antifungal resistance/tolerance in Candida biofilms.
Collapse
Affiliation(s)
| | | | - Peng Li
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing 100081, China; (P.Z.); (J.L.)
| | - Qingxian Luan
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing 100081, China; (P.Z.); (J.L.)
| |
Collapse
|
5
|
Wang Y, He Y, Cai T, Lei Z, Lei W, Cao Y, Wu J. A mechanism study on the synergistic effects of rifapentine and fluconazole against fluconazole-resistant Candida albicans in vitro. Heliyon 2024; 10:e27346. [PMID: 38515731 PMCID: PMC10955295 DOI: 10.1016/j.heliyon.2024.e27346] [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: 09/17/2023] [Revised: 01/07/2024] [Accepted: 02/28/2024] [Indexed: 03/23/2024] Open
Abstract
Candida albicans (C. albicans) is one of the most common clinical isolates of systemic fungal infection. Long-term and inappropriate use of antifungal drugs can cause fungal resistance, which poses a great challenge to the clinical treatment of fungal infections. The combination of antifungal drugs and non-antifungal drugs to overcome the problem of fungal resistance has become a research hotspot in recent years. Our previous study found that the combination of rifapentine (RFT) and fluconazole (FLC) has a significant synergistic against FLC-resistant C. albicans. The present study aimed to further verify the synergistic effect between FLC and RFT against the FLC-resistant C. albicans 100, and explore the underlying mechanism. The growth curve and spot assay test not only showed the synergistic effect of FLC and RFT on FLC-resistant C. albicans in vitro but exhibited a dose-dependent effect on RFT, indicating that RFT may play a principal role in the synergic effect of the two drugs. Flow cytometry showed that the combined use of RFT and FLC arrested cells in the G2/M phase, inhibiting the normal division and proliferation of FLC-resistant C. albicans. Transmission electron microscopy (TEM) demonstrated that FLC at a low concentration could still cause a certain degree of damage to the cell membrane in the FLC-resistant C. albicans, as represented by irregular morphologic changes and some defects observed in the cell membrane. When FLC was used in combination with RFT, the nuclear membrane was dissolved and the nucleus was condensed into a mass. Detection of the intracellular drug concentration of fungi revealed that the intracellular concentration of RFT was 31-195 fold that of RFT alone when it was concomitantly used with FLC. This indicated that FLC could significantly increase the concentration of RFT in cells, which may be due to the damage caused to the fungal cell membrane by FLC. In short, the present study revealed a synergistic mechanism in the combined use of RFT and FLC, which may provide a novel strategy for the clinical treatment of FLC-resistant C. albicans.
Collapse
Affiliation(s)
- Yulian Wang
- Department of Dermatology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yufei He
- Department of Dermatology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Tongkai Cai
- Department of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhongwei Lei
- Department of Rehabilitation, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Wenzhi Lei
- Department of Dermatology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yongbing Cao
- Department of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianhua Wu
- Department of Dermatology, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
6
|
Gao L, Xia X, Gong X, Zhang H, Sun Y. In vitro interactions of proton pump inhibitors and azoles against pathogenic fungi. Front Cell Infect Microbiol 2024; 14:1296151. [PMID: 38304196 PMCID: PMC10831725 DOI: 10.3389/fcimb.2024.1296151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 01/02/2024] [Indexed: 02/03/2024] Open
Abstract
Introduction Azole resistance has been increasingly reported and become an issue for clinical managements of invasive mycoses. New strategy with combination therapy arises as a valuable and promising alternative option. The aim of the present study is to investigate the in vitro combinational effect of proton pump inhibitors (PPIs) and azoles against pathogenic fungi. Methods In vitro interactions of PPIs including omeprazole (OME), lansoprazole (LAN), pantoprazole (PAN), and rabeprazole (RAB), and commonly used azoles including itraconazole (ITC), posaconazole (POS), voriconazole (VRC) and fluconazole (FLC), were investigated via broth microdilution chequerboard procedure adapted from the CLSI M27-A3 and M38-A2. A total of 67 clinically isolated strains, namely 27 strains of Aspergillus spp., 16 strains of Candida spp., and 24 strains of dematiaceous fungi, were studied. C. parapsilosis (ATCC 22019) and A. flavus (ATCC 204304) was included to ensure quality control. Results PPIs individually did not exert any significant antifungal activity. The combination of OME with ITC, POS, or VRC showed synergism against 77.6%, 86.6%, and 4% strains of tested pathogenic fungi, respectively, while synergism of OME/FLC was observed in 50% strains of Candida spp. Synergism between PAN and ITC, POS, or VRC was observed against 47.8%, 77.6% and 1.5% strains of tested fungi, respectively, while synergism of PNA/FLC was observed in 50% strains of Candida spp. Synergism of LAN with ITC, POS, or VRC was observed against 86.6%, 86.6%, and 3% of tested strains, respectively, while synergism of LAN/FLC was observed in 31.3% strains of Candida spp. Synergy of the combination of RAB with ITC, POS, or VRC was observed against 25.4%, 64.2%, and 4.5% of tested strains, respectively, while synergism of RAB/FLC was observed in 12.5% of Candida spp.. Among PPIs, synergism was least observed between RAB and triazoles, while among triazoles, synergism was least observed between VRC and PPIs. Among species, synergy was much more frequently observed in Aspergillus spp. and dematiaceous fungi as compared to Candida spp. Antagonism between PPIs with ITC or VRC was occasionally observed in Aspergillus spp. and dematiaceous fungi. It is notable that PPIs combined with azoles showed synergy against azole resistant A. fumigatus, and resulted in category change of susceptibility of ITC and POS against Candida spp. Discussion The results suggested that PPIs combined with azoles has the potential to enhance the susceptibilities of azoles against multiple pathogenic fungi and could be a promising strategy to overcome azole resistance issues. However, further investigations are warranted to study the combinational efficacy in more isolates and more species, to investigate the underlying mechanism of interaction and to evaluate the potential for concomitant use of these agents in human.
Collapse
Affiliation(s)
- Lujuan Gao
- Department of Dermatology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, China
- Department of Dermatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xuqiong Xia
- Department of Dermatology, Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Gong
- Department of Dermatology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, China
| | - Heng Zhang
- Department of Dermatology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, China
| | - Yi Sun
- Department of Dermatology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
7
|
Li X, Kong B, Sun Y, Sun F, Yang H, Zheng S. Synergistic potential of teriflunomide with fluconazole against resistant Candida albicans in vitro and in vivo. Front Cell Infect Microbiol 2023; 13:1282320. [PMID: 38169891 PMCID: PMC10758495 DOI: 10.3389/fcimb.2023.1282320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
Introduction Candida albicans is the primary cause of systemic candidiasis, which is involved in high morbidity and mortality. Drug resistance exacerbates these problems. In addition, there are limited antifungal drugs available. In order to solve these problems, combination therapy has aroused great interest. Teriflunomide is an immunosuppressant. In the present work, we aimed to identify whether teriflunomide can reverse the resistance of Candida albicans in the presence of sub-inhibitory concentrations of fluconazole in vitro and in vivo. Methods Seven Candida albicans isolates were used in this study. Susceptibility of Candida albicans in vitro to the drugs was determined using a checkerboard microdilution assay in accordance with the recommendations of the Clinical and Laboratory Standards Institute. The effects of drugs on biofilm biomass of Candida albicans were determined by crystal violet staining. The development ability of Candida albicans hyphae was performed using a modified broth microdilution method. Galleria mellonella was used for testing the in vivo efficacy of the combination therapies. Results We found that the combination of teriflunomide (64 µg/mL) and fluconazole (0.5-1 µg/mL) has a significant synergistic effect in all resistant Candida albicans isolates (n=4). Also, this drug combination could inhibit the immature biofilm biomass and hyphae formation of resistant Candida albicans. Galleria mellonella was used for testing the in vivo efficacy of this combination therapies. As for the Galleria mellonella larvae infected by resistant Candida albicans, teriflunomide (1.6 µg/larvae) combined with fluconazole (1.6 µg/larvae) significantly increased their survival rates, and reduced the fungal burden, as well as damage of tissue in comparison to that in the control group or drug monotherapy group. Conclusion These results expand our knowledge about the antifungal potential of teriflunomide as an adjuvant of existing antifungal drugs, and also open new perspectives in the treatment of resistant Candida albicans based on repurposing clinically available nonantifungal drugs.
Collapse
Affiliation(s)
- Xiuyun Li
- Maternal and Child Health Development Research Center, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Bing Kong
- Department of Critical Care Medicine, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| | - Yaqiong Sun
- Obstetrics Department, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| | - Fenghua Sun
- Radiology Department, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| | - Huijun Yang
- Reproductive Medicine Center, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| | - Shicun Zheng
- Maternal and Child Health Development Research Center, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, Shandong, China
| |
Collapse
|
8
|
Abstract
Fungal infections are rising, with over 1.5 billion cases and more than 1 million deaths recorded each year. Among these, Candida infections are frequent in at-risk populations and the rapid development of drug resistance and tolerance contributes to their clinical persistence. Few antifungal drugs are available, and their efficacy is declining due to the environmental overuse and the expansion of multidrug-resistant species. One way to prolong their utility is by applying them in combination therapy. Here, we highlight recently described azole potentiators belonging to different categories: natural, repurposed, or novel compounds. We showcase examples of molecules and discuss their identified or proposed mode of action. We also emphasise the challenges in azole potentiator development, compounded by the lack of animal testing, the overreliance on Candida albicans and Candida auris, as well as the limited understanding of compound efficacy.
Collapse
Affiliation(s)
| | - Iuliana V. Ene
- Institut Pasteur, Université Paris Cité, Fungal Heterogeneity Group, Paris, France
| |
Collapse
|
9
|
Liu Y, Ma L, Cheng J, Su J. Effects of Omeprazole on Recurrent Clostridioides difficile Infection Caused by ST81 Strains and Their Potential Mechanisms. Antimicrob Agents Chemother 2023; 67:e0022123. [PMID: 37223895 PMCID: PMC10269155 DOI: 10.1128/aac.00221-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/12/2023] [Indexed: 05/25/2023] Open
Abstract
Clostridioides difficile infection (CDI) is associated with high recurrence rates that have substantial effects on patients' quality of life. To investigate the risk factors and potential mechanisms contributing to recurrent CDI (rCDI), a total of 243 cases were enrolled in this study. The history of omeprazole (OME) medication and ST81 strain infection were considered the two independent risks with the highest odds ratios in rCDI. In the presence of OME, we detected concentration-dependent increases in the MIC values of fluoroquinolone antibiotics against ST81 strains. Mechanically, OME facilitated ST81 strain sporulation and spore germination by blocking the pathway of purine metabolism and also promoted an increase in cell motility and toxin production by turning the flagellar switch to the ON state. In conclusion, OME affects several biological processes during C difficile growth, which have fundamental impacts on the development of rCDI caused by ST81 strains. Programmed OME administration and stringent surveillance of the emerging ST81 genotype are matters of considerable urgency and significance in rCDI prevention.
Collapse
Affiliation(s)
- Yifeng Liu
- Clinical Laboratory Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Liyan Ma
- Clinical Laboratory Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jingwei Cheng
- Clinical Laboratory Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jianrong Su
- Clinical Laboratory Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
Toepfer S, Lackner M, Keniya MV, Zenz LM, Friemert M, Bracher F, Monk BC. Clorgyline Analogs Synergize with Azoles against Drug Efflux in Candida auris. J Fungi (Basel) 2023; 9:663. [PMID: 37367600 DOI: 10.3390/jof9060663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 06/28/2023] Open
Abstract
Concern about the global emergence of multidrug-resistant fungal pathogens led us to explore the use of combination therapy to combat azole resistance in Candida auris. Clorgyline had previously been shown to be a multi-target inhibitor of Cdr1 and Mdr1 efflux pumps of Candida albicans and Candida glabrata. A screen for antifungal sensitizers among synthetic analogs of Clorgyline detected interactions with the C. auris efflux pump azole substrates Posaconazole and Voriconazole. Of six Clorgyline analogs, M19 and M25 were identified as potential sensitizers of azole resistance. M19 and M25 were found to act synergistically with azoles against resistant C. auris clade I isolates and recombinant Saccharomyces cerevisiae strains overexpressing C. auris efflux pumps. Nile Red assays with the recombinant strains showed M19 and M25 inhibited the activity of Cdr1 and Mdr1 efflux pumps that are known to play key roles in azole resistance in C. auris clades I, III, and IV. While Clorgyline, M19 and M25 uncoupled the Oligomycin-sensitive ATPase activity of Cdr1 from C. albicans and C. auris, their mode of action is yet to be fully elucidated. The experimental combinations described herein provides a starting point to combat azole resistance dominated by overexpression of CauCdr1 in C. auris clades I and IV and CauMdr1 in C. auris clade III.
Collapse
Affiliation(s)
- Stephanie Toepfer
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
| | - Michaela Lackner
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Mikhail V Keniya
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
- Hackensack Meridian Health Center for Discovery and Innovation, Nutley, NJ 07110, USA
| | - Lisa-Maria Zenz
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Marianne Friemert
- Center for Drug Research, Department of Pharmacy, Ludwig-Maximilian University of Munich, 81377 Munich, Germany
| | - Franz Bracher
- Center for Drug Research, Department of Pharmacy, Ludwig-Maximilian University of Munich, 81377 Munich, Germany
| | - Brian C Monk
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
| |
Collapse
|
11
|
Abduljalil H, Bakri A, Albashaireh K, Alshanta OA, Brown JL, Sherry L, Kean R, Nile C, McLean W, Ramage G. Screening the Tocriscreen™ bioactive compound library in search for inhibitors of
Candida
biofilm formation. APMIS 2022; 130:568-577. [PMID: 35791082 PMCID: PMC9541805 DOI: 10.1111/apm.13260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/29/2022] [Indexed: 11/28/2022]
Abstract
Biofilms formed by Candida species present a significant clinical problem due to the ineffectiveness of many conventional antifungal agents, in particular the azole class. We urgently require new and clinically approved antifungal agents quickly for treatment of critically ill patients. To improve efficiency in antifungal drug development, we utilized a library of 1280 biologically active molecules within the Tocriscreen 2.0 Micro library. Candida aurisNCPF 8973 and Candida albicansSC5314 were initially screened for biofilm inhibitory activity using metabolic and biomass quantitative assessment methods, followed up by targeted evaluation of five selected hits. The initial screening (80% metabolic inhibition rate) revealed that there was 90 and 87 hits (approx. 7%) for C. albicans and C. auris, respectively. Additionally, all five compounds selected from the initial hits exhibited a biofilm inhibition effect against several key Candida species tested, including C. glabrata and C. krusei. Toyocamycin displayed the most potent activity at concentrations as low as 0.5 μg/mL, though was limited to inhibition. Darapladib demonstrated an efficacy for biofilm inhibition and treatment at a concentration range from 8 to 32 μg/mL and from 16 to 256 μg/mL, respectively. Combinational testing with conventional antifungals against C. albicans strains demonstrated a range of synergies for planktonic cells, and notably an anti‐biofilm synergy for darapladib and caspofungin. Together, these data provide new insights into antifungal management possibilities for Candida biofilms.
Collapse
Affiliation(s)
- Hafsa Abduljalil
- Glasgow Biofilm Research Group School of Medicine Dentistry and Nursing College of Medical, Veterinary and Life Sciences University of Glasgow Glasgow UK
| | - Ahmed Bakri
- Glasgow Biofilm Research Group School of Medicine Dentistry and Nursing College of Medical, Veterinary and Life Sciences University of Glasgow Glasgow UK
| | - Khawlah Albashaireh
- Glasgow Biofilm Research Group School of Medicine Dentistry and Nursing College of Medical, Veterinary and Life Sciences University of Glasgow Glasgow UK
| | - Om Alkhir Alshanta
- Glasgow Biofilm Research Group School of Medicine Dentistry and Nursing College of Medical, Veterinary and Life Sciences University of Glasgow Glasgow UK
| | - Jason L. Brown
- Glasgow Biofilm Research Group School of Medicine Dentistry and Nursing College of Medical, Veterinary and Life Sciences University of Glasgow Glasgow UK
| | - Leighann Sherry
- Glasgow Biofilm Research Group School of Medicine Dentistry and Nursing College of Medical, Veterinary and Life Sciences University of Glasgow Glasgow UK
| | - Ryan Kean
- Department of Biological and Biomedical Sciences Glasgow Caledonian University
| | | | - William McLean
- Glasgow Biofilm Research Group School of Medicine Dentistry and Nursing College of Medical, Veterinary and Life Sciences University of Glasgow Glasgow UK
| | - Gordon Ramage
- Glasgow Biofilm Research Group School of Medicine Dentistry and Nursing College of Medical, Veterinary and Life Sciences University of Glasgow Glasgow UK
| |
Collapse
|
12
|
Ben Ghezala I, Luu M, Bardou M. An update on drug-drug interactions associated with proton pump inhibitors. Expert Opin Drug Metab Toxicol 2022; 18:337-346. [PMID: 35787720 DOI: 10.1080/17425255.2022.2098107] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Proton pump inhibitors (PPIs) block the gastric H/K-ATPase, therefore inhibiting acid gastric secretion, leading to an increased pH (>4). They account for an extremely high number of prescriptions worldwide. Numerous drug-drug interactions have been described with PPIs, but all the described interactions do not have clinical significance. AREAS COVERED This review will discuss the latest updates on drug-drug interactions with PPIs, focusing on the last ten-year publications in the following areas: anti-infective agents, anticancer drugs, antiplatelet agents and anticoagulants, and antidiabetics. EXPERT OPINION Although pharmacokinetic interactions of PPIs have been described with many drugs, their clinical relevance remains controversial. However, given the extremely high number of people being treated with PPIs, clinicians should remain vigilant for interactions that may be clinically significant and require dose adjustment or therapeutic monitoring. Interestingly, not all PPIs have the same pharmacokinetic and pharmacodynamic profile, with some having a strong potential to inhibit CYP2C19, such as omeprazole, esomeprazole and lansoprazole, while others, pantoprazole, rabeprazole and dexlansoprazole, are weak CYP2C19 inhibitors. These may be preferred depending on co-prescribed treatments.In addition, new formulations have been developed to prevent some of the gastric pH-dependent drug interactions and should be evaluated in further large-scale prospective comparative studies.
Collapse
Affiliation(s)
- Inès Ben Ghezala
- INSERM, CIC1432, Plurithematic Unit, 21079 Dijon, France.,Centre d'Investigations Cliniques, Dijon Bourgogne University Hospital, 21079 Dijon, France.,Ophthalmology Department, Dijon Bourgogne University Hospital, 21079 Dijon, France
| | - Maxime Luu
- INSERM, CIC1432, Plurithematic Unit, 21079 Dijon, France.,Centre d'Investigations Cliniques, Dijon Bourgogne University Hospital, 21079 Dijon, France
| | - Marc Bardou
- INSERM, CIC1432, Plurithematic Unit, 21079 Dijon, France.,Centre d'Investigations Cliniques, Dijon Bourgogne University Hospital, 21079 Dijon, France.,Gastroenterology Department, Dijon Bourgogne University Hospital, 21079 Dijon, France
| |
Collapse
|
13
|
The plasma membrane H +-ATPase is critical for cell growth and pathogenicity in Penicillium digitatum. Appl Microbiol Biotechnol 2022; 106:5123-5136. [PMID: 35771244 DOI: 10.1007/s00253-022-12036-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/11/2022] [Accepted: 06/16/2022] [Indexed: 11/02/2022]
Abstract
The plasma membrane H+-ATPase (PMA1) is a major cytosolic pH regulator and a potential candidate for antifungal drug discovery due to its fungal specificity and criticality. In this study, the function of Penicillum digitatum PMA1 was characterized through RNA interference (RNAi) and overexpression technology. The results showed that silencing the PMA1 gene reduces cell growth and pathogenicity, and increases susceptibility of P. digitatum to proton pump inhibitors (PPIs). Under scanning electron microscopy (SEM) and transmission electron microscopy (TEM) examination, cell morphology was significantly altered in the PMA1- silenced mutant (si57). When compared with wild type (WT) and the overexpressed mutant (oe9), the cell walls of the si57 mutant were thicker and their cell membrane damage manifested particularly at sites of polarized growth. Consistent with the morphological change on the cell wall, chitin and glucan content of the cell wall of si57 were significantly lower and accompanied with increased activities of chitinase and glucanase. The lower ergosterol content in the si57 mutant then increased cell membrane permeability, ultimately leading to leakage of cytoplasmic contents such as ions, reduced sugars and soluble proteins. Furthermore, significantly decreased activity of cell wall degrading enzymes of si57 during citrus fruit infections indicates a reduced pathogenicity in this mutant. We conclude that PMA1 in P. digitatum plays an important role in maintaining pathogenesis and PMA1 could be a candidate novel fungicidal drug discovery for citrus green mold. KEY POINTS: Silencing PMA1 gene decreased the growth and pathogenicity of P. digitatum. Silencing PMA1 gene damaged cell wall and cell membrane integrity of P. digitatum. PMA1 appears to be a suitable fungicidal target against citrus green mold.
Collapse
|
14
|
Kane A, Carter DA. Augmenting Azoles with Drug Synergy to Expand the Antifungal Toolbox. Pharmaceuticals (Basel) 2022; 15:482. [PMID: 35455479 PMCID: PMC9027798 DOI: 10.3390/ph15040482] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 03/25/2022] [Accepted: 03/26/2022] [Indexed: 12/23/2022] Open
Abstract
Fungal infections impact the lives of at least 12 million people every year, killing over 1.5 million. Wide-spread use of fungicides and prophylactic antifungal therapy have driven resistance in many serious fungal pathogens, and there is an urgent need to expand the current antifungal arsenal. Recent research has focused on improving azoles, our most successful class of antifungals, by looking for synergistic interactions with secondary compounds. Synergists can co-operate with azoles by targeting steps in related pathways, or they may act on mechanisms related to resistance such as active efflux or on totally disparate pathways or processes. A variety of sources of potential synergists have been explored, including pre-existing antimicrobials, pharmaceuticals approved for other uses, bioactive natural compounds and phytochemicals, and novel synthetic compounds. Synergy can successfully widen the antifungal spectrum, decrease inhibitory dosages, reduce toxicity, and prevent the development of resistance. This review highlights the diversity of mechanisms that have been exploited for the purposes of azole synergy and demonstrates that synergy remains a promising approach for meeting the urgent need for novel antifungal strategies.
Collapse
Affiliation(s)
| | - Dee A. Carter
- School of Life and Environmental Sciences and Sydney ID, University of Sydney, Camperdown, NSW 2006, Australia;
| |
Collapse
|
15
|
El-Ganiny AM, Kamel HA, Yossef NE, Mansour B, El-Baz AM. Repurposing pantoprazole and haloperidol as efflux pump inhibitors in azole resistant clinical Candida albicans and non-albicans isolates. Saudi Pharm J 2022; 30:245-255. [PMID: 35498219 PMCID: PMC9051972 DOI: 10.1016/j.jsps.2022.01.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/12/2022] [Indexed: 12/17/2022] Open
Abstract
Candida species have a major role in nosocomial infections leading to high morbidity and mortality. Increased resistance to various antifungals, especially azoles is a significant problem. One of the main mechanisms for azole resistance is the up-regulation of efflux pump genes including CDR1 and MDR1. In the current study, clinical Candida isolates were identified to the species level and the antifungal susceptibility (AFS) of different Candida species was determined by disk diffusion method. Furthermore, the main mechanisms of azole resistance were investigated. Finally, haloperidol and pantoprazole were tested for their potential synergistic effect against fluconazole-resistant isolates. One hundred and twenty-two Candida clinical isolates were used in this study. 70 isolates were Candida albicans (57.4%), the non-albicans Candida species include: C. krusei (20.5%), C. tropicalis (6.6%), C. parapsilosis (5.7%), C. dubliniensis (4.9%) and C. glabrata (4.9%). The AFS testing showed that resistance to fluconazole and voriconazole were 13.1% (n = 16) and 9.8% (n = 12), respectively. Among the 16 resistant isolates, eight isolates (50%) were strong biofilm producers, seven (43.8 %) formed intermediate biofilm and one had no biofilm. All resistant strains overexpressed efflux pumps. Using RT-PCR, the efflux genes CDR1, MDR1 and ABC2 were over-expressed in azole resistant isolates. Haloperidol-fluconazole and pantoprazole-fluconazole combinations reduced the MIC of fluconazole in resistant isolates. The current study showed an increase in azole resistance of Candida species. The majority of resistant isolates form biofilm, and overexpress efflux pumps. Pantoprazole and Haloperidol showed a noteworthy effect as efflux pump inhibitors which oppose the fluconazole resistance in different Candida species.
Collapse
Affiliation(s)
- Amira M. El-Ganiny
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, 44519 Zagazig, Egypt
| | - Hend A. Kamel
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, 44519 Zagazig, Egypt
- Microbiology Department, Faculty of Pharmacy and Pharmaceutical Industries, Sinai University, Kantara, Egypt
| | - Nehal E. Yossef
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, 44519 Zagazig, Egypt
| | - Basem Mansour
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt
| | - Ahmed M. El-Baz
- Microbiology and Biotechnology Department, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt
| |
Collapse
|
16
|
Chen X, Wu J, Sun L, Nie J, Su S, Sun S. Antifungal Effects and Potential Mechanisms of Benserazide Hydrochloride Alone and in Combination with Fluconazole Against Candida albicans. Drug Des Devel Ther 2021; 15:4701-4711. [PMID: 34815665 PMCID: PMC8605804 DOI: 10.2147/dddt.s336667] [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: 08/29/2021] [Accepted: 10/29/2021] [Indexed: 01/23/2023] Open
Abstract
Purpose The resistance of C. albicans to traditional antifungal drugs brings a great challenge to clinical treatment. To overcome the resistance, developing antifungal agent sensitizers has attracted considerable attention. This study aimed to determine the anti-Candida activity of BEH alone or BEH–FLC combination and to explore the underlying mechanisms. Materials and Methods In vitro antifungal effects were performed by broth microdilution assay and XTT reduction assay. Infected Galleria mellonella larvae model was used to determine the antifungal effects in vivo. Probes Fluo-3/AM, FITC-VAD-FMK and rhodamine 6G were used to study the influence of BEH and FLC on intracellular calcium concentration, metacaspase activity and drug efflux of C. albicans. Results BEH alone exhibited obvious antifungal activities against C. albicans. BEH plus FLC not only showed synergistic effects against planktonic cells and preformed biofilms within 8 h but also enhanced the antifungal activity in infected G. mellonella larvae. Mechanistic studies indicated that antifungal effects of drugs might be associated with the increasement of calcium concentration, activation of metacaspase activity to reduce virulence and anti-biofilms, but were not related to drug efflux. Conclusion BEH alone or combined with FLC displayed potent antifungal activity both in vitro and in vivo, and the underlying mechanisms were related to reduced virulence factors.
Collapse
Affiliation(s)
- Xueqi Chen
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, People's Republic of China.,Department of Pharmacy, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Jiyong Wu
- Department of Pharmacy, Shandong Second Provincial General Hospital, Jinan, People's Republic of China
| | - Lei Sun
- Department of Pharmacy, Shandong Second Provincial General Hospital, Jinan, People's Republic of China
| | - Jing Nie
- Department of Pharmacy, Shandong Second Provincial General Hospital, Jinan, People's Republic of China
| | - Shan Su
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, People's Republic of China.,Department of Pharmacy, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Shujuan Sun
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, People's Republic of China.,Department of Pharmacy, Shandong Second Provincial General Hospital, Jinan, People's Republic of China
| |
Collapse
|
17
|
Pantoprazole ameliorates liver fibrosis and suppresses hepatic stellate cell activation in bile duct ligation rats by promoting YAP degradation. Acta Pharmacol Sin 2021; 42:1808-1820. [PMID: 34465912 PMCID: PMC8563954 DOI: 10.1038/s41401-021-00754-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 07/27/2021] [Indexed: 02/07/2023] Open
Abstract
Liver fibrosis is one of the most severe pathologic consequences of chronic liver diseases, and effective therapeutic strategies are urgently needed. Proton pump inhibitors (PPIs) are H+/K+-ATPase inhibitors and currently used to treat acid-related diseases such as gastric ulcers, which have shown other therapeutic effects in addition to inhibiting acid secretion. However, few studies have focused on PPIs from the perspective of inhibiting hepatic fibrosis. In the present study, we investigated the effects of pantoprazole (PPZ), a PPI, against liver fibrosis in a bile duct ligation (BDL) rat model, human hepatic stellate cell (HSC) line LX-2 and mouse primary HSCs (pHSCs), and explored the potential mechanisms underlying the effects of PPZ in vitro and in vivo. In BDL rats, administration of PPZ (150 mg· kg-1· d-1, i.p. for 14 d) significantly attenuated liver histopathological injury, collagen accumulation, and inflammatory responses, and suppressed fibrogenesis-associated gene expression including Col1a1, Acta2, Tgfβ1, and Mmp-2. In LX-2 cells and mouse pHSCs, PPZ (100-300 μM) dose-dependently suppressed the levels of fibrogenic markers. We conducted transcriptome analysis and subsequent validation in PPZ-treated LX-2 cells, and revealed that PPZ inhibited the expression of Yes-associated protein (YAP) and its downstream targets such as CTGF, ID1, survivin, CYR61, and GLI2. Using YAP overexpression and silencing, we demonstrated that PPZ downregulated hepatic fibrogenic gene expression via YAP. Furthermore, we showed that PPZ promoted the proteasome-dependent degradation and ubiquitination of YAP, thus inhibiting HSC activation. Additionally, we showed that PPZ destabilized YAP by disrupting the interaction between a deubiquitinating enzyme OTUB2 and YAP, and subsequently blocked the progression of hepatic fibrosis.
Collapse
|
18
|
Sayed SA, Hassan EAB, Abdel Hameed MR, Agban MN, Mohammed Saleh MF, Mohammed HH, Abdel-Aal ABM, Elgendy SG. Ketorolac-fluconazole: A New Combination Reverting Resistance in Candida albicans from Acute Myeloid Leukemia Patients on Induction Chemotherapy: In vitro Study. J Blood Med 2021; 12:465-474. [PMID: 34163275 PMCID: PMC8214543 DOI: 10.2147/jbm.s302158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/20/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Candida albicans is a significant source of morbidity and mortality for patients with acute myeloid leukemia (AML). Prolonged use of fluconazole as empirical antifungal prophylaxis in AML patients leads to overexpression of efflux pump genes that resulted in the emergence of azole-resistant species. Consequently, the introduction of a new strategy to improve the management of C. albicans infections is an urgent need. Nonsteroidal anti-inflammatory drug (NSAID) ketorolac is associated with a reduction in cancer relapses. The present study was performed to investigate the use of ketorolac-fluconazole combination to reverse fluconazole resistance in C. albicans isolated from AML patients on induction chemotherapy. PATIENTS AND METHODS One hundred and seventy AML patients were evaluated. Fifty C. albicans were isolated and subjected to disc diffusion assay and broth microdilution for fluconazole alone and combined with different concentrations of ketorolac. Efflux pump gene (CDR1, CDR2, and MDR1) expressions were quantified by real-time PCR. RESULTS The tested ketorolac acted synergistically with fluconazole against resistant C. albicans with the minimum inhibitory concentration (MIC) of fluconazole decreased from >160 μg/mL to 0.3-1.25 μg/mL in (93.8%) of resistant isolates with fractional inhibitory concentration index (FICI) value of 0.25. The majority of the resistant isolates overexpressed CDR1 (71.1%) and MDR1 (60%). CONCLUSION Ketorolac-fluconazole in vitro combination would be a promising strategy for further clinical in vivo trials to overcome fluconazole resistance in AML patients on induction chemotherapy.
Collapse
Affiliation(s)
- Shereen A Sayed
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Ehsan A B Hassan
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Muhamad R Abdel Hameed
- Department of Internal Medicine & Hematology Unit, Assiut University Hospitals and Bone Marrow Transplantation Unit, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Michael N Agban
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Mostafa F Mohammed Saleh
- Department of Internal Medicine & Hematology Unit, Assiut University Hospitals and Bone Marrow Transplantation Unit, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Hayam H Mohammed
- Department of Clinical Pathology, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Abu-Baker M Abdel-Aal
- Department of Organic Chemistry, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Sherein G Elgendy
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
19
|
Galdiero E, Salvatore MM, Maione A, Carraturo F, Galdiero S, Falanga A, Andolfi A, Salvatore F, Guida M. Impact of the Peptide WMR-K on Dual-Species Biofilm Candida albicans/Klebsiella pneumoniae and on the Untargeted Metabolomic Profile. Pathogens 2021; 10:214. [PMID: 33669279 PMCID: PMC7920046 DOI: 10.3390/pathogens10020214] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/08/2021] [Accepted: 02/12/2021] [Indexed: 01/09/2023] Open
Abstract
In recent years, the scientific community has focused on the development of new antibiotics to address the difficulties linked to biofilm-forming microorganisms and drug-resistant infections. In this respect, synthetic antimicrobial peptides (AMPs) are particularly regarded for their therapeutic potential against a broad spectrum of pathogens. In this work, the antimicrobial and antibiofilm activities of the peptide WMR-K towards single and dual species cultures of Candida albicans and Klebsiella pneumoniae were investigated. We found minimum inhibitory concentration (MIC) values for WMR-K of 10 µM for K. pneumoniae and of 200 µM for C. albicans. Furthermore, sub-MIC concentrations of peptide showed an in vitro inhibition of biofilm formation of mono and polymicrobial systems and also a good biofilm eradication even if higher concentrations of it are needed. In order to provide additional evidence for the effect of the examined peptide, a study of changes in extracellular metabolites excreted and/or uptaken from the culture medium (metabolomic footprinting) in the poly-microbial association of C. albicans and K. pneumoniae in presence and absence of WMR-K was performed. Comparing to the untreated dual species biofilm culture, the metabolomic profile of the WMR-K treated culture appears significantly altered. The differentially expressed compounds are mainly related to the primary metabolic pathways, including amino acids, trehalose, pyruvic acid, glycerol and vitamin B6.
Collapse
Affiliation(s)
- Emilia Galdiero
- Department of Biology, University of Naples ‘Federico II’, via Cinthia, 80126 Naples, Italy; (E.G.); (A.M.); (F.C.); (M.G.)
| | - Maria Michela Salvatore
- Department of Chemical Sciences, University of Naples ‘Federico II’, via Cinthia, 80126 Naples, Italy; (A.A.); (F.S.)
| | - Angela Maione
- Department of Biology, University of Naples ‘Federico II’, via Cinthia, 80126 Naples, Italy; (E.G.); (A.M.); (F.C.); (M.G.)
| | - Federica Carraturo
- Department of Biology, University of Naples ‘Federico II’, via Cinthia, 80126 Naples, Italy; (E.G.); (A.M.); (F.C.); (M.G.)
| | - Stefania Galdiero
- Department of Pharmacy, School of Medicine, University of Naples ‘Federico II’, Via Domenico Montesano 49, 80131 Naples, Italy;
| | - Annarita Falanga
- Department of Agricultural Science, University of Naples ‘Federico II’, Via dell’ Università 100, 80055 Naples, Italy;
| | - Anna Andolfi
- Department of Chemical Sciences, University of Naples ‘Federico II’, via Cinthia, 80126 Naples, Italy; (A.A.); (F.S.)
- BAT Center—Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Naples ‘Federico II’, 80055 Naples, Italy
| | - Francesco Salvatore
- Department of Chemical Sciences, University of Naples ‘Federico II’, via Cinthia, 80126 Naples, Italy; (A.A.); (F.S.)
| | - Marco Guida
- Department of Biology, University of Naples ‘Federico II’, via Cinthia, 80126 Naples, Italy; (E.G.); (A.M.); (F.C.); (M.G.)
| |
Collapse
|
20
|
Bidaud AL, Schwarz P, Herbreteau G, Dannaoui E. Techniques for the Assessment of In Vitro and In Vivo Antifungal Combinations. J Fungi (Basel) 2021; 7:jof7020113. [PMID: 33557026 PMCID: PMC7913650 DOI: 10.3390/jof7020113] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 01/28/2021] [Accepted: 02/02/2021] [Indexed: 12/18/2022] Open
Abstract
Systemic fungal infections are associated with high mortality rates despite adequate treatment. Moreover, acquired resistance to antifungals is increasing, which further complicates the therapeutic management. One strategy to overcome antifungal resistance is to use antifungal combinations. In vitro, several techniques are used to assess drug interactions, such as the broth microdilution checkerboard, agar-diffusion methods, and time-kill curves. Currently, the most widely used technique is the checkerboard method. The aim of all these techniques is to determine if the interaction between antifungal agents is synergistic, indifferent, or antagonistic. However, the interpretation of the results remains difficult. Several methods of analysis can be used, based on different theories. The most commonly used method is the calculation of the fractional inhibitory concentration index. Determination of the usefulness of combination treatments in patients needs well-conducted clinical trials, which are difficult. It is therefore important to study antifungal combinations in vivo, in experimental animal models of fungal infections. Although mammalian models have mostly been used, new alternative animal models in invertebrates look promising. To evaluate the antifungal efficacy, the most commonly used criteria are the mortality rate and the fungal load in the target organs.
Collapse
Affiliation(s)
- Anne-Laure Bidaud
- Parasitology-Mycology Unit, Microbiology Department, APHP, European Georges Pompidou Hospital, Paris-Descartes University, F-75015 Paris, France;
| | - Patrick Schwarz
- Department of Internal Medicine, Respiratory and Critical Care Medicine, University Hospital Marburg, Baldingerstraße, D-35043 Marburg, Germany;
- Center for Invasive Mycoses and Antifungals, Philipps University Marburg, D-35037 Marburg, Germany
| | | | - Eric Dannaoui
- Parasitology-Mycology Unit, Microbiology Department, APHP, European Georges Pompidou Hospital, Paris-Descartes University, F-75015 Paris, France;
- Dynamyc Research Group, Paris Est Créteil University (UPEC, EnvA), F-94010 Paris, France
- Correspondence: ; Tel.: +33-1-56-09-39-48; Fax: +33-1-56-09-24-46
| |
Collapse
|
21
|
Vega-Chacón Y, de Albuquerque MC, Pavarina AC, Goldman GH, Mima EGDO. Verapamil inhibits efflux pumps in Candida albicans, exhibits synergism with fluconazole, and increases survival of Galleria mellonella. Virulence 2021; 12:231-243. [PMID: 33410730 PMCID: PMC8923067 DOI: 10.1080/21505594.2020.1868814] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The emergence of resistance requires alternative methods to treat Candida albicans infections. We evaluated efficacy of the efflux pump inhibitor (EPI) verapamil (VER) with fluconazole (FLC) against FLC-resistant (CaR) and -susceptible C. albicans (CaS). The susceptibility of both strains to VER and FLC was determined, as well as the synergism of VER with FLC. Experiments were performed in vitro for planktonic cultures and biofilms and in vivo using Galleria mellonella. Larval survival and fungal recovery were evaluated after treatment with VER and FLC. Data were analyzed by analysis of variance and Kaplan-Meier tests. The combination of VER with FLC at sub-lethal concentrations reduced fungal growth. VER inhibited the efflux of rhodamine 123 and showed synergism with FLC against CaR. For biofilms, FLC and VER alone reduced fungal viability. The combination of VER with FLC at sub-lethal concentrations also reduced biofilm viability. In the in vivo assays, VER and FLC used alone or in combination increased the survival of larvae infected with CaR. Reduction of fungal recovery was observed only for larvae infected with CaR and treated with VER with FLC. VER reverted the FLC-resistance of C. albicans. Based on the results obtained, VER reverted the FLC-resistance of C. albicans and showed synergism with FLC against CaR. VER also increased the survival of G. mellonella infected with CaR and reduced the fungal recovery.
Collapse
Affiliation(s)
- Yuliana Vega-Chacón
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, São Paulo State University (UNESP), School of Dentistry , Araraquara, Brazil
| | - Maria Carolina de Albuquerque
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, São Paulo State University (UNESP), School of Dentistry , Araraquara, Brazil
| | - Ana Cláudia Pavarina
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, São Paulo State University (UNESP), School of Dentistry , Araraquara, Brazil
| | - Gustavo Henrique Goldman
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo (USP) , Ribeirão Preto, Brazil
| | - Ewerton Garcia de Oliveira Mima
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, São Paulo State University (UNESP), School of Dentistry , Araraquara, Brazil
| |
Collapse
|
22
|
Tits J, Cammue BPA, Thevissen K. Combination Therapy to Treat Fungal Biofilm-Based Infections. Int J Mol Sci 2020; 21:ijms21228873. [PMID: 33238622 PMCID: PMC7700406 DOI: 10.3390/ijms21228873] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/21/2022] Open
Abstract
An increasing number of people is affected by fungal biofilm-based infections, which are resistant to the majority of currently-used antifungal drugs. Such infections are often caused by species from the genera Candida, Aspergillus or Cryptococcus. Only a few antifungal drugs, including echinocandins and liposomal formulations of amphotericin B, are available to treat such biofilm-based fungal infections. This review discusses combination therapy as a novel antibiofilm strategy. More specifically, in vitro methods to discover new antibiofilm combinations will be discussed. Furthermore, an overview of the main modes of action of promising antibiofilm combination treatments will be provided as this knowledge may facilitate the optimization of existing antibiofilm combinations or the development of new ones with a similar mode of action.
Collapse
|
23
|
Lund PA, De Biase D, Liran O, Scheler O, Mira NP, Cetecioglu Z, Fernández EN, Bover-Cid S, Hall R, Sauer M, O'Byrne C. Understanding How Microorganisms Respond to Acid pH Is Central to Their Control and Successful Exploitation. Front Microbiol 2020; 11:556140. [PMID: 33117305 PMCID: PMC7553086 DOI: 10.3389/fmicb.2020.556140] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 08/21/2020] [Indexed: 12/20/2022] Open
Abstract
Microbes from the three domains of life, Bacteria, Archaea, and Eukarya, share the need to sense and respond to changes in the external and internal concentrations of protons. When the proton concentration is high, acidic conditions prevail and cells must respond appropriately to ensure that macromolecules and metabolic processes are sufficiently protected to sustain life. While, we have learned much in recent decades about the mechanisms that microbes use to cope with acid, including the unique challenges presented by organic acids, there is still much to be gained from developing a deeper understanding of the effects and responses to acid in microbes. In this perspective article, we survey the key molecular mechanisms known to be important for microbial survival during acid stress and discuss how this knowledge might be relevant to microbe-based applications and processes that are consequential for humans. We discuss the research approaches that have been taken to investigate the problem and highlight promising new avenues. We discuss the influence of acid on pathogens during the course of infections and highlight the potential of using organic acids in treatments for some types of infection. We explore the influence of acid stress on photosynthetic microbes, and on biotechnological and industrial processes, including those needed to produce organic acids. We highlight the importance of understanding acid stress in controlling spoilage and pathogenic microbes in the food chain. Finally, we invite colleagues with an interest in microbial responses to low pH to participate in the EU-funded COST Action network called EuroMicropH and contribute to a comprehensive database of literature on this topic that we are making publicly available.
Collapse
Affiliation(s)
- Peter A Lund
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Daniela De Biase
- Department of Medico-Surgical Sciences and Biotechnologies, Laboratory affiliated to the Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Sapienza University of Rome, Latina, Italy
| | - Oded Liran
- Department of Plant Sciences, MIGAL - Galilee Research Institute, Kiryat-Shemona, Israel
| | - Ott Scheler
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Nuno Pereira Mira
- Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Zeynep Cetecioglu
- Department of Chemical Engineering, KTH Royal Institute of Technology, Stockholm, Sweden
| | | | - Sara Bover-Cid
- IRTA, Food Safety Programme, Finca Camps i Armet, Monells, Spain
| | - Rebecca Hall
- School of Biosciences, Kent Fungal Group, University of Kent, Canterbury, United Kingdom
| | - Michael Sauer
- Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria
| | - Conor O'Byrne
- Bacterial Stress Response Group, Microbiology, School of Natural Sciences, NUI Galway, Galway, Ireland
| |
Collapse
|
24
|
Galleria mellonella for the Evaluation of Antifungal Efficacy against Medically Important Fungi, a Narrative Review. Microorganisms 2020. [DOI: 10.3390/microorganisms8030390
expr 890942362 + 917555800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
The treatment of invasive fungal infections remains challenging and the emergence of new fungal pathogens as well as the development of resistance to the main antifungal drugs highlight the need for novel therapeutic strategies. Although in vitro antifungal susceptibility testing has come of age, the proper evaluation of therapeutic efficacy of current or new antifungals is dependent on the use of animal models. Mammalian models, particularly using rodents, are the cornerstone for evaluation of antifungal efficacy, but are limited by increased costs and ethical considerations. To circumvent these limitations, alternative invertebrate models, such as Galleria mellonella, have been developed. Larvae of G. mellonella have been widely used for testing virulence of fungi and more recently have proven useful for evaluation of antifungal efficacy. This model is suitable for infection by different fungal pathogens including yeasts (Candida, Cryptococcus, Trichosporon) and filamentous fungi (Aspergillus, Mucorales). Antifungal efficacy may be easily estimated by fungal burden or mortality rate in infected and treated larvae. The aim of the present review is to summarize the actual data about the use of G. mellonella for testing the in vivo efficacy of licensed antifungal drugs, new drugs, and combination therapies.
Collapse
|
25
|
Jemel S, Guillot J, Kallel K, Botterel F, Dannaoui E. Galleria mellonella for the Evaluation of Antifungal Efficacy against Medically Important Fungi, a Narrative Review. Microorganisms 2020; 8:microorganisms8030390. [PMID: 32168839 PMCID: PMC7142887 DOI: 10.3390/microorganisms8030390] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/05/2020] [Accepted: 03/08/2020] [Indexed: 12/26/2022] Open
Abstract
The treatment of invasive fungal infections remains challenging and the emergence of new fungal pathogens as well as the development of resistance to the main antifungal drugs highlight the need for novel therapeutic strategies. Although in vitro antifungal susceptibility testing has come of age, the proper evaluation of therapeutic efficacy of current or new antifungals is dependent on the use of animal models. Mammalian models, particularly using rodents, are the cornerstone for evaluation of antifungal efficacy, but are limited by increased costs and ethical considerations. To circumvent these limitations, alternative invertebrate models, such as Galleria mellonella, have been developed. Larvae of G. mellonella have been widely used for testing virulence of fungi and more recently have proven useful for evaluation of antifungal efficacy. This model is suitable for infection by different fungal pathogens including yeasts (Candida, Cryptococcus, Trichosporon) and filamentous fungi (Aspergillus, Mucorales). Antifungal efficacy may be easily estimated by fungal burden or mortality rate in infected and treated larvae. The aim of the present review is to summarize the actual data about the use of G. mellonella for testing the in vivo efficacy of licensed antifungal drugs, new drugs, and combination therapies.
Collapse
Affiliation(s)
- Sana Jemel
- EA Dynamyc UPEC, EnvA, USC Anses, Faculté de Médecine de Créteil, 94000 Créteil, France; (S.J.); (J.G.); (F.B.)
- Université Tunis EL Manar, Faculté de médecine de Tunis, Tunis 1007, Tunisie;
- UR17SP03, centre hospitalo-universitaire La Rabta, Jabbari, Tunis 1007, Tunisie
| | - Jacques Guillot
- EA Dynamyc UPEC, EnvA, USC Anses, Faculté de Médecine de Créteil, 94000 Créteil, France; (S.J.); (J.G.); (F.B.)
| | - Kalthoum Kallel
- Université Tunis EL Manar, Faculté de médecine de Tunis, Tunis 1007, Tunisie;
- UR17SP03, centre hospitalo-universitaire La Rabta, Jabbari, Tunis 1007, Tunisie
| | - Françoise Botterel
- EA Dynamyc UPEC, EnvA, USC Anses, Faculté de Médecine de Créteil, 94000 Créteil, France; (S.J.); (J.G.); (F.B.)
| | - Eric Dannaoui
- EA Dynamyc UPEC, EnvA, USC Anses, Faculté de Médecine de Créteil, 94000 Créteil, France; (S.J.); (J.G.); (F.B.)
- Hôpital Européen Georges Pompidou, APHP, Unité de Parasitologie-Mycologie, Service de Microbiologie, 75015 Paris, France
- Université René Descartes, Faculté de médecine, 75006 Paris, France
- Correspondence: ; Tel.: +33-1-56-09-39-48; Fax: +33-1-56-09-24-46
| |
Collapse
|