Current Medicinal Chemistry - Volume 32, Issue 37, 2025
Volume 32, Issue 37, 2025
-
-
The Gut Microbiota-anxiety Connection: Evidence, Mechanisms, and Therapeutic Strategies
More LessAuthors: Geir Bjørklund, Monica Butnariu, Maryam Dadar and Yuliya SemenovaThe gut-brain axis (GBA), a bidirectional communication system between the gut and the brain, has emerged as a critical player in mental health. The interest in the connection between anxiety disorders (AD) and the gut microbiota is growing. This paper provides an overview of gut microbiota’s role in dysregulation in anxiety, including alterations in gut microbiota (dysbiosis), leaky gut, metabolic endotoxemia, and the effect of antipsychotic medications. The mechanisms underlying the gut microbiota-anxiety (GMA) connection, such as neurotransmitter production, immune dysregulation, and GBA communication, are discussed. Furthermore, the paper explores gut microbiota-based therapeutic strategies, including probiotics, prebiotics, symbiotics, fecal microbiota transplantation, and dietary interventions, as potential approaches for anxiety management. This research field's clinical implications and future directions are also examined, underscoring that more studies are needed on gut microbiota’s role in anxiety disorders. The conclusion highlights the importance of this ongoing research and the potential for personalized therapeutic interventions, instilling hope and optimism for the future of anxiety management and providing reassurance about the potential for personalized therapeutic interventions in this field.
-
-
-
Effect of Biomedical Hydrogels on Exercise-induced Muscle Damage: A Narrative Review
More LessAuthors: Jie Cai, Mingbo Fan, Ailin Yu and Chenghu WuExercise-induced muscle damage (EIMD) is a common occurrence among athletes and individuals engaged in physical fitness activities. Muscle strains result from excessive or repetitive muscle tension, leading to tissue damage, inflammation, and pain. These strains can range from mild discomfort to severe damage, resulting in pain, inflammation, and reduced functionality. Effective management of muscle damage is crucial for promoting recovery and returning individuals to their desired level of activity. Conventional treatment modalities such as rest, ice, compression, and elevation (RICE), physical therapy, and nonsteroidal anti-inflammatory drugs (NSAIDs) have limitations in terms of efficacy and long-term outcomes. Consequently, there is a need for innovative approaches that not only address the symptoms but also promote healing and prevention of future injuries. Hydrogels are three-dimensional crosslinked networks of hydrophilic polymers that have gained significant attention in the field of biomedicine. Their unique properties, drug-delivery capabilities, and capacity to provide mechanical support make them promising tools in muscle damage management. Biomedical hydrogels hold significant potential as a preventive or alleviative approach for EIMD. This review provides a comprehensive overview of biomedical hydrogels as a promising approach for preventing and alleviating EIMD, addressing current challenges, and outlining future directions for research and development in the field.
-
-
-
Trends on Potential Boron-containing Drugs by Advancements in Ligand-protein Crystals Complexes
More LessBackgroundBoron-containing compounds (BCC) are attracting attention in drug design. Certain chemical features invite the exploration of efficacious interactions on known and potential drug targets for human use.
ObjectiveThe objective of this study is to analyze the reported crystal structure studies to determine trends resulting from the inclusion of boron atoms in potential drugs.
MethodsPublished data in the Protein Data Bank (PDB) with at least one BCC were analyzed; both ligands and targets were analyzed to describe the inferred or reported biological activity and the potential application as a drug in the treatment of human diseases.
ResultsData from the PDB indicated targets for certain infectious diseases and cancers; however, potential treatments may extend to many other human pathologies as a consequence of the careful analysis of BCCs with proteins. All classes of enzymes and receptors have been crystallized with BCCs as ligands with most complexes demonstrating interactions in the regions known as relevant to protein function.
ConclusionThe number of crystallized BCC-proteins complexes is increasing, and the variability of proteins expands the possibilities of medical applications. Currently, most systems are related to cancer growth and treatment, but deeper analysis may expand BCC utility and efficacy to many other chronic and degenerative diseases.
-
-
-
Exploring the Multitarget Therapeutic Potential of Mangostin Derivatives
More LessAuthors: Jordan Joon-Yip Lew and Yeun-Mun ChooMangostins and their derivatives exhibit broad therapeutic potential, with structural modifications enhancing their efficacy against cancer, inflammation, neurodegenerative disorders, oxidative stress, and microbial infections. Modified derivatives have demonstrated improved effectiveness in cancer treatment. They exhibit potent anti-inflammatory effects for conditions like pulmonary fibrosis and Parkinson’s disease and neuroprotective benefits through cholinesterase inhibition and protection against oxidative damage. For example, structural modifications of α-mangostin (1) significantly enhanced its cytotoxicity, with the 3,6-dibenzylated (4) derivative achieving three times greater efficacy against HL-60 cells and diacetyl (8) and benzoyl (9) derivatives and two- and four-fold improvements against HT-29 cells. The enhanced antioxidant properties of these derivatives improve radical scavenging, lipid protection, and metal ion binding. They possess antimicrobial properties against multidrug-resistant bacteria and fungi, with several derivatives exhibiting high membrane selectivity, low toxicity, and strong in vivo efficacy. Their antimalarial, antiparasitic, and antiviral activities further expand their therapeutic uses, including inhibition of viral proteases. Structural modifications of α-mangostin (1) show promising clinical applications, including enhanced cytotoxicity in cancer therapy with the 3,6-dibenzylated (4), diacetyl (8), and benzoyl (9) derivatives, potent anti-inflammatory activity with PDE4-targeting compound (43), and effective antimicrobial properties in derivatives (18 and 22) against multidrug-resistant infections.
-
-
-
Lysyl Oxidase as a Target to Reduce Graft Failure Post Solid Organ Transplantation, a Potential Target for Novel Treatment
More LessThe present review was undertaken to clarify the potential role of the lysyl oxidase (Lox) family of enzymes in delaying graft dysfunction. Delayed graft failure is a well-known event that occurs post-transplantation period. Ischemia and trauma to the graft tissue before or during the operation procedures are likely to be the most important etiological causes of this complication. The lox proteins family including Lox and Lox- like proteins (LoxL1-4) are copper-dependent enzymes that catalyze the cross-linking of collagens to stabilize extracellular matrix (ECM). Hypoxia-induced factor 1-α (HIF-1α) and transforming growth factor β (TGF-β) are two upstream regulators of the Lox proteins family whose expression increased following hypoxia and tissue injury. Lox proteins’ overactivation upregulates several intracellular transduction pathways to promote oxidative stress (OS), ECM proteins accumulation, and epithelial to mesenchymal transition (EMT) contribute to vascular stiffness and tissue fibrogenesis, which increase the risk of graft failure post solid organ transplantation (SOT). Preclinical studies have shown that Lox protein inhibitors have the potential to prevent organ fibrosis. Regarding the molecular effects of Lox proteins in causing tissue fibrosis, these molecules can be further investigated as a drug target in reducing the possibility of organ fibrosis after allograft transplantation.
-
-
-
Computational Model to Predict Potential Therapeutic Targets Employing Generative Adversarial Networks for Analysis of Proteins Involved in Mycobacterium fortuitum Biofilm Formation
More LessAuthors: Shan Ghai, Rahul Shrivastava and Shruti JainA planktonic population of bacteria can form a biofilm by adhesion and colonization. Proteins known as “adhesins” can bind to certain environmental structures, such as sugars, which will cause the bacteria to attach to the substrate. Quorum sensing is used to establish the population is dense enough to form a biofilm. This paper presents a comprehensive overview of our investigation into these processes, specifically focusing on Mycobacterium fortuitum, an emerging pathogen of increasing clinical relevance. In our study, we detailed the methodology employed for the proteomic analysis of M. fortuitum, as well as our innovative application of Generative Adversarial Networks (GANs). These advanced computational tools allow us to analyze complex data sets and identify patterns that might otherwise remain obscured. With a particular focus on the effectiveness of GAN, the identified proteins and their potential roles in the context of M. fortuitum's pathogenesis were discussed. The insights gained from this study can significantly contribute to our understanding of this emerging pathogen and pave the way for developing targeted interventions, potentially leading to improved diagnostic tools and more effective therapeutic strategies against M. fortuitum infection. The authors achieved 95.43% accuracy for the generator and 87.89% for the discriminator. The model was validated by considering different Machine learning algorithms, reinforcing that integrating computational techniques with microbiological investigations can significantly enhance our understanding of emerging pathogens. Overall, this study emphasizes the importance of exploring the molecular mechanisms behind biofilm formation and pathogenicity, providing a foundation for future research that could lead to innovative solutions in combating infections caused by M. fortuitum and other similar pathogens.
-
-
-
Poly Lactic Co-glycolic Acid d-α-tocopheryl Polyethylene Glycol 1000 Succinate Fabricated Polyethylene Glycol Hybrid Nanoparticles of Imatinib Mesylate for the Treatment of Glioblastoma Multiforme
More LessAuthors: Sankha Bhattacharya, Prafull Shinde, Amit Page and Bhupendra G. PrajapatiAimsThis study aimed to develop Imatinib Mesylate (IMT)-loaded Poly Lactic-co-Glycolic Acid (PLGA)-D-α-tocopheryl polyethylene glycol succinate (TPGS)- Polyethylene glycol (PEG) hybrid nanoparticles (CSLHNPs) with optimized physicochemical properties for targeted delivery to glioblastoma multiforme.
BackgroundGlioblastoma multiforme (GBM) is the most destructive type of brain tumor with several complications. Currently, most treatments for drug delivery for this disease face challenges due to the poor blood-brain barrier (BBB) and lack of site-specific delivery. Imatinib Mesylate (IMT) is one of the most effective drugs for GBM, but its primary issue is low bioavailability. Therefore, nanotechnology presents a promising solution for targeted IMT delivery to GBM. This article primarily explores the fabrication of IMT-loaded core-shell lipid-polymer hybrid nanoparticles (CSLHNPs) to achieve enhanced brain delivery with therapeutic efficacy.
ObjectiveThe primary objective of this study is to develop optimized, stable IMT-loaded hybrid nanoparticles with an encapsulated polymer matrix and to evaluate these nanoparticles using sophisticated instruments such as SEM and TEM to achieve smooth, spherical nanoparticles in a monodispersed phase.
MethodsThe enhanced stable formulation yielded a notable increase in entrapment efficiency, reaching 58.89 ± 0.5%. The physical stability analysis of nanoparticles was assessed over 30 days under conditions of 25 ± 2°C and 60 ± 5% relative humidity. Hemolytic assays affirmed the biocompatibility and safety profile of the nanoparticles. In vitro drug release kinetics revealed a sustained IMT release over 48 hours.
ResultsThe formulated CSLHNPs achieved a narrow size distribution with a mean vesicle diameter of 155.03 ± 2.41 nm and a low polydispersity index (PDI) of 0.23 ± 0.4, indicating monodispersity. A high negative zeta potential of -23.89 ± 3.47 mV ensured excellent colloidal stability in physiological conditions. XRD analysis confirmed the successful encapsulation of IMT within the nanoparticle matrix, with the drug transitioning to an amorphous state for enhanced dissolution. During cell-cell viability assays on LN229, glioblastoma cells were treated with IMT-loaded nanoparticles and showed a significantly enhanced inhibitory effect compared to free IMT. These hybrid nanoparticles demonstrated potential in reducing oxidative stress-induced cellular damage by mitigating reactive oxygen species (ROS). Thus, the prepared IMT hybrid nanoparticles showed higher cellular uptake and superior cytotoxicity compared to the plain drug.
ConclusionThis study posits the IMT-PLGA-TPGS-DSPE PEG 2000-CSPLHNPs as a formidable and innovative drug delivery system for Glioblastoma Multiforme (GBM) treatment, warranting further exploration into their clinical application potential. Future work could involve conducting in vivo studies to evaluate the pharmacokinetics, biodistribution, and therapeutic efficacy of the IMT-PLGA-TPGS-DSPE PEG 2000-CSPLHNPs in animal models of Glioblastoma Multiforme (GBM). Additionally, further research may focus on optimizing the nanoparticle formulation for enhanced targeting capabilities, investigating long-term stability under varied storage conditions, exploring potential combination therapies to synergize with the nanoparticles, and assessing the scalability and manufacturability of the developed drug delivery system for potential clinical translation. Integration of advanced imaging techniques for real- time tracking and visualization of nanoparticle distribution within tumours could also be a promising direction for future investigations.
-
-
-
Analysis of the Mechanism Underlying Radiotherapy Resistance Caused by Oligodendroglia Cells in Glioblastoma by Applying the Single-cell RNA Sequencing Technology
More LessAuthors: Qinghua Yuan, Weida Gao, Mian Guo and Bo LiuBackgroundGlioblastoma (GBM) is an aggressive malignancy. The inherent resistance of GBM to radiotherapy poses great challenges for clinical treatment.
ObjectivesThe primary objective of this study is to explore the molecular mechanisms of radiotherapy resistance in GBM and identify the key influencing factors that contribute to this phenomenon.
MethodsThe single-cell RNA sequencing (scRNA-seq) data of GBM were downloaded from the Gene Expression Omnibus (GEO) database. Cells were clustered using the Seurat R package, and the clusters were annotated using the CellMarker database. Pseudotime analysis was conducted using Monocle2. Marker scores were calculated based on the RNA-seq data of GBM from the UCSC database, and the enrichment of Hallmark gene sets was measured with the AUCell package. Furthermore, the most frequently mutated genes were identified using the simple nucleotide variation data from The Cancer Genome Atlas (TCGA) applying the maftools package.
ResultsThis study identified two oligodendrocyte subsets (ODC3 and ODC4) as radiotherapy-resistant groups in GBM. Enrichment and Pseudotime analysis revealed that the inflammatory response and immune activation pathways were enriched in ODC3, while the cell division and interferon response pathways were enriched in ODC4. The enrichment scores of hallmark gene sets further confirmed that ODC3 and ODC4 subpopulations developed radiotherapy resistance via distinct molecular mechanisms. Analysis of gene mutation frequencies showed that TP53 exhibited the most significant change in mutation frequency, indicating that it was an important risk factor involved in radiotherapy resistance in GBM.
ConclusionWe identified two ODC subpopulations that exhibited resistance to radiotherapy, providing a new perspective and potential targets for personalized treatment strategies for GBM.
-
-
-
Identification of Key Genes and Pathways in Lenvatinib-resistant Hepatocellular Carcinoma using Bioinformatic Analysis and Experimental Validation
More LessAuthors: Ming Yang, ZhaoYue Wang, Riga Su, Dongbing Li and Jun ZhouBackgroundResistance to lenvatinib poses a serious threat to the therapy of patients with Hepatocellular Carcinoma (HCC). The mechanism by which HCC develops resistance to lenvatinib is currently unknown.
ObjectiveThe aim of this study was to identify key genes and pathways involved in lenvatinib resistance in HCC using bioinformatic analysis and experimental validation.
MethodsDifferentially expressed genes (DEGs) were identified from the GSE186191 gene expression profile, comparing HCC cell lines with lenvatinib-resistant HCC cell lines. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses were then carried out using DAVID. A protein-protein interaction network was constructed to visualize DEGs and identify hub genes. The expression and prognostic significance of these hub genes were further examined. Additionally, genomic enrichment analysis (GSEA) was utilized to investigate the potential functions of key genes. Following this, the presence of AHSG was validated in both the original Huh7 cells and the lenvatinib-resistant Huh7 (Huh7LR) cells resistant to lenvatinib through the utilization of quantitative real-time PCR (qRT-PCR).
ResultsA total of 232 DEGs were identified between HCC cell lines and those that are resistant to lenvatinib. These DEGs were significantly associated with arrhythmogenic right ventricular cardiomyopathy, hypertrophic cardiomyopathy, dilated cardiomyopathy, and mucin-type O-glycan biosynthesis. Three hub genes, including AHSG, C6, and ORM1, were identified. The low expression of AHSG showed a poorer prognosis in HCC. GSEA demonstrated a significant correlation between low AHSG expression and pathways involving fatty acid metabolism, ribosome function, glycine, serine, and threonine metabolism, peroxisome activity, and bile acid biosynthesis. The expression of AHSG was notably reduced in Huh7LR cells (p = 0.006) compared to Huh7 cells.
ConclusionDiminished AHSG expression is strongly associated with lenvatinib resistance in HCC, suggesting that it may have implications for developing effective strategies to overcome this resistance.
-
-
-
The Effect of Gallic Acid on the Alleviation of the Chemotherapyinduced Myelosuppression
More LessAuthors: Junyi Luo, Zhaoxia Zhang, Liming Jin, Zhaoying Wang, Qiuyue Sun and Dawei HeObjectiveThis study aims to investigate the effect of Gallic Acid (GA) on the alleviation of chemotherapy-induced bone marrow suppression, with a comparison to Diyu sheng bai tablets (DYSB) and RhG-CSF.
MethodsA mouse model of bone marrow suppression was established in BALB/c mice using intraperitoneal injections of cyclophosphamide (CTX). All procedures were performed after obtaining ethical clearance from the institutional animal ethics committee. Mice were treated with low (100 mg/kg/d), medium (200 mg/kg/d), and high (400 mg/kg/d) doses of Gallic Acid (GA) to mitigate CTX-induced bone marrow suppression. In parallel, mice in the positive control group were also treated with DYSB and RhG-CSF at their respective standard doses (DYSB: 100 mg/kg/day, RhG-CSF: 125 mg/kg/day). The efficacy of GA in alleviating chemotherapy-induced bone marrow suppression was evaluated through blood cell counts, immune organ (thymus and spleen) indices, bone marrow nucleated cell (BMNC) counts, cell cycle analysis, apoptosis, histopathology of bone marrow and spleen, and analysis of splenic hematopoietic factors.
ResultsCTX induced a decrease in peripheral blood cells and BMNC counts, reduced spleen and thymus indices, and diminished abnormal pathology of bone marrow and spleen, as well as decreasing disturbances in hematopoietic factors. GA was able to alleviate these abnormalities in the bone marrow. It modulated cell proliferation and apoptosis, adjusted the proportion of cells in the G0/G1 phase, and reduced apoptosis in femoral bone marrow.
ConclusionGallic Acid (GA) alleviates chemotherapy-induced bone marrow suppression by improving immune organ function, promoting bone marrow cell recovery, and inhibiting apoptosis. These findings support GA as a potential adjunct therapy for chemotherapy, with promising clinical applications.
-
-
-
An Azomethine Derivative, 1-(4-nitrophenyl)-N-phenylmethanimine (BCS2) Ameliorated 7,12-dimethylbenz(a)anthracene-induced Mammary Carcinoma through Nrf2-Keap1-HO-1 Pathway
More LessAuthors: Reetuparna Acharya, Pran Kishore Deb and Shakti Prasad PattanayakAimsThe aim of this study is the evaluation of an Azomethine derivative, BCS2, for its antioxidant and anti-tumor activities against mammary carcinoma through the Nrf2-Keap1-HO-1 pathway.
BackgroundThe global prevalence of breast cancer is rising at an alarming rate. The facilitation of abnormal cell proliferation in mammary carcinoma occurs due to the disruption of signaling pathways that balance pro- and antioxidant status, thereby producing oxidative stress that disrupts genomic stability. Therefore, introducing a potent antioxidant molecule with antitumor activity is of paramount importance for treating breast cancer.
ObjectiveSynthesis, characterization, and in vitro, in vivo, and in silico evaluation of an Azomethine derivative, BCS2, for its antioxidant and anti-tumor activities against chemical carcinogen-induced mammary carcinogenesis in Sprague-Dawley rats.
MethodsAn azomethine derivative, 1-(4-nitrophenyl)-N-phenylmethanimine (BCS2), was synthesized and characterized based on its spectral data. The cytotoxic potential was observed on breast cancer cells, MCF-7, MDA-MB-231, and MDA-MB-468. The in vivo chemotherapeutic potential of BCS2 was established on 7,12-dimethylbenz(a)anthracene (DMBA) induced breast cancer in Sprague-Dawley (SD) rats. The effect of BCS2 on kelch-like ECH-associated protein-1 (Keap1), Nrf2, heme oxygenase-1 (HO-1), mitogen-activated protein kinase (MAPK), and nuclear factor kappa-light-chain-enhancer of activated-B (NF-κB) was evaluated through ELISA and qPCR techniques. Furthermore, the binding potential and stability of BCS2 with Keap-1, HO-1, and MAPK were predicted using in silico molecular docking and dynamics studies. Additionally, drug-likeness properties of BCS2 were evaluated using in silico ADMET tools.
ResultsBCS2 showed remarkable cytotoxic activity on MCF-7 cells followed by MDA-MB-231 and MDA-MB-468 cells having an IC50 of 2.368 µM, 4.843 µM and 6.472 µM respectively, without affecting normal breast cells, MCF-10A. In the DMBA-induced animal model, BCS2 showed potent antitumor potential and showed protective action on endogenous-enzymatic and non-enzymatic antioxidants in cancer-bearing animals. Marked improvement in cellular architecture and ultrastructure of breast/tumor tissues excised from experimental animals was noted through histopathological and field emission scanning electron microscopy (FESEM) analyses. Significant upregulation of antioxidant proteins, Keap1 and HO-1, and downregulation of inflammatory proteins, MAPK, and NF-κB was observed after BCS2 treatment. The in silico computational studies predicted the potent binding of BCS2 with the active pockets of Keap1, HO-1, and MAPK proteins that validated the biological findings.
ConclusionThe study revealed BCS2's potent antioxidant and antitumor potential against mammary carcinoma through the Nrf2-Keap1-HO-1 signaling pathway.
-
-
-
Integrated Transcriptome and Proteome Analyses Reveal Differentially Expressed Genes and Proteins in Granulosa Cells from Female Patients with Metabolic Syndrome-associated Infertility
More LessAuthors: Fangli Dong, Wanjun Zhang, Bo Sun, Wenbin Niu, Jun Zhai, Yihong Guo and Fang WangBackgroundMetabolic Syndrome (MS) is a cluster of conditions that significantly increase the risk of infertility in women. Granulosa cells are crucial for ovarian folliculogenesis and fertility. Understanding molecular alterations in these cells can provide insights into MS-associated infertility.
ObjectiveThis study aimed to investigate Differentially Expressed Genes (DEGs) and Proteins (DEPs) in granulosa cells from female patients with MS-associated infertility.
MethodsTranscriptome and proteome analyses were integrated to compare granulosa cells from three MS patients with infertility to three control subjects. RNA sequencing and quantitative proteomics analyses were conducted, followed by differential expression analysis, Gene Set Enrichment Analysis (GSEA), and Protein-protein Interaction (PPI) network construction. Functional enrichment of overlapping DEGs and DEPs and potential drug-protein interactions were also explored. Hub genes identified by PPI were validated via quantitative Polymerase Chain Reaction (qPCR) and western blot assays.
ResultsPrincipal Component Analysis (PCA) demonstrated a distinct separation between MS and control groups, indicating significant differences in gene and protein expression. A total of 1,046 upregulated and 23 downregulated DEGs, along with 222 upregulated and 412 downregulated DEPs, were identified in the MS group. GSEA highlighted enrichment in processes, like the cell cycle and immune response. Venn diagram revealed 71 overlapping DEGs and DEPs, mainly related to immune regulation. Key hub proteins and potential therapeutic candidates were identified, with hub genes upregulated at the mRNA level, but downregulated at the protein level in granulosa cells of MS patients.
ConclusionThe integrative analyses revealed significant molecular alterations in granulosa cells from MS patients with infertility. Identified DEGs, DEPs, and hub proteins suggested potential therapeutic targets and pathways for addressing MS-associated infertility.
-
-
-
Ab-initio Molecular Dynamics and Density Functional Theory Study of Amodiaquine Analogues as Potential Inhibitors of β-haematin Crystallization
More LessAuthors: Pélagie Manwal A. Mekoung, Kevin A. Lobb and Ibrahim N. MbouombouoIntroductionPrevention of the formation of β-haematin is the target of several existing antimalarials drugs, most notably chloroquine. This target is therefore attractive for the development of new molecules with antimalarial potential.
MethodsIn this study, we have used a combination of ab-initio molecular dynamics and density functional tight-binding to examine the possible interaction mechanisms between five amodiaquine analogues and four conformations of haematin. Reactivity and stability of these complexes were investigated using bond length (Fe-N and Fe-O), energies (HOMO-LUMO) and molecular dynamics.
ResultsResults revealed a good interaction between haem and the compounds, stable geometries of complexes.
ConclusionThe findings from this study are valuable because they can aid the design and understanding of new therapeutic molecules that could be used to treat drug-resistant malaria, a global threat of today.
-
-
-
Azole Homodimers as Promising Antifungal Agents: Synthesis, Biological Activity Evaluation and Molecular Modeling
More LessIntroductionA new series of triazoles with antifungal activity have been synthesized in a one-step fashion by direct reaction of 2-(2,4-difluorophenyl)-2,3-epoxy-1-(1H-1,2,4-triazol-1-yl)propane with various diamines.
MethodsObtained compounds were profiled for biological activity against pathogenic strains of fungi C. albicans and A. niger. Molecular modeling was used to predict binding modes.
ResultsThe lead compound was 4 times more active against C. albicans than fluconazole and demonstrated a wider spectrum of activity, inhibiting the growth of A. niger.
ConclusionThe results presented herein can contribute to the development of novel antifungal therapeutic agents.
-
Volumes & issues
-
Volume 32 (2025)
-
Volume (2025)
-
Volume 31 (2024)
-
Volume 30 (2023)
-
Volume 29 (2022)
-
Volume 28 (2021)
-
Volume 27 (2020)
-
Volume 26 (2019)
-
Volume 25 (2018)
-
Volume 24 (2017)
-
Volume 23 (2016)
-
Volume 22 (2015)
-
Volume 21 (2014)
-
Volume 20 (2013)
-
Volume 19 (2012)
-
Volume 18 (2011)
-
Volume 17 (2010)
-
Volume 16 (2009)
-
Volume 15 (2008)
-
Volume 14 (2007)
-
Volume 13 (2006)
-
Volume 12 (2005)
-
Volume 11 (2004)
-
Volume 10 (2003)
-
Volume 9 (2002)
-
Volume 8 (2001)
-
Volume 7 (2000)
Most Read This Month