Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (22)

Search Parameters:
Keywords = reverse virtual docking

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
898 KB  
Proceeding Paper
CRISPR-Cas as a Chemically Programmable System: Advances in Modulation and Delivery
by Yukti Sabikhi, Anshika Singh, Chhavi Dudeja, Sameen Masroor and Richa Gupta
Chem. Proc. 2025, 18(1), 69; https://doi.org/10.3390/ecsoc-29-26883 - 13 Nov 2025
Viewed by 182
Abstract
CRISPR-Cas systems have transformed genome engineering with their exceptional precision, programmability, and affordability. Although they originate from microbial defense mechanisms, expanding their use, especially in therapeutics, requires a chemically oriented framework that allows for tunable, reversible, and safe gene editing. This review offers [...] Read more.
CRISPR-Cas systems have transformed genome engineering with their exceptional precision, programmability, and affordability. Although they originate from microbial defense mechanisms, expanding their use, especially in therapeutics, requires a chemically oriented framework that allows for tunable, reversible, and safe gene editing. This review offers a multidisciplinary look at recent progress in the structural, synthetic, and computational aspects of CRISPR-Cas technologies. Structural analyses examine the domain architectures of Cas enzymes, including the recognition (REC), nuclease (HNH and RuvC), and PAM-interacting domains, emphasizing the catalytic importance of divalent metal ions. Comparative insights into Cas9, Cas12, and Cas13 demonstrate functional diversity across DNA- and RNA-targeting systems, supported by high-resolution structural data on guide RNA pairing and conformational dynamics. The review highlights advances in chemical modulation, such as anti-CRISPR proteins, small-molecule inhibitors, and stimuli-responsive switches, focusing on structure–activity relationships. Additionally, bioorganic delivery systems like lipid nanoparticles, polymers, and cell-penetrating peptides are discussed for their role in improving in vivo delivery through formulation chemistry. Computational chemistry methods—molecular docking, molecular dynamics simulations, and virtual screening—are identified as critical tools for discovering and optimizing modulators. The use of AI-driven tools is proposed as a promising direction for rational CRISPR design. Overall, this chemistry-focused perspective emphasizes the importance of molecular control in developing the next generation of programmable and safe CRISPR-based therapies. Full article
Show Figures

Figure 1

30 pages, 10669 KB  
Article
Integration of Untargeted Metabolomics, Network Pharmacology, Single-Cell RNA Sequencing, and Molecular Dynamics Simulation Reveals GOT1, CYP1A2, and CA2 as Potential Targets of Huang Qin Decoction Preventing Colorectal Cancer Liver Metastasis
by Tiegang Li, Zheng Yan, Mingxuan Zhou, Wenyi Zhao, Fang Zhang, Silin Lv, Yufang Hou, Zifan Zeng, Liu Yang, Yixin Zhou, Zengni Zhu, Xinyi Ren and Min Yang
Pharmaceuticals 2025, 18(7), 1052; https://doi.org/10.3390/ph18071052 - 17 Jul 2025
Cited by 1 | Viewed by 1913
Abstract
Background: Huang Qin Decoction (HQD) is a well-established Traditional Chinese Medicine (TCM) formulation recognized for its application in the treatment of colorectal cancer (CRC). However, the precise therapeutic mechanisms remain inadequately defined. Methods: This study integrates metabolomics from a mouse model and network [...] Read more.
Background: Huang Qin Decoction (HQD) is a well-established Traditional Chinese Medicine (TCM) formulation recognized for its application in the treatment of colorectal cancer (CRC). However, the precise therapeutic mechanisms remain inadequately defined. Methods: This study integrates metabolomics from a mouse model and network pharmacology to screen potential targets and bio-active ingredients of HQD. The pharmacological activity of HQD for CRC was evidenced via single-cell RNA sequencing (scRNA-seq), molecular docking, and molecular dynamics simulations. Atomic force microscopy (AFM) assays and cellular experimental validation were used to confirm the relative mechanisms. Results: The metabolite profile undergoes significant alterations, with metabolic reprogramming evident during the malignant progression of CRC liver metastasis. Network pharmacology analysis identified that HQD regulates several metabolic pathways, including arginine biosynthesis, alanine, aspartate, and glutamate metabolism, nitrogen metabolism, phenylalanine metabolism, and linoleic acid metabolism, by targeting key proteins such as aspartate aminotransferase (GOT1), cytochrome P450 1A2 (CYP1A2), and carbonic anhydrase 2 (CA2). ScRNA-seq analysis indicated that HQD may enhance the functionality of cytotoxic T cells, thereby reversing the immunosuppressive microenvironment. Virtual verification revealed a strong binding affinity between the identified hub targets and active constituents of HQD, a finding subsequently corroborated by AFM assays. Cellular experiments confirmed that naringenin treatment inhibits the proliferation, migration, and invasion of CRC cells by downregulating GOT1 expression and disrupting glutamine metabolism. Conclusions: Computational prediction and in vitro validation reveal the active ingredients, potential targets, and molecular mechanisms of HQD against CRC liver metastasis, thereby providing a scientific foundation for the application of TCM in CRC treatment. Full article
(This article belongs to the Section Natural Products)
Show Figures

Figure 1

29 pages, 6460 KB  
Article
Flipping the Target: Evaluating Natural LDHA Inhibitors for Selective LDHB Modulation
by Amanda El Khoury and Christos Papaneophytou
Molecules 2025, 30(14), 2923; https://doi.org/10.3390/molecules30142923 - 10 Jul 2025
Cited by 3 | Viewed by 3458
Abstract
Lactate dehydrogenase (LDH) catalyzes the reversible interconversion of pyruvate and lactate, coupled with the redox cycling of NADH and NAD+. While LDHA has been extensively studied as a therapeutic target, particularly in cancer, due to its role in the Warburg effect, [...] Read more.
Lactate dehydrogenase (LDH) catalyzes the reversible interconversion of pyruvate and lactate, coupled with the redox cycling of NADH and NAD+. While LDHA has been extensively studied as a therapeutic target, particularly in cancer, due to its role in the Warburg effect, LDHB remains underexplored, despite its involvement in the metabolic reprogramming of specific cancer types, including breast and lung cancers. Most known LDH inhibitors are designed against the LDHA isoform and act competitively at the active site. In contrast, LDHB exhibits distinct kinetic properties, substrate preferences, and structural features, warranting isoform-specific screening strategies. In this study, 115 natural compounds previously reported as LDHA inhibitors were systematically evaluated for LDHB inhibition using an integrated in silico and in vitro approach. Virtual screening identified 16 lead phytochemicals, among which luteolin and quercetin exhibited uncompetitive inhibition of LDHB, as demonstrated by enzyme kinetic assays. These findings were strongly supported by molecular docking analyses, which revealed that both compounds bind at an allosteric site located at the dimer interface, closely resembling the binding mode of the established LDHB uncompetitive inhibitor AXKO-0046. In contrast, comparative docking against LDHA confirmed their active-site binding and competitive inhibition, underscoring their isoform-specific behavior. Our findings highlight the necessity of assay conditions tailored to LDHB’s physiological role and demonstrate the application of a previously validated colorimetric assay for high-throughput screening. This work lays the foundation for the rational design of selective LDHB inhibitors from natural product libraries. Full article
Show Figures

Graphical abstract

18 pages, 15637 KB  
Article
Molecular Mechanisms of Reversal of Multidrug Resistance in Breast Cancer by Inhibition of P-gp by Cytisine N-Isoflavones Derivatives Explored Through Network Pharmacology, Molecular Docking, and Molecular Dynamics
by Chuangchuang Xiao, Xiaoying Yin, Rui Xi, Chunping Yuan and Yangsheng Ou
Int. J. Mol. Sci. 2025, 26(8), 3813; https://doi.org/10.3390/ijms26083813 - 17 Apr 2025
Viewed by 1508
Abstract
The compound CNI1, identified as a novel antitumor agent based on the cytisine N-isoflavones scaffold, and its series of cytisine N-isoflavones derivatives (CNI2, CNI3, and CNI4), were first isolated from bitter bean seeds, a traditional Chinese medicinal source, by our research team. Cellular [...] Read more.
The compound CNI1, identified as a novel antitumor agent based on the cytisine N-isoflavones scaffold, and its series of cytisine N-isoflavones derivatives (CNI2, CNI3, and CNI4), were first isolated from bitter bean seeds, a traditional Chinese medicinal source, by our research team. Cellular activity assays combined with virtual screening targeting P-gp revealed that CNI1, along with the three cytisine N-isoflavones derivatives, CNI2, CNI3, and CNI4, exhibited significant multidrug resistance (MDR) reversal activity in breast cancer. Despite this promising outcome, the precise molecular mechanisms and key targets involved in the MDR reversal of these compounds remain to be elucidated. To explore potential mechanisms, targets for CNI1, CNII2, CNI3, and CNI4 (CNI1-4) were predicted using SwissTargetPrediction and Pharmmapper databases, while MDR-related targets in breast cancer were retrieved from OMIM and GeneCards. The overlapping targets were utilized to construct a protein–protein interaction (PPI) network to identify core targets. Additionally, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted using the DAVID database to identify relevant signaling pathways. Molecular docking simulations were employed to evaluate the binding sites and energies of CNI1-4 with the identified key targets, with the highest binding energy complexes selected for subsequent molecular dynamics simulations. This study identified 81 intersecting multidrug resistance (MDR) targets and 19 core targets in breast cancer. GO and KEGG pathway enrichment analyses revealed that MDR was primarily mediated by genes involved in cellular processes, apoptosis, protein phosphorylation, as well as the MAPK and PI3K-Akt signaling pathways. Molecular docking studies demonstrated that the binding energies of P-gp, AKT1, and SRC to CNI1-4 were all lower than −10 kcal/mol, indicating strong binding affinities. Molecular dynamics simulations further confirmed the stable and favorable binding interactions of CNI1-4 with AKT1 and P-gp. This study provides preliminary insights into the potential targets and molecular mechanisms of cytisine N-isoflavones compounds in reversing MDR in breast cancer, offering crucial data for the pharmacological investigation of CNI1-4 and supporting the development of P-gp inhibitors. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Figure 1

18 pages, 4829 KB  
Article
Identification of Two Flavonoids as New and Safe Inhibitors of Kynurenine Aminotransferase II via Computational and In Vitro Study
by Redouane Rebai, Luc Jasmin and Abdennacer Boudah
Pharmaceuticals 2025, 18(1), 76; https://doi.org/10.3390/ph18010076 - 10 Jan 2025
Cited by 4 | Viewed by 1399
Abstract
Background/Objectives: Kynurenine aminotransferase II (KAT-II) is a target for treating several diseases characterized by an excess of kynurenic acid (KYNA). Although KAT-II inactivators are available, they often lead to adverse side effects due to their irreversible inhibition mechanism. This study aimed to identify [...] Read more.
Background/Objectives: Kynurenine aminotransferase II (KAT-II) is a target for treating several diseases characterized by an excess of kynurenic acid (KYNA). Although KAT-II inactivators are available, they often lead to adverse side effects due to their irreversible inhibition mechanism. This study aimed to identify potent and safe inhibitors of KAT-II using computational and in vitro approaches. Methods: Virtual screening, MM/GBSA, and molecular dynamics simulations were conducted to identify the top drug candidates, followed by kinetic measurements and in vitro cytotoxicity evaluation. Results: The study showed that two compounds, herbacetin and (-)-Epicatechin exhibited the best scores. Their Glide docking scores are −8.66 kcal/mol and −8.16 kcal/mol, respectively, and their MM/GBSA binding energies are −50.30 kcal/mol and −51.35 kcal/mol, respectively. These scores are superior to those of the standard inhibitor, PF-04859989, which has docking scores of −7.12 kcal/mol and binding energy of −38.41 kcal/mol. ADMET analysis revealed that the selected compounds have favorable pharmacokinetic parameters, moderate bioavailability, and a safe toxicity profile, which supports their potential use. Further, the kinetic study showed that herbacetin and (-)-Epicatechin are reversible KAT-II inhibitors and exhibit a competitive inhibition mechanism. Their half-maximal inhibitory concentrations (IC50) are 5.98 ± 0.18 µM and 8.76 ± 0.76 µM, respectively. The MTT assay for cell toxicity indicated that the two compounds do not affect HepG2 cell viability at the necessary concentration for KAT-II inhibition. Conclusions: These results suggest that herbacetin and (-)-Epicatechin are suitable for KAT-II inhibition and are promising candidates for further development of KAT-II inhibitors. Full article
Show Figures

Figure 1

20 pages, 5989 KB  
Article
Drug Target Investigation of N-p-Coumaroyl-N’-Caffeoylputrescine, a Naturally-Occurring Alkaloid Derived from Saxifraga tangutica
by Chuang Liu, Jun Dang and Minchen Wu
Antioxidants 2025, 14(1), 12; https://doi.org/10.3390/antiox14010012 - 25 Dec 2024
Cited by 2 | Viewed by 1494
Abstract
The exploration of drug targets has always been a priority in new drug research, and this work is even more essential for natural active compounds. Saxifraga tangutica is a traditional Tibetan medicine with excellent antioxidant properties. In this study, an alkaloid, N- [...] Read more.
The exploration of drug targets has always been a priority in new drug research, and this work is even more essential for natural active compounds. Saxifraga tangutica is a traditional Tibetan medicine with excellent antioxidant properties. In this study, an alkaloid, N-p-coumaroyl-N’-caffeoylputrescine (PCC), was first isolated from the plant, Saxifraga tangutica, with a DPPH scavenging rate of 0.936 μg/mL. To further identify its target, the drug affinity responsive target stability technique and multiple public databases were integrated to retrieve a total of 317 common targets from comprehensive screening. A further bioinformatics analysis not only identified 13 hub targets but also indicated PCC as having biological activities against cancer and affecting metabolic diseases. Integrating reverse virtual docking, molecular dynamics simulations, and cellular thermal shift assays ultimately focused on HSP90AA1 as the target of PCC. An in vitro study on liver (HepG2) cells and breast (MCF-7) cancer cells revealed that PCC modulates HSP90AA1, subsequently affecting Mut-p53 expression, triggering a cascade effect that reduced adriamycin-induced drug resistance in cells. Furthermore, a prediction of the absorption, distribution, metabolism, excretion, and toxicity was also applied to evaluate the drug-like properties of PCC. Overall, the integrated strategy used in this study successfully identified the target of PCC, providing a valuable paradigm for future research on the action targets of natural products. Full article
Show Figures

Figure 1

16 pages, 3715 KB  
Article
Screening for Potential Antiviral Compounds from Cyanobacterial Secondary Metabolites Using Machine Learning
by Tingrui Zhang, Geyao Sun, Xueyu Cheng, Cheng Cao, Zhonghua Cai and Jin Zhou
Mar. Drugs 2024, 22(11), 501; https://doi.org/10.3390/md22110501 - 5 Nov 2024
Cited by 4 | Viewed by 2358
Abstract
The secondary metabolites of seawater and freshwater blue-green algae are a rich natural product pool containing diverse compounds with various functions, including antiviral compounds; however, high-efficiency methods to screen such compounds are lacking. Advanced virtual screening techniques can significantly reduce the time and [...] Read more.
The secondary metabolites of seawater and freshwater blue-green algae are a rich natural product pool containing diverse compounds with various functions, including antiviral compounds; however, high-efficiency methods to screen such compounds are lacking. Advanced virtual screening techniques can significantly reduce the time and cost of novel antiviral drug identification. In this study, we used a cyanobacterial secondary metabolite library as an example and trained three models to identify compounds with potential antiviral activity using a machine learning method based on message-passing neural networks. Using this method, 364 potential antiviral compounds were screened from >2000 cyanobacterial secondary metabolites, with amides predominating (area under the receiver operating characteristic curve value: 0.98). To verify the actual effectiveness of the candidate antiviral compounds, HIV virus reverse transcriptase (HIV-1 RT) was selected as a target to evaluate their antiviral potential. Molecular docking experiments demonstrated that candidate compounds, including kororamide, mollamide E, nostopeptolide A3, anachelin-H, and kasumigamide, produced relatively robust non-covalent bonding interactions with the RNase H active site on HIV-1 RT, supporting the effectiveness of the proposed screening model. Our data demonstrate that artificial intelligence-based screening methods are effective tools for mining potential antiviral compounds, which can facilitate the exploration of various natural product libraries. Full article
(This article belongs to the Special Issue Marine Drug Discovery through Molecular Docking)
Show Figures

Figure 1

22 pages, 7689 KB  
Article
Identification of Potent Acetylcholinesterase Inhibitors as New Candidates for Alzheimer Disease via Virtual Screening, Molecular Docking, Dynamic Simulation, and Molecular Mechanics–Poisson–Boltzmann Surface Area Calculations
by Hind Yassmine Chennai, Salah Belaidi, Lotfi Bourougaa, Mebarka Ouassaf, Leena Sinha, Abdelouahid Samadi and Samir Chtita
Molecules 2024, 29(6), 1232; https://doi.org/10.3390/molecules29061232 - 10 Mar 2024
Cited by 20 | Viewed by 5692
Abstract
Huperzine A (HUP) plays a crucial role in Alzheimer’s therapy by enhancing cognitive function through increased cholinergic activity as a reversible acetylcholinesterase (AChE) inhibitor. Despite some limitations being seen in AChE inhibitors, ongoing research remains dedicated to finding innovative and more effective treatments [...] Read more.
Huperzine A (HUP) plays a crucial role in Alzheimer’s therapy by enhancing cognitive function through increased cholinergic activity as a reversible acetylcholinesterase (AChE) inhibitor. Despite some limitations being seen in AChE inhibitors, ongoing research remains dedicated to finding innovative and more effective treatments for Alzheimer’s disease. To achieve the goal of the discovery of potential HUP analogues with improved physicochemical properties, less toxic properties, and high biological activity, many in silico methods were applied. Based on the acetylcholinesterase–ligand complex, an e-pharmacophore model was developed. Subsequently, a virtual screening involving a collection of 1762 natural compounds, sourced from the PubChem database, was performed. This screening yielded 131 compounds that exhibited compatibility with the established pharmacophoric hypothesis. These selected ligands were then subjected to molecular docking within the active site of the 4EY5 receptor. As a result, we identified four compounds that displayed remarkable docking scores and exhibited low free binding energy to the target. These top four compounds, CID_162895946, CID_44461278, CID_44285285, and CID_81108419, were submitted to ADMET prediction and molecular dynamic simulations, yielding encouraging findings in terms of their pharmacokinetic characteristics and stability. Finally, the molecular dynamic simulation, cross-dynamic correlation matrix, free energy landscape, and MM-PBSA calculations demonstrated that two ligands from the selected ligands formed very resilient complexes with the enzyme acetylcholinesterase, with significant binding affinity. Therefore, these two compounds are recommended for further experimental research as possible (AChE) inhibitors. Full article
Show Figures

Figure 1

19 pages, 4367 KB  
Article
Targeted Affinity Purification and Mechanism of Action of Angiotensin-Converting Enzyme (ACE) Inhibitory Peptides from Sea Cucumber Gonads
by Yangduo Wang, Shicheng Chen, Wenzheng Shi, Shuji Liu, Xiaoting Chen, Nan Pan, Xiaoyan Wang, Yongchang Su and Zhiyu Liu
Mar. Drugs 2024, 22(2), 90; https://doi.org/10.3390/md22020090 - 16 Feb 2024
Cited by 10 | Viewed by 3932
Abstract
Protein hydrolysates from sea cucumber (Apostichopus japonicus) gonads are rich in active materials with remarkable angiotensin-converting enzyme (ACE) inhibitory activity. Alcalase was used to hydrolyze sea cucumber gonads, and the hydrolysate was separated by the ultrafiltration membrane to produce a low-molecular-weight [...] Read more.
Protein hydrolysates from sea cucumber (Apostichopus japonicus) gonads are rich in active materials with remarkable angiotensin-converting enzyme (ACE) inhibitory activity. Alcalase was used to hydrolyze sea cucumber gonads, and the hydrolysate was separated by the ultrafiltration membrane to produce a low-molecular-weight peptide component (less than 3 kDa) with good ACE inhibitory activity. The peptide component (less than 3 kDa) was isolated and purified using a combination method of ACE gel affinity chromatography and reverse high-performance liquid chromatography. The purified fractions were identified by liquid chromatography–tandem mass spectrometry (LC–MS/MS), and the resulting products were filtered using structure-based virtual screening (SBVS) to obtain 20 peptides. Of those, three noncompetitive inhibitory peptides (DDQIHIF with an IC50 value of 333.5 μmol·L−1, HDWWKER with an IC50 value of 583.6 μmol·L−1, and THDWWKER with an IC50 value of 1291.8 μmol·L−1) were further investigated based on their favorable pharmacochemical properties and ACE inhibitory activity. Molecular docking studies indicated that the three peptides were entirely enclosed within the ACE protein cavity, improving the overall stability of the complex through interaction forces with the ACE active site. The total free binding energies (ΔGtotal) for DDQIHIF, HDWWKER, and THDWWKER were −21.9 Kcal·mol−1, −71.6 Kcal·mol−1, and −69.1 Kcal·mol−1, respectively. Furthermore, a short-term assay of antihypertensive activity in spontaneously hypertensive rats (SHRs) revealed that HDWWKER could significantly decrease the systolic blood pressure (SBP) of SHRs after intravenous administration. The results showed that based on the better antihypertensive activity of the peptide in SHRs, the feasibility of targeted affinity purification and computer-aided drug discovery (CADD) for the efficient screening and preparation of ACE inhibitory peptide was verified, which provided a new idea of modern drug development method for clinical use. Full article
Show Figures

Graphical abstract

21 pages, 8858 KB  
Article
LSD1-Based Reversible Inhibitors Virtual Screening and Binding Mechanism Computational Study
by Zhili Yin, Shaohui Liu, Xiaoyue Yang, Mengguo Chen, Jiangfeng Du, Hongmin Liu and Longhua Yang
Molecules 2023, 28(14), 5315; https://doi.org/10.3390/molecules28145315 - 10 Jul 2023
Cited by 8 | Viewed by 3536
Abstract
As one of the crucial targets of epigenetics, histone lysine-specific demethylase 1 (LSD1) is significant in the occurrence and development of various tumors. Although several irreversible covalent LSD1 inhibitors have entered clinical trials, the large size and polarity of the FAD-binding pocket and [...] Read more.
As one of the crucial targets of epigenetics, histone lysine-specific demethylase 1 (LSD1) is significant in the occurrence and development of various tumors. Although several irreversible covalent LSD1 inhibitors have entered clinical trials, the large size and polarity of the FAD-binding pocket and undesired toxicity have focused interest on developing reversible LSD1 inhibitors. In this study, targeting the substrate-binding pocket of LSD1, structure-based and ligand-based virtual screenings were adopted to expand the potential novel structures with molecular docking and pharmacophore model strategies, respectively. Through drug-likeness evaluation, ADMET screening, molecular dynamics simulations, and binding free energy screening, we screened out one and four hit compounds from the databases of 2,029,554 compounds, respectively. Generally, these hit compounds can be divided into two categories, amide (Lig2 and Comp2) and 1,2,4-triazolo-4,3-α-quinazoline (Comp3, Comp4, Comp7). Among them, Comp4 exhibits the strongest binding affinity. Finally, the binding mechanisms of the hit compounds were further calculated in detail by the residue free energy decomposition. It was found that van der Waals interactions contribute most to the binding, and FAD is also helpful in stabilizing the binding and avoiding off-target effects. We believe this work not only provides a solid theoretical foundation for the design of LSD1 substrate reversible inhibitors, but also expands the diversity of parent nucleus, offering new insights for synthetic chemists. Full article
(This article belongs to the Special Issue Role of Computer Aided Drug Design in Drug Development)
Show Figures

Graphical abstract

20 pages, 2379 KB  
Article
Molecular Multi-Target Approach for Human Acetylcholinesterase, Butyrylcholinesterase and β-Secretase 1: Next Generation for Alzheimer’s Disease Treatment
by Géssica Oliveira Mendes, Samuel Silva da Rocha Pita, Paulo Batista de Carvalho, Michel Pires da Silva, Alex Gutterres Taranto and Franco Henrique Andrade Leite
Pharmaceuticals 2023, 16(6), 880; https://doi.org/10.3390/ph16060880 - 15 Jun 2023
Cited by 12 | Viewed by 3244
Abstract
Alzheimer’s Disease (AD) is a neurodegenerative condition characterized by progressive memory loss and other affected cognitive functions. Pharmacological therapy of AD relies on inhibitors of the enzymes acetylcholinesterase (AChE) and butyrylcholinesterase (BChE), offering only a palliative effect and being incapable of stopping or [...] Read more.
Alzheimer’s Disease (AD) is a neurodegenerative condition characterized by progressive memory loss and other affected cognitive functions. Pharmacological therapy of AD relies on inhibitors of the enzymes acetylcholinesterase (AChE) and butyrylcholinesterase (BChE), offering only a palliative effect and being incapable of stopping or reversing the neurodegenerative process. However, recent studies have shown that inhibiting the enzyme β-secretase 1 (BACE-1) may be able to stop neurodegeneration, making it a promising target. Considering these three enzymatic targets, it becomes feasible to apply computational techniques to guide the identification and planning of molecules capable of binding to all of them. After virtually screening 2119 molecules from a library, 13 hybrids were built and further screened by triple pharmacophoric model, molecular docking, and molecular dynamics (t = 200 ns). The selected hybrid G meets all stereo-electronic requirements to bind to AChE, BChE, and BACE-1 and offers a promising structure for future synthesis, enzymatic testing, and validation. Full article
(This article belongs to the Special Issue New Perspectives on Chemoinformatics and Drug Design)
Show Figures

Figure 1

20 pages, 9147 KB  
Article
Exploiting Blood Transport Proteins as Carborane Supramolecular Vehicles for Boron Neutron Capture Therapy
by Tainah Dorina Marforio, Edoardo Jun Mattioli, Francesco Zerbetto and Matteo Calvaresi
Nanomaterials 2023, 13(11), 1770; https://doi.org/10.3390/nano13111770 - 31 May 2023
Cited by 8 | Viewed by 3015
Abstract
Carboranes are promising agents for applications in boron neutron capture therapy (BNCT), but their hydrophobicity prevents their use in physiological environments. Here, by using reverse docking and molecular dynamics (MD) simulations, we identified blood transport proteins as candidate carriers of carboranes. Hemoglobin showed [...] Read more.
Carboranes are promising agents for applications in boron neutron capture therapy (BNCT), but their hydrophobicity prevents their use in physiological environments. Here, by using reverse docking and molecular dynamics (MD) simulations, we identified blood transport proteins as candidate carriers of carboranes. Hemoglobin showed a higher binding affinity for carboranes than transthyretin and human serum albumin (HSA), which are well-known carborane-binding proteins. Myoglobin, ceruloplasmin, sex hormone-binding protein, lactoferrin, plasma retinol-binding protein, thyroxine-binding globulin, corticosteroid-binding globulin and afamin have a binding affinity comparable to transthyretin/HSA. The carborane@protein complexes are stable in water and characterized by favorable binding energy. The driving force in the carborane binding is represented by the formation of hydrophobic interactions with aliphatic amino acids and BH-π and CH-π interactions with aromatic amino acids. Dihydrogen bonds, classical hydrogen bonds and surfactant-like interactions also assist the binding. These results (i) identify the plasma proteins responsible for binding carborane upon their intravenous administration, and (ii) suggest an innovative formulation for carboranes based on the formation of a carborane@protein complex prior to the administration. Full article
Show Figures

Figure 1

13 pages, 4512 KB  
Article
Screening of the Active Compounds against Neural Oxidative Damage from Ginseng Phloem Using UPLC-Q-Exactive-MS/MS Coupled with the Content-Effect Weighted Method
by Xiao-Chen Gao, Nan-Xi Zhang, Jia-Ming Shen, Jing-Wei Lv, Kai-Yue Zhang, Yao Sun, Hang Li, Yue-Long Wang, Duan-Duan Cheng, Meng-Ya Zhao, Hui Zhang, Chun-Nan Li and Jia-Ming Sun
Molecules 2022, 27(24), 9061; https://doi.org/10.3390/molecules27249061 - 19 Dec 2022
Cited by 6 | Viewed by 2397
Abstract
The neuroprotective properties of ginsenosides have been found to reverse the neurological damage caused by oxidation in many neurodegenerative diseases. However, the distribution of ginsenosides in different tissues of the main root, which was regarded as the primary medicinal portion in clinical practice [...] Read more.
The neuroprotective properties of ginsenosides have been found to reverse the neurological damage caused by oxidation in many neurodegenerative diseases. However, the distribution of ginsenosides in different tissues of the main root, which was regarded as the primary medicinal portion in clinical practice was different, the specific parts and specific components against neural oxidative damage were not clear. The present study aims to screen and determine the potential compounds in different parts of the main root in ginseng. Comparison of the protective effects in the main root, phloem and xylem of ginseng on hydrogen peroxide-induced cell death of SH-SY5Y neurons was investigated. UPLC-Q-Exactive-MS/MS was used to quickly and comprehensively characterize the chemical compositions of the active parts. Network pharmacology combined with a molecular docking approach was employed to virtually screen for disease-related targets and potential active compounds. By comparing the changes before and after Content-Effect weighting, the compounds with stronger anti-nerve oxidative damage activity were screened out more accurately. Finally, the activity of the selected monomer components was verified. The results suggested that the phloem of ginseng was the most effective part. There were 19 effective compounds and 14 core targets, and enriched signaling pathway and biological functions were predicted. After Content-Effect weighting, compounds Ginsenosides F1, Ginsenosides Rf, Ginsenosides Rg1 and Ginsenosides Rd were screened out as potential active compounds against neural oxidative damage. The activity verification study indicated that all four predicted ginsenosides were effective in protecting SH-SY5Y cells from oxidative injury. The four compounds can be further investigated as potential lead compounds for neurodegenerative diseases. This also provides a combined virtual and practical method for the simple and rapid screening of active ingredients in natural products. Full article
Show Figures

Figure 1

20 pages, 4469 KB  
Article
The Discovery of Small Allosteric and Active Site Inhibitors of the SARS-CoV-2 Main Protease via Structure-Based Virtual Screening and Biological Evaluation
by Radwa E. Mahgoub, Feda E. Mohamed, Lara Alzyoud, Bassam R. Ali, Juliana Ferreira, Wael M. Rabeh, Shaikha S. AlNeyadi, Noor Atatreh and Mohammad A. Ghattas
Molecules 2022, 27(19), 6710; https://doi.org/10.3390/molecules27196710 - 9 Oct 2022
Cited by 6 | Viewed by 3412
Abstract
The main protease enzyme (Mpro) of SARS-CoV-2 is one of the most promising targets for COVID-19 treatment. Accordingly, in this work, a structure-based virtual screening of 3.8 million ligand libraries was carried out. After rigorous filtering, docking, and post screening assessments, [...] Read more.
The main protease enzyme (Mpro) of SARS-CoV-2 is one of the most promising targets for COVID-19 treatment. Accordingly, in this work, a structure-based virtual screening of 3.8 million ligand libraries was carried out. After rigorous filtering, docking, and post screening assessments, 78 compounds were selected for biological evaluation, 3 of which showed promising inhibition of the Mpro enzyme. The obtained hits (CB03, GR04, and GR20) had reasonable potencies with Ki values in the medium to high micromolar range. Interestingly, while our most potent hit, GR20, was suggested to act via a reversible covalent mechanism, GR04 was confirmed as a noncompetitive inhibitor that seems to be one of a kind when compared to the other allosteric inhibitors discovered so far. Moreover, all three compounds have small sizes (~300 Da) with interesting fittings in their relevant binding sites, and they possess lead-like characteristics that can introduce them as very attractive candidates for the future development of COVID-19 treatments. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Figure 1

23 pages, 6630 KB  
Article
Design, Synthesis, Bioactivity Evaluation, Crystal Structures, and In Silico Studies of New α-Amino Amide Derivatives as Potential Histone Deacetylase 6 Inhibitors
by Yangrong Xu, Hangjun Tang, Yijie Xu, Jialin Guo, Xu Zhao, Qingguo Meng and Junhai Xiao
Molecules 2022, 27(10), 3335; https://doi.org/10.3390/molecules27103335 - 22 May 2022
Cited by 7 | Viewed by 4046
Abstract
Hydroxamate, as a zinc-binding group (ZBG), prevails in the design of histone deacetylase 6(HDAC6) inhibitors due to its remarkable zinc-chelating capability. However, hydroxamate-associated genotoxicity and mutagenicity have limited the widespread application of corresponding HDAC6 inhibitors in the treatment of human diseases. To avoid [...] Read more.
Hydroxamate, as a zinc-binding group (ZBG), prevails in the design of histone deacetylase 6(HDAC6) inhibitors due to its remarkable zinc-chelating capability. However, hydroxamate-associated genotoxicity and mutagenicity have limited the widespread application of corresponding HDAC6 inhibitors in the treatment of human diseases. To avoid such side effects, researchers are searching for novel ZBGs that may be used for the synthesis of HDAC6 inhibitors. In this study, a series of stereoisomeric compounds were designed and synthesized to discover non-hydroxamate HDAC6 inhibitors using α-amino amide as zinc-ion-chelating groups, along with a pair of enantiomeric isomers with inverted L-shaped vertical structure as cap structures. The anti-proliferative activities were determined against HL-60, Hela, and RPMI 8226 cells, and 7a and its stereoisomer 13a exhibited excellent activities against Hela cells with IC50 = 0.31 µM and IC50 = 5.19 µM, respectively. Interestingly, there is a significant difference between the two stereoisomers. Moreover, an evaluation of cytotoxicity toward human normal liver cells HL-7702 indicated its safety for normal cells. X-ray single crystal diffraction was employed to increase insights into molecule structure and activities. It was found that the carbonyl of the amide bond is on the different side from the amino and pyridine nitrogen atoms. To identify possible protein targets to clarify the mechanism of action and biological activity of 7a, a small-scale virtual screen using reverse docking for HDAC isoforms (1–10) was performed and the results showed that HDAC6 was the best receptor for 7a, suggesting that HDAC6 may be a potential target for 7a. The interaction pattern analysis showed that the α-amino amide moiety of 7a coordinated with the zinc ion of HDAC6 in a bidentate chelate manner, which is similar to the chelation pattern of hydroxamic acid. Finally, the molecular dynamics simulation approaches were used to assess the docked complex’s conformational stability. In this work, we identified 7a as a potential HDAC6 inhibitor and provide some references for the discovery of non-hydroxamic acid HDAC6 inhibitors. Full article
Show Figures

Figure 1

Back to TopTop