Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.470
Filtrar
Más filtros











Intervalo de año de publicación
1.
Int J Mol Sci ; 25(9)2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38731835

RESUMEN

Combining new therapeutics with all-trans-retinoic acid (ATRA) could improve the efficiency of acute myeloid leukemia (AML) treatment. Modeling the process of ATRA-induced differentiation based on the transcriptomic profile of leukemic cells resulted in the identification of key targets that can be used to increase the therapeutic effect of ATRA. The genome-scale transcriptome analysis revealed the early molecular response to the ATRA treatment of HL-60 cells. In this study, we performed the transcriptomic profiling of HL-60, NB4, and K562 cells exposed to ATRA for 3-72 h. After treatment with ATRA for 3, 12, 24, and 72 h, we found 222, 391, 359, and 1032 differentially expressed genes (DEGs) in HL-60 cells, as well as 641, 1037, 1011, and 1499 DEGs in NB4 cells. We also found 538 and 119 DEGs in K562 cells treated with ATRA for 24 h and 72 h, respectively. Based on experimental transcriptomic data, we performed hierarchical modeling and determined cyclin-dependent kinase 6 (CDK6), tumor necrosis factor alpha (TNF-alpha), and transcriptional repressor CUX1 as the key regulators of the molecular response to the ATRA treatment in HL-60, NB4, and K562 cell lines, respectively. Mapping the data of TMT-based mass-spectrometric profiling on the modeling schemes, we determined CDK6 expression at the proteome level and its down-regulation at the transcriptome and proteome levels in cells treated with ATRA for 72 h. The combination of therapy with a CDK6 inhibitor (palbociclib) and ATRA (tretinoin) could be an alternative approach for the treatment of acute myeloid leukemia (AML).


Asunto(s)
Leucemia Mieloide Aguda , Biología de Sistemas , Tretinoina , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Tretinoina/farmacología , Biología de Sistemas/métodos , Células HL-60 , Perfilación de la Expresión Génica , Células K562 , Descubrimiento de Drogas/métodos , Transcriptoma , Línea Celular Tumoral , Quinasa 6 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/genética , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
2.
Int J Mol Sci ; 25(9)2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38731911

RESUMEN

In drug discovery, selecting targeted molecules is crucial as the target could directly affect drug efficacy and the treatment outcomes. As a member of the CCN family, CTGF (also known as CCN2) is an essential regulator in the progression of various diseases, including fibrosis, cancer, neurological disorders, and eye diseases. Understanding the regulatory mechanisms of CTGF in different diseases may contribute to the discovery of novel drug candidates. Summarizing the CTGF-targeting and -inhibitory drugs is also beneficial for the analysis of the efficacy, applications, and limitations of these drugs in different disease models. Therefore, we reviewed the CTGF structure, the regulatory mechanisms in various diseases, and drug development in order to provide more references for future drug discovery.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo , Descubrimiento de Drogas , Humanos , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Descubrimiento de Drogas/métodos , Animales , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Oftalmopatías/tratamiento farmacológico , Oftalmopatías/metabolismo , Fibrosis , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos
3.
PLoS One ; 19(5): e0302276, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38713692

RESUMEN

Based on topological descriptors, QSPR analysis is an incredibly helpful statistical method for examining many physical and chemical properties of compounds without demanding costly and time-consuming laboratory tests. Firstly, we discuss and provide research on kidney cancer drugs using topological indices and done partition of the edges of kidney cancer drugs which are based on the degree. Secondly, we examine the attributes of nineteen drugs casodex, eligard, mitoxanrone, rubraca, and zoladex, etc and among others, using linear QSPR model. The study in the article not only demonstrates a good correlation between TIs and physical characteristics with the QSPR model being the most suitable for predicting complexity, enthalpy, molar refractivity, and other factors and a best-fit model is attained in this study. This theoretical approach might benefit chemists and professionals in the pharmaceutical industry to forecast the characteristics of kidney cancer therapies. This leads towards new opportunities to paved the way for drug discovery and the formation of efficient and suitable treatment options in therapeutic targeting. We also employed multicriteria decision making techniques like COPRAS and PROMETHEE-II for ranking of said disease treatment drugs and physicochemical characteristics.


Asunto(s)
Antineoplásicos , Neoplasias Renales , Relación Estructura-Actividad Cuantitativa , Neoplasias Renales/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Antineoplásicos/química , Humanos , Toma de Decisiones , Descubrimiento de Drogas/métodos
4.
Nat Commun ; 15(1): 3636, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38710699

RESUMEN

Polypharmacology drugs-compounds that inhibit multiple proteins-have many applications but are difficult to design. To address this challenge we have developed POLYGON, an approach to polypharmacology based on generative reinforcement learning. POLYGON embeds chemical space and iteratively samples it to generate new molecular structures; these are rewarded by the predicted ability to inhibit each of two protein targets and by drug-likeness and ease-of-synthesis. In binding data for >100,000 compounds, POLYGON correctly recognizes polypharmacology interactions with 82.5% accuracy. We subsequently generate de-novo compounds targeting ten pairs of proteins with documented co-dependency. Docking analysis indicates that top structures bind their two targets with low free energies and similar 3D orientations to canonical single-protein inhibitors. We synthesize 32 compounds targeting MEK1 and mTOR, with most yielding >50% reduction in each protein activity and in cell viability when dosed at 1-10 µM. These results support the potential of generative modeling for polypharmacology.


Asunto(s)
Simulación del Acoplamiento Molecular , Humanos , Serina-Treonina Quinasas TOR/metabolismo , Polifarmacología , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 1/química , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/química , Unión Proteica , Descubrimiento de Drogas/métodos , Diseño de Fármacos , Supervivencia Celular/efectos de los fármacos
5.
J Vis Exp ; (206)2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38738886

RESUMEN

Monoclonal antibody-based immunotherapy targeting tumor antigens is now a mainstay of cancer treatment. One of the clinically relevant mechanisms of action of the antibodies is antibody-dependent cellular cytotoxicity (ADCC), where the antibody binds to the cancer cells and engages the cellular component of the immune system, e.g., natural killer (NK) cells, to kill the tumor cells. The effectiveness of these therapies could be improved by identifying adjuvant compounds that increase the sensitivity of the cancer cells or the potency of the immune cells. In addition, undiscovered drug interactions in cancer patients co-medicated for previous conditions or cancer-associated symptoms may determine the success of the antibody therapy; therefore, such unwanted drug interactions need to be eliminated. With these goals in mind, we created a cancer ADCC model and describe here a simple protocol to find ADCC-modulating drugs. Since 3D models such as cancer cell spheroids are superior to 2D cultures in predicting in vivo responses of tumors to anticancer therapies, spheroid co-cultures of EGFP-expressing HER2+ JIMT-1 breast cancer cells and the NK92.CD16 cell lines were set up and induced with Trastuzumab, a monoclonal antibody clinically approved against HER2-positive breast cancer. JIMT-1 spheroids were allowed to form in cell-repellent U-bottom 96-well plates. On day 3, NK cells and Trastuzumab were added. The spheroids were then stained with Annexin V-Alexa 647 to measure apoptotic cell death, which was quantitated in the peripheral zone of the spheroids with an automated microscope. The applicability of our assay to identify ADCC-modulating molecules is demonstrated by showing that Sunitinib, a receptor tyrosine kinase inhibitor approved by the FDA against metastatic cancer, almost completely abolishes ADCC. The generation of the spheroids and image acquisition and analysis pipelines are compatible with high-throughput screening for ADCC-modulating compounds in cancer cell spheroids.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos , Esferoides Celulares , Humanos , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/inmunología , Descubrimiento de Drogas/métodos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/efectos de los fármacos , Línea Celular Tumoral , Receptores de IgG/inmunología , Antineoplásicos Inmunológicos/farmacología , Trastuzumab/farmacología
6.
J Chem Inf Model ; 64(9): 3826-3840, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38696451

RESUMEN

Recent advances in computational methods provide the promise of dramatically accelerating drug discovery. While mathematical modeling and machine learning have become vital in predicting drug-target interactions and properties, there is untapped potential in computational drug discovery due to the vast and complex chemical space. This paper builds on our recently published computational fragment-based drug discovery (FBDD) method called fragment databases from screened ligand drug discovery (FDSL-DD). FDSL-DD uses in silico screening to identify ligands from a vast library, fragmenting them while attaching specific attributes based on predicted binding affinity and interaction with the target subdomain. In this paper, we further propose a two-stage optimization method that utilizes the information from prescreening to optimize computational ligand synthesis. We hypothesize that using prescreening information for optimization shrinks the search space and focuses on promising regions, thereby improving the optimization for candidate ligands. The first optimization stage assembles these fragments into larger compounds using genetic algorithms, followed by a second stage of iterative refinement to produce compounds with enhanced bioactivity. To demonstrate broad applicability, the methodology is demonstrated on three diverse protein targets found in human solid cancers, bacterial antimicrobial resistance, and the SARS-CoV-2 virus. Combined, the proposed FDSL-DD and a two-stage optimization approach yield high-affinity ligand candidates more efficiently than other state-of-the-art computational FBDD methods. We further show that a multiobjective optimization method accounting for drug-likeness can still produce potential candidate ligands with a high binding affinity. Overall, the results demonstrate that integrating detailed chemical information with a constrained search framework can markedly optimize the initial drug discovery process, offering a more precise and efficient route to developing new therapeutics.


Asunto(s)
Descubrimiento de Drogas , Ligandos , Descubrimiento de Drogas/métodos , Humanos , SARS-CoV-2/metabolismo , Algoritmos , Tratamiento Farmacológico de COVID-19 , COVID-19/virología
7.
PLoS Comput Biol ; 20(4): e1011945, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38578805

RESUMEN

Early identification of safe and efficacious disease targets is crucial to alleviating the tremendous cost of drug discovery projects. However, existing experimental methods for identifying new targets are generally labor-intensive and failure-prone. On the other hand, computational approaches, especially machine learning-based frameworks, have shown remarkable application potential in drug discovery. In this work, we propose Progeni, a novel machine learning-based framework for target identification. In addition to fully exploiting the known heterogeneous biological networks from various sources, Progeni integrates literature evidence about the relations between biological entities to construct a probabilistic knowledge graph. Graph neural networks are then employed in Progeni to learn the feature embeddings of biological entities to facilitate the identification of biologically relevant target candidates. A comprehensive evaluation of Progeni demonstrated its superior predictive power over the baseline methods on the target identification task. In addition, our extensive tests showed that Progeni exhibited high robustness to the negative effect of exposure bias, a common phenomenon in recommendation systems, and effectively identified new targets that can be strongly supported by the literature. Moreover, our wet lab experiments successfully validated the biological significance of the top target candidates predicted by Progeni for melanoma and colorectal cancer. All these results suggested that Progeni can identify biologically effective targets and thus provide a powerful and useful tool for advancing the drug discovery process.


Asunto(s)
Biología Computacional , Descubrimiento de Drogas , Aprendizaje Automático , Redes Neurales de la Computación , Humanos , Biología Computacional/métodos , Descubrimiento de Drogas/métodos , Algoritmos , Melanoma , Probabilidad , Neoplasias Colorrectales
8.
Methods Mol Biol ; 2806: 19-30, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38676793

RESUMEN

Patient-derived xenografts (PDXs), established by implanting patient tumor cells into immunodeficient mice, offer a platform for faithfully replicating human tumors. They closely mimic the histopathology, genomics, and drug sensitivity of patient tumors. This chapter highlights the versatile applications of PDXs, including studying tumor biology, metastasis, and chemoresistance, as well as their use in biomarker identification, drug screening, and personalized medicine. It also addresses challenges in using PDXs in cancer research, including variations in metastatic potential, lengthy establishment timelines, stromal changes, and limitations in immunocompromised models. Despite these challenges, PDXs remain invaluable tools guiding patient treatment and advancing preclinical drug development.


Asunto(s)
Biomarcadores de Tumor , Medicina de Precisión , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Humanos , Ratones , Biomarcadores de Tumor/metabolismo , Medicina de Precisión/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Neoplasias/metabolismo , Desarrollo de Medicamentos/métodos , Descubrimiento de Drogas/métodos , Modelos Animales de Enfermedad , Antineoplásicos/farmacología
9.
Expert Opin Drug Discov ; 19(5): 617-629, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38595031

RESUMEN

INTRODUCTION: ω-3 Polyunsaturated fatty acids (PUFAs) have a range of health benefits, including anticancer activity, and are converted to lipid mediators that could be adapted into pharmacological strategies. However, the stability of these mediators must be improved, and they may require formulation to achieve optimal tissue concentrations. AREAS COVERED: Herein, the author reviews the literature around chemical stabilization and formulation of ω-3 PUFA mediators and their application in anticancer drug discovery. EXPERT OPINION: Aryl-urea bioisosteres of ω-3 PUFA epoxides that killed cancer cells targeted the mitochondrion by a novel dual mechanism: as protonophoric uncouplers and as inhibitors of electron transport complex III that activated ER-stress and disrupted mitochondrial integrity. In contrast, aryl-ureas that contain electron-donating substituents prevented cancer cell migration. Thus, aryl-ureas represent a novel class of agents with tunable anticancer properties. Stabilized analogues of other ω-3 PUFA-derived mediators could also be adapted into anticancer strategies. Indeed, a cocktail of agents that simultaneously promote cell killing, inhibit metastasis and angiogenesis, and that attenuate the pro-inflammatory microenvironment is a novel future anticancer strategy. Such regimen may enhance anticancer drug efficacy, minimize the development of anticancer drug resistance and enhance outcomes.


Asunto(s)
Antineoplásicos , Descubrimiento de Drogas , Ácidos Grasos Omega-3 , Neoplasias , Humanos , Ácidos Grasos Omega-3/farmacología , Antineoplásicos/farmacología , Descubrimiento de Drogas/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Animales , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo
10.
J Bioinform Comput Biol ; 22(1): 2450003, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38567386

RESUMEN

In this paper, we propose a novel approach for predicting the activity/inactivity of molecules with the BRCA1 gene by combining pharmacophore modeling and deep learning techniques. Initially, we generated 3D pharmacophore fingerprints using a pharmacophore model, which captures the essential features and spatial arrangements critical for biological activity. These fingerprints served as informative representations of the molecular structures. Next, we employed deep learning algorithms to train a predictive model using the generated pharmacophore fingerprints. The deep learning model was designed to learn complex patterns and relationships between the pharmacophore features and the corresponding activity/inactivity labels of the molecules. By utilizing this integrated approach, we aimed to enhance the accuracy and efficiency of activity prediction. To validate the effectiveness of our approach, we conducted experiments using a dataset of known molecules with BRCA1 gene activity/inactivity from diverse sources. Our results demonstrated promising predictive performance, indicating the successful integration of pharmacophore modeling and deep learning. Furthermore, we utilized the trained model to predict the activity/inactivity of unknown molecules extracted from the ChEMBL database. The predictions obtained from the ChEMBL database were assessed and compared against experimentally determined values to evaluate the reliability and generalizability of our model. Overall, our proposed approach showcased significant potential in accurately predicting the activity/inactivity of molecules with the BRCA1 gene, thus enabling the identification of potential candidates for further investigation in drug discovery and development processes.


Asunto(s)
Aprendizaje Profundo , Farmacóforo , Genes BRCA1 , Reproducibilidad de los Resultados , Descubrimiento de Drogas/métodos
11.
Life Sci ; 347: 122642, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38641047

RESUMEN

Drug repurposing involves the investigation of existing drugs for new indications. It offers a great opportunity to quickly identify a new drug candidate at a lower cost than novel discovery and development. Despite the importance and potential role of drug repurposing, there is no specific definition that healthcare providers and the World Health Organization credit. Unfortunately, many similar and interchangeable concepts are being used in the literature, making it difficult to collect and analyze uniform data on repurposed drugs. This research was conducted based on understanding general criteria for drug repurposing, concentrating on liver diseases. Many drugs have been investigated for their effect on liver diseases even though they were originally approved (or on their way to being approved) for other diseases. Some of the hypotheses for drug repurposing were first captured from the literature and then processed further to test the hypothesis. Recently, with the revolution in bioinformatics techniques, scientists have started to use drug libraries and computer systems that can analyze hundreds of drugs to give a short list of candidates to be analyzed pharmacologically. However, this study revealed that drug repurposing is a potential aid that may help deal with liver diseases. It provides available or under-investigated drugs that could help treat hepatitis, liver cirrhosis, Wilson disease, liver cancer, and fatty liver. However, many further studies are needed to ensure the efficacy of these drugs on a large scale.


Asunto(s)
Reposicionamiento de Medicamentos , Hepatopatías , Reposicionamiento de Medicamentos/métodos , Humanos , Hepatopatías/tratamiento farmacológico , Biología Computacional/métodos , Descubrimiento de Drogas/métodos
12.
Drug Discov Today ; 29(5): 103950, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38514040

RESUMEN

Drugs targeting the µ-opioid receptor (MOR) remain the most efficacious analgesics for the treatment of pain, but activation of MOR with current opioid analgesics also produces harmful side effects, notably physical dependence, addiction, and respiratory depression. Opioid peptides have been accepted as promising candidates for the development of safer and more efficacious analgesics. To develop peptide-based opioid analgesics, strategies such as modification of endogenous opioid peptides, development of multifunctional opioid peptides, G protein-biased opioid peptides, and peripherally restricted opioid peptides have been reported. This review seeks to provide an overview of the opioid peptides that produce potent antinociception with much reduced side effects in animal models and highlight the potential advantages of peptides as safer opioid analgesics.


Asunto(s)
Analgésicos Opioides , Descubrimiento de Drogas , Péptidos Opioides , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/farmacología , Animales , Humanos , Ligandos , Descubrimiento de Drogas/métodos , Dolor/tratamiento farmacológico , Receptores Opioides mu/metabolismo , Péptidos/farmacología , Péptidos/uso terapéutico
13.
Int J Mol Sci ; 25(5)2024 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-38473785

RESUMEN

Deep learning is a machine learning technique to model high-level abstractions in data by utilizing a graph composed of multiple processing layers that experience various linear and non-linear transformations. This technique has been shown to perform well for applications in drug discovery, utilizing structural features of small molecules to predict activity. Here, we report a large-scale study to predict the activity of small molecules across the human kinome-a major family of drug targets, particularly in anti-cancer agents. While small-molecule kinase inhibitors exhibit impressive clinical efficacy in several different diseases, resistance often arises through adaptive kinome reprogramming or subpopulation diversity. Polypharmacology and combination therapies offer potential therapeutic strategies for patients with resistant diseases. Their development would benefit from a more comprehensive and dense knowledge of small-molecule inhibition across the human kinome. Leveraging over 650,000 bioactivity annotations for more than 300,000 small molecules, we evaluated multiple machine learning methods to predict the small-molecule inhibition of 342 kinases across the human kinome. Our results demonstrated that multi-task deep neural networks outperformed classical single-task methods, offering the potential for conducting large-scale virtual screening, predicting activity profiles, and bridging the gaps in the available data.


Asunto(s)
Aprendizaje Profundo , Humanos , Fosfotransferasas , Descubrimiento de Drogas/métodos , Polifarmacología , Aprendizaje Automático
14.
Mol Pharm ; 21(4): 1563-1590, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38466810

RESUMEN

Understanding protein sequence and structure is essential for understanding protein-protein interactions (PPIs), which are essential for many biological processes and diseases. Targeting protein binding hot spots, which regulate signaling and growth, with rational drug design is promising. Rational drug design uses structural data and computational tools to study protein binding sites and protein interfaces to design inhibitors that can change these interactions, thereby potentially leading to therapeutic approaches. Artificial intelligence (AI), such as machine learning (ML) and deep learning (DL), has advanced drug discovery and design by providing computational resources and methods. Quantum chemistry is essential for drug reactivity, toxicology, drug screening, and quantitative structure-activity relationship (QSAR) properties. This review discusses the methodologies and challenges of identifying and characterizing hot spots and binding sites. It also explores the strategies and applications of artificial-intelligence-based rational drug design technologies that target proteins and protein-protein interaction (PPI) binding hot spots. It provides valuable insights for drug design with therapeutic implications. We have also demonstrated the pathological conditions of heat shock protein 27 (HSP27) and matrix metallopoproteinases (MMP2 and MMP9) and designed inhibitors of these proteins using the drug discovery paradigm in a case study on the discovery of drug molecules for cancer treatment. Additionally, the implications of benzothiazole derivatives for anticancer drug design and discovery are deliberated.


Asunto(s)
Inteligencia Artificial , Descubrimiento de Drogas , Descubrimiento de Drogas/métodos , Diseño de Fármacos , Aprendizaje Automático , Relación Estructura-Actividad Cuantitativa
15.
J Biol Chem ; 300(4): 107133, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38432632

RESUMEN

Protein mechanical stability determines the function of a myriad of proteins, especially proteins from the extracellular matrix. Failure to maintain protein mechanical stability may result in diseases and disorders such as cancer, cardiomyopathies, or muscular dystrophy. Thus, developing mutation-free approaches to enhance and control the mechanical stability of proteins using pharmacology-based methods may have important implications in drug development and discovery. Here, we present the first approach that employs computational high-throughput virtual screening and molecular docking to search for small molecules in chemical libraries that function as mechano-regulators of the stability of human cluster of differentiation 4, receptor of HIV-1. Using single-molecule force spectroscopy, we prove that these small molecules can increase the mechanical stability of CD4D1D2 domains over 4-fold in addition to modifying the mechanical unfolding pathways. Our experiments demonstrate that chemical libraries are a source of mechanoactive molecules and that drug discovery approaches provide the foundation of a new type of molecular function, that is, mechano-regulation, paving the way toward mechanopharmacology.


Asunto(s)
Antígenos CD4 , Descubrimiento de Drogas , Bibliotecas de Moléculas Pequeñas , Humanos , Antígenos CD4/metabolismo , Antígenos CD4/química , Descubrimiento de Drogas/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , VIH-1/metabolismo , VIH-1/química , Simulación del Acoplamiento Molecular , Estabilidad Proteica , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología
16.
Bioorg Chem ; 145: 107172, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38340475

RESUMEN

The exploration of hybridization emerges as a potent tool in advancing drug discovery research, with a significant emphasis on carbohydrate-containing hybrid scaffolds. Evidence indicates that linking carbohydrate molecules to privileged bioactive scaffolds enhances the bioactivity of drug molecules. This synergy results in a diverse range of activities, making carbohydrate scaffolds pivotal for synthesizing compound libraries with significant functional and structural diversity. Beyond their synthesis utility, these scaffolds offer applications in screening bioactive molecules, presenting alternative avenues for drug development. This comprehensive review spanning 2015 to 2023 focuses on synthesized glycohybrid molecules, revealing their bioactivity in areas such as anti-microbial, anti-cancer, anti-diabetic, anti-inflammatory activities, enzyme inhibition and pesticides. Numerous novel glycohybrids surpass positive control drugs in biological activity. This focused study not only highlights the diverse bioactivities of glycohybrids but also underscores their promising role in innovative drug development strategies.


Asunto(s)
Carbohidratos , Descubrimiento de Drogas , Descubrimiento de Drogas/métodos
17.
Adv Sci (Weinh) ; 11(11): e2307245, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38204214

RESUMEN

One of the main challenges in small molecule drug discovery is finding novel chemical compounds with desirable activity. Traditional drug development typically begins with target selection, but the correlation between targets and disease remains to be further investigated, and drugs designed based on targets may not always have the desired drug efficacy. The emergence of machine learning provides a powerful tool to overcome the challenge. Herein, a machine learning-based strategy is developed for de novo generation of novel compounds with drug efficacy termed DTLS (Deep Transfer Learning-based Strategy) by using dataset of disease-direct-related activity as input. DTLS is applied in two kinds of disease: colorectal cancer (CRC) and Alzheimer's disease (AD). In each case, novel compound is discovered and identified in in vitro and in vivo disease models. Their mechanism of actionis further explored. The experimental results reveal that DTLS can not only realize the generation and identification of novel compounds with drug efficacy but also has the advantage of identifying compounds by focusing on protein targets to facilitate the mechanism study. This work highlights the significant impact of machine learning on the design of novel compounds with drug efficacy, which provides a powerful new approach to drug discovery.


Asunto(s)
Descubrimiento de Drogas , Aprendizaje Automático , Descubrimiento de Drogas/métodos , Proteínas
18.
J Chem Inf Model ; 64(5): 1433-1455, 2024 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-38294194

RESUMEN

Solute carrier transporters (SLCs) are a class of important transmembrane proteins that are involved in the transportation of diverse solute ions and small molecules into cells. There are approximately 450 SLCs within the human body, and more than a quarter of them are emerging as attractive therapeutic targets for multiple complex diseases, e.g., depression, cancer, and diabetes. However, only 44 unique transporters (∼9.8% of the SLC superfamily) with 3D structures and specific binding sites have been reported. To design innovative and effective drugs targeting diverse SLCs, there are a number of obstacles that need to be overcome. However, computational chemistry, including physics-based molecular modeling and machine learning- and deep learning-based artificial intelligence (AI), provides an alternative and complementary way to the classical drug discovery approach. Here, we present a comprehensive overview on recent advances and existing challenges of the computational techniques in structure-based drug design of SLCs from three main aspects: (i) characterizing multiple conformations of the proteins during the functional process of transportation, (ii) identifying druggability sites especially the cryptic allosteric ones on the transporters for substrates and drugs binding, and (iii) discovering diverse small molecules or synthetic protein binders targeting the binding sites. This work is expected to provide guidelines for a deep understanding of the structure and function of the SLC superfamily to facilitate rational design of novel modulators of the transporters with the aid of state-of-the-art computational chemistry technologies including artificial intelligence.


Asunto(s)
Inteligencia Artificial , Química Computacional , Humanos , Proteínas de Transporte de Membrana/química , Diseño de Fármacos , Descubrimiento de Drogas/métodos
19.
J Chem Inf Model ; 64(7): 2695-2704, 2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38293736

RESUMEN

Predicting compound activity in assays is a long-standing challenge in drug discovery. Computational models based on compound-induced gene expression signatures from a single profiling assay have shown promise toward predicting compound activity in other, seemingly unrelated, assays. Applications of such models include predicting mechanisms-of-action (MoA) for phenotypic hits, identifying off-target activities, and identifying polypharmacologies. Here, we introduce transcriptomics-to-activity transformer (TAT) models that leverage gene expression profiles observed over compound treatment at multiple concentrations to predict the compound activity in other biochemical or cellular assays. We built TAT models based on gene expression data from a RASL-seq assay to predict the activity of 2692 compounds in 262 dose-response assays. We obtained useful models for 51% of the assays, as determined through a realistic held-out set. Prospectively, we experimentally validated the activity predictions of a TAT model in a malaria inhibition assay. With a 63% hit rate, TAT successfully identified several submicromolar malaria inhibitors. Our results thus demonstrate the potential of transcriptomic responses over compound concentration and the TAT modeling framework as a cost-efficient way to identify the bioactivities of promising compounds across many assays.


Asunto(s)
Aprendizaje Profundo , Malaria , Humanos , Transcriptoma , Descubrimiento de Drogas/métodos , Perfilación de la Expresión Génica
20.
Curr Opin Struct Biol ; 84: 102771, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38215530

RESUMEN

In drug discovery, targeted polypharmacology, i.e., targeting multiple molecular targets with a single drug, is redefining therapeutic design to address complex diseases. Pre-selected pharmacological profiles, as exemplified in kinase drugs, promise enhanced efficacy and reduced toxicity. Historically, many of such drugs were discovered serendipitously, limiting predictability and efficacy, but currently artificial intelligence (AI) offers a transformative solution. Machine learning and deep learning techniques enable modeling protein structures, generating novel compounds, and decoding their polypharmacological effects, opening an avenue for more systematic and predictive multi-target drug design. This review explores the use of AI in identifying synergistic co-targets and delineating them from anti-targets that lead to adverse effects, and then discusses advances in AI-enabled docking, generative chemistry, and proteochemometric modeling of proteome-wide compound interactions, in the context of polypharmacology. We also provide insights into challenges ahead.


Asunto(s)
Inteligencia Artificial , Polifarmacología , Descubrimiento de Drogas/métodos , Diseño de Fármacos , Aprendizaje Automático
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA