compound screening
Recently Published Documents


TOTAL DOCUMENTS

254
(FIVE YEARS 97)

H-INDEX

27
(FIVE YEARS 3)

2021 ◽  
Author(s):  
Francesca Murganti ◽  
Wouter Derks ◽  
Marion Baniol ◽  
Irina Simonova ◽  
Katrin Neumann ◽  
...  

One of the major goals in cardiac regeneration research is to replace lost ventricular tissue with new cardiomyocytes. However, cardiomyocyte proliferation drops to low levels in neonatal hearts and is no longer efficient in compensating for the loss of functional myocardium in heart disease. We generated a human induced pluripotent stem cell (iPSC)-derived cardiomyocyte-specific cell cycle indicator system (TNNT2-FUCCI) to characterize regular and aberrant cardiomyocyte cycle dynamics. We visualized cell cycle progression in TNNT2-FUCCI and found G2 cycle arrest in endoreplicating cardiomyocytes. Moreover, we devised a live-cell compound screening platform to identify pro-proliferative drug candidates. We found that the alpha-adrenergic receptor agonist clonidine induced cardiomyocyte proliferation in vitro and increased cardiomyocyte cell cycle entry in neonatal mice. In conclusion, the TNNT2-FUCCI system is a valuable tool to characterize cardiomyocyte cell cycle dynamics and identify pro-proliferative candidates with regenerative potential in the mammalian heart.


2021 ◽  
Author(s):  
Catherine Margaret Moore ◽  
Jigang Wang ◽  
Qingsong Lin ◽  
Pedro Eduardo Ferreira ◽  
Mitchell A Avery ◽  
...  

Treatment failures with artemisinin combination therapies (ACTs) threaten global efforts to eradicate malaria. They highlight the importance of identifying drug targets and new inhibitors and of studying how existing antimalarial classes work. Herein we report the successful development of an heterologous expression-based compound screening tool. Validated drug target P. falciparum calcium ATPase6 (PfATP6) and a mammalian ortholog (SERCA1a) were functionally expressed in yeast providing a robust, sensitive, and specific screening tool. Whole-cell and in vitro assays consistently demonstrated inhibition and labelling of PfATP6 by artemisinins. Mutations in PfATP6 resulted in fitness costs that were ameliorated in the presence of artemisinin derivatives when studied in the yeast model. As previously hypothesised, PfATP6 is a target of artemisinins. Mammalian SERCA1a can be mutated to become more susceptible to artemisinins. The inexpensive, low technology yeast screening platform has identified unrelated classes of druggable PfATP6 inhibitors. Resistance to artemisinins may depend on mechanisms that can concomitantly address multi-targeting by artemisinins and fitness costs of mutations that reduce artemisinin susceptibility.


2021 ◽  
Author(s):  
Michael Moret ◽  
Francesca Grisoni ◽  
Cyrill Brunner ◽  
Gisbert Schneider

Generative chemical language models (CLMs) can be used for de novo molecular structure generation. These CLMs learn from the structural information of known molecules to generate new ones. In this paper, we show that “hybrid” CLMs can additionally leverage the bioactivity information available for the training compounds. To computationally design ligands of phosphoinositide 3-kinase gamma (PI3Kγ), we created a large collection of virtual molecules with a generative CLM. This primary virtual compound library was further refined using a CLM-based classifier for bioactivity prediction. This second hybrid CLM was pretrained with patented molecular structures and fine-tuned with known PI3Kγ binders and non-binders by transfer learning. Several of the computer-generated molecular designs were commercially available, which allowed for fast prescreening and preliminary experimental validation. A new PI3Kγ ligand with sub-micromolar activity was identified. The results positively advocate hybrid CLMs for virtual compound screening and activity-focused molecular design in low-data situations.


2021 ◽  
Author(s):  
Jonathan Smith ◽  
Hao Xu ◽  
Xinran Li ◽  
Laurence Yang ◽  
Jahir M. Gutierrez

AbstractDeep learning provides a tool for improving screening of candidates for drug re-purposing to treat neglected diseases. We show how a new pipeline can be developed to address the needs of repurposing for Leishmaniasis. In combination with traditional molecular docking techniques, this allows top candidates to be selected and analyzed, including for molecular descriptor similarity.


2021 ◽  
Author(s):  
Alessio Mascolini ◽  
Dario Cardamone ◽  
Francesco Ponzio ◽  
Santa Di Cataldo ◽  
Elisa Ficarra

Abstract Computer-aided analysis of biological images typically requires extensive training on large-scale annotated datasets, which is not viable in many situations. In this paper, we present GAN-DL, a Discriminator Learner based on the StyleGAN2 architecture, which we employ for self-supervised image representation learning in the case of fluorescent biological images. We show that Wasserstein Generative Adversarial Networks combined with linear Support Vector Machines enable high-throughput compound screening based on raw images. We demonstrate this by classifying active and inactive compounds tested for the inhibition of SARS-CoV-2 infection in VERO and HRCE cell lines. In contrast to previous methods, our deep learning-based approach does not require any annotation besides the one that is normally collected during the sample preparation process. We test our technique on the RxRx19a Sars-CoV-2 image collection. The dataset consists of fluorescent images that were generated to assess the ability of regulatory-approved or late-stage clinical trials compounds to modulate the in vitro infection from SARS-CoV-2 in both VERO and HRCE cell lines. We show that our technique can be exploited not only for classification tasks but also to effectively derive a dose-response curve for the tested treatments, in a self-supervised manner. Lastly, we demonstrate its generalization capabilities by successfully addressing a zero-shot learning task, consisting of the categorization of four different cell types of the RxRx1 fluorescent images collection.


2021 ◽  
Author(s):  
Qiao Liu ◽  
Yue Qiu ◽  
Lei Xie

Chemical phenomics which measures multiplex chemical-induced phenotypic response of cells or patients, particularly dose-dependent transcriptomics and drug-response curves, provides new opportunities for in silico mechanism-driven phenotype-based drug discovery. However, state-of-the-art computational methods only focus on predicting a single phenotypic readout and are less successful in screening compounds for novel cells or individual patients. We designed a new deep learning model, MultiDCP, to enable high-throughput compound screening based on multiplex chemical phenomics for the first time, and further expand the scope of chemical phenomics to unexplored cells and patients. The novelties of MultiDCP lie in a multi-task learning framework with a novel knowledge-driven autoencoder to integrate incoherent labeled and unlabeled omics data, and a teacher-student training strategy to exploit unreliable data. MultiDCP significantly outperforms the state-of-the-art for novel cell lines. The predicted chemical transcriptomics demonstrate a stronger predictive power than noisy experimental data for downstream tasks. We applied MultiDCP to repurpose individualized drugs for Alzheimer's disease, suggesting that MultiDCP is a potentially powerful tool for personalized medicine.


2021 ◽  
pp. 247255522110360
Author(s):  
Eun Jeong Cho ◽  
Kevin N. Dalby

Luminescence is characterized by the spontaneous emission of light resulting from either chemical or biological reactions. Because of their high sensitivity, reduced background interference, and applicability to numerous situations, luminescence-based assay strategies play an essential role in early-stage drug discovery. Newer developments in luminescence-based technologies have dramatically affected the ability of researchers to investigate molecular binding events. At the forefront of these developments are the nano bioluminescence resonance energy transfer (NanoBRET) and amplified luminescent proximity homogeneous assay (Alpha) technologies. These technologies have opened up numerous possibilities for analyzing the molecular biophysical properties of complexes in environments such as cell lysates. Moreover, NanoBRET enables the validation and quantitation of the interactions between therapeutic targets and small molecules in live cells, representing an essential benchmark for preclinical drug discovery. Both techniques involve proximity-based luminescence energy transfer, in which excited-state energy is transferred from a donor to an acceptor, where the efficiency of transfer depends on proximity. Both approaches can be applied to high-throughput compound screening in biological samples, with the NanoBRET assay providing opportunities for live-cell screening. Representative applications of both technologies for assessing physical interactions and associated challenges are discussed.


2021 ◽  
pp. 247255522110262
Author(s):  
Jonathan Choy ◽  
Yanqing Kan ◽  
Steve Cifelli ◽  
Josephine Johnson ◽  
Michelle Chen ◽  
...  

High-throughput phenotypic screening is a key driver for the identification of novel chemical matter in drug discovery for challenging targets, especially for those with an unclear mechanism of pathology. For toxic or gain-of-function proteins, small-molecule suppressors are a targeting/therapeutic strategy that has been successfully applied. As with other high-throughput screens, the screening strategy and proper assays are critical for successfully identifying selective suppressors of the target of interest. We executed a small-molecule suppressor screen to identify compounds that specifically reduce apolipoprotein L1 (APOL1) protein levels, a genetically validated target associated with increased risk of chronic kidney disease. To enable this study, we developed homogeneous time-resolved fluorescence (HTRF) assays to measure intracellular APOL1 and apolipoprotein L2 (APOL2) protein levels and miniaturized them to 1536-well format. The APOL1 HTRF assay served as the primary assay, and the APOL2 and a commercially available p53 HTRF assay were applied as counterscreens. Cell viability was also measured with CellTiter-Glo to assess the cytotoxicity of compounds. From a 310,000-compound screening library, we identified 1490 confirmed primary hits with 12 different profiles. One hundred fifty-three hits selectively reduced APOL1 in 786-O, a renal cell adenocarcinoma cell line. Thirty-one of these selective suppressors also reduced APOL1 levels in conditionally immortalized human podocytes. The activity and specificity of seven resynthesized compounds were validated in both 786-O and podocytes.


Author(s):  
Ashrulochan Sahoo ◽  
Ghulam Mehdi Dar

The 21st century is witnessing immense achievements in human history, starting from home science to space science. Artificial Intelligence (AI) is a salient one among these feats, the critical factor of the 4th industrial revolution. Health is the primary and essential asset for the continuity of human civilization on this planet. Not only must we address the deadly existing diseases like Cancer, AIDS, Alzheimer's, heart diseases, gastrointestinal diseases, etc., but on top of that, we must effectively predict, prevent and respond to potential pathogens capable of causing havoc like the recent outbreak caused by SARS-CoV-2. AI-enabled technology with the computational capacity of a computer and reasoning ability of humans saves surplus labor and time that is majorly consumed in target validation, lead optimization, molecular representation, and designing reaction pathways, which traditionally is a decade-long way of searching, visualizing, studying, imagining, experimenting and maintaining a ton of data. This article would focus on how AI will help find the drug-like properties in the compound screening phase predicting the Structure-Activity Relationship (SAR) and ADMET properties in lead identification and optimization phases, sustainable development of chemicals in the synthesis phases up to AI's assistance in the successful conduct of clinical trials and repurposing.


2021 ◽  
Vol 1 ◽  
Author(s):  
Yang Liu ◽  
You Wu ◽  
Xiaoke Shen ◽  
Lei Xie

The life-threatening disease COVID-19 has inspired significant efforts to discover novel therapeutic agents through repurposing of existing drugs. Although multi-targeted (polypharmacological) therapies are recognized as the most efficient approach to system diseases such as COVID-19, computational multi-targeted compound screening has been limited by the scarcity of high-quality experimental data and difficulties in extracting information from molecules. This study introduces MolGNN, a new deep learning model for molecular property prediction. MolGNN applies a graph neural network to computational learning of chemical molecule embedding. Comparing to state-of-the-art approaches heavily relying on labeled experimental data, our method achieves equivalent or superior prediction performance without manual labels in the pretraining stage, and excellent performance on data with only a few labels. Our results indicate that MolGNN is robust to scarce training data, and hence a powerful few-shot learning tool. MolGNN predicted several multi-targeted molecules against both human Janus kinases and the SARS-CoV-2 main protease, which are preferential targets for drugs aiming, respectively, at alleviating cytokine storm COVID-19 symptoms and suppressing viral replication. We also predicted molecules potentially inhibiting cell death induced by SARS-CoV-2. Several of MolGNN top predictions are supported by existing experimental and clinical evidence, demonstrating the potential value of our method.


Sign in / Sign up

Export Citation Format

Share Document