Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Comput Biol Med ; 171: 108211, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38422960

ABSTRACT

Single-cell RNA sequencing (scRNA-seq) has emerged as a transformative technology, offering unparalleled insights into the intricate landscape of cellular diversity and gene expression dynamics. scRNA-seq analysis represents a challenging and cutting-edge frontier within the field of biological research. Differential geometry serves as a powerful mathematical tool in various applications of scientific research. In this study, we introduce, for the first time, a multiscale differential geometry (MDG) strategy for addressing the challenges encountered in scRNA-seq data analysis. We assume that intrinsic properties of cells lie on a family of low-dimensional manifolds embedded in the high-dimensional space of scRNA-seq data. Multiscale cell-cell interactive manifolds are constructed to reveal complex relationships in the cell-cell network, where curvature-based features for cells can decipher the intricate structural and biological information. We showcase the utility of our novel approach by demonstrating its effectiveness in classifying cell types. This innovative application of differential geometry in scRNA-seq analysis opens new avenues for understanding the intricacies of biological networks and holds great potential for network analysis in other fields.


Subject(s)
Data Analysis , Gene Expression Profiling , Sequence Analysis, RNA/methods , Gene Expression Profiling/methods , Cluster Analysis
2.
Pain ; 165(4): 908-921, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-37851391

ABSTRACT

ABSTRACT: Pain is a significant global health issue, and the current treatment options for pain management have limitations in terms of effectiveness, side effects, and potential for addiction. There is a pressing need for improved pain treatments and the development of new drugs. Voltage-gated sodium channels, particularly Nav1.3, Nav1.7, Nav1.8, and Nav1.9, play a crucial role in neuronal excitability and are predominantly expressed in the peripheral nervous system. Targeting these channels may provide a means to treat pain while minimizing central and cardiac adverse effects. In this study, we construct protein-protein interaction (PPI) networks based on pain-related sodium channels and develop a corresponding drug-target interaction network to identify potential lead compounds for pain management. To ensure reliable machine learning predictions, we carefully select 111 inhibitor data sets from a pool of more than 1000 targets in the PPI network. We employ 3 distinct machine learning algorithms combined with advanced natural language processing (NLP)-based embeddings, specifically pretrained transformer and autoencoder representations. Through a systematic screening process, we evaluate the side effects and repurposing potential of more than 150,000 drug candidates targeting Nav1.7 and Nav1.8 sodium channels. In addition, we assess the ADMET (absorption, distribution, metabolism, excretion, and toxicity) properties of these candidates to identify leads with near-optimal characteristics. Our strategy provides an innovative platform for the pharmacological development of pain treatments, offering the potential for improved efficacy and reduced side effects.


Subject(s)
Voltage-Gated Sodium Channels , Humans , Voltage-Gated Sodium Channels/metabolism , Pain/drug therapy , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.7 Voltage-Gated Sodium Channel/metabolism
3.
J Chem Inf Model ; 63(22): 7189-7209, 2023 Nov 27.
Article in English | MEDLINE | ID: mdl-37956228

ABSTRACT

The birth of ChatGPT, a cutting-edge language model-based chatbot developed by OpenAI, ushered in a new era in AI. However, due to potential pitfalls, its role in rigorous scientific research is not clear yet. This paper vividly showcases its innovative application within the field of drug discovery. Focused specifically on developing anticocaine addiction drugs, the study employs GPT-4 as a virtual guide, offering strategic and methodological insights to researchers working on generative models for drug candidates. The primary objective is to generate optimal drug-like molecules with desired properties. By leveraging the capabilities of ChatGPT, the study introduces a novel approach to the drug discovery process. This symbiotic partnership between AI and researchers transforms how drug development is approached. Chatbots become facilitators, steering researchers toward innovative methodologies and productive paths for creating effective drug candidates. This research sheds light on the collaborative synergy between human expertise and AI assistance, wherein ChatGPT's cognitive abilities enhance the design and development of pharmaceutical solutions. This paper not only explores the integration of advanced AI in drug discovery but also reimagines the landscape by advocating for AI-powered chatbots as trailblazers in revolutionizing therapeutic innovation.


Subject(s)
Drug Development , Substance-Related Disorders , Humans , Drug Discovery , Language , Research Personnel
4.
J Med Chem ; 66(17): 12479-12498, 2023 09 14.
Article in English | MEDLINE | ID: mdl-37623046

ABSTRACT

Opioid use disorder (OUD) has emerged as a significant global public health issue, necessitating the discovery of new medications. In this study, we propose a deep generative model that combines a stochastic differential equation (SDE)-based diffusion model with a pretrained autoencoder. The molecular generator enables efficient generation of molecules that target multiple opioid receptors, including mu, kappa, and delta. Additionally, we assess the ADMET (absorption, distribution, metabolism, excretion, and toxicity) properties of the generated molecules to identify druglike compounds. We develop a molecular optimization approach to enhance the pharmacokinetic properties of some lead compounds. Advanced binding affinity predictors were built using molecular fingerprints, including autoencoder embeddings, transformer embeddings, and topological Laplacians. Our process yields druglike molecules that can be used in highly focused experimental studies to further evaluate their pharmacological effects. Our machine learning platform serves as a valuable tool for designing effective molecules to address OUD.


Subject(s)
Machine Learning , Opioid-Related Disorders , Humans , Diffusion , Opioid-Related Disorders/drug therapy , Receptors, Opioid
5.
ArXiv ; 2023 Oct 19.
Article in English | MEDLINE | ID: mdl-37645039

ABSTRACT

The birth of ChatGPT, a cutting-edge language model-based chatbot developed by OpenAI, ushered in a new era in AI. However, due to potential pitfalls, its role in rigorous scientific research is not clear yet. This paper vividly showcases its innovative application within the field of drug discovery. Focused specifically on developing anti-cocaine addiction drugs, the study employs GPT-4 as a virtual guide, offering strategic and methodological insights to researchers working on generative models for drug candidates. The primary objective is to generate optimal drug-like molecules with desired properties. By leveraging the capabilities of ChatGPT, the study introduces a novel approach to the drug discovery process. This symbiotic partnership between AI and researchers transforms how drug development is approached. Chatbots become facilitators, steering researchers towards innovative methodologies and productive paths for creating effective drug candidates. This research sheds light on the collaborative synergy between human expertise and AI assistance, wherein ChatGPT's cognitive abilities enhance the design and development of potential pharmaceutical solutions. This paper not only explores the integration of advanced AI in drug discovery but also reimagines the landscape by advocating for AI-powered chatbots as trailblazers in revolutionizing therapeutic innovation.

6.
ArXiv ; 2023 Jun 13.
Article in English | MEDLINE | ID: mdl-37396606

ABSTRACT

Opioid Use Disorder (OUD) has emerged as a significant global public health issue, with complex multifaceted conditions. Due to the lack of effective treatment options for various conditions, there is a pressing need for the discovery of new medications. In this study, we propose a deep generative model that combines a stochastic differential equation (SDE)-based diffusion modeling with the latent space of a pretrained autoencoder model. The molecular generator enables efficient generation of molecules that are effective on multiple targets, specifically the mu, kappa, and delta opioid receptors. Furthermore, we assess the ADMET (absorption, distribution, metabolism, excretion, and toxicity) properties of the generated molecules to identify drug-like compounds. To enhance the pharmacokinetic properties of some lead compounds, we employ a molecular optimization approach. We obtain a diverse set of drug-like molecules. We construct binding affinity predictors by integrating molecular fingerprints derived from autoencoder embeddings, transformer embeddings, and topological Laplacians with advanced machine learning algorithms. Further experimental studies are needed to evaluate the pharmacological effects of these drug-like compounds for OUD treatment. Our machine learning platform serves as a valuable tool in designing and optimizing effective molecules for addressing OUD.

7.
ArXiv ; 2023 Jul 11.
Article in English | MEDLINE | ID: mdl-37502628

ABSTRACT

Pain is a significant global health issue, and the current treatment options for pain management have limitations in terms of effectiveness, side effects, and potential for addiction. There is a pressing need for improved pain treatments and the development of new drugs. Voltage-gated sodium channels, particularly Nav1.3, Nav1.7, Nav1.8, and Nav1.9, play a crucial role in neuronal excitability and are predominantly expressed in the peripheral nervous system. Targeting these channels may provide a means to treat pain while minimizing central and cardiac adverse effects. In this study, we construct protein-protein interaction (PPI) networks based on pain-related sodium channels and develop a corresponding drug-target interaction (DTI) network to identify potential lead compounds for pain management. To ensure reliable machine learning predictions, we carefully select 111 inhibitor datasets from a pool of over 1,000 targets in the PPI network. We employ three distinct machine learning algorithms combined with advanced natural language processing (NLP)-based embeddings, specifically pre-trained transformer and autoencoder representations. Through a systematic screening process, we evaluate the side effects and repurposing potential of over 150,000 drug candidates targeting Nav1.7 and Nav1.8 sodium channels. Additionally, we assess the ADMET (absorption, distribution, metabolism, excretion, and toxicity) properties of these candidates to identify leads with near-optimal characteristics. Our strategy provides an innovative platform for the pharmacological development of pain treatments, offering the potential for improved efficacy and reduced side effects.

8.
Commun Biol ; 6(1): 536, 2023 05 18.
Article in English | MEDLINE | ID: mdl-37202415

ABSTRACT

Virtual screening (VS) is a critical technique in understanding biomolecular interactions, particularly in drug design and discovery. However, the accuracy of current VS models heavily relies on three-dimensional (3D) structures obtained through molecular docking, which is often unreliable due to the low accuracy. To address this issue, we introduce a sequence-based virtual screening (SVS) as another generation of VS models that utilize advanced natural language processing (NLP) algorithms and optimized deep K-embedding strategies to encode biomolecular interactions without relying on 3D structure-based docking. We demonstrate that SVS outperforms state-of-the-art performance for four regression datasets involving protein-ligand binding, protein-protein, protein-nucleic acid binding, and ligand inhibition of protein-protein interactions and five classification datasets for protein-protein interactions in five biological species. SVS has the potential to transform current practices in drug discovery and protein engineering.


Subject(s)
Algorithms , Proteins , Molecular Docking Simulation , Ligands , Proteins/metabolism , Drug Discovery/methods
9.
Comput Biol Med ; 160: 106921, 2023 06.
Article in English | MEDLINE | ID: mdl-37178605

ABSTRACT

Opioid use disorder (OUD) is a chronic and relapsing condition that involves the continued and compulsive use of opioids despite harmful consequences. The development of medications with improved efficacy and safety profiles for OUD treatment is urgently needed. Drug repurposing is a promising option for drug discovery due to its reduced cost and expedited approval procedures. Computational approaches based on machine learning enable the rapid screening of DrugBank compounds, identifying those with the potential to be repurposed for OUD treatment. We collected inhibitor data for four major opioid receptors and used advanced machine learning predictors of binding affinity that fuse the gradient boosting decision tree algorithm with two natural language processing (NLP)-based molecular fingerprints and one traditional 2D fingerprint. Using these predictors, we systematically analyzed the binding affinities of DrugBank compounds on four opioid receptors. Based on our machine learning predictions, we were able to discriminate DrugBank compounds with various binding affinity thresholds and selectivities for different receptors. The prediction results were further analyzed for ADMET (absorption, distribution, metabolism, excretion, and toxicity), which provided guidance on repurposing DrugBank compounds for the inhibition of selected opioid receptors. The pharmacological effects of these compounds for OUD treatment need to be tested in further experimental studies and clinical trials. Our machine learning studies provide a valuable platform for drug discovery in the context of OUD treatment.


Subject(s)
Drug Repositioning , Opioid-Related Disorders , Humans , Machine Learning , Opioid-Related Disorders/drug therapy
10.
ArXiv ; 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36911277

ABSTRACT

Opioid use disorder (OUD) is a chronic and relapsing condition that involves the continued and compulsive use of opioids despite harmful consequences. The development of medications with improved efficacy and safety profiles for OUD treatment is urgently needed. Drug repurposing is a promising option for drug discovery due to its reduced cost and expedited approval procedures. Computational approaches based on machine learning enable the rapid screening of DrugBank compounds, identifying those with the potential to be repurposed for OUD treatment. We collected inhibitor data for four major opioid receptors and used advanced machine learning predictors of binding affinity that fuse the gradient boosting decision tree algorithm with two natural language processing (NLP)-based molecular fingerprints and one traditional 2D fingerprint. Using these predictors, we systematically analyzed the binding affinities of DrugBank compounds on four opioid receptors. Based on our machine learning predictions, we were able to discriminate DrugBank compounds with various binding affinity thresholds and selectivities for different receptors. The prediction results were further analyzed for ADMET (absorption, distribution, metabolism, excretion, and toxicity), which provided guidance on repurposing DrugBank compounds for the inhibition of selected opioid receptors. The pharmacological effects of these compounds for OUD treatment need to be tested in further experimental studies and clinical trials. Our machine learning studies provide a valuable platform for drug discovery in the context of OUD treatment.

11.
Comput Biol Med ; 157: 106745, 2023 05.
Article in English | MEDLINE | ID: mdl-36924727

ABSTRACT

Opioid use disorder (OUD) continuously poses major public health challenges and social implications worldwide with dramatic rise of opioid dependence leading to potential abuse. Despite that a few pharmacological agents have been approved for OUD treatment, the efficacy of said agents for OUD requires further improvement in order to provide safer and more effective pharmacological and psychosocial treatments. Proteins including mu, delta, kappa, nociceptin, and zeta opioid receptors are the direct targets of opioids and play critical roles in therapeutic treatments. The protein-protein interaction (PPI) networks of the these receptors increase the complexity in the drug development process for an effective opioid addiction treatment. The report below presents a PPI-network informed machine-learning study of OUD. We have examined more than 500 proteins in the five opioid receptor networks and subsequently collected 74 inhibitor datasets. Machine learning models were constructed by pairing gradient boosting decision tree (GBDT) algorithm with two advanced natural language processing (NLP)-based autoencoder and Transformer fingerprints for molecules. With these models, we systematically carried out evaluations of screening and repurposing potential of more than 120,000 drug candidates for four opioid receptors. In addition, absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties were also considered in the screening of potential drug candidates. Our machine-learning tools determined a few inhibitor compounds with desired potency and ADMET properties for nociceptin opioid receptors. Our approach offers a valuable and promising tool for the pharmacological development of OUD treatments.


Subject(s)
Opioid-Related Disorders , Receptors, Opioid, mu , Humans , Receptors, Opioid, mu/metabolism , Receptors, Opioid , Opioid-Related Disorders/drug therapy , Nociceptin Receptor , Machine Learning , Receptors, Opioid, kappa/metabolism
12.
J Chem Inf Model ; 63(5): 1472-1489, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36826415

ABSTRACT

Drug addiction is a global public health crisis, and the design of antiaddiction drugs remains a major challenge due to intricate mechanisms. Since experimental drug screening and optimization are too time-consuming and expensive, there is urgent need to develop innovative artificial intelligence (AI) methods for addressing the challenge. We tackle this challenge by topology-inferred drug addiction learning (TIDAL) built from integrating multiscale topological Laplacians, deep bidirectional transformer, and ensemble-assisted neural networks (EANNs). Multiscale topological Laplacians are a novel class of algebraic topology tools that embed molecular topological invariants and algebraic invariants into its harmonic spectra and nonharmonic spectra, respectively. These invariants complement sequence information extracted from a bidirectional transformer. We validate the proposed TIDAL framework on 22 drug addiction related, 4 hERG, and 12 DAT data sets, which suggests that the proposed TIDAL is a state-of-the-art framework for the modeling and analysis of drug addiction data. We carry out cross-target analysis of the current drug addiction candidates to alert their side effects and identify their repurposing potentials. Our analysis reveals drug-mediated linear and bilinear target correlations. Finally, TIDAL is applied to shed light on relative efficacy, repurposing potential, and potential side effects of 12 existing antiaddiction medications. Our results suggest that TIDAL provides a new computational strategy for pressingly needed antisubstance addiction drug development.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Substance-Related Disorders , Humans , Artificial Intelligence , Algorithms , Neural Networks, Computer , Drug Development
13.
ArXiv ; 2023 Jan 12.
Article in English | MEDLINE | ID: mdl-36713254

ABSTRACT

Opioid use disorder (OUD) continuously poses major public health challenges and social implications worldwide with dramatic rise of opioid dependence leading to potential abuse. Despite that a few pharmacological agents have been approved for OUD treatment, the efficacy of said agents for OUD requires further improvement in order to provide safer and more effective pharmacological and psychosocial treatments. Preferable therapeutic treatments of OUD rely on the advances in understanding the neurobiological mechanism of opioid dependence. Proteins including mu, delta, kappa, nociceptin, and zeta opioid receptors are the direct targets of opioids. Each receptor has a large protein-protein interaction (PPI) network, that behaves differently when subjected to various treatments, thus increasing the complexity in the drug development process for an effective opioid addiction treatment. The report below analyzes the work by presenting a PPI-network informed machine-learning study of OUD. We have examined more than 500 proteins in the five opioid receptor networks and subsequently collected 74 inhibitor datasets. Machine learning models were constructed by pairing gradient boosting decision tree (GBDT) algorithm with two advanced natural language processing (NLP)-based molecular fingerprints. With these models, we systematically carried out evaluations of screening and repurposing potential of drug candidates for four opioid receptors. In addition, absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties were also considered in the screening of potential drug candidates. Our study can be a valuable and promising tool of pharmacological development for OUD treatments.

14.
Comput Biol Med ; 153: 106491, 2023 02.
Article in English | MEDLINE | ID: mdl-36599209

ABSTRACT

The human ether-a-go-go (hERG) potassium channel (Kv11.1) plays a critical role in mediating cardiac action potential. The blockade of this ion channel can potentially lead fatal disorder and/or long QT syndrome. Many drugs have been withdrawn because of their serious hERG-cardiotoxicity. It is crucial to assess the hERG blockade activity in the early stage of drug discovery. We are particularly interested in the hERG-cardiotoxicity of compounds collected in the DrugBank database considering that many DrugBank compounds have been approved for therapeutic treatments or have high potential to become drugs. Machine learning-based in silico tools offer a rapid and economical platform to virtually screen DrugBank compounds. We design accurate and robust classifiers for blockers/non-blockers and then build regressors to quantitatively analyze the binding potency of the DrugBank compounds on the hERG channel. Molecular sequences are embedded with two natural language processing (NLP) methods, namely, autoencoder and transformer. Complementary three-dimensional (3D) molecular structures are embedded with two advanced mathematical approaches, i.e., topological Laplacians and algebraic graphs. With our state-of-the-art tools, we reveal that 227 out of the 8641 DrugBank compounds are potential hERG blockers, suggesting serious drug safety problems. Our predictions provide guidance for the further experimental interrogation of DrugBank compounds' hERG-cardiotoxicity.


Subject(s)
Cardiotoxicity , Ether-A-Go-Go Potassium Channels , Humans , Ether-A-Go-Go Potassium Channels/chemistry , Ether-A-Go-Go Potassium Channels/metabolism , Ether , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/chemistry , Machine Learning , Ethyl Ethers , Ethers
15.
J Chem Theory Comput ; 18(4): 2703-2719, 2022 Apr 12.
Article in English | MEDLINE | ID: mdl-35294204

ABSTRACT

Cocaine addiction is a psychosocial disorder induced by the chronic use of cocaine and causes a large number of deaths around the world. Despite decades of effort, no drugs have been approved by the Food and Drug Administration (FDA) for the treatment of cocaine dependence. Cocaine dependence is neurological and involves many interacting proteins in the interactome. Among them, the dopamine (DAT), serotonin (SERT), and norepinephrine (NET) transporters are three major targets. Each of these targets has a large protein-protein interaction (PPI) network, which must be considered in the anticocaine addiction drug discovery. This work presents DAT, SERT, and NET interactome network-informed machine learning/deep learning (ML/DL) studies of cocaine addiction. We collected and analyzed 61 protein targets out of 460 proteins in the DAT, SERT, and NET PPI networks that have sufficiently large existing inhibitor datasets. Utilizing autoencoder (AE) and other ML/DL algorithms, including gradient boosting decision tree (GBDT) and multitask deep neural network (MT-DNN), we built predictive models for these targets with 115 407 inhibitors to predict drug repurposing potential and possible side effects. We further screened their absorption, distribution, metabolism, and excretion, and toxicity (ADMET) properties to search for leads having potential for developing treatments for cocaine addiction. Our approach offers a new systematic protocol for artificial intelligence (AI)-based anticocaine addiction lead discovery.


Subject(s)
Cocaine-Related Disorders , Artificial Intelligence , Cocaine-Related Disorders/drug therapy , Cocaine-Related Disorders/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Humans , Machine Learning , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins , United States
SELECTION OF CITATIONS
SEARCH DETAIL
...