Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.724
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 177(4): 999-1009.e10, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-31051108

RESUMEN

What specific features should visual neurons encode, given the infinity of real-world images and the limited number of neurons available to represent them? We investigated neuronal selectivity in monkey inferotemporal cortex via the vast hypothesis space of a generative deep neural network, avoiding assumptions about features or semantic categories. A genetic algorithm searched this space for stimuli that maximized neuronal firing. This led to the evolution of rich synthetic images of objects with complex combinations of shapes, colors, and textures, sometimes resembling animals or familiar people, other times revealing novel patterns that did not map to any clear semantic category. These results expand our conception of the dictionary of features encoded in the cortex, and the approach can potentially reveal the internal representations of any system whose input can be captured by a generative model.


Asunto(s)
Red Nerviosa/fisiología , Lóbulo Temporal/fisiología , Percepción Visual/fisiología , Algoritmos , Animales , Corteza Cerebral/fisiología , Macaca mulatta/fisiología , Masculino , Neuronas/metabolismo , Neuronas/fisiología
2.
Cell ; 178(1): 91-106.e23, 2019 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-31178116

RESUMEN

Alternative polyadenylation (APA) is a major driver of transcriptome diversity in human cells. Here, we use deep learning to predict APA from DNA sequence alone. We trained our model (APARENT, APA REgression NeT) on isoform expression data from over 3 million APA reporters. APARENT's predictions are highly accurate when tasked with inferring APA in synthetic and human 3'UTRs. Visualizing features learned across all network layers reveals that APARENT recognizes sequence motifs known to recruit APA regulators, discovers previously unknown sequence determinants of 3' end processing, and integrates these features into a comprehensive, interpretable, cis-regulatory code. We apply APARENT to forward engineer functional polyadenylation signals with precisely defined cleavage position and isoform usage and validate predictions experimentally. Finally, we use APARENT to quantify the impact of genetic variants on APA. Our approach detects pathogenic variants in a wide range of disease contexts, expanding our understanding of the genetic origins of disease.


Asunto(s)
Aprendizaje Profundo , Modelos Genéticos , Poliadenilación/genética , Regiones no Traducidas 3'/genética , Secuencia de Bases/genética , Bases de Datos Genéticas , Expresión Génica/genética , Células HEK293 , Humanos , Mutagénesis/genética , División del ARN/genética , ARN Mensajero/genética , RNA-Seq , Biología Sintética , Transcriptoma
3.
Annu Rev Pharmacol Toxicol ; 64: 527-550, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-37738505

RESUMEN

Drug discovery is adapting to novel technologies such as data science, informatics, and artificial intelligence (AI) to accelerate effective treatment development while reducing costs and animal experiments. AI is transforming drug discovery, as indicated by increasing interest from investors, industrial and academic scientists, and legislators. Successful drug discovery requires optimizing properties related to pharmacodynamics, pharmacokinetics, and clinical outcomes. This review discusses the use of AI in the three pillars of drug discovery: diseases, targets, and therapeutic modalities, with a focus on small-molecule drugs. AI technologies, such as generative chemistry, machine learning, and multiproperty optimization, have enabled several compounds to enter clinical trials. The scientific community must carefully vet known information to address the reproducibility crisis. The full potential of AI in drug discovery can only be realized with sufficient ground truth and appropriate human intervention at later pipeline stages.


Asunto(s)
Inteligencia Artificial , Médicos , Animales , Humanos , Reproducibilidad de los Resultados , Descubrimiento de Drogas , Tecnología
4.
Proc Natl Acad Sci U S A ; 121(5): e2311436121, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38266050

RESUMEN

Manifold fitting, which offers substantial potential for efficient and accurate modeling, poses a critical challenge in nonlinear data analysis. This study presents an approach that employs neural networks to fit the latent manifold. Leveraging the generative adversarial framework, this method learns smooth mappings between low-dimensional latent space and high-dimensional ambient space, echoing the Riemannian exponential and logarithmic maps. The well-trained neural networks provide estimations for the latent manifold, facilitate data projection onto the manifold, and even generate data points that reside directly within the manifold. Through an extensive series of simulation studies and real data experiments, we demonstrate the effectiveness and accuracy of our approach in capturing the inherent structure of the underlying manifold within the ambient space data. Notably, our method exceeds the computational efficiency limitations of previous approaches and offers control over the dimensionality and smoothness of the resulting manifold. This advancement holds significant potential in the fields of statistics and computer science. The seamless integration of powerful neural network architectures with generative adversarial techniques unlocks possibilities for manifold fitting, thereby enhancing data analysis. The implications of our findings span diverse applications, from dimensionality reduction and data visualization to generating authentic data. Collectively, our research paves the way for future advancements in nonlinear data analysis and offers a beacon for subsequent scholarly pursuits.

5.
Proc Natl Acad Sci U S A ; 121(23): e2322376121, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38809705

RESUMEN

In this article, we develop CausalEGM, a deep learning framework for nonlinear dimension reduction and generative modeling of the dependency among covariate features affecting treatment and response. CausalEGM can be used for estimating causal effects in both binary and continuous treatment settings. By learning a bidirectional transformation between the high-dimensional covariate space and a low-dimensional latent space and then modeling the dependencies of different subsets of the latent variables on the treatment and response, CausalEGM can extract the latent covariate features that affect both treatment and response. By conditioning on these features, one can mitigate the confounding effect of the high dimensional covariate on the estimation of the causal relation between treatment and response. In a series of experiments, the proposed method is shown to achieve superior performance over existing methods in both binary and continuous treatment settings. The improvement is substantial when the sample size is large and the covariate is of high dimension. Finally, we established excess risk bounds and consistency results for our method, and discuss how our approach is related to and improves upon other dimension reduction approaches in causal inference.

6.
Proc Natl Acad Sci U S A ; 121(27): e2311500121, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38916999

RESUMEN

Proteins mediate their functions through chemical interactions; modeling these interactions, which are typically through sidechains, is an important need in protein design. However, constructing an all-atom generative model requires an appropriate scheme for managing the jointly continuous and discrete nature of proteins encoded in the structure and sequence. We describe an all-atom diffusion model of protein structure, Protpardelle, which represents all sidechain states at once as a "superposition" state; superpositions defining a protein are collapsed into individual residue types and conformations during sample generation. When combined with sequence design methods, our model is able to codesign all-atom protein structure and sequence. Generated proteins are of good quality under the typical quality, diversity, and novelty metrics, and sidechains reproduce the chemical features and behavior of natural proteins. Finally, we explore the potential of our model to conduct all-atom protein design and scaffold functional motifs in a backbone- and rotamer-free way.


Asunto(s)
Modelos Moleculares , Conformación Proteica , Proteínas , Proteínas/química , Secuencia de Aminoácidos
7.
Proc Natl Acad Sci U S A ; 121(18): e2307304121, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38640257

RESUMEN

Over the past few years, machine learning models have significantly increased in size and complexity, especially in the area of generative AI such as large language models. These models require massive amounts of data and compute capacity to train, to the extent that concerns over the training data (such as protected or private content) cannot be practically addressed by retraining the model "from scratch" with the questionable data removed or altered. Furthermore, despite significant efforts and controls dedicated to ensuring that training corpora are properly curated and composed, the sheer volume required makes it infeasible to manually inspect each datum comprising a training corpus. One potential approach to training corpus data defects is model disgorgement, by which we broadly mean the elimination or reduction of not only any improperly used data, but also the effects of improperly used data on any component of an ML model. Model disgorgement techniques can be used to address a wide range of issues, such as reducing bias or toxicity, increasing fidelity, and ensuring responsible use of intellectual property. In this paper, we survey the landscape of model disgorgement methods and introduce a taxonomy of disgorgement techniques that are applicable to modern ML systems. In particular, we investigate the various meanings of "removing the effects" of data on the trained model in a way that does not require retraining from scratch.


Asunto(s)
Lenguaje , Aprendizaje Automático
8.
Proc Natl Acad Sci U S A ; 121(21): e2314021121, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38722813

RESUMEN

Generative AI that can produce realistic text, images, and other human-like outputs is currently transforming many different industries. Yet it is not yet known how such tools might influence social science research. I argue Generative AI has the potential to improve survey research, online experiments, automated content analyses, agent-based models, and other techniques commonly used to study human behavior. In the second section of this article, I discuss the many limitations of Generative. I examine how bias in the data used to train these tools can negatively impact social science research-as well as a range of other challenges related to ethics, replication, environmental impact, and the proliferation of low-quality research. I conclude by arguing that social scientists can address many of these limitations by creating open-source infrastructure for research on human behavior. Such infrastructure is not only necessary to ensure broad access to high-quality research tools, I argue, but also because the progress of AI will require deeper understanding of the social forces that guide human behavior.


Asunto(s)
Inteligencia Artificial , Ciencias Sociales , Humanos
9.
Brief Bioinform ; 25(4)2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38980369

RESUMEN

Recent studies have extensively used deep learning algorithms to analyze gene expression to predict disease diagnosis, treatment effectiveness, and survival outcomes. Survival analysis studies on diseases with high mortality rates, such as cancer, are indispensable. However, deep learning models are plagued by overfitting owing to the limited sample size relative to the large number of genes. Consequently, the latest style-transfer deep generative models have been implemented to generate gene expression data. However, these models are limited in their applicability for clinical purposes because they generate only transcriptomic data. Therefore, this study proposes ctGAN, which enables the combined transformation of gene expression and survival data using a generative adversarial network (GAN). ctGAN improves survival analysis by augmenting data through style transformations between breast cancer and 11 other cancer types. We evaluated the concordance index (C-index) enhancements compared with previous models to demonstrate its superiority. Performance improvements were observed in nine of the 11 cancer types. Moreover, ctGAN outperformed previous models in seven out of the 11 cancer types, with colon adenocarcinoma (COAD) exhibiting the most significant improvement (median C-index increase of ~15.70%). Furthermore, integrating the generated COAD enhanced the log-rank p-value (0.041) compared with using only the real COAD (p-value = 0.797). Based on the data distribution, we demonstrated that the model generated highly plausible data. In clustering evaluation, ctGAN exhibited the highest performance in most cases (89.62%). These findings suggest that ctGAN can be meaningfully utilized to predict disease progression and select personalized treatments in the medical field.


Asunto(s)
Aprendizaje Profundo , Humanos , Análisis de Supervivencia , Algoritmos , Neoplasias/genética , Neoplasias/mortalidad , Perfilación de la Expresión Génica/métodos , Redes Neurales de la Computación , Biología Computacional/métodos , Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Femenino , Regulación Neoplásica de la Expresión Génica
10.
Brief Bioinform ; 25(4)2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-39007594

RESUMEN

Artificial intelligence (AI)-driven methods can vastly improve the historically costly drug design process, with various generative models already in widespread use. Generative models for de novo drug design, in particular, focus on the creation of novel biological compounds entirely from scratch, representing a promising future direction. Rapid development in the field, combined with the inherent complexity of the drug design process, creates a difficult landscape for new researchers to enter. In this survey, we organize de novo drug design into two overarching themes: small molecule and protein generation. Within each theme, we identify a variety of subtasks and applications, highlighting important datasets, benchmarks, and model architectures and comparing the performance of top models. We take a broad approach to AI-driven drug design, allowing for both micro-level comparisons of various methods within each subtask and macro-level observations across different fields. We discuss parallel challenges and approaches between the two applications and highlight future directions for AI-driven de novo drug design as a whole. An organized repository of all covered sources is available at https://github.com/gersteinlab/GenAI4Drug.


Asunto(s)
Inteligencia Artificial , Diseño de Fármacos , Proteínas , Proteínas/química , Humanos , Biología Computacional/métodos
11.
Brief Bioinform ; 25(3)2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38647154

RESUMEN

Molecular generative models have exhibited promising capabilities in designing molecules from scratch with high binding affinities in a predetermined protein pocket, offering potential synergies with traditional structural-based drug design strategy. However, the generative processes of such models are random and the atomic interaction information between ligand and protein are ignored. On the other hand, the ligand has high propensity to bind with residues called hotspots. Hotspot residues contribute to the majority of the binding free energies and have been recognized as appealing targets for designed molecules. In this work, we develop an interaction prompt guided diffusion model, InterDiff to deal with the challenges. Four kinds of atomic interactions are involved in our model and represented as learnable vector embeddings. These embeddings serve as conditions for individual residue to guide the molecular generative process. Comprehensive in silico experiments evince that our model could generate molecules with desired ligand-protein interactions in a guidable way. Furthermore, we validate InterDiff on two realistic protein-based therapeutic agents. Results show that InterDiff could generate molecules with better or similar binding mode compared to known targeted drugs.


Asunto(s)
Proteínas , Proteínas/química , Proteínas/metabolismo , Ligandos , Unión Proteica , Diseño de Fármacos , Modelos Moleculares , Algoritmos , Sitios de Unión , Simulación por Computador
12.
Brief Bioinform ; 25(2)2024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-38385880

RESUMEN

We present a language model Affordable Cancer Interception and Diagnostics (ACID) that can achieve high classification performance in the diagnosis of cancer exclusively from using raw cfDNA sequencing reads. We formulate ACID as an autoregressive language model. ACID is pretrained with language sentences that are obtained from concatenation of raw sequencing reads and diagnostic labels. We benchmark ACID against three methods. On testing set subjected to whole-genome sequencing, ACID significantly outperforms the best benchmarked method in diagnosis of cancer [Area Under the Receiver Operating Curve (AUROC), 0.924 versus 0.853; P < 0.001] and detection of hepatocellular carcinoma (AUROC, 0.981 versus 0.917; P < 0.001). ACID can achieve high accuracy with just 10 000 reads per sample. Meanwhile, ACID achieves the best performance on testing sets that were subjected to bisulfite sequencing compared with benchmarked methods. In summary, we present an affordable, simple yet efficient end-to-end paradigm for cancer detection using raw cfDNA sequencing reads.


Asunto(s)
Carcinoma Hepatocelular , Ácidos Nucleicos Libres de Células , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/genética , Área Bajo la Curva , Ácidos Nucleicos Libres de Células/genética , Lenguaje , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/genética
13.
Brief Bioinform ; 25(3)2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38581415

RESUMEN

Discovering hit molecules with desired biological activity in a directed manner is a promising but profound task in computer-aided drug discovery. Inspired by recent generative AI approaches, particularly Diffusion Models (DM), we propose Graph Latent Diffusion Model (GLDM)-a latent DM that preserves both the effectiveness of autoencoders of compressing complex chemical data and the DM's capabilities of generating novel molecules. Specifically, we first develop an autoencoder to encode the molecular data into low-dimensional latent representations and then train the DM on the latent space to generate molecules inducing targeted biological activity defined by gene expression profiles. Manipulating DM in the latent space rather than the input space avoids complicated operations to map molecule decomposition and reconstruction to diffusion processes, and thus improves training efficiency. Experiments show that GLDM not only achieves outstanding performances on molecular generation benchmarks, but also generates samples with optimal chemical properties and potentials to induce desired biological activity.


Asunto(s)
Benchmarking , Descubrimiento de Drogas , Difusión
14.
Brief Bioinform ; 25(3)2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38742520

RESUMEN

The dynamic evolution of the severe acute respiratory syndrome coronavirus 2 virus is primarily driven by mutations in its genetic sequence, culminating in the emergence of variants with increased capability to evade host immune responses. Accurate prediction of such mutations is fundamental in mitigating pandemic spread and developing effective control measures. This study introduces a robust and interpretable deep-learning approach called PRIEST. This innovative model leverages time-series viral sequences to foresee potential viral mutations. Our comprehensive experimental evaluations underscore PRIEST's proficiency in accurately predicting immune-evading mutations. Our work represents a substantial step in utilizing deep-learning methodologies for anticipatory viral mutation analysis and pandemic response.


Asunto(s)
COVID-19 , Evasión Inmune , Mutación , SARS-CoV-2 , SARS-CoV-2/genética , SARS-CoV-2/inmunología , Humanos , COVID-19/virología , COVID-19/inmunología , COVID-19/genética , Evasión Inmune/genética , Aprendizaje Profundo , Evolución Molecular , Pandemias
15.
Brief Bioinform ; 25(4)2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38960404

RESUMEN

Recent advances in microfluidics and sequencing technologies allow researchers to explore cellular heterogeneity at single-cell resolution. In recent years, deep learning frameworks, such as generative models, have brought great changes to the analysis of transcriptomic data. Nevertheless, relying on the potential space of these generative models alone is insufficient to generate biological explanations. In addition, most of the previous work based on generative models is limited to shallow neural networks with one to three layers of latent variables, which may limit the capabilities of the models. Here, we propose a deep interpretable generative model called d-scIGM for single-cell data analysis. d-scIGM combines sawtooth connectivity techniques and residual networks, thereby constructing a deep generative framework. In addition, d-scIGM incorporates hierarchical prior knowledge of biological domains to enhance the interpretability of the model. We show that d-scIGM achieves excellent performance in a variety of fundamental tasks, including clustering, visualization, and pseudo-temporal inference. Through topic pathway studies, we found that d-scIGM-learned topics are better enriched for biologically meaningful pathways compared to the baseline models. Furthermore, the analysis of drug response data shows that d-scIGM can capture drug response patterns in large-scale experiments, which provides a promising way to elucidate the underlying biological mechanisms. Lastly, in the melanoma dataset, d-scIGM accurately identified different cell types and revealed multiple melanin-related driver genes and key pathways, which are critical for understanding disease mechanisms and drug development.


Asunto(s)
Aprendizaje Profundo , RNA-Seq , Análisis de la Célula Individual , Análisis de la Célula Individual/métodos , Humanos , RNA-Seq/métodos , Biología Computacional/métodos , Algoritmos , Análisis de Secuencia de ARN/métodos , Redes Neurales de la Computación , Análisis de Expresión Génica de una Sola Célula
16.
Brief Bioinform ; 25(4)2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38856172

RESUMEN

With their diverse biological activities, peptides are promising candidates for therapeutic applications, showing antimicrobial, antitumour and hormonal signalling capabilities. Despite their advantages, therapeutic peptides face challenges such as short half-life, limited oral bioavailability and susceptibility to plasma degradation. The rise of computational tools and artificial intelligence (AI) in peptide research has spurred the development of advanced methodologies and databases that are pivotal in the exploration of these complex macromolecules. This perspective delves into integrating AI in peptide development, encompassing classifier methods, predictive systems and the avant-garde design facilitated by deep-generative models like generative adversarial networks and variational autoencoders. There are still challenges, such as the need for processing optimization and careful validation of predictive models. This work outlines traditional strategies for machine learning model construction and training techniques and proposes a comprehensive AI-assisted peptide design and validation pipeline. The evolving landscape of peptide design using AI is emphasized, showcasing the practicality of these methods in expediting the development and discovery of novel peptides within the context of peptide-based drug discovery.


Asunto(s)
Inteligencia Artificial , Descubrimiento de Drogas , Péptidos , Péptidos/química , Péptidos/uso terapéutico , Péptidos/farmacología , Descubrimiento de Drogas/métodos , Humanos , Diseño de Fármacos , Aprendizaje Automático , Biología Computacional/métodos
17.
Brief Bioinform ; 25(2)2024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-38314912

RESUMEN

Increasing volumes of biomedical data are amassing in databases. Large-scale analyses of these data have wide-ranging applications in biology and medicine. Such analyses require tools to characterize and process entries at scale. However, existing tools, mainly centered on extracting predefined fields, often fail to comprehensively process database entries or correct evident errors-a task humans can easily perform. These tools also lack the ability to reason like domain experts, hindering their robustness and analytical depth. Recent advances with large language models (LLMs) provide a fundamentally new way to query databases. But while a tool such as ChatGPT is adept at answering questions about manually input records, challenges arise when scaling up this process. First, interactions with the LLM need to be automated. Second, limitations on input length may require a record pruning or summarization pre-processing step. Third, to behave reliably as desired, the LLM needs either well-designed, short, 'few-shot' examples, or fine-tuning based on a larger set of well-curated examples. Here, we report ChIP-GPT, based on fine-tuning of the generative pre-trained transformer (GPT) model Llama and on a program prompting the model iteratively and handling its generation of answer text. This model is designed to extract metadata from the Sequence Read Archive, emphasizing the identification of chromatin immunoprecipitation (ChIP) targets and cell lines. When trained with 100 examples, ChIP-GPT demonstrates 90-94% accuracy. Notably, it can seamlessly extract data from records with typos or absent field labels. Our proposed method is easily adaptable to customized questions and different databases.


Asunto(s)
Medicina , Humanos , Línea Celular , Inmunoprecipitación de Cromatina , Bases de Datos Factuales , Lenguaje
18.
Proc Natl Acad Sci U S A ; 120(31): e2305273120, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37487072

RESUMEN

Spider webs are incredible biological structures, comprising thin but strong silk filament and arranged into complex hierarchical architectures with striking mechanical properties (e.g., lightweight but high strength, achieving diverse mechanical responses). While simple 2D orb webs can easily be mimicked, the modeling and synthesis of 3D-based web structures remain challenging, partly due to the rich set of design features. Here, we provide a detailed analysis of the heterogeneous graph structures of spider webs and use deep learning as a way to model and then synthesize artificial, bioinspired 3D web structures. The generative models are conditioned based on key geometric parameters (including average edge length, number of nodes, average node degree, and others). To identify graph construction principles, we use inductive representation sampling of large experimentally determined spider web graphs, to yield a dataset that is used to train three conditional generative models: 1) an analog diffusion model inspired by nonequilibrium thermodynamics, with sparse neighbor representation; 2) a discrete diffusion model with full neighbor representation; and 3) an autoregressive transformer architecture with full neighbor representation. All three models are scalable, produce complex, de novo bioinspired spider web mimics, and successfully construct graphs that meet the design objectives. We further propose an algorithm that assembles web samples produced by the generative models into larger-scale structures based on a series of geometric design targets, including helical and parametric shapes, mimicking, and extending natural design principles toward integration with diverging engineering objectives. Several webs are manufactured using 3D printing and tested to assess mechanical properties.


Asunto(s)
Aprendizaje Profundo , Arañas , Animales , Algoritmos , Comercio , Citoesqueleto
19.
Proc Natl Acad Sci U S A ; 120(21): e2209124120, 2023 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-37192164

RESUMEN

Detecting differentially expressed genes is important for characterizing subpopulations of cells. In scRNA-seq data, however, nuisance variation due to technical factors like sequencing depth and RNA capture efficiency obscures the underlying biological signal. Deep generative models have been extensively applied to scRNA-seq data, with a special focus on embedding cells into a low-dimensional latent space and correcting for batch effects. However, little attention has been paid to the problem of utilizing the uncertainty from the deep generative model for differential expression (DE). Furthermore, the existing approaches do not allow for controlling for effect size or the false discovery rate (FDR). Here, we present lvm-DE, a generic Bayesian approach for performing DE predictions from a fitted deep generative model, while controlling the FDR. We apply the lvm-DE framework to scVI and scSphere, two deep generative models. The resulting approaches outperform state-of-the-art methods at estimating the log fold change in gene expression levels as well as detecting differentially expressed genes between subpopulations of cells.


Asunto(s)
ARN , Análisis de la Célula Individual , Teorema de Bayes , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Perfilación de la Expresión Génica/métodos
20.
Proc Natl Acad Sci U S A ; 120(41): e2311627120, 2023 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-37788311

RESUMEN

Political discourse is the soul of democracy, but misunderstanding and conflict can fester in divisive conversations. The widespread shift to online discourse exacerbates many of these problems and corrodes the capacity of diverse societies to cooperate in solving social problems. Scholars and civil society groups promote interventions that make conversations less divisive or more productive, but scaling these efforts to online discourse is challenging. We conduct a large-scale experiment that demonstrates how online conversations about divisive topics can be improved with AI tools. Specifically, we employ a large language model to make real-time, evidence-based recommendations intended to improve participants' perception of feeling understood. These interventions improve reported conversation quality, promote democratic reciprocity, and improve the tone, without systematically changing the content of the conversation or moving people's policy attitudes.


Asunto(s)
Lenguaje , Políticas , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA