4.6 Article

Artificial intelligence framework identifies candidate targets for drug repurposing in Alzheimer's disease

期刊

ALZHEIMERS RESEARCH & THERAPY
卷 14, 期 1, 页码 -

出版社

BMC
DOI: 10.1186/s13195-021-00951-z

关键词

Alzheimer's disease; Drug repurposing; Genome-wide association studies (GWAS); Multi-omics; Network medicine; Pioglitazone

资金

  1. National Institute of Aging (NIA) of the National Institutes of Health (NIH) [R01AG066707, U01AG073323, 3R01AG066707-01S1]
  2. NIH Research Grant [3R01AG066707-02S1]
  3. Office of Data Science Strategy (ODSS)
  4. NIA [R56AG063870, R35AG071476, R01AG069900]
  5. Translational Therapeutics Core of the Cleveland Alzheimer's Disease Research Center [P30AG072959]
  6. NIH-NINDS [U01NS093334]
  7. NIH-NIGMS [P20GM109025]
  8. NIH-NHGRI [U01HG009086]
  9. Brockman Foundation
  10. AHA/Allen Initiative [19PABH134580006]

向作者/读者索取更多资源

This study develops a network-based artificial intelligence framework that integrates multi-omics data with protein-protein interactome networks to accurately infer drug targets affected by GWAS-identified variants, leading to the discovery of new therapeutics. Through applying this approach to AD, 103 validated ARGs are identified, and three drugs are found to be significantly associated with decreased risk of AD.
Background Genome-wide association studies (GWAS) have identified numerous susceptibility loci for Alzheimer's disease (AD). However, utilizing GWAS and multi-omics data to identify high-confidence AD risk genes (ARGs) and druggable targets that can guide development of new therapeutics for patients suffering from AD has heretofore not been successful. Methods To address this critical problem in the field, we have developed a network-based artificial intelligence framework that is capable of integrating multi-omics data along with human protein-protein interactome networks to accurately infer accurate drug targets impacted by GWAS-identified variants to identify new therapeutics. When applied to AD, this approach integrates GWAS findings, multi-omics data from brain samples of AD patients and AD transgenic animal models, drug-target networks, and the human protein-protein interactome, along with large-scale patient database validation and in vitro mechanistic observations in human microglia cells. Results Through this approach, we identified 103 ARGs validated by various levels of pathobiological evidence in AD. Via network-based prediction and population-based validation, we then showed that three drugs (pioglitazone, febuxostat, and atenolol) are significantly associated with decreased risk of AD compared with matched control populations. Pioglitazone usage is significantly associated with decreased risk of AD (hazard ratio (HR) = 0.916, 95% confidence interval [CI] 0.861-0.974, P = 0.005) in a retrospective case-control validation. Pioglitazone is a peroxisome proliferator-activated receptor (PPAR) agonist used to treat type 2 diabetes, and propensity score matching cohort studies confirmed its association with reduced risk of AD in comparison to glipizide (HR = 0.921, 95% CI 0.862-0.984, P = 0.0159), an insulin secretagogue that is also used to treat type 2 diabetes. In vitro experiments showed that pioglitazone downregulated glycogen synthase kinase 3 beta (GSK3 beta) and cyclin-dependent kinase (CDK5) in human microglia cells, supporting a possible mechanism-of-action for its beneficial effect in AD. Conclusions In summary, we present an integrated, network-based artificial intelligence methodology to rapidly translate GWAS findings and multi-omics data to genotype-informed therapeutic discovery in AD.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.6
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据