1
|
Qi C, Li A, Su F, Wang Y, Zhou L, Tang C, Feng R, Mao R, Chen M, Chen L, Koppelman GH, Bourgonje AR, Zhou H, Hu S. An atlas of the shared genetic architecture between atopic and gastrointestinal diseases. Commun Biol 2024; 7:1696. [PMID: 39719505 DOI: 10.1038/s42003-024-07416-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 12/18/2024] [Indexed: 12/26/2024] Open
Abstract
Comorbidity among atopic diseases (ADs) and gastrointestinal diseases (GIDs) has been repeatedly demonstrated by epidemiological studies, whereas the shared genetic liability remains largely unknown. Here we establish an atlas of the shared genetic architecture between 10 ADs or related traits and 11 GIDs, comprehensively investigating the comorbidity-associated genomic regions, cell types, genes and genetically predicted causality. Although distinct genetic correlations between AD-GID are observed, including 14 genome-wide and 28 regional correlations, genetic factors of Crohn's disease (CD), ulcerative colitis (UC), celiac disease and asthma subtypes are converged on CD4+ T cells consistently across relevant tissues. Fourteen genes are associated with comorbidities, with three genes are known treatment targets, showing probabilities for drug repurposing. Lower expressions of WDR18 and GPX4 in PBMC CD4+ T cells predict decreased risk of CD and asthma, which could be novel drug targets. MR unveils certain ADs led to higher risk of GIDs or vice versa. Taken together, here we show distinct genetic correlations between AD-GID pairs, but the correlated genomic loci converge on the dysregulation of CD4+ T cells. Inhibiting WDR18 and GPX4 expressions might be candidate therapeutic strategies for CD and asthma. Estimated causality indicates potential guidance for preventing comorbidity.
Collapse
Affiliation(s)
- Cancan Qi
- Microbiome Medicine Center, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - An Li
- Department of Periodontology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Fengyuan Su
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yu Wang
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Longyuan Zhou
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Ce Tang
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Rui Feng
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Department of Gastroenterology, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-Sen University, Nanning, Guangxi, China
| | - Ren Mao
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Minhu Chen
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Lianmin Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China
- Cardiovascular Research Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Gerard H Koppelman
- University of Groningen University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
- University of Groningen University Medical Centre Groningen, Beatrix Children's Hospital, Department of Paediatric Pulmonology and Paediatric Allergology, Groningen, the Netherlands
| | - Arno R Bourgonje
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
- The Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Hongwei Zhou
- Microbiome Medicine Center, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Shixian Hu
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Sun TH, Wang CC, Liu TY, Lo SC, Huang YX, Chien SY, Chu YD, Tsai FJ, Hsu KC. Utility of polygenic scores across diverse diseases in a hospital cohort for predictive modeling. Nat Commun 2024; 15:3168. [PMID: 38609356 PMCID: PMC11014845 DOI: 10.1038/s41467-024-47472-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
Polygenic scores estimate genetic susceptibility to diseases. We systematically calculated polygenic scores across 457 phenotypes using genotyping array data from China Medical University Hospital. Logistic regression models assessed polygenic scores' ability to predict disease traits. The polygenic score model with the highest accuracy, based on maximal area under the receiver operating characteristic curve (AUC), is provided on the GeneAnaBase website of the hospital. Our findings indicate 49 phenotypes with AUC greater than 0.6, predominantly linked to endocrine and metabolic diseases. Notably, hyperplasia of the prostate exhibited the highest disease prediction ability (P value = 1.01 × 10-19, AUC = 0.874), highlighting the potential of these polygenic scores in preventive medicine and diagnosis. This study offers a comprehensive evaluation of polygenic scores performance across diverse human traits, identifying promising applications for precision medicine and personalized healthcare, thereby inspiring further research and development in this field.
Collapse
Affiliation(s)
- Ting-Hsuan Sun
- Artificial Intelligence Center, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Chia-Chun Wang
- Artificial Intelligence Center, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Ting-Yuan Liu
- Million-person Precision Medicine Initiative, Department of Medical Research, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Shih-Chang Lo
- Artificial Intelligence Center, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Yi-Xuan Huang
- Artificial Intelligence Center, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Shang-Yu Chien
- Artificial Intelligence Center, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Yu-De Chu
- Artificial Intelligence Center, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Fuu-Jen Tsai
- Department of Medical Research, China Medical University Hospital, Taichung, 40447, Taiwan.
- School of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan.
- Division of Pediatric Genetics, Children's Hospital of China Medical University, Taichung, 40447, Taiwan.
- Department of Biotechnology and Bioinformatics, Asia University, Taichung, 41354, Taiwan.
| | - Kai-Cheng Hsu
- Artificial Intelligence Center, China Medical University Hospital, Taichung, 40447, Taiwan.
- Department of Neurology, China Medical University Hospital, Taichung, 40447, Taiwan.
- Department of Medicine, China Medical University, Taichung, 40402, Taiwan.
| |
Collapse
|
3
|
John M, Lencz T. Potential application of elastic nets for shared polygenicity detection with adapted threshold selection. Int J Biostat 2023; 19:417-438. [PMID: 36327464 PMCID: PMC10154439 DOI: 10.1515/ijb-2020-0108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 10/05/2022] [Indexed: 11/06/2022]
Abstract
Current research suggests that hundreds to thousands of single nucleotide polymorphisms (SNPs) with small to modest effect sizes contribute to the genetic basis of many disorders, a phenomenon labeled as polygenicity. Additionally, many such disorders demonstrate polygenic overlap, in which risk alleles are shared at associated genetic loci. A simple strategy to detect polygenic overlap between two phenotypes is based on rank-ordering the univariate p-values from two genome-wide association studies (GWASs). Although high-dimensional variable selection strategies such as Lasso and elastic nets have been utilized in other GWAS analysis settings, they are yet to be utilized for detecting shared polygenicity. In this paper, we illustrate how elastic nets, with polygenic scores as the dependent variable and with appropriate adaptation in selecting the penalty parameter, may be utilized for detecting a subset of SNPs involved in shared polygenicity. We provide theory to better understand our approaches, and illustrate their utility using synthetic datasets. Results from extensive simulations are presented comparing the elastic net approaches with the rank ordering approach, in various scenarios. Results from simulations studies exhibit one of the elastic net approaches to be superior when the correlations among the SNPs are high. Finally, we apply the methods on two real datasets to illustrate further the capabilities, limitations and differences among the methods.
Collapse
Affiliation(s)
- Majnu John
- Institute of Behavioral Science, Feinstein Institutes of Medical Research, Manhasset, NY
- Division of Psychiatry Research, The Zucker Hillside Hospital, Northwell Health System, Glen Oaks, NY
- Departments of Psychiatry and of Mathematics, Hofstra University, Hempstead, NY
| | - Todd Lencz
- Institute of Behavioral Science, Feinstein Institutes of Medical Research, Manhasset, NY
- Division of Psychiatry Research, The Zucker Hillside Hospital, Northwell Health System, Glen Oaks, NY
- Departments of Psychiatry and of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY
| |
Collapse
|
4
|
Sánchez-Valle J, Valencia A. Molecular bases of comorbidities: present and future perspectives. Trends Genet 2023; 39:773-786. [PMID: 37482451 DOI: 10.1016/j.tig.2023.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/12/2023] [Accepted: 06/12/2023] [Indexed: 07/25/2023]
Abstract
Co-occurrence of diseases decreases patient quality of life, complicates treatment choices, and increases mortality. Analyses of electronic health records present a complex scenario of comorbidity relationships that vary by age, sex, and cohort under study. The study of similarities between diseases using 'omics data, such as genes altered in diseases, gene expression, proteome, and microbiome, are fundamental to uncovering the origin of, and potential treatment for, comorbidities. Recent studies have produced a first generation of genetic interpretations for as much as 46% of the comorbidities described in large cohorts. Integrating different sources of molecular information and using artificial intelligence (AI) methods are promising approaches for the study of comorbidities. They may help to improve the treatment of comorbidities, including the potential repositioning of drugs.
Collapse
Affiliation(s)
- Jon Sánchez-Valle
- Life Sciences Department, Barcelona Supercomputing Center, Barcelona, 08034, Spain.
| | - Alfonso Valencia
- Life Sciences Department, Barcelona Supercomputing Center, Barcelona, 08034, Spain; ICREA, Barcelona, 08010, Spain.
| |
Collapse
|
5
|
Li Q, Wang H, Wang H, Deng J, Cheng Z, Lin W, Zhu R, Chen S, Guo J, Tang LV, Hu Y. Association between serum alkaline phosphatase levels in late pregnancy and the incidence of venous thromboembolism postpartum: a retrospective cohort study. EClinicalMedicine 2023; 62:102088. [PMID: 37533415 PMCID: PMC10393549 DOI: 10.1016/j.eclinm.2023.102088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 08/04/2023] Open
Abstract
Background Two previous studies found alkaline phosphatase (ALP) levels were related with the development of venous thromboembolism (VTE) in hospitalised patients. VTE is a leading cause of death during pregnancy and postpartum. No prior study has investigated the associations of ALP levels and VTE postpartum, and the related mechanisms remain unclear. This study aimed to investigate the associations between ALP levels and VTE postpartum, and to reveal the potential mechanisms. Methods In this retrospective cohort study, we included pregnant women who planned to deliver at the Department of Obstetrics and Gynecology in the three designated hospitals in a multicentre cohort of pregnant women in Wuhan, China, during two recruitment periods of January 1, 2018 to December 31, 2019, and May 14, 2020 to March 25, 2022. A total of 10,044 participants with serum ALP and whole blood hemoglobin measurements in late pregnancy (median, 37 (35, 39) weeks) were enrolled. The participants' incidences of VTE (deep venous thrombosis and/or pulmonary embolism) postpartum were confirmed from the medical records. Pregnant women with new-onset VTE postpartum (within 6 weeks after delivery) were confirmed as VTE cases. Findings Approximately 0.8% (79/10,044) of the pregnant women were diagnosed with VTE postpartum. In the unadjusted model, pregnant women with the lowest quintile of serum ALP levels (≤116 U/L) in late pregnancy had higher risk of VTE postpartum compared with those with the highest quintile (≥199 U/L) (OR, 2.83 [1.32, 6.05]). After adjusting for covariates of demographic, life style, birth outcomes, and other liver enzymes, pregnant women with the lowest quintile of serum ALP levels (≤116 U/L) in late pregnancy had increased risk of VTE postpartum compared with those with the highest quintile (≥199 U/L) (OR, 2.48 [1.14, 5.40]). A one standard deviation decrease of ln-transformed ALP levels were associated with elevated risk of VTE postpartum (OR, 1.29 [1.02, 1.62]). Significant negative associations of ALP with VTE were found in the unadjusted and adjusted models. The negative associations between ALP and VTE remained consistent in sensitivity analyses among participants with non-GDM, single pregnancy, non-preeclampsia, non-postpartum hemorrhage, non-extremely/very preterm and cesarean delivery. Decreased serum ALP levels significantly (P < 0.05) related to decreased hemoglobin, which was significantly (P < 0.05) related to increased risk of VTE postpartum. Decreased hemoglobin significantly (P < 0.05) mediated 7.59% of ALP-associated VTE postpartum. Interpretation This study suggested that low serum ALP levels in late pregnancy were associated with increased risk of VTE postpartum, and the ALP-associated VTE risk may be partially mediated by hemoglobin, suggesting that serum ALP in late pregnancy could be a promising biomarker for the prediction of VTE postpartum. Funding The National Natural Science Foundation of China, and the Program for HUST Academic Frontier Youth Team.
Collapse
Affiliation(s)
- Qian Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongfei Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huafang Wang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jun Deng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhipeng Cheng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenyi Lin
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ruiqi Zhu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shi Chen
- Department of Biobank, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jinrong Guo
- Department of Medical Records Management and Statistics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Liang V. Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
6
|
Lang X, Liu J, Zhang G, Feng X, Dan W. Knowledge Mapping of Drug Repositioning's Theme and Development. Drug Des Devel Ther 2023; 17:1157-1174. [PMID: 37096060 PMCID: PMC10122475 DOI: 10.2147/dddt.s405906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 04/11/2023] [Indexed: 04/26/2023] Open
Abstract
Background In recent years, the emergence of new diseases and resistance to known diseases have led to increasing demand for new drugs. By means of bibliometric analysis, this paper studied the relevant articles on drug repositioning in recent years and analyzed the current research foci and trends. Methodology The Web of Science database was searched to collect all relevant literature on drug repositioning from 2001 to 2022. These data were imported into CiteSpace and bibliometric online analysis platforms for bibliometric analysis. The processed data and visualized images predict the development trends in the research field. Results The quality and quantity of articles published after 2011 have improved significantly, with 45 of them cited more than 100 times. Articles posted by journals from different countries have high citation values. Authors from other institutions have also collaborated to analyze drug rediscovery. Keywords found in the literature include molecular docking (N=223), virtual screening (N=170), drug discovery (N=126), machine learning (N=125), and drug-target interaction (N=68); these words represent the core content of drug repositioning. Conclusion The key focus of drug research and development is related to the discovery of new indications for drugs. Researchers are starting to retarget drugs after analyzing online databases and clinical trials. More and more drugs are being targeted at other diseases to treat more patients, based on saving money and time. It is worth noting that researchers need more financial and technical support to complete drug development.
Collapse
Affiliation(s)
- Xiaona Lang
- Pharmacy Department, Tianjin Hospital, Tianjin, People’s Republic of China
| | - Jinlei Liu
- Cardiology Department, Guang ‘anmen Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, People’s Republic of China
| | - Guangzhong Zhang
- Dermatological Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Xin Feng
- Pharmacy Department, Tianjin Hospital, Tianjin, People’s Republic of China
| | - Wenchao Dan
- Dermatological Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, People’s Republic of China
- Correspondence: Wenchao Dan, Dermatological Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, People’s Republic of China, Tel +86 13652001152, Email
| |
Collapse
|
7
|
Causal deep learning reveals the comparative effectiveness of antihyperglycemic treatments in poorly controlled diabetes. Nat Commun 2022; 13:6921. [PMID: 36376286 PMCID: PMC9663714 DOI: 10.1038/s41467-022-33732-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/30/2022] [Indexed: 11/16/2022] Open
Abstract
Type-2 diabetes is associated with severe health outcomes, the effects of which are responsible for approximately 1/4th of the total healthcare spending in the United States (US). Current treatment guidelines endorse a massive number of potential anti-hyperglycemic treatment options in various combinations. Strategies for optimizing treatment selection are lacking. Real-world data from a nationwide population of over one million high-risk diabetic patients (HbA1c ≥ 9%) in the US is analyzed to evaluate the comparative effectiveness for HbA1c reduction in this population of more than 80 different treatment strategies ranging from monotherapy up to combinations of five concomitant classes of drugs across each of 10 clinical cohorts defined by age, insulin dependence, and a number of other chronic conditions. A causal deep learning approach developed on such data allows for more personalized evaluation of treatment selection. An average confounder-adjusted reduction in HbA1c of 0.69% [-0.75, -0.65] is observed between patients receiving high vs low ranked treatments across cohorts for which the difference was significant. This method can be extended to explore treatment optimization for other chronic conditions.
Collapse
|
8
|
Paik H, Lee J, Jeong CS, Park JS, Lee JH, Rappoport N, Kim Y, Sohn HY, Jo C, Kim J, Cho SB. Identification of a pleiotropic effect of ADIPOQ on cardiac dysfunction and Alzheimer's disease based on genetic evidence and health care records. Transl Psychiatry 2022; 12:389. [PMID: 36114174 PMCID: PMC9481623 DOI: 10.1038/s41398-022-02144-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 08/21/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
Observations of comorbidity in heart diseases, including cardiac dysfunction (CD) are increasing, including and cognitive impairment, such as Alzheimer's disease and dementia (AD/D). This comorbidity might be due to a pleiotropic effect of genetic variants shared between CD and AD/D. Here, we validated comorbidity of CD and AD/D based on diagnostic records from millions of patients in Korea and the University of California, San Francisco Medical Center (odds ratio 11.5 [8.5-15.5, 95% Confidence Interval (CI)]). By integrating a comprehensive human disease-SNP association database (VARIMED, VARiants Informing MEDicine) and whole-exome sequencing of 50 brains from individuals with and without Alzheimer's disease (AD), we identified missense variants in coding regions including APOB, a known risk factor for CD and AD/D, which potentially have a pleiotropic role in both diseases. Of the identified variants, site-directed mutation of ADIPOQ (268 G > A; Gly90Ser) in neurons produced abnormal aggregation of tau proteins (p = 0.02), suggesting a functional impact for AD/D. The association of CD and ADIPOQ variants was confirmed based on domain deletion in cardiac cells. Using the UK Biobank including data from over 500000 individuals, we examined a pleiotropic effect of the ADIPOQ variant by comparing CD- and AD/D-associated phenotypic evidence, including cardiac hypertrophy and cognitive degeneration. These results indicate that convergence of health care records and genetic evidences may help to dissect the molecular underpinnings of heart disease and associated cognitive impairment, and could potentially serve a prognostic function. Validation of disease-disease associations through health care records and genomic evidence can determine whether health conditions share risk factors based on pleiotropy.
Collapse
Affiliation(s)
- Hyojung Paik
- Division of Supercomputing, Korea Institute of Science and Technology Information, Daejeon, 34141, Republic of Korea
- Bakar Computational Health Sciences Institute, University of California San Francisco, 550 16th Street, San Francisco, CA, 94143, USA
- Department of Pediatrics, School of Medicine, University of California San Francisco, 550 16th Street, San Francisco, CA, 94143, USA
- Department of Data and HPC Science, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Junehawk Lee
- Division of Supercomputing, Korea Institute of Science and Technology Information, Daejeon, 34141, Republic of Korea
| | - Chan-Seok Jeong
- Division of Supercomputing, Korea Institute of Science and Technology Information, Daejeon, 34141, Republic of Korea
| | - Jun Sung Park
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jeong Ho Lee
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Nadav Rappoport
- Bakar Computational Health Sciences Institute, University of California San Francisco, 550 16th Street, San Francisco, CA, 94143, USA
- Departement of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beersheba, 8410501, Israel
| | - Younghoon Kim
- Division of Supercomputing, Korea Institute of Science and Technology Information, Daejeon, 34141, Republic of Korea
| | - Hee-Young Sohn
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Chungcheongbuk-do, 28159, Republic of Korea
| | - Chulman Jo
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Chungcheongbuk-do, 28159, Republic of Korea
| | - Jimin Kim
- Division of Supercomputing, Korea Institute of Science and Technology Information, Daejeon, 34141, Republic of Korea
| | - Seong Beom Cho
- Department of Bio-Medical Informatics, Gachon University, College of Medicine, Incheon, 21565, Republic of Korea.
| |
Collapse
|
9
|
Jamal QMS, Alharbi AH. Molecular docking and dynamics studies of cigarette smoke carcinogens interacting with acetylcholinesterase and butyrylcholinesterase enzymes of the central nervous system. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:61972-61992. [PMID: 34382170 DOI: 10.1007/s11356-021-15269-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 06/28/2021] [Indexed: 06/13/2023]
Abstract
The free radicals produced by cigarette smoking are responsible for tissue damage, heart and lung diseases, and carcinogenesis. The effect of tobacco on the central nervous system (CNS) has received increased attention nowadays in research. Therefore, to explore the molecular interaction of cigarette smoke carcinogens (CSC) 4-(methylnitrosamine)-1-(3-pyridyl)-1-butanol (NNAL), 4-(methylnitrosamine)-1-(3-pyridyl)-1-butanone (NNK), and N'-nitrosonornicotine (NNN) with well-known targets of CNS-related disorders, acetylcholinesterase (AChE), and butyrylcholinesterase (BuChE) enzymes, a cascade of the computational study was conducted including molecular docking and molecular dynamics simulations (MDS). The investigated results of NNAL+AChEcomplex, NNK+AChEcomplex, and NNK+BuChEcomplex based on intermolecular energies (∆G) were found to -8.57 kcal/mol, -8.21 kcal/mol, and -8.08 kcal/mol, respectively. MDS deviation and fluctuation plots of the NNAL and NNK interaction with AChE and BuChE have shown significant results. Further, Molecular Mechanics Poisson-Boltzmann Surface Area (MM-PBSA) results shown the best total binding energy (Binding∆G) -87.381 (+/-13.119) kJ/mol during NNK interaction with AChE. Our study suggests that CSC is well capable of altering the normal biomolecular mechanism of CNS; thus, obtained data could be useful to design extensive wet laboratory experimentation to know the effects of CSC on human CNS.
Collapse
Affiliation(s)
- Qazi Mohammad Sajid Jamal
- Department of Health Informatics, College of Public Health and Health Informatics, Qassim University, Al Bukayriyah, Saudi Arabia.
| | - Ali H Alharbi
- Department of Health Informatics, College of Public Health and Health Informatics, Qassim University, Al Bukayriyah, Saudi Arabia
| |
Collapse
|
10
|
Besson C, Moore A, Wu W, Vajdic CM, de Sanjose S, Camp NJ, Smedby KE, Shanafelt TD, Morton LM, Brewer JD, Zablotska L, Engels EA, Cerhan JR, Slager SL, Han J, Berndt SI. Common genetic polymorphisms contribute to the association between chronic lymphocytic leukaemia and non-melanoma skin cancer. Int J Epidemiol 2021; 50:1325-1334. [PMID: 33748835 DOI: 10.1093/ije/dyab042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2021] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Epidemiological studies have demonstrated a positive association between chronic lymphocytic leukaemia (CLL) and non-melanoma skin cancer (NMSC). We hypothesized that shared genetic risk factors between CLL and NMSC could contribute to the association observed between these diseases. METHODS We examined the association between (i) established NMSC susceptibility loci and CLL risk in a meta-analysis including 3100 CLL cases and 7667 controls and (ii) established CLL loci and NMSC risk in a study of 4242 basal cell carcinoma (BCC) cases, 825 squamous cell carcinoma (SCC) cases and 12802 controls. Polygenic risk scores (PRS) for CLL, BCC and SCC were constructed using established loci. Logistic regression was used to estimate odds ratios (ORs) and 95% confidence intervals (CIs). RESULTS Higher CLL-PRS was associated with increased BCC risk (OR4th-quartile-vs-1st-quartile = 1.13, 95% CI: 1.02-1.24, Ptrend = 0.009), even after removing the shared 6p25.3 locus. No association was observed with BCC-PRS and CLL risk (Ptrend = 0.68). These findings support a contributory role for CLL in BCC risk, but not for BCC in CLL risk. Increased CLL risk was observed with higher SCC-PRS (OR4th-quartile-vs-1st-quartile = 1.22, 95% CI: 1.08-1.38, Ptrend = 1.36 × 10-5), which was driven by shared genetic susceptibility at the 6p25.3 locus. CONCLUSION These findings highlight the role of pleiotropy regarding the pathogenesis of CLL and NMSC and shows that a single pleiotropic locus, 6p25.3, drives the observed association between genetic susceptibility to SCC and increased CLL risk. The study also provides evidence that genetic susceptibility for CLL increases BCC risk.
Collapse
Affiliation(s)
- Caroline Besson
- Service d'hématologie et Oncologie, Centre Hospitalier de Versailles, Le Chesnay; Université Paris-Saclay, UVSQ, Inserm, Équipe "Exposome et Hérédité", CESP, 94805, Villejuif, France
| | - Amy Moore
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Wenting Wu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, USA
| | - Claire M Vajdic
- Centre for Big Data Research in Health, University of New South Wales, Sydney, New South Wales, Australia
| | | | - Nicola J Camp
- Department of Internal Medicine and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Karin E Smedby
- Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Tait D Shanafelt
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Lindsay M Morton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jerry D Brewer
- Department of Dermatology, Mayo Clinic, Rochester, MN, USA
| | - Lydia Zablotska
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Eric A Engels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - James R Cerhan
- Service d'hématologie et Oncologie, Centre Hospitalier de Versailles, Le Chesnay; Université Paris-Saclay, UVSQ, Inserm, Équipe "Exposome et Hérédité", CESP, 94805, Villejuif, France
| | - Susan L Slager
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Jiali Han
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | | |
Collapse
|
11
|
Jabalameli N, Rajabi F, Firooz A, Rezaei N. The Overlap between Genetic Susceptibility to COVID-19 and Skin Diseases. Immunol Invest 2021; 51:1087-1094. [PMID: 33494631 DOI: 10.1080/08820139.2021.1876086] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Navid Jabalameli
- Network of Dermatology Research (NDR), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Fateme Rajabi
- Network of Dermatology Research (NDR), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Center for Research & Training in Skin Diseases & Leprosy, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Firooz
- Center for Research & Training in Skin Diseases & Leprosy, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Sheffield, UK
| |
Collapse
|
12
|
Melamed RD. Using indication embeddings to represent patient health for drug safety studies. JAMIA Open 2020; 3:422-430. [PMID: 33376961 PMCID: PMC7751136 DOI: 10.1093/jamiaopen/ooaa040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/11/2020] [Indexed: 11/12/2022] Open
Abstract
Objective The electronic health record is a rising resource for quantifying medical practice, discovering the adverse effects of drugs, and studying comparative effectiveness. One of the challenges of applying these methods to health care data is the high dimensionality of the health record. Methods to discover the effects of drugs in health data must account for tens of thousands of potentially relevant confounders. Our goal in this work is to reduce the dimensionality of the health data with the aim of accelerating the application of retrospective cohort studies to this data. Materials and methods Here, we develop indication embeddings, a way to reduce the dimensionality of health data while capturing information relevant to treatment decisions. We evaluate these embeddings using external data on drug indications. Then, we use the embeddings as a substitute for medical history to match patients and develop evaluation metrics for these matches. Results We demonstrate that these embeddings recover the therapeutic uses of drugs. We use embeddings as an informative representation of relationships between drugs, between health history events and drug prescriptions, and between patients at a particular time in their health history. We show that using embeddings to match cohorts improves the balance of the cohorts, even in terms of poorly measured risk factors like smoking. Discussion and conclusion Unlike other embeddings inspired by word2vec, indication embeddings are specifically designed to capture the medical history leading to the prescription of a new drug. For retrospective cohort studies, our low-dimensional representation helps in finding comparator drugs and constructing comparator cohorts.
Collapse
Affiliation(s)
- Rachel D Melamed
- Department of Biological Sciences, University of Massachusetts, Lowell, 198 Riverside St, Lowell, Massachusetts, USA.,Lowell Department of Medicine, University of Chicago, 900 E 57 St, Chicago, Illinois, USA
| |
Collapse
|
13
|
Garcin LM, Gelot A, Gomez RR, Gusto G, Boutron-Ruault MC, Kvaskoff M, Severi G, Besson C. Pigmentary traits, sun exposure, and risk of non-Hodgkin's lymphoma/chronic lymphocytic leukemia: A study within the French E3N prospective cohort. Cancer Med 2020; 10:297-304. [PMID: 33219744 PMCID: PMC7826467 DOI: 10.1002/cam4.3586] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/09/2020] [Accepted: 10/14/2020] [Indexed: 11/13/2022] Open
Abstract
To investigate whether risk factors for keratinocyte carcinomas (KCs), namely pigmentary traits and sun exposure, are associated with risk of non‐Hodgkin's lymphomas (NHL) and chronic lymphocytic leukemia (CLL). E3N is a prospective cohort of French women aged 40–65 years at inclusion in 1990. Cancer data were collected at baseline and updated every 2–3 years. Hazard Ratios (HRs) and 95% confidence intervals (CIs) for associations between pigmentary traits and sun exposure, and risk of CLL/NHL were estimated using Cox models. With a median follow‐up of 24 years, 622 incident cases of CLL/NHL were ascertained among the 92,097 included women. The presence of nevi was associated with CLL/NHL risk: HR for “many or very many nevi” relative to “no nevi”: 1.56 [1.15; 2.11]. Such association with number of nevi appears to be mostly limited to risk of CLL: HR for “many or very many nevi”: 3.00 [1.38; 6.52]; versus 1.32 [0.94; 1.84] for NHL. Women whose skin was highly sensitive to sunburn also had a higher risk of CLL: HR = 1.96 [1.21; 3.18], while no increase in risk of NHL was observed. Skin or hair color, number of freckles, and average daily ultraviolet (UV) dose during spring and summer in location of residence at birth or at inclusion (kJ/m2) were not associated with CLL/NHL risk. Some pigmentary traits (presence of nevi and skin sensitivity), but not sun exposure, were associated with CLL/NHL. These observations suggest that CLL may share some constitutional risk factors with keratinocyte cancers.
Collapse
Affiliation(s)
- Louis-Marie Garcin
- Department of Medical Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France.,Hematology-Oncology Department, Centre Hospitalier de Versailles, Le Chesnay, France
| | - Amandine Gelot
- CESP, Université Paris-Saclay, Univ. Paris-Sud, UVSQ, INSERM, Villejuif, France.,Gustave Roussy, Villejuif, France
| | - Roselyn-Rima Gomez
- CESP, Université Paris-Saclay, Univ. Paris-Sud, UVSQ, INSERM, Villejuif, France.,Gustave Roussy, Villejuif, France
| | - Gaëlle Gusto
- CESP, Université Paris-Saclay, Univ. Paris-Sud, UVSQ, INSERM, Villejuif, France.,Gustave Roussy, Villejuif, France
| | | | - Marina Kvaskoff
- CESP, Université Paris-Saclay, Univ. Paris-Sud, UVSQ, INSERM, Villejuif, France.,Gustave Roussy, Villejuif, France
| | - Gianluca Severi
- CESP, Université Paris-Saclay, Univ. Paris-Sud, UVSQ, INSERM, Villejuif, France.,Gustave Roussy, Villejuif, France.,Departement of Statistics, Computer Science and Applications (DISIA), University of Florence, Florence, Italy
| | - Caroline Besson
- Hematology-Oncology Department, Centre Hospitalier de Versailles, Le Chesnay, France.,CESP, Université Paris-Saclay, Univ. Paris-Sud, UVSQ, INSERM, Villejuif, France
| |
Collapse
|
14
|
Faulkner E, Holtorf AP, Walton S, Liu CY, Lin H, Biltaj E, Brixner D, Barr C, Oberg J, Shandhu G, Siebert U, Snyder SR, Tiwana S, Watkins J, IJzerman MJ, Payne K. Being Precise About Precision Medicine: What Should Value Frameworks Incorporate to Address Precision Medicine? A Report of the Personalized Precision Medicine Special Interest Group. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2020; 23:529-539. [PMID: 32389217 DOI: 10.1016/j.jval.2019.11.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 11/18/2019] [Accepted: 11/25/2019] [Indexed: 06/11/2023]
Abstract
Precision medicine is a dynamic area embracing a diverse and increasing type of approaches that allow the targeting of new medicines, screening programs or preventive healthcare strategies, which include the use of biologic markers or complex tests driven by algorithms also potentially taking account of patient preferences. The International Society for Pharmacoeconomics and Outcome Research expanded its current work around precision medicine to (1) describe the evolving paradigm of precision medicine with examples of current and evolving applications, (2) describe key stakeholders perspectives on the value of precision medicine in their respective domains, and (3) define the core factors that should be considered in a value assessment framework for precision medicine. With the ultimate goal of improving health of well-defined patient groups, precision medicine will affect all stakeholders in the healthcare system at multiple levels spanning the individual perspective to the societal perspective. For an efficient, timely and practical precision medicine value assessment framework, it will be important to address these multiple perspectives through building consensus among the stakeholders for robust procedures and measures of value aspects, including performance of precision mechanism; aligned reimbursement processes of precision mechanism and subsequent treatment; transparent expectations for evidence requirements and study designs adequately matched to the intended use of the precision mechanism and to the smaller target patient populations; recognizing the potential range of value-generation such as ruling-in and ruling-out decisions.
Collapse
Affiliation(s)
- Eric Faulkner
- Evidera, Bethesda, MD, USA; University of North Carolina at Chapel Hill, Chapel Hill, NC; National Association of Managed Care Physicians, Glen Allen, VA, USA.
| | | | - Surrey Walton
- University of Illinois at Chicago, Chicago, IL, USA; Second City Outcomes Research, LLC, Chicago, IL, USA
| | | | - Hwee Lin
- National University of Singapore, Singapore
| | | | | | | | | | | | - Uwe Siebert
- University for Health Sciences, Medical Informatics, and Technology, Hall in Tirol, Austria; Harvard School of Public Health and Harvard Medical School, Boston, MA, USA; ONCOTYROL Center for Personalized Cancer Medicine, Innsbruck, Austria
| | | | | | | | - Maarten J IJzerman
- University of Melbourne Centre for Cancer Research, Parkville, Australia
| | | |
Collapse
|
15
|
Kosnik MB, Reif DM. Determination of chemical-disease risk values to prioritize connections between environmental factors, genetic variants, and human diseases. Toxicol Appl Pharmacol 2019; 379:114674. [PMID: 31323264 PMCID: PMC6708494 DOI: 10.1016/j.taap.2019.114674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/05/2019] [Accepted: 07/15/2019] [Indexed: 12/18/2022]
Abstract
Traditional methods for chemical risk assessment are too time-consuming and resource-intensive to characterize either the diversity of chemicals to which humans are exposed or how that diversity may manifest in population susceptibility differences. The advent of novel toxicological data sources and their integration with bioinformatic databases affords opportunities for modern approaches that consider gene-environment (GxE) interactions in population risk assessment. Here, we present an approach that systematically links multiple data sources to relate chemical risk values to diseases and gene-disease variants. These data sources include high-throughput screening (HTS) results from Tox21/ToxCast, chemical-disease relationships from the Comparative Toxicogenomics Database (CTD), hazard data from resources like the Integrated Risk Information System, exposure data from the ExpoCast initiative, and gene-variant-disease information from the DisGeNET database. We use these integrated data to identify variants implicated in chemical-disease enrichments and develop a new value that estimates the risk of these associations toward differential population responses. Finally, we use this value to prioritize chemical-disease associations by exploring the genomic distribution of variants implicated in high-risk diseases. We offer this modular approach, termed DisQGOS (Disease Quotient Genetic Overview Score), for relating overall chemical-disease risk to potential for population variable responses, as a complement to methods aiming to modernize aspects of risk assessment.
Collapse
Affiliation(s)
- Marissa B Kosnik
- Toxicology Program, North Carolina State University, Raleigh, NC 27695-7617, United States of America; Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27695-7617, United States of America; Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7617, United States of America.
| | - David M Reif
- Toxicology Program, North Carolina State University, Raleigh, NC 27695-7617, United States of America; Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27695-7617, United States of America; Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7617, United States of America; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695-7617, United States of America.
| |
Collapse
|
16
|
Li H, Fan J, Vitali F, Berghout J, Aberasturi D, Li J, Wilson L, Chiu W, Pumarejo M, Han J, Kenost C, Koripella PC, Pouladi N, Billheimer D, Bedrick EJ, Lussier YA. Novel disease syndromes unveiled by integrative multiscale network analysis of diseases sharing molecular effectors and comorbidities. BMC Med Genomics 2018; 11:112. [PMID: 30598089 PMCID: PMC6311938 DOI: 10.1186/s12920-018-0428-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Background Forty-two percent of patients experience disease comorbidity, contributing substantially to mortality rates and increased healthcare costs. Yet, the possibility of underlying shared mechanisms for diseases remains not well established, and few studies have confirmed their molecular predictions with clinical datasets. Methods In this work, we integrated genome-wide association study (GWAS) associating diseases and single nucleotide polymorphisms (SNPs) with transcript regulatory activity from expression quantitative trait loci (eQTL). This allowed novel mechanistic insights for noncoding and intergenic regions. We then analyzed pairs of SNPs across diseases to identify shared molecular effectors robust to multiple test correction (False Discovery Rate FDReRNA < 0.05). We hypothesized that disease pairs found to be molecularly convergent would also be significantly overrepresented among comorbidities in clinical datasets. To assess our hypothesis, we used clinical claims datasets from the Healthcare Cost and Utilization Project (HCUP) and calculated significant disease comorbidities (FDRcomorbidity < 0.05). We finally verified if disease pairs resulting molecularly convergent were also statistically comorbid more than by chance using the Fisher’s Exact Test. Results Our approach integrates: (i) 6175 SNPs associated with 238 diseases from ~ 1000 GWAS, (ii) eQTL associations from 19 tissues, and (iii) claims data for 35 million patients from HCUP. Logistic regression (controlled for age, gender, and race) identified comorbidities in HCUP, while enrichment analyses identified cis- and trans-eQTL downstream effectors of GWAS-identified variants. Among ~ 16,000 combinations of diseases, 398 disease-pairs were prioritized by both convergent eQTL-genetics (RNA overlap enrichment, FDReRNA < 0.05) and clinical comorbidities (OR > 1.5, FDRcomorbidity < 0.05). Case studies of comorbidities illustrate specific convergent noncoding regulatory elements. An intergenic architecture of disease comorbidity was unveiled due to GWAS and eQTL-derived convergent mechanisms between distinct diseases being overrepresented among observed comorbidities in clinical datasets (OR = 8.6, p-value = 6.4 × 10− 5 FET). Conclusions These comorbid diseases with convergent eQTL genetic mechanisms suggest clinical syndromes. While it took over a decade to confirm the genetic underpinning of the metabolic syndrome, this study is likely highlighting hundreds of new ones. Further, this knowledge may improve the clinical management of comorbidities with precision and shed light on novel approaches of drug repositioning or SNP-guided precision molecular therapy inclusive of intergenic risks. Electronic supplementary material The online version of this article (10.1186/s12920-018-0428-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Haiquan Li
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA. .,Department of Medicine at the College of Medicine-Tucson, The University of Arizona, Tucson, AZ, 85721, USA. .,Graduate Interdisciplinary Program in Statistics, The University of Arizona, Tucson, AZ, 85721, USA. .,Department of Biosystems Engineering, The University of Arizona, Tucson, AZ, 85721, USA.
| | - Jungwei Fan
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA.,Department of Medicine at the College of Medicine-Tucson, The University of Arizona, Tucson, AZ, 85721, USA
| | - Francesca Vitali
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA.,Department of Medicine at the College of Medicine-Tucson, The University of Arizona, Tucson, AZ, 85721, USA.,University of Arizona Health Sciences, The University of Arizona, Tucson, AZ, 85721, USA
| | - Joanne Berghout
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA.,Department of Medicine at the College of Medicine-Tucson, The University of Arizona, Tucson, AZ, 85721, USA.,The Center for Applied Genetics and Genomics Medicine, The University of Arizona, Tucson, AZ, 85721, USA.,The Center for Innovation in Brain Science, The University of Arizona, Tucson, AZ, 85721, USA
| | - Dillon Aberasturi
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA.,Department of Medicine at the College of Medicine-Tucson, The University of Arizona, Tucson, AZ, 85721, USA.,Graduate Interdisciplinary Program in Statistics, The University of Arizona, Tucson, AZ, 85721, USA
| | - Jianrong Li
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA.,Department of Medicine at the College of Medicine-Tucson, The University of Arizona, Tucson, AZ, 85721, USA.,University of Arizona Health Sciences, The University of Arizona, Tucson, AZ, 85721, USA
| | - Liam Wilson
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA
| | - Wesley Chiu
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA
| | - Minsu Pumarejo
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA
| | - Jiali Han
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA.,Department of Medicine at the College of Medicine-Tucson, The University of Arizona, Tucson, AZ, 85721, USA.,Department of Systems & Industrial Engineering, The University of Arizona, Tucson, AZ, 85721, USA
| | - Colleen Kenost
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA.,Department of Medicine at the College of Medicine-Tucson, The University of Arizona, Tucson, AZ, 85721, USA
| | - Pradeep C Koripella
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA.,Department of Medicine at the College of Medicine-Tucson, The University of Arizona, Tucson, AZ, 85721, USA
| | - Nima Pouladi
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA.,Department of Medicine at the College of Medicine-Tucson, The University of Arizona, Tucson, AZ, 85721, USA
| | - Dean Billheimer
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA.,Graduate Interdisciplinary Program in Statistics, The University of Arizona, Tucson, AZ, 85721, USA.,University of Arizona Health Sciences, The University of Arizona, Tucson, AZ, 85721, USA.,Epidemiology and Biostatistics Department, College of Public Health, The University of Arizona, Tucson, AZ, 85721, USA
| | - Edward J Bedrick
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA.,Graduate Interdisciplinary Program in Statistics, The University of Arizona, Tucson, AZ, 85721, USA.,University of Arizona Health Sciences, The University of Arizona, Tucson, AZ, 85721, USA.,Epidemiology and Biostatistics Department, College of Public Health, The University of Arizona, Tucson, AZ, 85721, USA
| | - Yves A Lussier
- Center for Biomedical Informatics and Biostatistics, The University of Arizona, Tucson, AZ, 85721, USA. .,Department of Medicine at the College of Medicine-Tucson, The University of Arizona, Tucson, AZ, 85721, USA. .,Graduate Interdisciplinary Program in Statistics, The University of Arizona, Tucson, AZ, 85721, USA. .,The Center for Applied Genetics and Genomics Medicine, The University of Arizona, Tucson, AZ, 85721, USA. .,The Center for Innovation in Brain Science, The University of Arizona, Tucson, AZ, 85721, USA. .,UA Cancer Center, The University of Arizona, Tucson, AZ, 85721, USA. .,University of Arizona Health Sciences, The University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
17
|
Shameer K, Glicksberg BS, Hodos R, Johnson KW, Badgeley MA, Readhead B, Tomlinson MS, O’Connor T, Miotto R, Kidd BA, Chen R, Ma’ayan A, Dudley JT. Systematic analyses of drugs and disease indications in RepurposeDB reveal pharmacological, biological and epidemiological factors influencing drug repositioning. Brief Bioinform 2018; 19:656-678. [PMID: 28200013 PMCID: PMC6192146 DOI: 10.1093/bib/bbw136] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 11/29/2016] [Indexed: 12/22/2022] Open
Abstract
Increase in global population and growing disease burden due to the emergence of infectious diseases (Zika virus), multidrug-resistant pathogens, drug-resistant cancers (cisplatin-resistant ovarian cancer) and chronic diseases (arterial hypertension) necessitate effective therapies to improve health outcomes. However, the rapid increase in drug development cost demands innovative and sustainable drug discovery approaches. Drug repositioning, the discovery of new or improved therapies by reevaluation of approved or investigational compounds, solves a significant gap in the public health setting and improves the productivity of drug development. As the number of drug repurposing investigations increases, a new opportunity has emerged to understand factors driving drug repositioning through systematic analyses of drugs, drug targets and associated disease indications. However, such analyses have so far been hampered by the lack of a centralized knowledgebase, benchmarking data sets and reporting standards. To address these knowledge and clinical needs, here, we present RepurposeDB, a collection of repurposed drugs, drug targets and diseases, which was assembled, indexed and annotated from public data. RepurposeDB combines information on 253 drugs [small molecules (74.30%) and protein drugs (25.29%)] and 1125 diseases. Using RepurposeDB data, we identified pharmacological (chemical descriptors, physicochemical features and absorption, distribution, metabolism, excretion and toxicity properties), biological (protein domains, functional process, molecular mechanisms and pathway cross talks) and epidemiological (shared genetic architectures, disease comorbidities and clinical phenotype similarities) factors mediating drug repositioning. Collectively, RepurposeDB is developed as the reference database for drug repositioning investigations. The pharmacological, biological and epidemiological principles of drug repositioning identified from the meta-analyses could augment therapeutic development.
Collapse
Affiliation(s)
- Khader Shameer
- Institute of Next Generation Healthcare, Mount Sinai Health System, New York,
NY, USA
| | - Benjamin S Glicksberg
- Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York,
NY, USA
| | - Rachel Hodos
- Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York,
NY, USA
- New York University, New York, NY, USA
| | - Kipp W Johnson
- Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York,
NY, USA
| | - Marcus A Badgeley
- Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York,
NY, USA
| | - Ben Readhead
- Institute of Next Generation Healthcare, Mount Sinai Health System, New York,
NY, USA
| | - Max S Tomlinson
- Institute of Next Generation Healthcare, Mount Sinai Health System, New York,
NY, USA
| | | | - Riccardo Miotto
- Institute of Next Generation Healthcare, Mount Sinai Health System, New York,
NY, USA
| | - Brian A Kidd
- Institute of Next Generation Healthcare, Mount Sinai Health System, New York,
NY, USA
| | - Rong Chen
- Clinical Genome Informatics, Icahn Institute of Genetics and Multiscale
Biology, Mount Sinai Health System, New York, NY
| | - Avi Ma’ayan
- Mount Sinai Center for Bioinformatics, Mount Sinai Health System, New York,
NY
| | - Joel T Dudley
- Institute of Next Generation Healthcare, Mount Sinai Health System, New York,
NY, USA
- Department of Genetics and Genomic Sciences, Mount Sinai Health System, New
York, NY, USA
- Department of Population Health Science and Policy, Mount Sinai Health System,
New York, NY, USA
- Director of Biomedical Informatics, Icahn School of Medicine at Mount Sinai,
Mount Sinai Health System, New York, NY
| |
Collapse
|
18
|
Ohn JH. The landscape of genetic susceptibility correlations among diseases and traits. J Am Med Inform Assoc 2018; 24:921-926. [PMID: 28371808 DOI: 10.1093/jamia/ocx026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 03/01/2017] [Indexed: 11/12/2022] Open
Abstract
Objective The aim of the study was to comprehensively explore the genetic susceptibility correlations among diseases and traits from large-scale individual genotype data. Materials and Methods Based on a knowledge base of genetic variants significantly (P < 5 × 10 -8 ) linked with human phenotypes, genetic risk scores (GRSs) of diseases or traits were calculated for 2504 individuals with whole-genome sequencing data from the 1000 Genomes Project. Associations between diseases/traits were statistically evaluated by pairwise correlation analysis of GRSs. Overlaps between the genetic susceptibility correlations and disease comorbidity associations from hospital claims data in more than 30 million patients in United States were assessed. Results Correlation analysis of GRSs revealed 823 significant correlations among 78 diseases and 89 traits (false discovery rate adjusted P -value or Q -value < 0.01). It is noticeable that GRSs were correlated in 464 associations (56.4%) even if they were combinations of distinct sets of risk variants without chromosomal linkage, suggesting the presence of genetic interactions beyond chromosome position. When 312 significant genetic susceptibility correlations between diseases were compared to nationwide disease comorbidity correlations obtained from data from 32 million Medicare claims in the United States, 108 overlaps (34.6%) were found that had both genetic susceptibility and epidemiologic comorbid correlations. Conclusion The study suggests that common genetic background exists between diseases and traits with epidemiologic associations. The GRS correlation approach provides a rich source of candidate associations among diseases and traits from the genetic perspective, warranting further epidemiologic studies.
Collapse
Affiliation(s)
- Jung Hun Ohn
- Division of General Internal Medicine, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| |
Collapse
|
19
|
Chesmore K, Bartlett J, Williams SM. The ubiquity of pleiotropy in human disease. Hum Genet 2017; 137:39-44. [DOI: 10.1007/s00439-017-1854-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 11/14/2017] [Indexed: 02/07/2023]
|
20
|
Glicksberg BS, Li L, Badgeley MA, Shameer K, Kosoy R, Beckmann ND, Pho N, Hakenberg J, Ma M, Ayers KL, Hoffman GE, Dan Li S, Schadt EE, Patel CJ, Chen R, Dudley JT. Comparative analyses of population-scale phenomic data in electronic medical records reveal race-specific disease networks. Bioinformatics 2017; 32:i101-i110. [PMID: 27307606 PMCID: PMC4908366 DOI: 10.1093/bioinformatics/btw282] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Motivation: Underrepresentation of racial groups represents an important challenge and major gap in phenomics research. Most of the current human phenomics research is based primarily on European populations; hence it is an important challenge to expand it to consider other population groups. One approach is to utilize data from EMR databases that contain patient data from diverse demographics and ancestries. The implications of this racial underrepresentation of data can be profound regarding effects on the healthcare delivery and actionability. To the best of our knowledge, our work is the first attempt to perform comparative, population-scale analyses of disease networks across three different populations, namely Caucasian (EA), African American (AA) and Hispanic/Latino (HL). Results: We compared susceptibility profiles and temporal connectivity patterns for 1988 diseases and 37 282 disease pairs represented in a clinical population of 1 025 573 patients. Accordingly, we revealed appreciable differences in disease susceptibility, temporal patterns, network structure and underlying disease connections between EA, AA and HL populations. We found 2158 significantly comorbid diseases for the EA cohort, 3265 for AA and 672 for HL. We further outlined key disease pair associations unique to each population as well as categorical enrichments of these pairs. Finally, we identified 51 key ‘hub’ diseases that are the focal points in the race-centric networks and of particular clinical importance. Incorporating race-specific disease comorbidity patterns will produce a more accurate and complete picture of the disease landscape overall and could support more precise understanding of disease relationships and patient management towards improved clinical outcomes. Contacts: rong.chen@mssm.edu or joel.dudley@mssm.edu Supplementary information:Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Benjamin S Glicksberg
- Department of Genetics and Genomic Sciences Icahn Institute for Genomics and Multiscale Biology Harris Center for Precision Wellness, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA
| | - Li Li
- Department of Genetics and Genomic Sciences Icahn Institute for Genomics and Multiscale Biology Harris Center for Precision Wellness, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA
| | - Marcus A Badgeley
- Department of Genetics and Genomic Sciences Icahn Institute for Genomics and Multiscale Biology Harris Center for Precision Wellness, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA
| | - Khader Shameer
- Department of Genetics and Genomic Sciences Icahn Institute for Genomics and Multiscale Biology Harris Center for Precision Wellness, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA
| | - Roman Kosoy
- Department of Genetics and Genomic Sciences Icahn Institute for Genomics and Multiscale Biology Harris Center for Precision Wellness, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA
| | - Noam D Beckmann
- Department of Genetics and Genomic Sciences Icahn Institute for Genomics and Multiscale Biology
| | - Nam Pho
- Department of Biomedical Informatics, Harvard Medical School, Boston, 02115 MA, USA
| | - Jörg Hakenberg
- Department of Genetics and Genomic Sciences Icahn Institute for Genomics and Multiscale Biology
| | - Meng Ma
- Department of Genetics and Genomic Sciences Icahn Institute for Genomics and Multiscale Biology
| | - Kristin L Ayers
- Department of Genetics and Genomic Sciences Icahn Institute for Genomics and Multiscale Biology
| | - Gabriel E Hoffman
- Department of Genetics and Genomic Sciences Icahn Institute for Genomics and Multiscale Biology
| | - Shuyu Dan Li
- Department of Genetics and Genomic Sciences Icahn Institute for Genomics and Multiscale Biology
| | - Eric E Schadt
- Department of Genetics and Genomic Sciences Icahn Institute for Genomics and Multiscale Biology
| | - Chirag J Patel
- Department of Biomedical Informatics, Harvard Medical School, Boston, 02115 MA, USA
| | - Rong Chen
- Department of Genetics and Genomic Sciences Icahn Institute for Genomics and Multiscale Biology
| | - Joel T Dudley
- Department of Genetics and Genomic Sciences Icahn Institute for Genomics and Multiscale Biology Harris Center for Precision Wellness, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA Department of Population Health Science and Policy, New York City, NY 10029, USA
| |
Collapse
|
21
|
Clifton DA, Niehaus KE, Charlton P, Colopy GW. Health Informatics via Machine Learning for the Clinical Management of Patients. Yearb Med Inform 2017; 10:38-43. [PMID: 26293849 DOI: 10.15265/iy-2015-014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
OBJECTIVES To review how health informatics systems based on machine learning methods have impacted the clinical management of patients, by affecting clinical practice. METHODS We reviewed literature from 2010-2015 from databases such as Pubmed, IEEE xplore, and INSPEC, in which methods based on machine learning are likely to be reported. We bring together a broad body of literature, aiming to identify those leading examples of health informatics that have advanced the methodology of machine learning. While individual methods may have further examples that might be added, we have chosen some of the most representative, informative exemplars in each case. RESULTS Our survey highlights that, while much research is taking place in this high-profile field, examples of those that affect the clinical management of patients are seldom found. We show that substantial progress is being made in terms of methodology, often by data scientists working in close collaboration with clinical groups. CONCLUSIONS Health informatics systems based on machine learning are in their infancy and the translation of such systems into clinical management has yet to be performed at scale.
Collapse
Affiliation(s)
- D A Clifton
- David A. Clifton, Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK, E-mail:
| | | | | | | |
Collapse
|
22
|
Yu ACS, Li JW, Chan TF. Using genetics to inform new therapeutics for diabetes. Expert Rev Endocrinol Metab 2017; 12:159-169. [PMID: 30063460 DOI: 10.1080/17446651.2017.1323631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The genetic architecture of diabetes has been extensively studied. Numerous genetic markers for diabetes have been reported. However, the translation of such knowledge into clinical interventions has been inadequate. Areas covered: We performed a literature search on various frontiers in diabetes treatment that could be improved using genetic information: (1) understanding the mechanisms of existing antidiabetic drugs, (2) repurposing existing drugs for the treatment of diabetes, (3) complementing clinical trial findings; (4) finding novel treatment approaches; (5) better estimation of the efficacy of metabolic surgery. Expert commentary: The translation of genetic information to clinical intervention requires further study, including the development of an appropriate genetic risk score algorithm for type 2 diabetes. Genomic studies provide empirical explanations for clinical trial findings. Moreover, the mechanisms of antidiabetic drugs should be thoroughly investigated to enable clinical trials and pharmacogenomics studies of these drugs. As metabolic surgery becomes more prevalent for the treatment of diabetes, genetic approaches may improve patient prioritization.
Collapse
Affiliation(s)
- Allen Chi-Shing Yu
- a School of Life Sciences , The Chinese University of Hong Kong , Shatin , Hong Kong SAR
| | - Jing-Woei Li
- a School of Life Sciences , The Chinese University of Hong Kong , Shatin , Hong Kong SAR
- b Faculty of Medicine , The Chinese University of Hong Kong , Shatin , Hong Kong SAR
| | - Ting-Fung Chan
- a School of Life Sciences , The Chinese University of Hong Kong , Shatin , Hong Kong SAR
- c CUHK-BGI Innovation Institute of Transomics , The Chinese University of Hong Kong , Shatin , Hong Kong SAR
- d Hong Kong Institute of Diabetes and Obesity , The Chinese University of Hong Kong , Shatin , Hong Kong SAR
| |
Collapse
|
23
|
Shameer K, Badgeley MA, Miotto R, Glicksberg BS, Morgan JW, Dudley JT. Translational bioinformatics in the era of real-time biomedical, health care and wellness data streams. Brief Bioinform 2017; 18:105-124. [PMID: 26876889 PMCID: PMC5221424 DOI: 10.1093/bib/bbv118] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 11/27/2015] [Indexed: 01/01/2023] Open
Abstract
Monitoring and modeling biomedical, health care and wellness data from individuals and converging data on a population scale have tremendous potential to improve understanding of the transition to the healthy state of human physiology to disease setting. Wellness monitoring devices and companion software applications capable of generating alerts and sharing data with health care providers or social networks are now available. The accessibility and clinical utility of such data for disease or wellness research are currently limited. Designing methods for streaming data capture, real-time data aggregation, machine learning, predictive analytics and visualization solutions to integrate wellness or health monitoring data elements with the electronic medical records (EMRs) maintained by health care providers permits better utilization. Integration of population-scale biomedical, health care and wellness data would help to stratify patients for active health management and to understand clinically asymptomatic patients and underlying illness trajectories. In this article, we discuss various health-monitoring devices, their ability to capture the unique state of health represented in a patient and their application in individualized diagnostics, prognosis, clinical or wellness intervention. We also discuss examples of translational bioinformatics approaches to integrating patient-generated data with existing EMRs, personal health records, patient portals and clinical data repositories. Briefly, translational bioinformatics methods, tools and resources are at the center of these advances in implementing real-time biomedical and health care analytics in the clinical setting. Furthermore, these advances are poised to play a significant role in clinical decision-making and implementation of data-driven medicine and wellness care.
Collapse
Affiliation(s)
| | - Marcus A Badgeley
- Harris Center for Precision Wellness, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Riccardo Miotto
- Harris Center for Precision Wellness, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Benjamin S Glicksberg
- Harris Center for Precision Wellness, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Joseph W Morgan
- Harris Center for Precision Wellness, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Joel T Dudley
- Harris Center for Precision Wellness, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Health Evidence and Policy, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
24
|
Paik H, Chen B, Sirota M, Hadley D, Butte AJ. Integrating Clinical Phenotype and Gene Expression Data to Prioritize Novel Drug Uses. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2016; 5:599-607. [PMID: 27860440 PMCID: PMC5192994 DOI: 10.1002/psp4.12108] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 08/05/2016] [Indexed: 12/22/2022]
Abstract
Drug repositioning has been based largely on genomic signatures of drugs and diseases. One challenge in these efforts lies in connecting the molecular signatures of drugs into clinical responses, including therapeutic and side effects, to the repurpose of drugs. We addressed this challenge by evaluating drug‐drug relationships using a phenotypic and molecular‐based approach that integrates therapeutic indications, side effects, and gene expression profiles induced by each drug. Using cosine similarity, relationships between 445 drugs were evaluated based on high‐dimensional spaces consisting of phenotypic terms of drugs and genomic signatures, respectively. One hundred fifty‐one of 445 drugs comprising 450 drug pairs displayed significant similarities in both phenotypic and genomic signatures (P value < 0.05). We also found that similar gene expressions of drugs do indeed yield similar clinical phenotypes. We generated similarity matrixes of drugs using the expression profiles they induce in a cell line and phenotypic effects.
Collapse
Affiliation(s)
- H Paik
- Institute for Computational Health Sciences, School of Medicine, University of California San Francisco, San Francisco, California, USA.,Department of Pediatrics, School of Medicine, University of California - San Francisco, San Francisco, California, USA
| | - B Chen
- Institute for Computational Health Sciences, School of Medicine, University of California San Francisco, San Francisco, California, USA.,Department of Pediatrics, School of Medicine, University of California - San Francisco, San Francisco, California, USA
| | - M Sirota
- Institute for Computational Health Sciences, School of Medicine, University of California San Francisco, San Francisco, California, USA.,Department of Pediatrics, School of Medicine, University of California - San Francisco, San Francisco, California, USA
| | - D Hadley
- Institute for Computational Health Sciences, School of Medicine, University of California San Francisco, San Francisco, California, USA.,Department of Pediatrics, School of Medicine, University of California - San Francisco, San Francisco, California, USA
| | - A J Butte
- Institute for Computational Health Sciences, School of Medicine, University of California San Francisco, San Francisco, California, USA.,Department of Pediatrics, School of Medicine, University of California - San Francisco, San Francisco, California, USA
| |
Collapse
|
25
|
Pouladi N, Achour I, Li H, Berghout J, Kenost C, Gonzalez-Garay ML, Lussier YA. Biomechanisms of Comorbidity: Reviewing Integrative Analyses of Multi-omics Datasets and Electronic Health Records. Yearb Med Inform 2016; 25:194-206. [PMID: 27830251 PMCID: PMC5171562 DOI: 10.15265/iy-2016-040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES Disease comorbidity is a pervasive phenomenon impacting patients' health outcomes, disease management, and clinical decisions. This review presents past, current and future research directions leveraging both phenotypic and molecular information to uncover disease similarity underpinning the biology and etiology of disease comorbidity. METHODS We retrieved ~130 publications and retained 59, ranging from 2006 to 2015, that comprise a minimum number of five diseases and at least one type of biomolecule. We surveyed their methods, disease similarity metrics, and calculation of comorbidities in the electronic health records, if present. RESULTS Among the surveyed studies, 44% generated or validated disease similarity metrics in context of comorbidity, with 60% being published in the last two years. As inputs, 87% of studies utilized intragenic loci and proteins while 13% employed RNA (mRNA, LncRNA or miRNA). Network modeling was predominantly used (35%) followed by statistics (28%) to impute similarity between these biomolecules and diseases. Studies with large numbers of biomolecules and diseases used network models or naïve overlap of disease-molecule associations, while machine learning, statistics, and information retrieval were utilized in smaller and moderate sized studies. Multiscale computations comprising shared function, network topology, and phenotypes were performed exclusively on proteins. CONCLUSION This review highlighted the growing methods for identifying the molecular mechanisms underpinning comorbidities that leverage multiscale molecular information and patterns from electronic health records. The survey unveiled that intergenic polymorphisms have been overlooked for similarity imputation compared to their intragenic counterparts, offering new opportunities to bridge the mechanistic and similarity gaps of comorbidity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Y A Lussier
- Dr. Yves A. Lussier, The University of Arizona, Bio5 Building, 1657 East Helen Street, Tucson, AZ 85721, USA, Fax: +1 520 626 4824, E-Mail:
| |
Collapse
|
26
|
Dixon P, Davey Smith G, von Hinke S, Davies NM, Hollingworth W. Estimating Marginal Healthcare Costs Using Genetic Variants as Instrumental Variables: Mendelian Randomization in Economic Evaluation. PHARMACOECONOMICS 2016; 34:1075-1086. [PMID: 27484822 PMCID: PMC5073110 DOI: 10.1007/s40273-016-0432-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Accurate measurement of the marginal healthcare costs associated with different diseases and health conditions is important, especially for increasingly prevalent conditions such as obesity. However, existing observational study designs cannot identify the causal impact of disease on healthcare costs. This paper explores the possibilities for causal inference offered by Mendelian randomization, a form of instrumental variable analysis that uses genetic variation as a proxy for modifiable risk exposures, to estimate the effect of health conditions on cost. Well-conducted genome-wide association studies provide robust evidence of the associations of genetic variants with health conditions or disease risk factors. The subsequent causal effects of these health conditions on cost can be estimated using genetic variants as instruments for the health conditions. This is because the approximately random allocation of genotypes at conception means that many genetic variants are orthogonal to observable and unobservable confounders. Datasets with linked genotypic and resource use information obtained from electronic medical records or from routinely collected administrative data are now becoming available and will facilitate this form of analysis. We describe some of the methodological issues that arise in this type of analysis, which we illustrate by considering how Mendelian randomization could be used to estimate the causal impact of obesity, a complex trait, on healthcare costs. We describe some of the data sources that could be used for this type of analysis. We conclude by considering the challenges and opportunities offered by Mendelian randomization for economic evaluation.
Collapse
Affiliation(s)
- Padraig Dixon
- School of Social and Community Medicine, University of Bristol, Canynge Hall, 39 Whatley Road, Bristol, BS8 2PS, UK.
| | - George Davey Smith
- School of Social and Community Medicine, University of Bristol, Canynge Hall, 39 Whatley Road, Bristol, BS8 2PS, UK
- MRC Integrative Epidemiology Unit at the University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
| | - Stephanie von Hinke
- School of Economics, Finance and Management, University of Bristol, 8 Woodland Road, Bristol, BS8 1TN, UK
| | - Neil M Davies
- School of Social and Community Medicine, University of Bristol, Canynge Hall, 39 Whatley Road, Bristol, BS8 2PS, UK
- MRC Integrative Epidemiology Unit at the University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
| | - William Hollingworth
- School of Social and Community Medicine, University of Bristol, Canynge Hall, 39 Whatley Road, Bristol, BS8 2PS, UK
| |
Collapse
|
27
|
He L, Kernogitski Y, Kulminskaya I, Loika Y, Arbeev KG, Loiko E, Bagley O, Duan M, Yashkin A, Ukraintseva SV, Kovtun M, Yashin AI, Kulminski AM. Pleiotropic Meta-Analyses of Longitudinal Studies Discover Novel Genetic Variants Associated with Age-Related Diseases. Front Genet 2016; 7:179. [PMID: 27790247 PMCID: PMC5061751 DOI: 10.3389/fgene.2016.00179] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 09/21/2016] [Indexed: 01/31/2023] Open
Abstract
Age-related diseases may result from shared biological mechanisms in intrinsic processes of aging. Genetic effects on age-related diseases are often modulated by environmental factors due to their little contribution to fitness or are mediated through certain endophenotypes. Identification of genetic variants with pleiotropic effects on both common complex diseases and endophenotypes may reveal potential conflicting evolutionary pressures and deliver new insights into shared genetic contribution to healthspan and lifespan. Here, we performed pleiotropic meta-analyses of genetic variants using five NIH-funded datasets by integrating univariate summary statistics for age-related diseases and endophenotypes. We investigated three groups of traits: (1) endophenotypes such as blood glucose, blood pressure, lipids, hematocrit, and body mass index, (2) time-to-event outcomes such as the age-at-onset of diabetes mellitus (DM), cancer, cardiovascular diseases (CVDs) and neurodegenerative diseases (NDs), and (3) both combined. In addition to replicating previous findings, we identify seven novel genome-wide significant loci (< 5e-08), out of which five are low-frequency variants. Specifically, from Group 2, we find rs7632505 on 3q21.1 in SEMA5B, rs460976 on 21q22.3 (1 kb from TMPRSS2) and rs12420422 on 11q24.1 predominantly associated with a variety of CVDs, rs4905014 in ITPK1 associated with stroke and heart failure, rs7081476 on 10p12.1 in ANKRD26 associated with multiple diseases including DM, CVDs, and NDs. From Group 3, we find rs8082812 on 18p11.22 and rs1869717 on 4q31.3 associated with both endophenotypes and CVDs. Our follow-up analyses show that rs7632505, rs4905014, and rs8082812 have age-dependent effects on coronary heart disease or stroke. Functional annotation suggests that most of these SNPs are within regulatory regions or DNase clusters and in linkage disequilibrium with expression quantitative trait loci, implying their potential regulatory influence on the expression of nearby genes. Our mediation analyses suggest that the effects of some SNPs are mediated by specific endophenotypes. In conclusion, these findings indicate that loci with pleiotropic effects on age-related disorders tend to be enriched in genes involved in underlying mechanisms potentially related to nervous, cardiovascular and immune system functions, stress resistance, inflammation, ion channels and hematopoiesis, supporting the hypothesis of shared pathological role of infection, and inflammation in chronic age-related diseases.
Collapse
Affiliation(s)
- Liang He
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke UniversityDurham, NC, USA
| | | | | | | | | | | | | | | | | | | | | | | | - Alexander M. Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke UniversityDurham, NC, USA
| |
Collapse
|
28
|
Pickrell JK, Berisa T, Liu JZ, Ségurel L, Tung JY, Hinds DA. Detection and interpretation of shared genetic influences on 42 human traits. Nat Genet 2016; 48:709-17. [PMID: 27182965 PMCID: PMC5207801 DOI: 10.1038/ng.3570] [Citation(s) in RCA: 761] [Impact Index Per Article: 84.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 04/20/2016] [Indexed: 12/14/2022]
Abstract
We performed a scan for genetic variants associated with multiple phenotypes by comparing large genome-wide association studies (GWAS) of 42 traits or diseases. We identified 341 loci (at a false discovery rate of 10%) associated with multiple traits. Several loci are associated with multiple phenotypes; for example, a nonsynonymous variant in the zinc transporter SLC39A8 influences seven of the traits, including risk of schizophrenia (rs13107325: log-transformed odds ratio (log OR) = 0.15, P = 2 × 10(-12)) and Parkinson disease (log OR = -0.15, P = 1.6 × 10(-7)), among others. Second, we used these loci to identify traits that have multiple genetic causes in common. For example, variants associated with increased risk of schizophrenia also tended to be associated with increased risk of inflammatory bowel disease. Finally, we developed a method to identify pairs of traits that show evidence of a causal relationship. For example, we show evidence that increased body mass index causally increases triglyceride levels.
Collapse
Affiliation(s)
- Joseph K Pickrell
- New York Genome Center, New York, New York, USA
- Department of Biological Sciences, Columbia University, New York, New York, USA
| | | | - Jimmy Z Liu
- New York Genome Center, New York, New York, USA
| | - Laure Ségurel
- UMR 7206 Eco-Anthropologie et Ethnobiologie, CNRS, MNHN, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | | | | |
Collapse
|
29
|
Ramanan SV, Radhakrishna K, Waghmare A, Raj T, Nathan SP, Sreerama SM, Sampath S. Dense Annotation of Free-Text Critical Care Discharge Summaries from an Indian Hospital and Associated Performance of a Clinical NLP Annotator. J Med Syst 2016; 40:187. [PMID: 27342107 DOI: 10.1007/s10916-016-0541-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 06/08/2016] [Indexed: 10/21/2022]
Abstract
Electronic Health Record (EHR) use in India is generally poor, and structured clinical information is mostly lacking. This work is the first attempt aimed at evaluating unstructured text mining for extracting relevant clinical information from Indian clinical records. We annotated a corpus of 250 discharge summaries from an Intensive Care Unit (ICU) in India, with markups for diseases, procedures, and lab parameters, their attributes, as well as key demographic information and administrative variables such as patient outcomes. In this process, we have constructed guidelines for an annotation scheme useful to clinicians in the Indian context. We evaluated the performance of an NLP engine, Cocoa, on a cohort of these Indian clinical records. We have produced an annotated corpus of roughly 90 thousand words, which to our knowledge is the first tagged clinical corpus from India. Cocoa was evaluated on a test corpus of 50 documents. The overlap F-scores across the major categories, namely disease/symptoms, procedures, laboratory parameters and outcomes, are 0.856, 0.834, 0.961 and 0.872 respectively. These results are competitive with results from recent shared tasks based on US records. The annotated corpus and associated results from the Cocoa engine indicate that unstructured text mining is a viable method for cohort analysis in the Indian clinical context, where structured EHR records are largely absent.
Collapse
Affiliation(s)
- S V Ramanan
- RelAgent Technologies (P) Limited, IIT Madras Research Park, #14, 1st Floor, Taramani, Chennai, 600113, India.
| | - Kedar Radhakrishna
- Division of Medical Informatics, St. John's Research Institute, 100 Feet Road, Koramangala, Bangalore, 560034, India.
| | - Abijeet Waghmare
- Division of Medical Informatics, St. John's Research Institute, 100 Feet Road, Koramangala, Bangalore, 560034, India
| | - Tony Raj
- Division of Medical Informatics, St. John's Research Institute, 100 Feet Road, Koramangala, Bangalore, 560034, India
| | - Senthil P Nathan
- RelAgent Technologies (P) Limited, IIT Madras Research Park, #14, 1st Floor, Taramani, Chennai, 600113, India
| | - Sai Madhukar Sreerama
- Division of Medical Informatics, St. John's Research Institute, 100 Feet Road, Koramangala, Bangalore, 560034, India
| | - Sriram Sampath
- Department of Critical Care Medicine, St. John's Medical College, Bangalore, India
| |
Collapse
|
30
|
Bagley SC, Sirota M, Chen R, Butte AJ, Altman RB. Constraints on Biological Mechanism from Disease Comorbidity Using Electronic Medical Records and Database of Genetic Variants. PLoS Comput Biol 2016; 12:e1004885. [PMID: 27115429 PMCID: PMC4846031 DOI: 10.1371/journal.pcbi.1004885] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 03/29/2016] [Indexed: 12/24/2022] Open
Abstract
Patterns of disease co-occurrence that deviate from statistical independence may represent important constraints on biological mechanism, which sometimes can be explained by shared genetics. In this work we study the relationship between disease co-occurrence and commonly shared genetic architecture of disease. Records of pairs of diseases were combined from two different electronic medical systems (Columbia, Stanford), and compared to a large database of published disease-associated genetic variants (VARIMED); data on 35 disorders were available across all three sources, which include medical records for over 1.2 million patients and variants from over 17,000 publications. Based on the sources in which they appeared, disease pairs were categorized as having predominant clinical, genetic, or both kinds of manifestations. Confounding effects of age on disease incidence were controlled for by only comparing diseases when they fall in the same cluster of similarly shaped incidence patterns. We find that disease pairs that are overrepresented in both electronic medical record systems and in VARIMED come from two main disease classes, autoimmune and neuropsychiatric. We furthermore identify specific genes that are shared within these disease groups.
Collapse
Affiliation(s)
- Steven C. Bagley
- Department of Genetics, Stanford University, Stanford, California, United States of America
- * E-mail:
| | - Marina Sirota
- UCSF Institute for Computational Health Sciences, San Francisco, California, United States of America
| | - Richard Chen
- Personalis, Inc., Menlo Park, California, United States of America
| | - Atul J. Butte
- UCSF Institute for Computational Health Sciences, San Francisco, California, United States of America
| | - Russ B. Altman
- Department of Genetics, Stanford University, Stanford, California, United States of America
- Department of Bioengineering, Stanford University, Stanford, California, United States of America
| |
Collapse
|
31
|
Huang BE, Mulyasasmita W, Rajagopal G. The path from big data to precision medicine. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2016. [DOI: 10.1080/23808993.2016.1157686] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
32
|
Abstract
Next-generation sequencing (NGS) approaches are highly applicable to clinical studies. We review recent advances in sequencing technologies, as well as their benefits and tradeoffs, to provide an overview of clinical genomics from study design to computational analysis. Sequencing technologies enable genomic, transcriptomic, and epigenomic evaluations. Studies that use a combination of whole genome, exome, mRNA, and bisulfite sequencing are now feasible due to decreasing sequencing costs. Single-molecule sequencing increases read length, with the MinIONTM nanopore sequencer, which offers a uniquely portable option at a lower cost. Many of the published comparisons we review here address the challenges associated with different sequencing methods. Overall, NGS techniques, coupled with continually improving analysis algorithms, are useful for clinical studies in many realms, including cancer, chronic illness, and neurobiology. We, and others in the field, anticipate the clinical use of NGS approaches will continue to grow, especially as we shift into an era of precision medicine.
Collapse
Affiliation(s)
- Priyanka Vijay
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York. Tri-Institutional Training Program in Computational Biology and Medicine, Weill Cornell Medical College, New York, New York
| | - Alexa B.R. McIntyre
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York. Tri-Institutional Training Program in Computational Biology and Medicine, Weill Cornell Medical College, New York, New York
| | - Christopher E. Mason
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York. Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York. Feil Family Brain and Mind Research Institute, New York, New York
| | - Jeffrey P. Greenfield
- Department of Neurological Surgery, New York-Presbyterian Hospital, Weill Cornell Medical College, New York, New York
| | - Sheng Li
- Department of Neurological Surgery, New York-Presbyterian Hospital, Weill Cornell Medical College, New York, New York
| |
Collapse
|
33
|
Smith MT, de la Rosa R, Daniels SI. Using exposomics to assess cumulative risks and promote health. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2015; 56:715-23. [PMID: 26475350 PMCID: PMC4636923 DOI: 10.1002/em.21985] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/21/2015] [Indexed: 05/10/2023]
Abstract
Under the exposome paradigm all nongenetic factors contributing to disease are considered to be 'environmental' including chemicals, drugs, infectious agents, and psychosocial stress. We can consider these collectively as environmental stressors. Exposomics is the comprehensive analysis of exposure to all environmental stressors and should yield a more thorough understanding of chronic disease development. We can operationalize exposomics by studying all the small molecules in the body and their influence on biological pathways that lead to impaired health. Here, we describe methods by which this may be achieved and discuss the application of exposomics to cumulative risk assessment in vulnerable populations. Since the goal of cumulative risk assessment is to analyze, characterize, and quantify the combined risks to health from exposures to multiple agents or stressors, it seems that exposomics is perfectly poised to advance this important area of environmental health science. We should therefore support development of tools for exposomic analysis and begin to engage impacted communities in participatory exposome research. A first step may be to apply exposomics to vulnerable populations already studied by more conventional cumulative risk approaches. We further propose that recent migrants, low socioeconomic groups with high environmental chemical exposures, and pregnant women should be high priority populations for study by exposomics. Moreover, exposomics allows us to study interactions between chronic stress and environmental chemicals that disrupt stress response pathways (i.e., 'stressogens'). Exploring the impact of early life exposures and maternal stress may be an interesting and accessible topic for investigation by exposomics using biobanked samples.
Collapse
Affiliation(s)
- Martyn T Smith
- Superfund Research Program, Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, California, 94720-7360
| | - Rosemarie de la Rosa
- Superfund Research Program, Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, California, 94720-7360
| | - Sarah I Daniels
- Superfund Research Program, Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, California, 94720-7360
| |
Collapse
|
34
|
Klosinski LP, Yao J, Yin F, Fonteh AN, Harrington MG, Christensen TA, Trushina E, Brinton RD. White Matter Lipids as a Ketogenic Fuel Supply in Aging Female Brain: Implications for Alzheimer's Disease. EBioMedicine 2015; 2:1888-904. [PMID: 26844268 PMCID: PMC4703712 DOI: 10.1016/j.ebiom.2015.11.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 10/24/2015] [Accepted: 11/02/2015] [Indexed: 01/28/2023] Open
Abstract
White matter degeneration is a pathological hallmark of neurodegenerative diseases including Alzheimer's. Age remains the greatest risk factor for Alzheimer's and the prevalence of age-related late onset Alzheimer's is greatest in females. We investigated mechanisms underlying white matter degeneration in an animal model consistent with the sex at greatest Alzheimer's risk. Results of these analyses demonstrated decline in mitochondrial respiration, increased mitochondrial hydrogen peroxide production and cytosolic-phospholipase-A2 sphingomyelinase pathway activation during female brain aging. Electron microscopic and lipidomic analyses confirmed myelin degeneration. An increase in fatty acids and mitochondrial fatty acid metabolism machinery was coincident with a rise in brain ketone bodies and decline in plasma ketone bodies. This mechanistic pathway and its chronologically phased activation, links mitochondrial dysfunction early in aging with later age development of white matter degeneration. The catabolism of myelin lipids to generate ketone bodies can be viewed as a systems level adaptive response to address brain fuel and energy demand. Elucidation of the initiating factors and the mechanistic pathway leading to white matter catabolism in the aging female brain provides potential therapeutic targets to prevent and treat demyelinating diseases such as Alzheimer's and multiple sclerosis. Targeting stages of disease and associated mechanisms will be critical. Mitochondrial dysfunction activates mechanisms for catabolism of myelin lipids to generate ketone bodies for ATP production. Mechanisms leading to ketone body driven energy production in brain coincide with stages of reproductive aging in females. Sequential activation of myelin catabolism pathway during aging provides multiple therapeutic targets and windows of efficacy.
The mechanisms underlying white matter degeneration, a hallmark of multiple neurodegenerative diseases including Alzheimer's, remain unclear. Herein we provide a mechanistic pathway, spanning multiple transitions of aging, that links mitochondrial dysfunction early in aging with later age white matter degeneration. Catabolism of myelin lipids to generate ketone bodies can be viewed as an adaptive survival response to address brain fuel and energy demand. Women are at greatest risk of late-onset-AD, thus, our analyses in female brain address mechanisms of AD pathology and therapeutic targets to prevent, delay and treat AD in the sex most affected with potential relevance to men.
Collapse
Key Words
- ABAD, Aβ-binding alcohol dehydrogenase
- ABAD, Aβ-binding-alcohol-dehydrogenase
- ACER3, alkaline ceramidase
- AD, Alzheimer's disease
- APO-ε4, apolipoprotein ε4
- APP, amyloid precursor protein
- Aging oxidative stress
- Alzheimer's disease
- BACE1, beta-secretase 1
- BBB, blood brain barrier
- CC, corpus callosum
- CMRglu, cerebral glucose metabolic rate
- COX, complex IV cytochrome c oxidase
- CPT1, carnitine palmitoyltransferase 1
- Cldn11, claudin 11
- Cyp2j6, arachidonic acid epoxygenase
- Cytosolic phospholipase A2
- DHA, docosahexaesnoic acid
- Erbb3, Erb-B2 receptor tyrosine kinase 3
- FDG-PET, 2-[18F]fluoro-2-deoxy-d-glucose
- GFAP, glial fibrillary acidic protein
- H2O2, hydrogen peroxide
- HADHA, hydroxyacyl-CoA dehydrogenase
- HK, hexokinase
- Ketone bodies
- LC MS, liquid chromatography mass spectrometer
- MAG, myelin associated glycoprotein
- MBP, myelin basic protein
- MCT1, monocarboxylate transporter 1
- MIB, mitochondrial isolation buffer
- MOG, myelin oligodendrocyte glycoprotein
- MTL, medial temporal lobe
- Mitochondria
- NEFA, nonesterified fatty acids
- Neurodegeneration
- OCR, oxygen consumption rate
- Olig2, oligodendrocyte transcription factor
- PB, phosphate buffer
- PCC, posterior cingulate
- PCR, polymerase chain reaction
- PDH, pyruvate dehydrogenase
- PEI, polyethyleneimine
- RCR, respiratory control ratio
- ROS, reactive oxygen species
- S1P, sphingosine
- TLDA, TaqMan low density array
- WM, white matter
- WT, wild type
- White matter
- cPLA2, cytosolic phospholipase A2
Collapse
Affiliation(s)
- Lauren P Klosinski
- Department of Neuroscience, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, USA
| | - Jia Yao
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Fei Yin
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | | | | | | | - Eugenia Trushina
- Department of Neurology, Mayo Clinic Rochester, MN, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Roberta Diaz Brinton
- Department of Neuroscience, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, USA; Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA; Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
35
|
Oellrich A, Collier N, Groza T, Rebholz-Schuhmann D, Shah N, Bodenreider O, Boland MR, Georgiev I, Liu H, Livingston K, Luna A, Mallon AM, Manda P, Robinson PN, Rustici G, Simon M, Wang L, Winnenburg R, Dumontier M. The digital revolution in phenotyping. Brief Bioinform 2015; 17:819-30. [PMID: 26420780 PMCID: PMC5036847 DOI: 10.1093/bib/bbv083] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Indexed: 12/22/2022] Open
Abstract
Phenotypes have gained increased notoriety in the clinical and biological domain owing to their application in numerous areas such as the discovery of disease genes and drug targets, phylogenetics and pharmacogenomics. Phenotypes, defined as observable characteristics of organisms, can be seen as one of the bridges that lead to a translation of experimental findings into clinical applications and thereby support 'bench to bedside' efforts. However, to build this translational bridge, a common and universal understanding of phenotypes is required that goes beyond domain-specific definitions. To achieve this ambitious goal, a digital revolution is ongoing that enables the encoding of data in computer-readable formats and the data storage in specialized repositories, ready for integration, enabling translational research. While phenome research is an ongoing endeavor, the true potential hidden in the currently available data still needs to be unlocked, offering exciting opportunities for the forthcoming years. Here, we provide insights into the state-of-the-art in digital phenotyping, by means of representing, acquiring and analyzing phenotype data. In addition, we provide visions of this field for future research work that could enable better applications of phenotype data.
Collapse
|
36
|
Wang L, Oehlers SH, Espenschied ST, Rawls JF, Tobin DM, Ko DC. CPAG: software for leveraging pleiotropy in GWAS to reveal similarity between human traits links plasma fatty acids and intestinal inflammation. Genome Biol 2015; 16:190. [PMID: 26374098 PMCID: PMC4570686 DOI: 10.1186/s13059-015-0722-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 07/09/2015] [Indexed: 12/31/2022] Open
Abstract
Meta-analyses of genome-wide association studies (GWAS) have demonstrated that the same genetic variants can be associated with multiple diseases and other complex traits. We present software called CPAG (Cross-Phenotype Analysis of GWAS) to look for similarities between 700 traits, build trees with informative clusters, and highlight underlying pathways. Clusters are consistent with pre-defined groups and literature-based validation but also reveal novel connections. We report similarity between plasma palmitoleic acid and Crohn's disease and find that specific fatty acids exacerbate enterocolitis in zebrafish. CPAG will become increasingly powerful as more genetic variants are uncovered, leading to a deeper understanding of complex traits. CPAG is freely available at www.sourceforge.net/projects/CPAG/.
Collapse
Affiliation(s)
- Liuyang Wang
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, 27710, USA.
| | - Stefan H Oehlers
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, 27710, USA.
| | - Scott T Espenschied
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, 27710, USA.
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, 27710, USA.
| | - David M Tobin
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, 27710, USA.
| | - Dennis C Ko
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, 27710, USA. .,Department of Medicine and the Center for Human Genome Variation, School of Medicine, Duke University, Durham, NC, 27710, USA.
| |
Collapse
|
37
|
Yang C, Li C, Wang Q, Chung D, Zhao H. Implications of pleiotropy: challenges and opportunities for mining Big Data in biomedicine. Front Genet 2015; 6:229. [PMID: 26175753 PMCID: PMC4485215 DOI: 10.3389/fgene.2015.00229] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 06/15/2015] [Indexed: 01/23/2023] Open
Abstract
Pleiotropy arises when a locus influences multiple traits. Rich GWAS findings of various traits in the past decade reveal many examples of this phenomenon, suggesting the wide existence of pleiotropic effects. What underlies this phenomenon is the biological connection among seemingly unrelated traits/diseases. Characterizing the molecular mechanisms of pleiotropy not only helps to explain the relationship between diseases, but may also contribute to novel insights concerning the pathological mechanism of each specific disease, leading to better disease prevention, diagnosis and treatment. However, most pleiotropic effects remain elusive because their functional roles have not been systematically examined. A systematic investigation requires availability of qualified measurements at multilayered biological processes (e.g., transcription and translation). The rise of Big Data in biomedicine, such as high-quality multi-omics data, biomedical imaging data and electronic medical records of patients, offers us an unprecedented opportunity to investigate pleiotropy. There will be a great need of computationally efficient and statistically rigorous methods for integrative analysis of these Big Data in biomedicine. In this review, we outline many opportunities and challenges in methodology developments for systematic analysis of pleiotropy, and highlight its implications on disease prevention, diagnosis and treatment.
Collapse
Affiliation(s)
- Can Yang
- Department of Mathematics, Hong Kong Baptist UniversityHong Kong, Hong Kong
- Hong Kong Baptist University Institute of Research and Continuing EducationShenzhen, China
| | - Cong Li
- Program in Computational Biology and Bioinformatics, Yale UniversityNew Haven, CT, USA
| | - Qian Wang
- Program in Computational Biology and Bioinformatics, Yale UniversityNew Haven, CT, USA
| | - Dongjun Chung
- Department of Public Health Sciences, Medical University of South CarolinaCharleston, SC, USA
| | - Hongyu Zhao
- Program in Computational Biology and Bioinformatics, Yale UniversityNew Haven, CT, USA
- Department of Biostatistics, Yale School of Public HealthNew Haven, CT, USA
- Department of Genetics, Yale School of MedicineNew Haven, CT, USA
- VA Cooperative Studies Program Coordinating CenterWest Haven, CT, USA
| |
Collapse
|
38
|
Repurpose terbutaline sulfate for amyotrophic lateral sclerosis using electronic medical records. Sci Rep 2015; 5:8580. [PMID: 25739475 PMCID: PMC4894399 DOI: 10.1038/srep08580] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 01/23/2015] [Indexed: 12/17/2022] Open
Abstract
Prediction of new disease indications for approved drugs by computational methods has been based largely on the genomics signatures of drugs and diseases. We propose a method for drug repositioning that uses the clinical signatures extracted from over 13 years of electronic medical records from a tertiary hospital, including >9.4 M laboratory tests from >530,000 patients, in addition to diverse genomics signatures. Cross-validation using over 17,000 known drug–disease associations shows this approach outperforms various predictive models based on genomics signatures and a well-known “guilt-by-association” method. Interestingly, the prediction suggests that terbutaline sulfate, which is widely used for asthma, is a promising candidate for amyotrophic lateral sclerosis for which there are few therapeutic options. In vivo tests using zebrafish models found that terbutaline sulfate prevents defects in axons and neuromuscular junction degeneration in a dose-dependent manner. A therapeutic potential of terbutaline sulfate was also observed when axonal and neuromuscular junction degeneration have already occurred in zebrafish model. Cotreatment with a β2-adrenergic receptor antagonist, butoxamine, suggests that the effect of terbutaline is mediated by activation of β2-adrenergic receptors.
Collapse
|