1
|
Jefferson W, Wang T, Padilla-Banks E, Williams C. Unexpected nuclear hormone receptor and chromatin dynamics regulate estrous cycle dependent gene expression. Nucleic Acids Res 2024; 52:10897-10917. [PMID: 39166489 PMCID: PMC11472041 DOI: 10.1093/nar/gkae714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 06/27/2024] [Accepted: 08/06/2024] [Indexed: 08/23/2024] Open
Abstract
Chromatin changes in response to estrogen and progesterone are well established in cultured cells, but how they control gene expression under physiological conditions is largely unknown. To address this question, we examined in vivo estrous cycle dynamics of mouse uterus hormone receptor occupancy, chromatin accessibility and chromatin structure by combining RNA-seq, ATAC-seq, HiC-seq and ChIP-seq. Two estrous cycle stages were chosen for these analyses, diestrus (highest estrogen) and estrus (highest progesterone). Unexpectedly, rather than alternating with each other, estrogen receptor alpha (ERα) and progesterone receptor (PGR) were co-bound during diestrus and lost during estrus. Motif analysis of open chromatin followed by hypoxia inducible factor 2A (HIF2A) ChIP-seq and conditional uterine deletion of this transcription factor revealed a novel role for HIF2A in regulating diestrus gene expression patterns that were independent of either ERα or PGR binding. Proteins in complex with ERα included PGR and cohesin, only during diestrus. Combined with HiC-seq analyses, we demonstrate that complex chromatin architecture changes including enhancer switching are coordinated with ERα and PGR co-binding during diestrus and non-hormone receptor transcription factors such as HIF2A during estrus to regulate most differential gene expression across the estrous cycle.
Collapse
Affiliation(s)
- Wendy N Jefferson
- Reproductive & Developmental Biology Laboratory, Research Triangle Park, NC 27709, USA
| | - Tianyuan Wang
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | | | - Carmen J Williams
- Reproductive & Developmental Biology Laboratory, Research Triangle Park, NC 27709, USA
| |
Collapse
|
2
|
Shen Y, Huang J, Jia L, Zhang C, Xu J. Bioinformatics and machine learning driven key genes screening for hepatocellular carcinoma. Biochem Biophys Rep 2024; 37:101587. [PMID: 38107663 PMCID: PMC10724547 DOI: 10.1016/j.bbrep.2023.101587] [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: 07/23/2023] [Revised: 11/01/2023] [Accepted: 11/17/2023] [Indexed: 12/19/2023] Open
Abstract
Liver cancer, a global menace, ranked as the sixth most prevalent and third deadliest cancer in 2020. The challenge of early diagnosis and treatment, especially for hepatocellular carcinoma (HCC), persists due to late-stage detections. Understanding HCC's complex pathogenesis is vital for advancing diagnostics and therapies. This study combines bioinformatics and machine learning, examining HCC comprehensively. Three datasets underwent meticulous scrutiny, employing various analytical tools such as Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, protein interaction assessment, and survival analysis. These rigorous investigations uncovered twelve pivotal genes intricately linked with HCC's pathophysiological intricacies. Among them, CYP2C8, CYP2C9, EPHX2, and ESR1 were significantly positively correlated with overall patient survival, while AKR1B10 and NQO1 displayed a negative correlation. Moreover, the Adaboost prediction model yielded an 86.8 % accuracy, showcasing machine learning's potential in deciphering complex dataset patterns for clinically relevant predictions. These findings promise to contribute valuable insights into the elusive mechanisms driving liver cancer (HCC). They hold the potential to guide the development of more precise diagnostic methods and treatment strategies in the future. In the fight against this global health challenge, unraveling HCC's intricacies is of paramount importance.
Collapse
Affiliation(s)
- Ye Shen
- Department of Radiology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213002, China
| | - Juanjie Huang
- Department of General Surgery, Dongguan Qingxi Hospital, Dongguan, 523660, China
| | - Lei Jia
- International Health Medicine Innovation Center, Shenzhen University, ShenZhen, 518060, China
| | - Chi Zhang
- Huaxia Eye Hospital of Foshan, Huaxia Eye Hospital Group, Foshan, Guangdong, 528000, China
| | - Jianxing Xu
- Department of Radiology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213002, China
- Department of Radiology, The Wujin Clinical College of Xuzhou Medical University, Changzhou, 213002, China
| |
Collapse
|
3
|
Zhao Z, Yang LL, Wang QL, Du JF, Zheng ZG, Jiang Y, Li P, Li HJ. Baohuoside I inhibits FXR signaling pathway to interfere with bile acid homeostasis via targeting ER α degradation. Cell Biol Toxicol 2023; 39:1215-1235. [PMID: 35802278 DOI: 10.1007/s10565-022-09737-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 06/20/2022] [Indexed: 11/30/2022]
Abstract
Epimedii folium (EF) is an effective herbal medicine in osteoporosis treatment, but the clinical utilization of EF has been limited due to potential hepatotoxicity. The previous studies identified that baohuoside I (BI), the main active component of EF, was relevant to EF-induced liver injury. However, the mechanisms of BI causing direct injury to hepatocytes remain unclear. Here, we reveal that BI inhibits FXR-mediated signaling pathway via targeting estrogen receptor α (ER α), leading to the accumulation of bile acids (BAs). Targeted bile acid analyses show BI alters the BA composition and distribution, resulting in impaired BA homeostasis. Mechanistically, BI induces FXR-dependent hepatotoxicity at transcriptional level. Additionally, ER α is predicted to bind to the FXR promoter region based on transcription factor binding sites databases and we further demonstrate that ER α positively regulates FXR promoter activity and affects the expression of target genes involved in BA metabolism. Importantly, we discover that ER α and its mediated FXR transcription regulation might be involved in BI-induced liver injury via ligand-dependent ER α degradation. Collectively, our findings indicate that FXR is a newly discovered target gene of ER α mediated BI-induced liver injury, and suggest BI may be responsible for EF-induced liver injury.
Collapse
Affiliation(s)
- Zhen Zhao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Lu-Lu Yang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Qiao-Lei Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Jin-Fa Du
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Zu-Guo Zheng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Yan Jiang
- College of Chemical Engineering, Nanjing Forestry University, 159 Longpan Road, Nanjing, 210037, China.
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Hui-Jun Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China.
| |
Collapse
|
4
|
Pakdel F. The Role of Estrogen Receptors in Health and Disease. Int J Mol Sci 2023; 24:11354. [PMID: 37511113 PMCID: PMC10378944 DOI: 10.3390/ijms241411354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Many biological and physiological events, including growth, development, and metabolism of reproductive and non-reproductive tissues in men and women, are regulated by estrogens and estrogen receptors (ERs) [...].
Collapse
Affiliation(s)
- Farzad Pakdel
- Irset (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, EHESP, Inserm, Université de Rennes, F-35000 Rennes, France
| |
Collapse
|
5
|
Isola JVV, Ko S, Ocañas SR, Stout MB. Role of Estrogen Receptor α in Aging and Chronic Disease. ADVANCES IN GERIATRIC MEDICINE AND RESEARCH 2023; 5:e230005. [PMID: 37425648 PMCID: PMC10327608 DOI: 10.20900/agmr20230005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Estrogen receptor alpha (ERα) plays a crucial role in reproductive function in both sexes. It also mediates cellular responses to estrogens in multiple nonreproductive organ systems, many of which regulate systemic metabolic homeostasis and inflammatory processes in mammals. The loss of estrogens and/or ERα agonism during aging is associated with the emergence of several comorbid conditions, particularly in females undergoing the menopausal transition. Emerging data also suggests that male mammals likely benefit from ERα agonism if done in a way that circumvents feminizing characteristics. This has led us, and others, to speculate that tissue-specific ERα agonism may hold therapeutic potential for curtailing aging and chronic disease burden in males and females that are at high-risk of cancer and/or cardiovascular events with traditional estrogen replacement therapies. In this mini-review, we emphasize the role of ERα in the brain and liver, summarizing recent evidence that indicates these two organs systems mediate the beneficial effects of estrogens on metabolism and inflammation during aging. We also discuss how 17α-estradiol administration elicits health benefits in an ERα-dependent manner, which provides proof-of-concept that ERα may be a druggable target for attenuating aging and age-related disease burden.
Collapse
Affiliation(s)
- José V. V. Isola
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Sunghwan Ko
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Sarah R. Ocañas
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
| | - Michael B. Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
6
|
Collins JM, Nworu AC, Mohammad SJ, Li L, Li C, Li C, Schwendeman E, Cefalu M, Abdel‐Rasoul M, Sun JW, Smith SA, Wang D. Regulatory variants in a novel distal enhancer regulate the expression of CYP3A4 and CYP3A5. Clin Transl Sci 2022; 15:2720-2731. [PMID: 36045613 PMCID: PMC9652438 DOI: 10.1111/cts.13398] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/11/2022] [Accepted: 08/23/2022] [Indexed: 01/26/2023] Open
Abstract
The cytochrome P450 3As (CYP3As) are abundantly expressed in the liver and metabolize many commonly prescribed medications. Their expression is highly variable between individuals with little known genetic cause. Despite extensive investigation, cis-acting genetic elements that control the expression of the CYP3As remain uncharacterized. Using chromatin conformation capture (4C assays), we detected reciprocal interaction between a distal regulatory region (DRR) and the CYP3A4 promoter. The DRR colocalizes with a variety of enhancer marks and was found to promote transcription in reporter assays. CRISPR-mediated deletion of the DRR decreased expression of CYP3A4, CYP3A5, and CYP3A7, supporting its role as a shared enhancer regulating the expression of three CYP3A genes. Using reporter gene assays, we identified two single-nucleotide polymorphisms (rs115025140 and rs776744/rs776742) that increased DRR-driven luciferase reporter expression. In a liver cohort (n = 246), rs115025140 was associated with increased expression of CYP3A4 mRNA (1.8-fold) and protein (1.6-fold) and rs776744/rs776742 was associated with 1.39-fold increased expression of CYP3A5 mRNA. The rs115025140 is unique to the African population and in a clinical cohort of African Americans taking statins for lipid control rs115025140 carriers showed a trend toward reduced statin-mediated lipid reduction. In addition, using a published cohort of Chinese patients who underwent renal transplantation taking tacrolimus, rs776744/rs776742 carriers were associated with reduced tacrolimus concentration after adjusting for CYP3A5*3. Our results elucidate a complex regulatory network controlling expression of three CYP3A genes and identify two novel regulatory variants with potential clinical relevance for predicting CYP3A4 and CYP3A5 expression.
Collapse
Affiliation(s)
- Joseph M. Collins
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Adaeze C. Nworu
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Somayya J. Mohammad
- Department of Internal Medicine, Division of Cardiology, College of MedicineThe Ohio State UniversityColumbusOhioUSA
| | - Liang Li
- Department of Medical Genetics, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Chengcheng Li
- Department of Medical Genetics, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Chuanjiang Li
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Ethan Schwendeman
- Department of Internal Medicine, Division of Cardiology, College of MedicineThe Ohio State UniversityColumbusOhioUSA
| | - Mattew Cefalu
- Department of Internal Medicine, Division of Cardiology, College of MedicineThe Ohio State UniversityColumbusOhioUSA
| | - Mahmoud Abdel‐Rasoul
- Center for Biostatistics, Department of Biomedical Informatics, College of MedicineThe Ohio State UniversityColumbusOhioUSA
| | - Jessie W. Sun
- Department of Internal Medicine, Division of Cardiology, College of MedicineThe Ohio State UniversityColumbusOhioUSA,School of Medicine and Health SciencesGeorge Washington UniversityWashingtonDCUSA
| | - Sakima A. Smith
- Department of Internal Medicine, Division of Cardiology, College of MedicineThe Ohio State UniversityColumbusOhioUSA
| | - Danxin Wang
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
7
|
Tantawy M, Collins JM, Wang D. Genome-wide microRNA profiles identify miR-107 as a top miRNA associating with expression of the CYP3As and other drug metabolizing cytochrome P450 enzymes in the liver. Front Pharmacol 2022; 13:943538. [PMID: 36059981 PMCID: PMC9428441 DOI: 10.3389/fphar.2022.943538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Cytochrome P450 (CYP) drug metabolizing enzymes are responsible for the metabolism of over 70% of currently used medications with the CYP3A family being the most important CYP enzymes in the liver. Large inter-person variability in expression/activity of the CYP3As greatly affects drug exposure and treatment outcomes, yet the cause of such variability remains elusive. Micro-RNAs (miRNAs) are small noncoding RNAs that negatively regulate gene expression and are involved in diverse cellular processes including metabolism of xenobiotics and therapeutic outcomes. Target prediction and in vitro functional assays have linked several miRNAs to the control of CYP3A4 expression. Yet, their co-expression with CYP3As in the liver remain unclear. In this study, we used genome-wide miRNA profiling in liver samples to identify miRNAs associated with the expression of the CYP3As. We identified and validated both miR-107 and miR-1260 as strongly associated with the expression of CYP3A4, CYP3A5, and CYP3A43. Moreover, we found associations between miR-107 and nine transcription factors (TFs) that regulate CYP3A expression, with estrogen receptor alpha (ESR1) having the largest effect size. Including ESR1 and the other TFs in the regression model either diminished or abolished the associations between miR-107 and the CYP3As, indicating that the role of miR-107 in CYP3A expression may be indirect and occur through these key TFs. Indeed, testing the other nine CYPs previously shown to be regulated by ESR1 identified similar miR-107 associations that were dependent on the exclusion of ESR1 and other key TFs in the regression model. In addition, we found significant differences in miRNA expression profiles in liver samples between race and sex. Together, our results identify miR-107 as a potential epigenetic regulator that is strongly associated with the expression of many CYPs, likely via impacting the CYP regulatory network controlled by ESR1 and other key TFs. Therefore, both genetic and epigenetic factors that alter the expression of miR-107 may have a broad influence on drug metabolism.
Collapse
|
8
|
Stokar J, Gurt I, Cohen-Kfir E, Yakubovsky O, Hallak N, Benyamini H, Lishinsky N, Offir N, Tam J, Dresner-Pollak R. Hepatic Adropin is Regulated by Estrogen and Contributes to Adverse Metabolic Phenotypes in Ovariectomized Mice. Mol Metab 2022; 60:101482. [PMID: 35364299 PMCID: PMC9044006 DOI: 10.1016/j.molmet.2022.101482] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/13/2022] [Accepted: 03/24/2022] [Indexed: 12/02/2022] Open
Abstract
Objective Menopause is associated with visceral adiposity, hepatic steatosis and increased risk for cardiovascular disease. As estrogen replacement therapy is not suitable for all postmenopausal women, a need for alternative therapeutics and biomarkers has emerged. Methods 9-week-old C57BL/6 J female mice were subjected to ovariectomy (OVX) or SHAM surgery (n = 10 per group), fed a standard diet and sacrificed 6- & 12 weeks post-surgery. Results Increased weight gain, hepatic triglyceride content and changes in hepatic gene expression of Cyp17a1, Rgs16, Fitm1 as well as Il18, Rares2, Retn, Rbp4 in mesenteric visceral adipose tissue (VAT) were observed in OVX vs. SHAM. Liver RNA-sequencing 6-weeks post-surgery revealed changes in genes and microRNAs involved in fat metabolism in OVX vs. SHAM mice. Energy Homeostasis Associated gene (Enho) coding for the hepatokine adropin was significantly reduced in OVX mice livers and strongly inversely correlated with weight gain (r = −0.7 p < 0.001) and liver triglyceride content (r = −0.4, p = 0.04), with a similar trend for serum adropin. In vitro, Enho expression was tripled by 17β-estradiol in BNL 1 ME liver cells with increased adropin in supernatant. Analysis of open-access datasets revealed increased hepatic Enho expression in estrogen treated OVX mice and estrogen dependent ERα binding to Enho. Treatment of 5-month-old OVX mice with Adropin (i.p. 450 nmol/kg/twice daily, n = 4,5 per group) for 6-weeks reversed adverse adipokine gene expression signature in VAT, with a trended increase in lean body mass and decreased liver TG content with upregulation of Rgs16. Conclusions OVX is sufficient to induce deranged metabolism in adult female mice. Hepatic adropin is regulated by estrogen, negatively correlated with adverse OVX-induced metabolic phenotypes, which were partially reversed with adropin treatment. Adropin should be further explored as a potential therapeutic target and biomarker for menopause-related metabolic derangement. OVX increased body weight, liver fat & adverse visceral fat adipokine signature. OVX altered liver transcriptome & miRNA profile including fat metabolism pathways. Enho was downregulated by OVX & inversely correlated with weight gain & liver fat. Hepatic adropin expression was upregulated by estrogen in-vitro & in-vivo. Adropin treatment partially reversed OVX induced adverse metabolic phenotypes.
Collapse
|
9
|
Collins JM, Lu R, Wang X, Zhu HJ, Wang D. Transcriptional Regulation of Carboxylesterase 1 in Human Liver: Role of the Nuclear Receptor Subfamily 1 Group H Member 3 and Its Splice Isoforms. Drug Metab Dispos 2022; 50:43-48. [PMID: 34697082 PMCID: PMC8969197 DOI: 10.1124/dmd.121.000649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/20/2021] [Indexed: 01/03/2023] Open
Abstract
Carboxylesterase 1 (CES1) is the predominant carboxylesterase in the human liver, involved in metabolism of both xenobiotics and endogenous substrates. Genetic or epigenetic factors that alter CES1 activity or expression are associated with changes in drug response, lipid, and glucose homeostasis. However, the transcriptional regulation of CES1 in the human liver remains uncertain. By applying both the random forest and Sobol's Sensitivity Indices (SSI) to analyze existing liver RNA expression microarray data (GSE9588), we identified nuclear receptor subfamily 1 group H member 3 (NR1H3) liver X receptor (LXR)α as a key factor regulating constitutive CES1 expression. This model prediction was validated using small interfering RNA (siRNA) knockdown and CRISPR-mediated transcriptional activation of NR1H3 in Huh7 and HepG2 cells. We found that NR1H3's activation of CES1 is splice isoform-specific, namely that increased expression of the NR1H3-211 isoform increased CES1 expression whereas NR1H3-201 did not. Also, in human liver samples, expression of NR1H3-211 and CES1 are correlated, whereas NR1H3-201 and CES1 are not. This trend also occurs during differentiation of induced pluripotent stem cells (iPSCs) to hepatocytes, where only expression of the NR1H3-211 isoform parallels expression of CES1 Moreover, we found that treatment with the NR1H3 agonist T0901317 in HepG2 cells had no effect on CES1 expression. Overall, our results demonstrate a key role of NR1H3 in maintaining the constitutive expression of CES1 in the human liver. Furthermore, our results support that the effect of NR1H3 is splice isoform-specific and appears to be ligand independent. SIGNIFICANCE STATEMENT: Despite the central role of carboxylesterase 1 (CES1) in metabolism of numerous medications, little is known about its transcriptional regulation. This study identifies nuclear receptor subfamily 1 group H member 3 as a key regulator of constitutive CES1 expression and therefore is a potential target for future studies to understand interperson variabilities in CES1 activity and drug metabolism.
Collapse
Affiliation(s)
- Joseph M Collins
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.C., D.W.); The Quantitative Sciences Unit, Division of Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, California (R.L.); Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, (X.W.); and Department of Clinical Pharmacy, University of Michigan, Ann Arbor, Michigan (H.-J.Z.)
| | - Rong Lu
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.C., D.W.); The Quantitative Sciences Unit, Division of Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, California (R.L.); Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, (X.W.); and Department of Clinical Pharmacy, University of Michigan, Ann Arbor, Michigan (H.-J.Z.)
| | - Xinwen Wang
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.C., D.W.); The Quantitative Sciences Unit, Division of Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, California (R.L.); Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, (X.W.); and Department of Clinical Pharmacy, University of Michigan, Ann Arbor, Michigan (H.-J.Z.)
| | - Hao-Jie Zhu
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.C., D.W.); The Quantitative Sciences Unit, Division of Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, California (R.L.); Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, (X.W.); and Department of Clinical Pharmacy, University of Michigan, Ann Arbor, Michigan (H.-J.Z.)
| | - Danxin Wang
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, Florida (J.M.C., D.W.); The Quantitative Sciences Unit, Division of Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, California (R.L.); Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, (X.W.); and Department of Clinical Pharmacy, University of Michigan, Ann Arbor, Michigan (H.-J.Z.)
| |
Collapse
|