Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Suzette J. Bielinski is active.

Publication


Featured researches published by Suzette J. Bielinski.


Nature Biotechnology | 2013

Systematic comparison of phenome-wide association study of electronic medical record data and genome-wide association study data

Joshua C. Denny; Marylyn D. Ritchie; Robert J. Carroll; Raquel Zink; Jonathan D. Mosley; Julie R. Field; Jill M. Pulley; Andrea H. Ramirez; Erica Bowton; Melissa A. Basford; David Carrell; Peggy L. Peissig; Abel N. Kho; Jennifer A. Pacheco; Luke V. Rasmussen; David R. Crosslin; Paul K. Crane; Jyotishman Pathak; Suzette J. Bielinski; Sarah A. Pendergrass; Hua Xu; Lucia A. Hindorff; Rongling Li; Teri A. Manolio; Christopher G. Chute; Rex L. Chisholm; Eric B. Larson; Gail P. Jarvik; Murray H. Brilliant; Catherine A. McCarty

Candidate gene and genome-wide association studies (GWAS) have identified genetic variants that modulate risk for human disease; many of these associations require further study to replicate the results. Here we report the first large-scale application of the phenome-wide association study (PheWAS) paradigm within electronic medical records (EMRs), an unbiased approach to replication and discovery that interrogates relationships between targeted genotypes and multiple phenotypes. We scanned for associations between 3,144 single-nucleotide polymorphisms (previously implicated by GWAS as mediators of human traits) and 1,358 EMR-derived phenotypes in 13,835 individuals of European ancestry. This PheWAS replicated 66% (51/77) of sufficiently powered prior GWAS associations and revealed 63 potentially pleiotropic associations with P < 4.6 × 10−6 (false discovery rate < 0.1); the strongest of these novel associations were replicated in an independent cohort (n = 7,406). These findings validate PheWAS as a tool to allow unbiased interrogation across multiple phenotypes in EMR-based cohorts and to enhance analysis of the genomic basis of human disease.


Science Translational Medicine | 2011

Electronic Medical Records for Genetic Research: Results of the eMERGE Consortium

Abel N. Kho; Jennifer A. Pacheco; Peggy L. Peissig; Luke V. Rasmussen; Katherine M. Newton; Noah Weston; Paul K. Crane; Jyotishman Pathak; Christopher G. Chute; Suzette J. Bielinski; Iftikhar J. Kullo; Rongling Li; Teri A. Manolio; Rex L. Chisholm; Joshua C. Denny

Clinical data captured in electronic medical records accurately identify cases and controls for genome-wide association studies. Where Electronic Records and Genomics Meet There has been a surge of interest in using electronic medical records in hospitals and clinics to capture information about patients that is normally buried in doctors’ handwritten notes. Indeed, the U.S. government has made the implementation of electronic medical records a priority area and has instigated standards for the recording and use of these records. The clinical data captured in electronic medical records including diagnoses, medical tests, and medications provide accurate clinical information that will improve patient care. With the ability to sequence the genomes of individuals faster and cheaper than ever before, it may be possible in the future to include the genome sequences of patients in their electronic medical records. A consortium called the Electronic Medical Records and Genomics Network (eMERGE) has set out to investigate whether clinical data captured in electronic medical records could be used to accurately identify patients with particular diseases for inclusion in genome-wide association studies (GWAS). GWAS scrutinize the genomes of individuals with particular diseases to identify tiny genetic variations that are associated with the risk of developing that disease. Here, the eMERGE consortium reports its study of the electronic medical records from five clinical centers and how accurately it identified patients with one of five diseases: dementia, cataracts, peripheral arterial disease, type 2 diabetes, and cardiac conduction defects. The investigators show that even though the electronic medical records were of different types and did not all use natural language processing to extract information from the records, they were able to obtain robust positive and negative values for identifying patients with these diseases with sufficient accuracy for use in GWAS. They conclude that widespread adoption of electronic medical records will provide real-world clinical data that will be valuable for GWAS and other types of genetic research. Clinical data in electronic medical records (EMRs) are a potential source of longitudinal clinical data for research. The Electronic Medical Records and Genomics Network (eMERGE) investigates whether data captured through routine clinical care using EMRs can identify disease phenotypes with sufficient positive and negative predictive values for use in genome-wide association studies (GWAS). Using data from five different sets of EMRs, we have identified five disease phenotypes with positive predictive values of 73 to 98% and negative predictive values of 98 to 100%. Most EMRs captured key information (diagnoses, medications, laboratory tests) used to define phenotypes in a structured format. We identified natural language processing as an important tool to improve case identification rates. Efforts and incentives to increase the implementation of interoperable EMRs will markedly improve the availability of clinical data for genomics research.


PLOS Genetics | 2009

NRXN3 Is a Novel Locus for Waist Circumference: A Genome-Wide Association Study from the CHARGE Consortium

Nancy L. Heard-Costa; M. Carola Zillikens; Keri L. Monda; Åsa Johansson; Tamara B. Harris; Mao Fu; Talin Haritunians; Mary F. Feitosa; Thor Aspelund; Gudny Eiriksdottir; Melissa Garcia; Lenore J. Launer; Albert V. Smith; Braxton D. Mitchell; Patrick F. McArdle; Alan R. Shuldiner; Suzette J. Bielinski; Eric Boerwinkle; Fred Brancati; Ellen W. Demerath; James S. Pankow; Alice M. Arnold; Yii-Der I. Chen; Nicole L. Glazer; Barbara McKnight; Bruce M. Psaty; Jerome I. Rotter; Najaf Amin; Harry Campbell; Ulf Gyllensten

Central abdominal fat is a strong risk factor for diabetes and cardiovascular disease. To identify common variants influencing central abdominal fat, we conducted a two-stage genome-wide association analysis for waist circumference (WC). In total, three loci reached genome-wide significance. In stage 1, 31,373 individuals of Caucasian descent from eight cohort studies confirmed the role of FTO and MC4R and identified one novel locus associated with WC in the neurexin 3 gene [NRXN3 (rs10146997, p = 6.4×10−7)]. The association with NRXN3 was confirmed in stage 2 by combining stage 1 results with those from 38,641 participants in the GIANT consortium (p = 0.009 in GIANT only, p = 5.3×10−8 for combined analysis, n = 70,014). Mean WC increase per copy of the G allele was 0.0498 z-score units (0.65 cm). This SNP was also associated with body mass index (BMI) [p = 7.4×10−6, 0.024 z-score units (0.10 kg/m2) per copy of the G allele] and the risk of obesity (odds ratio 1.13, 95% CI 1.07–1.19; p = 3.2×10−5 per copy of the G allele). The NRXN3 gene has been previously implicated in addiction and reward behavior, lending further evidence that common forms of obesity may be a central nervous system-mediated disorder. Our findings establish that common variants in NRXN3 are associated with WC, BMI, and obesity.


Journal of the American Medical Informatics Association | 2012

Use of diverse electronic medical record systems to identify genetic risk for type 2 diabetes within a genome-wide association study

Abel N. Kho; M. Geoffrey Hayes; Laura J. Rasmussen-Torvik; Jennifer A. Pacheco; William K. Thompson; Loren L. Armstrong; Joshua C. Denny; Peggy L. Peissig; Aaron W. Miller; Wei Qi Wei; Suzette J. Bielinski; Christopher G. Chute; Cynthia L. Leibson; Gail P. Jarvik; David R. Crosslin; Christopher S. Carlson; Katherine M. Newton; Wendy A. Wolf; Rex L. Chisholm; William L. Lowe

OBJECTIVE Genome-wide association studies (GWAS) require high specificity and large numbers of subjects to identify genotype-phenotype correlations accurately. The aim of this study was to identify type 2 diabetes (T2D) cases and controls for a GWAS, using data captured through routine clinical care across five institutions using different electronic medical record (EMR) systems. MATERIALS AND METHODS An algorithm was developed to identify T2D cases and controls based on a combination of diagnoses, medications, and laboratory results. The performance of the algorithm was validated at three of the five participating institutions compared against clinician review. A GWAS was subsequently performed using cases and controls identified by the algorithm, with samples pooled across all five institutions. RESULTS The algorithm achieved 98% and 100% positive predictive values for the identification of diabetic cases and controls, respectively, as compared against clinician review. By standardizing and applying the algorithm across institutions, 3353 cases and 3352 controls were identified. Subsequent GWAS using data from five institutions replicated the TCF7L2 gene variant (rs7903146) previously associated with T2D. DISCUSSION By applying stringent criteria to EMR data collected through routine clinical care, cases and controls for a GWAS were identified that subsequently replicated a known genetic variant. The use of standard terminologies to define data elements enabled pooling of subjects and data across five different institutions to achieve the robust numbers required for GWAS. CONCLUSIONS An algorithm using commonly available data from five different EMR can accurately identify T2D cases and controls for genetic study across multiple institutions.


Journal of the American Medical Informatics Association | 2013

Validation of electronic medical record-based phenotyping algorithms: results and lessons learned from the eMERGE network

Katherine M. Newton; Peggy L. Peissig; Abel N. Kho; Suzette J. Bielinski; Richard L. Berg; Vidhu Choudhary; Melissa A. Basford; Christopher G. Chute; Iftikhar J. Kullo; Rongling Li; Jennifer A. Pacheco; Luke V. Rasmussen; Leslie Spangler; Joshua C. Denny

BACKGROUND Genetic studies require precise phenotype definitions, but electronic medical record (EMR) phenotype data are recorded inconsistently and in a variety of formats. OBJECTIVE To present lessons learned about validation of EMR-based phenotypes from the Electronic Medical Records and Genomics (eMERGE) studies. MATERIALS AND METHODS The eMERGE network created and validated 13 EMR-derived phenotype algorithms. Network sites are Group Health, Marshfield Clinic, Mayo Clinic, Northwestern University, and Vanderbilt University. RESULTS By validating EMR-derived phenotypes we learned that: (1) multisite validation improves phenotype algorithm accuracy; (2) targets for validation should be carefully considered and defined; (3) specifying time frames for review of variables eases validation time and improves accuracy; (4) using repeated measures requires defining the relevant time period and specifying the most meaningful value to be studied; (5) patient movement in and out of the health plan (transience) can result in incomplete or fragmented data; (6) the review scope should be defined carefully; (7) particular care is required in combining EMR and research data; (8) medication data can be assessed using claims, medications dispensed, or medications prescribed; (9) algorithm development and validation work best as an iterative process; and (10) validation by content experts or structured chart review can provide accurate results. CONCLUSIONS Despite the diverse structure of the five EMRs of the eMERGE sites, we developed, validated, and successfully deployed 13 electronic phenotype algorithms. Validation is a worthwhile process that not only measures phenotype performance but also strengthens phenotype algorithm definitions and enhances their inter-institutional sharing.


Mayo Clinic Proceedings | 2014

Preemptive genotyping for personalized medicine: design of the right drug, right dose, right time-using genomic data to individualize treatment protocol.

Suzette J. Bielinski; Janet E. Olson; Jyotishman Pathak; Richard M. Weinshilboum; Liewei Wang; Kelly Lyke; Euijung Ryu; Paul V. Targonski; Michael D. Van Norstrand; Matthew A. Hathcock; Paul Y. Takahashi; Jennifer B. McCormick; Kiley J. Johnson; Karen J. Maschke; Carolyn R. Rohrer Vitek; Marissa S. Ellingson; Eric D. Wieben; Gianrico Farrugia; Jody A. Morrisette; Keri J. Kruckeberg; Jamie K. Bruflat; Lisa M. Peterson; Joseph H. Blommel; Jennifer M. Skierka; Matthew J. Ferber; John L. Black; Linnea M. Baudhuin; Eric W. Klee; Jason L. Ross; Tamra L. Veldhuizen

OBJECTIVE To report the design and implementation of the Right Drug, Right Dose, Right Time-Using Genomic Data to Individualize Treatment protocol that was developed to test the concept that prescribers can deliver genome-guided therapy at the point of care by using preemptive pharmacogenomics (PGx) data and clinical decision support (CDS) integrated into the electronic medical record (EMR). PATIENTS AND METHODS We used a multivariate prediction model to identify patients with a high risk of initiating statin therapy within 3 years. The model was used to target a study cohort most likely to benefit from preemptive PGx testing among the Mayo Clinic Biobank participants, with a recruitment goal of 1000 patients. We used a Cox proportional hazards model with variables selected through the Lasso shrinkage method. An operational CDS model was adapted to implement PGx rules within the EMR. RESULTS The prediction model included age, sex, race, and 6 chronic diseases categorized by the Clinical Classifications Software for International Classification of Diseases, Ninth Revision codes (dyslipidemia, diabetes, peripheral atherosclerosis, disease of the blood-forming organs, coronary atherosclerosis and other heart diseases, and hypertension). Of the 2000 Biobank participants invited, 1013 (51%) provided blood samples, 256 (13%) declined participation, 555 (28%) did not respond, and 176 (9%) consented but did not provide a blood sample within the recruitment window (October 4, 2012, through March 20, 2013). Preemptive PGx testing included CYP2D6 genotyping and targeted sequencing of 84 PGx genes. Synchronous real-time CDS was integrated into the EMR and flagged potential patient-specific drug-gene interactions and provided therapeutic guidance. CONCLUSION This translational project provides an opportunity to begin to evaluate the impact of preemptive sequencing and EMR-driven genome-guided therapy. These interventions will improve understanding and implementation of genomic data in clinical practice.


Circulation | 2011

Atrial Fibrillation and Death After Myocardial Infarction A Community Study

Patricia Jabre; Xavier Jouven; Frédéric Adnet; Gabriel Thabut; Suzette J. Bielinski; Susan A. Weston; Véronique L. Roger

Background— Atrial fibrillation (AF) often coexists with myocardial infarction (MI), yet its prognostic influence is disputed. Prior reports studied the relationship of AF during early hospitalization for acute MI to the risk of death and could not address the timing of AF in relation to the MI (ie, before, during, after). Furthermore, as data come mostly from clinical trials, their applicability to the community is uncertain. The aims of our study were to assess the occurrence of AF among MI patients, determine whether it has changed over time, and quantify its impact and the impact of its timing on mortality after MI. Methods and Results— This was a community-based cohort of 3220 patients hospitalized with incident (first-ever) MI from 1983 to 2007 in Olmsted County, MN. Atrial fibrillation was identified by diagnostic codes and ECG. Outcomes were all-cause and cardiovascular death. Atrial fibrillation before MI was identified in 304 patients, and 729 developed AF after MI (218 [30%] within 2 days, 119 [16%] between 3 and 30 days, and 392 [54%] >30 days post-MI). The cumulative incidence of AF after MI at 5 years was 19% and did not change over the calendar year of MI (the incidence of AF was the same regardless of when the MI occurred). During a mean follow-up of 6.6 years, 1638 deaths occurred. AF was associated with an increased risk of death (hazard ratio [95% confidence interval] 3.77 [3.37 to 4.21]), independently of clinical characteristics at the time of MI and heart failure. This risk differed markedly according to the timing of AF, and was the greatest for AF occurring >30 days post MI (hazard ratio [95% confidence interval] 1.63 [1.37 to 1.93] for AF within 2 days, 1.81 [0.45 to 2.27] for AF between 3 and 30 days, and 2.58 [2.21 to 3.00] for AF >30 days post MI). Conclusions— In the community, AF is frequent in the setting of MI. Atrial fibrillation carries an excess risk of death, which is the highest for AF developing >30 days after MI.


Clinical Pharmacology & Therapeutics | 2014

Design and anticipated outcomes of the eMERGE-PGx project: a multicenter pilot for preemptive pharmacogenomics in electronic health record systems.

Laura J. Rasmussen-Torvik; Sarah Stallings; Adam S. Gordon; Berta Almoguera; Melissa A. Basford; Suzette J. Bielinski; Ariel Brautbar; Murray H. Brilliant; David Carrell; John J. Connolly; David R. Crosslin; Kimberly F. Doheny; Carlos J. Gallego; Omri Gottesman; Daniel Seung Kim; Kathleen A. Leppig; Rongling Li; Simon Lin; Shannon Manzi; Ana R. Mejia; Jennifer A. Pacheco; Vivian Pan; Jyotishman Pathak; Cassandra Perry; Josh F. Peterson; Cynthia A. Prows; James D. Ralston; Luke V. Rasmussen; Marylyn D. Ritchie; Senthilkumar Sadhasivam

We describe here the design and initial implementation of the eMERGE‐PGx project. eMERGE‐PGx, a partnership of the Electronic Medical Records and Genomics Network and the Pharmacogenomics Research Network, has three objectives: (i) to deploy PGRNseq, a next‐generation sequencing platform assessing sequence variation in 84 proposed pharmacogenes, in nearly 9,000 patients likely to be prescribed drugs of interest in a 1‐ to 3‐year time frame across several clinical sites; (ii) to integrate well‐established clinically validated pharmacogenetic genotypes into the electronic health record with associated clinical decision support and to assess process and clinical outcomes of implementation; and (iii) to develop a repository of pharmacogenetic variants of unknown significance linked to a repository of electronic health record–based clinical phenotype data for ongoing pharmacogenomics discovery. We describe site‐specific project implementation and anticipated products, including genetic variant and phenotype data repositories, novel variant association studies, clinical decision support modules, clinical and process outcomes, approaches to managing incidental findings, and patient and clinician education methods.


JAMA | 2016

Association of Arrhythmia-Related Genetic Variants With Phenotypes Documented in Electronic Medical Records.

Sara L. Van Driest; Quinn S. Wells; Sarah Stallings; William S. Bush; Adam S. Gordon; Deborah A. Nickerson; Jerry H. Kim; David R. Crosslin; Gail P. Jarvik; David Carrell; James D. Ralston; Eric B. Larson; Suzette J. Bielinski; Janet E. Olson; Zi Ye; Iftikhar J. Kullo; Noura S. Abul-Husn; Stuart A. Scott; Erwin P. Bottinger; Berta Almoguera; John J. Connolly; Rosetta M. Chiavacci; Hakon Hakonarson; Laura J. Rasmussen-Torvik; Vivian Pan; Stephen D. Persell; Maureen E. Smith; Rex L. Chisholm; Terrie Kitchner; Max M. He

IMPORTANCE Large-scale DNA sequencing identifies incidental rare variants in established Mendelian disease genes, but the frequency of related clinical phenotypes in unselected patient populations is not well established. Phenotype data from electronic medical records (EMRs) may provide a resource to assess the clinical relevance of rare variants. OBJECTIVE To determine the clinical phenotypes from EMRs for individuals with variants designated as pathogenic by expert review in arrhythmia susceptibility genes. DESIGN, SETTING, AND PARTICIPANTS This prospective cohort study included 2022 individuals recruited for nonantiarrhythmic drug exposure phenotypes from October 5, 2012, to September 30, 2013, for the Electronic Medical Records and Genomics Network Pharmacogenomics project from 7 US academic medical centers. Variants in SCN5A and KCNH2, disease genes for long QT and Brugada syndromes, were assessed for potential pathogenicity by 3 laboratories with ion channel expertise and by comparison with the ClinVar database. Relevant phenotypes were determined from EMRs, with data available from 2002 (or earlier for some sites) through September 10, 2014. EXPOSURES One or more variants designated as pathogenic in SCN5A or KCNH2. MAIN OUTCOMES AND MEASURES Arrhythmia or electrocardiographic (ECG) phenotypes defined by International Classification of Diseases, Ninth Revision (ICD-9) codes, ECG data, and manual EMR review. RESULTS Among 2022 study participants (median age, 61 years [interquartile range, 56-65 years]; 1118 [55%] female; 1491 [74%] white), a total of 122 rare (minor allele frequency <0.5%) nonsynonymous and splice-site variants in 2 arrhythmia susceptibility genes were identified in 223 individuals (11% of the study cohort). Forty-two variants in 63 participants were designated potentially pathogenic by at least 1 laboratory or ClinVar, with low concordance across laboratories (Cohen κ = 0.26). An ICD-9 code for arrhythmia was found in 11 of 63 (17%) variant carriers vs 264 of 1959 (13%) of those without variants (difference, +4%; 95% CI, -5% to +13%; P = .35). In the 1270 (63%) with ECGs, corrected QT intervals were not different in variant carriers vs those without (median, 429 vs 439 milliseconds; difference, -10 milliseconds; 95% CI, -16 to +3 milliseconds; P = .17). After manual review, 22 of 63 participants (35%) with designated variants had any ECG or arrhythmia phenotype, and only 2 had corrected QT interval longer than 500 milliseconds. CONCLUSIONS AND RELEVANCE Among laboratories experienced in genetic testing for cardiac arrhythmia disorders, there was low concordance in designating SCN5A and KCNH2 variants as pathogenic. In an unselected population, the putatively pathogenic genetic variants were not associated with an abnormal phenotype. These findings raise questions about the implications of notifying patients of incidental genetic findings.


Journal of the American College of Cardiology | 2012

Electrophysiological and hemodynamic characteristics associated with obesity in patients with atrial fibrillation.

Thomas M. Munger; Ying Xue Dong; Mitsuru Masaki; Jae K. Oh; Sunil Mankad; Barry A. Borlaug; Samuel J. Asirvatham; Win Kuang Shen; Hon Chi Lee; Suzette J. Bielinski; David O. Hodge; Regina M. Herges; Traci L. Buescher; Jia Hui Wu; Chang-Sheng Ma; Yanhua Zhang; Peng Sheng Chen; Douglas L. Packer; Yong Mei Cha

OBJECTIVES The authors sought to characterize the left atrial (LA) and pulmonary vein (PV) electrophysiological and hemodynamic features in obese patients with atrial fibrillation (AF). BACKGROUND Obesity is associated with increased risk for AF. METHODS A total of 63 consecutive patients with AF who had normal left ventricular (LV) ejection fraction and who underwent catheter ablation were studied. Atrial and PV electrophysiological studies were performed at the time of ablation with hemodynamic assessment by cardiac catheterization, and LA/LV structure and function by echocardiography. Patients were compared on the basis of body mass index (BMI): <25 kg/m(2) (n = 19) and BMI ≥30 kg/m(2) (n = 44). RESULTS At a 600-ms pacing cycle length, obese patients had shorter effective refractory period (ERP) in the left atrium (251 ± 25 ms vs. 233 ± 32 ms, p = 0.04), and in the proximal (207 ± 33 ms vs. 248 ± 34 ms, p < 0.001) and distal (193 ± 33 ms vs. 248 ± 44 ms, p < 0.001) PV than normal BMI patients. Obese patients had higher mean LA pressure (15 ± 5 mm Hg vs. 10 ± 5 mm Hg, p < 0.001) and LA volume index (28 ± 12 ml/m(2) vs. 21 ± 14 ml/m(2), p = 0.006), and lower LA strain (5.5 ± 3.1% vs. 8.8 ± 2.8%; p < 0.001) than normal BMI patients. CONCLUSIONS Increased LA pressure and volume, and shortened ERP in the left atrium and PV are potential factors facilitating and perpetuating AF in obese patients with AF.

Collaboration


Dive into the Suzette J. Bielinski's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge