Network


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

Hotspot


Dive into the research topics where Li-Shiun Chen is active.

Publication


Featured researches published by Li-Shiun Chen.


American Journal of Psychiatry | 2012

Interplay of genetic risk factors (CHRNA5-CHRNA3-CHRNB4) and cessation treatments in smoking cessation success.

Li-Shiun Chen; Timothy B. Baker; Megan E. Piper; Naomi Breslau; Dale S. Cannon; Kimberly F. Doheny; Stephanie M. Gogarten; Eric O. Johnson; Nancy L. Saccone; Jen C. Wang; Robert B. Weiss; Alison M. Goate; Laura J. Bierut

OBJECTIVE Smoking is highly intractable, and the genetic influences on cessation are unclear. Identifying the genetic factors affecting smoking cessation could elucidate the nature of tobacco dependence, enhance risk assessment, and support development of treatment algorithms. This study tested whether variants in the nicotinic receptor gene cluster CHRNA5-CHRNA3-CHRNB4 predict age at smoking cessation and relapse after an attempt to quit smoking. METHOD In a community-based, crosssectional study (N=5,216) and a randomized comparative effectiveness smoking cessation trial (N=1,073), the authors used Cox proportional hazard models and logistic regression to model the relationships of smoking cessation (self-reported quit age in the community study and point-prevalence abstinence at the end of treatment in the clinical trial) to three common haplotypes in the CHRNA5-CHRNA3-CHRNB4 region defined by rs16969968 and rs680244. RESULTS The genetic variants in the CHRNA5-CHRNA3-CHRNB4 region that predict nicotine dependence also predicted a later age at smoking cessation in the community sample. In the smoking cessation trial, haplotype predicted abstinence at end of treatment in individuals receiving placebo but not among individuals receiving active medication. Haplotype interacted with treatment in affecting cessation success. CONCLUSIONS Smokers with the high-risk haplotype were three times as likely to respond to pharmacologic cessation treatments as were smokers with the low-risk haplotype. The high-risk haplotype increased the risk of cessation failure, and this increased risk was ameliorated by cessation pharmacotherapy. By identifying a high-risk genetic group with heightened response to smoking cessation pharmacotherapy, this work may support the development of personalized cessation treatments.


Cancer Epidemiology, Biomarkers & Prevention | 2009

Association of folate-pathway gene polymorphisms with the risk of prostate cancer: a population-based nested case-control study, systematic review, and meta-analysis.

Simon M Collin; Chris Metcalfe; Luisa Zuccolo; Sarah J Lewis; Li-Shiun Chen; Angela Cox; M. Davis; J A Lane; Jenny Donovan; George Davey Smith; David E. Neal; F C Hamdy; Julius Gudmundsson; Patrick Sulem; Thorunn Rafnar; K R Benediktsdottir; Rosalind Eeles; Michelle Guy; Zsofia Kote-Jarai; Jonathan Morrison; Ali Amin Al Olama; Kari Stefansson; Doug Easton; Richard M. Martin

Folate-pathway gene polymorphisms have been implicated in several cancers and investigated inconclusively in relation to prostate cancer. We conducted a systematic review, which identified nine case-control studies (eight included, one excluded). We also included data from four genome-wide association studies and from a case-control study nested within the UK population–based Prostate Testing for Cancer and Treatment study. We investigated by meta-analysis the effects of eight polymorphisms: MTHFR C677T (rs1801133; 12 studies; 10,745 cases; 40,158 controls), MTHFR A1298C (rs1801131; 5 studies; 3,176 cases; 4,829 controls), MTR A2756G (rs1805087; 8 studies; 7,810 cases; 37,543 controls), MTRR A66G (rs1801394; 4 studies; 3,032 cases; 4,515 controls), MTHFD1 G1958A (rs2236225; 6 studies; 7,493 cases; 36,941 controls), SLC19A1/RFC1 G80A (rs1051266; 4 studies; 6,222 cases; 35,821 controls), SHMT1 C1420T (rs1979277; 2 studies; 2,689 cases; 4,110 controls), and FOLH1 T1561C (rs202676; 5 studies; 6,314 cases; 35,190 controls). The majority (10 of 13) of eligible studies had 100% Caucasian subjects; only one study had <90% Caucasian subjects. We found weak evidence of dominant effects of two alleles: MTR 2756A>G [random effects pooled odds ratio, 1.06 (1.00-1.12); P = 0.06 (P = 0.59 for heterogeneity across studies)] and SHMT1 1420C>T [random effects pooled odds ratio, 1.11 (1.00-1.22); P = 0.05 (P = 0.38 for heterogeneity across studies)]. We found no effect of MTHFR 677C>T or any of the other alleles in dominant, recessive or additive models, or in comparing a/a versus A/A homozygous. Neither did we find any difference in effects on advanced or localized cancers. Our meta-analysis suggests that known common folate-pathway single nucleotide polymorphisms do not have significant effects on susceptibility to prostate cancer.(Cancer Epidemiol Biomarkers Prev 2009;18(9):2528–39)


Genetic Epidemiology | 2012

Smoking and genetic risk variation across populations of European, Asian, and African American ancestry--a meta-analysis of chromosome 15q25.

Li-Shiun Chen; Nancy L. Saccone; Robert Culverhouse; Paige M. Bracci; Chien-Hsiun Chen; Nicole Dueker; Younghun Han; Hongyan Huang; Guangfu Jin; Takashi Kohno; Jennie Z. Ma; Thomas R. Przybeck; Alan R. Sanders; Jennifer A. Smith; Yun Ju Sung; Angie S. Wenzlaff; Chen Wu; Dankyu Yoon; Ying Ting Chen; Yu Ching Cheng; Yoon Shin Cho; Sean P. David; Jubao Duan; Charles B. Eaton; Helena Furberg; Alison Goate; Dongfeng Gu; Helen M. Hansen; Sarah M. Hartz; Zhibin Hu

Recent meta‐analyses of European ancestry subjects show strong evidence for association between smoking quantity and multiple genetic variants on chromosome 15q25. This meta‐analysis extends the examination of association between distinct genes in the CHRNA5‐CHRNA3‐CHRNB4 region and smoking quantity to Asian and African American populations to confirm and refine specific reported associations. Association results for a dichotomized cigarettes smoked per day phenotype in 27 datasets (European ancestry (N = 14,786), Asian (N = 6,889), and African American (N = 10,912) for a total of 32,587 smokers) were meta‐analyzed by population and results were compared across all three populations. We demonstrate association between smoking quantity and markers in the chromosome 15q25 region across all three populations, and narrow the region of association. Of the variants tested, only rs16969968 is associated with smoking (P < 0.01) in each of these three populations (odds ratio [OR] = 1.33, 95% CI = 1.25–1.42, P = 1.1 × 10−17 in meta‐analysis across all population samples). Additional variants displayed a consistent signal in both European ancestry and Asian datasets, but not in African Americans. The observed consistent association of rs16969968 with heavy smoking across multiple populations, combined with its known biological significance, suggests rs16969968 is most likely a functional variant that alters risk for heavy smoking. We interpret additional association results that differ across populations as providing evidence for additional functional variants, but we are unable to further localize the source of this association. Using the cross‐population study paradigm provides valuable insights to narrow regions of interest and inform future biological experiments. Genet. Epidemiol. 36:340–351, 2012.


Addiction | 2014

Pharmacotherapy effects on smoking cessation vary with nicotine metabolism gene (CYP2A6)

Li-Shiun Chen; A. Joseph Bloom; Timothy B. Baker; Stevens S. Smith; Megan E. Piper; Maribel Martinez; Nancy L. Saccone; Dorothy K. Hatsukami; Alison Goate; Laura J. Bierut

BACKGROUND AND AIMS Evidence suggests that both the nicotinic receptor α5 subunit (CHRNA5) and Cytochrome P450 2A6 (CYP2A6) genotypes influence smoking cessation success and response to pharmacotherapy. We examine the effect of CYP2A6 genotype on smoking cessation success and response to cessation pharmacotherapy, and combine these effects with those of CHRNA5 genotypes. DESIGN Placebo-controlled randomized smoking cessation trial. SETTING Ambulatory care facility in Wisconsin, USA. PARTICIPANTS Smokers (n = 709) of European ancestry were randomized to placebo, bupropion, nicotine replacement therapy or combined bupropion and nicotine replacement therapy. MEASUREMENTS Survival analysis was used to model time to relapse using nicotine metabolism derived from CYP2A6 genotype-based estimates. Slow metabolism is defined as the lowest quartile of estimated metabolic function. FINDINGS CYP2A6-defined nicotine metabolic function moderated the effect of smoking cessation pharmacotherapy on smoking relapse over 90 days [hazard ratio (HR) = 2.81, 95% confidence interval (CI) = 1.32-5.99, P = 0.0075], with pharmacotherapy significantly slowing relapse in fast (HR = 0.39, 95% CI = 0.28-0.55, P = 1.97 × 10(-8)), but not slow metabolizers (HR = 1.09, 95% CI = 0.55-2.17, P = 0.80). Further, only the effect of nicotine replacement, and not bupropion, varies with CYP2A6-defined metabolic function. The effect of nicotine replacement on continuous abstinence is moderated by the combined genetic risks from CYP2A6 and CHRNA5 (Wald = 7.44, d.f. = 1, P = 0.0064). CONCLUSIONS Nicotine replacement therapy is effective among individuals with fast, but not slow, CYP2A6-defined nicotine metabolism. The effect of bupropion on relapse likelihood is unlikely to be affected by nicotine metabolism as estimated from CYP2A6 genotype. The variation in treatment responses among smokers with genes may guide future personalized smoking cessation interventions.


Journal of the National Cancer Institute | 2015

CHRNA5 Risk Variant Predicts Delayed Smoking Cessation and Earlier Lung Cancer Diagnosis—A Meta-Analysis

Li-Shiun Chen; Rayjean J. Hung; Timothy B. Baker; Amy C. Horton; Rob Culverhouse; Nancy L. Saccone; Iona Cheng; Bo Deng; Younghun Han; Helen M. Hansen; Janet Horsman; Claire H. Kim; Sharon M. Lutz; Albert Rosenberger; Katja K. Aben; Angeline S. Andrew; Naomi Breslau; Shen Chih Chang; Aida Karina Dieffenbach; Hendrik Dienemann; Brittni Frederiksen; Jiali Han; Dorothy K. Hatsukami; Eric O. Johnson; Mala Pande; Margaret Wrensch; John McLaughlin; Vidar Skaug; Henricus F. M. van der Heijden; Jason A. Wampfler

BACKGROUND Recent meta-analyses show strong evidence of associations among genetic variants in CHRNA5 on chromosome 15q25, smoking quantity, and lung cancer. This meta-analysis tests whether the CHRNA5 variant rs16969968 predicts age of smoking cessation and age of lung cancer diagnosis. METHODS Meta-analyses examined associations between rs16969968, age of quitting smoking, and age of lung cancer diagnosis in 24 studies of European ancestry (n = 29 072). In each dataset, we used Cox regression models to evaluate the association between rs16969968 and the two primary phenotypes (age of smoking cessation among ever smokers and age of lung cancer diagnosis among lung cancer case patients) and the secondary phenotype of smoking duration. Heterogeneity across studies was assessed with the Cochran Q test. All statistical tests were two-sided. RESULTS The rs16969968 allele (A) was associated with a lower likelihood of smoking cessation (hazard ratio [HR] = 0.95, 95% confidence interval [CI] = 0.91 to 0.98, P = .0042), and the AA genotype was associated with a four-year delay in median age of quitting compared with the GG genotype. Among smokers with lung cancer diagnoses, the rs16969968 genotype (AA) was associated with a four-year earlier median age of diagnosis compared with the low-risk genotype (GG) (HR = 1.08, 95% CI = 1.04 to 1.12, P = 1.1*10(-5)). CONCLUSION These data support the clinical significance of the CHRNA5 variant rs16969968. It predicts delayed smoking cessation and an earlier age of lung cancer diagnosis in this meta-analysis. Given the existing evidence that this CHRNA5 variant predicts favorable response to cessation pharmacotherapy, these findings underscore the potential clinical and public health importance of rs16969968 in CHRNA5 in relation to smoking cessation success and lung cancer risk.


Nicotine & Tobacco Research | 2011

Dissection of the Phenotypic and Genotypic Associations With Nicotinic Dependence

Li-Shiun Chen; Timothy B. Baker; Richard A. Grucza; Jen C. Wang; Eric O. Johnson; Naomi Breslau; Dorothy K. Hatsukami; Stevens S. Smith; Nancy L. Saccone; Scott F. Saccone; John P. Rice; Alison Goate; Laura J. Bierut

INTRODUCTION Strong evidence demonstrates that nicotine dependence is associated with 4 genetic variants rs16969968, rs6474412, rs3733829, and rs1329650 in large-scale Genome-Wide Association Studies. We examined how these identified genetic variants relate to nicotine dependence defined by different categorical and dimensional measures. METHODS Four genetic variants were analyzed in 2,047 subjects of European descent (1,062 cases and 985 controls). Nicotine dependence was assessed with multiple smoking measures, including the Fagerström Test for Nicotine Dependence, the Diagnostic and Statistical Manual for Mental Disorders-IV (DSM-IV) nicotine dependence, the Nicotine Dependence Syndrome Scale, and the Wisconsin Inventory of Smoking Dependence Motives. Single-item measures of cigarettes per day (CPD) and time to first cigarette (TTF) in the morning were also examined. RESULTS Among the variants, association effect sizes were largest for rs16969968, with measures of craving and heavy smoking, especially cigarettes smoked per day, showing the largest effects. Significant but weaker associations were found for rs6474412 and rs3733729 but not for rs1329650. None of the more comprehensive measures of smoking behaviors yielded stronger genetic associations with these variants than did CPD. CONCLUSIONS CPD is an important simple measure that captures in part the genetic associations of CHRNA5 and nicotine dependence, even when other more comprehensive measures of smoking behaviors are examined. The CHRNA5 gene is associated with heavy compulsive smoking and craving; this should inform the mission to improve the diagnostic validity of DSM-V.


Addiction Biology | 2010

Incorporating age at onset of smoking into genetic models for nicotine dependence: evidence for interaction with multiple genes.

Richard A. Grucza; Eric O. Johnson; Robert F. Krueger; Naomi Breslau; Nancy L. Saccone; Li-Shiun Chen; Jaime Derringer; Arpana Agrawal; Michael T. Lynskey; Laura J. Bierut

Nicotine dependence is moderately heritable, but identified genetic associations explain only modest portions of this heritability. We analyzed 3369 SNPs from 349 candidate genes and investigated whether incorporation of SNP‐by‐environment interaction into association analyses might bolster gene discovery efforts and prediction of nicotine dependence. Specifically, we incorporated the interaction between allele count and age at onset of regular smoking (AOS) into association analyses of nicotine dependence. Subjects were from the Collaborative Genetic Study of Nicotine Dependence and included 797 cases ascertained for Fagerström nicotine dependence and 811 non‐nicotine‐dependent smokers as controls, all of European descent. Compared with main effect models, SNP × AOS interaction models resulted in higher numbers of nominally significant tests, increased predictive utility at individual SNPs and higher predictive utility in a multi‐locus model. Some SNPs previously documented in main effect analyses exhibited improved fits in the joint analysis, including rs16969968 from CHRNA5 and rs2314379 from MAP3K4. CHRNA5 exhibited larger effects in later‐onset smokers, in contrast with a previous report that suggested the opposite interaction ( Weiss et al. 2008 ). However, a number of SNPs that did not emerge in main effect analyses were among the strongest findings in the interaction analyses. These include SNPs located in GRIN2B (P = 1.5 × 10−5), which encodes a subunit of the N‐methyl‐D‐aspartate receptor channel, a key molecule in mediating age‐dependent synaptic plasticity. Incorporation of logically chosen interaction parameters, such as AOS, into genetic models of substance use disorders may increase the degree of explained phenotypic variation and constitutes a promising avenue for gene discovery.


Annals of the American Thoracic Society | 2014

Beyond Cigarettes Per Day. A Genome-Wide Association Study of the Biomarker Carbon Monoxide

A. Joseph Bloom; Sarah M. Hartz; Timothy B. Baker; Li-Shiun Chen; Megan E. Piper; Louis Fox; Maribel Martinez; Dorothy K. Hatsukami; Eric O. Johnson; Cathy C. Laurie; Nancy L. Saccone; Alison Goate; Laura J. Bierut

RATIONALE The CHRNA5-CHRNA3-CHRNB4 locus is associated with self-reported smoking behavior and also harbors the strongest genetic associations with chronic obstructive pulmonary disease (COPD) and lung cancer. Because the associations with lung disease remain after adjustment for self-reported smoking behaviors, it has been asserted that CHRNA5-CHRNA3-CHRNB4 variants increase COPD and lung cancer susceptibility independently of their effects on smoking. OBJECTIVES To compare the genetic associations of exhaled carbon monoxide (CO), a biomarker of current cigarette exposure, with self-reported smoking behaviors. METHODS A total of 1,521 European American and 247 African American current smokers recruited into smoking cessation studies were assessed for CO at intake before smoking cessation. DNA samples were genotyped using the Illumina Omni2.5 microarray. Genetic associations with CO and smoking behaviors (cigarettes smoked per day, Fagerstrom test for nicotine dependence) were studied. MEASUREMENTS AND MAIN RESULTS Variants in the CHRNA5-CHRNA3-CHRNB4 locus, including rs16969968, a nonsynonymous variant in CHRNA5, are genomewide association study-significantly associated with CO (β = 2.66; 95% confidence interval [CI], 1.74-3.58; P = 1.65 × 10(-8)), and this association remains strong after adjusting for smoking behavior (β = 2.18; 95% CI, 1.32-3.04; P = 7.47 × 10(-7)). The correlation between CO and cigarettes per day is statistically significantly lower (z = 3.43; P = 6.07 × 10(-4)) in African Americans (r = 0.14; 95% CI, 0.02-0.26; P = 0.003) than in European-Americans (r = 0.36; 95% CI, 0.31-0.40; P = 0.0001). CONCLUSIONS Exhaled CO, a biomarker that is simple to measure, captures aspects of cigarette smoke exposure in current smokers beyond the number of cigarettes smoked per day. Behavioral measures of smoking are therefore insufficient indices of cigarette smoke exposure, suggesting that genetic associations with COPD or lung cancer that persist after adjusting for self-reported smoking behavior may still reflect genetic effects on smoking exposure.


Molecular Psychiatry | 2016

Rare, low frequency and common coding variants in CHRNA5 and their contribution to nicotine dependence in European and African Americans

Emily Olfson; Nancy L. Saccone; E. O. Johnson; Li-Shiun Chen; Robert Culverhouse; Kimberly F. Doheny; S. M. Foltz; Louis Fox; Stephanie M. Gogarten; Sarah M. Hartz; K. Hetrick; Cathy C. Laurie; B. Marosy; Najaf Amin; Donna K. Arnett; R. G. Barr; Traci M. Bartz; Sarah Bertelsen; Ingrid B. Borecki; Michael R. Brown; Daniel I. Chasman; C. M. van Duijn; Mary F. Feitosa; Ervin R. Fox; Nora Franceschini; Oscar H. Franco; Megan L. Grove; Xiuqing Guo; A. Hofman; Sharon L.R. Kardia

The common nonsynonymous variant rs16969968 in the α5 nicotinic receptor subunit gene (CHRNA5) is the strongest genetic risk factor for nicotine dependence in European Americans and contributes to risk in African Americans. To comprehensively examine whether other CHRNA5 coding variation influences nicotine dependence risk, we performed targeted sequencing on 1582 nicotine-dependent cases (Fagerström Test for Nicotine Dependence score⩾4) and 1238 non-dependent controls, with independent replication of common and low frequency variants using 12 studies with exome chip data. Nicotine dependence was examined using logistic regression with individual common variants (minor allele frequency (MAF)⩾0.05), aggregate low frequency variants (0.05>MAF⩾0.005) and aggregate rare variants (MAF<0.005). Meta-analysis of primary results was performed with replication studies containing 12 174 heavy and 11 290 light smokers. Next-generation sequencing with 180 × coverage identified 24 nonsynonymous variants and 2 frameshift deletions in CHRNA5, including 9 novel variants in the 2820 subjects. Meta-analysis confirmed the risk effect of the only common variant (rs16969968, European ancestry: odds ratio (OR)=1.3, P=3.5 × 10−11; African ancestry: OR=1.3, P=0.01) and demonstrated that three low frequency variants contributed an independent risk (aggregate term, European ancestry: OR=1.3, P=0.005; African ancestry: OR=1.4, P=0.0006). The remaining 22 rare coding variants were associated with increased risk of nicotine dependence in the European American primary sample (OR=12.9, P=0.01) and in the same risk direction in African Americans (OR=1.5, P=0.37). Our results indicate that common, low frequency and rare CHRNA5 coding variants are independently associated with nicotine dependence risk. These newly identified variants likely influence the risk for smoking-related diseases such as lung cancer.


Drug and Alcohol Dependence | 2015

Genetic variation (CHRNA5), medication (combination nicotine replacement therapy vs. varenicline), and smoking cessation

Li-Shiun Chen; Timothy B. Baker; Douglas E. Jorenby; Megan E. Piper; Nancy L. Saccone; Eric O. Johnson; Naomi Breslau; Dorothy K. Hatsukami; Robert M. Carney; Laura J. Bierut

OBJECTIVE Recent evidence suggests that the efficacy of smoking cessation pharmacotherapy can vary across patients based on their genotypes. This study tests whether the coding variant rs16969968 in the CHRNA5 nicotinic receptor gene predicts the effects of combination nicotine replacement therapy (cNRT) and varenicline on treatment outcomes. METHOD In two randomized smoking cessation trials comparing cNRT vs. placebo, and varenicline vs. placebo, we used logistic regression to model associations between CHRNA5 rs16969968 and abstinence at end of treatment. RESULTS For abstinence at end of treatment, there was an interaction between cNRT and rs16969968 (X(2)=8.15, df=2, omnibus-p=0.017 for the interaction); individuals with the high-risk AA genotype were more likely to benefit from cNRT. In contrast, varenicline increased abstinence, but its effect did not vary with CHRNA5. However, the genetic effects differed between the placebo control groups across two trials (wald=3.94, df=1, p=0.047), this non-replication can alter the interpretation of pharmacogenetic findings. CONCLUSIONS Results from two complementary smoking cessation trials demonstrate inconsistent genetic results in the placebo arms. This evidence highlights the need to compare the most effective pharmacotherapies with the same placebo control to establish pharmacogenetic evidence to aid decisions on medication choice for patients trying to quit smoking.

Collaboration


Dive into the Li-Shiun Chen's collaboration.

Top Co-Authors

Avatar

Laura J. Bierut

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Nancy L. Saccone

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Eric O. Johnson

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Timothy B. Baker

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Sarah M. Hartz

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Naomi Breslau

Michigan State University

View shared research outputs
Top Co-Authors

Avatar

Richard A. Grucza

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Robert Culverhouse

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Alison Goate

Icahn School of Medicine at Mount Sinai

View shared research outputs
Researchain Logo
Decentralizing Knowledge