Network


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

Hotspot


Dive into the research topics where Nan Hu is active.

Publication


Featured researches published by Nan Hu.


Nature Genetics | 2010

A shared susceptibility locus in PLCE1 at 10q23 for gastric adenocarcinoma and esophageal squamous cell carcinoma

Christian C. Abnet; Neal D. Freedman; Nan Hu; Zhaoming Wang; Kai Yu; Xiao-Ou Shu; Jian-Min Yuan; Wei Zheng; Sanford M. Dawsey; Linda M. Dong; Maxwell P. Lee; Ti Ding; You-Lin Qiao; Yu-Tang Gao; Woon-Puay Koh; Yong Bing Xiang; Ze Zhong Tang; Jin Hu Fan; Chaoyu Wang; William Wheeler; Mitchell H. Gail; Meredith Yeager; Jeff Yuenger; Amy Hutchinson; Kevin B. Jacobs; Carol Giffen; Laurie Burdett; Joseph F. Fraumeni; Margaret A. Tucker; Wong Ho Chow

We conducted a genome-wide association study of gastric cancer and esophageal squamous cell carcinoma (ESCC) in ethnic Chinese subjects in which we genotyped 551,152 SNPs. We report a combined analysis of 2,240 gastric cancer cases, 2,115 ESCC cases and 3,302 controls drawn from five studies. In logistic regression models adjusted for age, sex and study, multiple variants at 10q23 had genome-wide significance for gastric cancer and ESCC independently. A notable signal was rs2274223, a nonsynonymous SNP located in PLCE1, for gastric cancer (P = 8.40 × 10−9; per-allele odds ratio (OR) = 1.31) and ESCC (P = 3.85 × 10−9; OR = 1.34). The association with gastric cancer differed by anatomic subsite. For tumors in the cardia the association was stronger (P = 4.19 × 10−15; OR = 1.57), and for those in the noncardia stomach it was absent (P = 0.44; OR = 1.05). Our findings at 10q23 could provide insight into the high incidence of both cancers in China.


Nature Genetics | 2012

Genome-wide association analyses of esophageal squamous cell carcinoma in Chinese identify multiple susceptibility loci and gene-environment interactions

Chen Wu; Peter Kraft; Kan Zhai; Jiang Chang; Zhaoming Wang; Yun Li; Zhibin Hu; Zhonghu He; Weihua Jia; Christian C. Abnet; Liming Liang; Nan Hu; Xiaoping Miao; Yifeng Zhou; Zhihua Liu; Qimin Zhan; Yu Liu; Yan Qiao; Yuling Zhou; Guangfu Jin; Chuanhai Guo; Changdong Lu; Haijun Yang; Jianhua Fu; Dianke Yu; Neal D. Freedman; Ti Ding; Wen Tan; Alisa M. Goldstein; Tangchun Wu

We conducted a genome-wide association study (GWAS) and a genome-wide gene-environment interaction analysis of esophageal squamous-cell carcinoma (ESCC) in 2,031 affected individuals (cases) and 2,044 controls with independent validation in 8,092 cases and 8,620 controls. We identified six new ESCC susceptibility loci, of which four, at chromosomes 4q23, 16q12.1, 22q12 and 3q27 had a significant marginal effect (P = 1.78 × 10−39 to P = 2.49 × 10−11) and two of which, at 2q22 and 13q33, had a significant association only in the gene–alcohol drinking interaction (gene-environment interaction P (PG × E) = 4.39 × 10−11 and PG × E = 4.80 × 10−8, respectively). Variants at the 4q23 locus, which includes the ADH cluster, each had a significant interaction with alcohol drinking in their association with ESCC risk (PG × E = 2.54 × 10−7 to PG × E = 3.23 × 10−2). We confirmed the known association of the ALDH2 locus on 12q24 to ESCC, and a joint analysis showed that drinkers with both of the ADH1B and ALDH2 risk alleles had a fourfold increased risk for ESCC compared to drinkers without these risk alleles. Our results underscore the direct genetic contribution to ESCC risk, as well as the genetic contribution to ESCC through interaction with alcohol consumption.


Gut | 2014

Improved survival of gastric cancer with tumour Epstein–Barr virus positivity: an international pooled analysis

M. Constanza Camargo; Woo Ho Kim; Anna Maria Chiaravalli; Kyoung Mee Kim; Alejandro H. Corvalan; Keitaro Matsuo; Jun Yu; Joseph J.Y. Sung; Roberto Herrera-Goepfert; Fernando Meneses-Gonzalez; Yuko Kijima; Shoji Natsugoe; Linda M. Liao; Jolanta Lissowska; Sung Kim; Nan Hu; Carlos A. González; Y. Yatabe; Chihaya Koriyama; Stephen M. Hewitt; Suminori Akiba; Margaret L. Gulley; Philip R. Taylor; Charles S. Rabkin

Background and objective About 9% of gastric carcinomas have Epstein–Barr virus (EBV) in the tumour cells, but it is unclear whether viral presence influences clinical progression. We therefore examined a large multicentre case series for the association of tumour EBV status with survival after gastric cancer diagnosis, accounting for surgical stage and other prognostic factors. Methods We combined individual-level data on 4599 gastric cancer patients diagnosed between 1976 and 2010 from 13 studies in Asia (n=8), Europe (n=3), and Latin America (n=2). EBV positivity of tumours was assessed by in situ hybridisation. Mortality HRs for EBV positivity were estimated by Cox regression models stratified by study, adjusted for distributions of sex (71% male), age (mean 58u2005years), stage (52% tumour-node-metastasis stages III or IV), tumour histology (49% poorly differentiated, 57% Lauren intestinal-type), anatomic subsite (70% non-cardia) and year of diagnosis. Variations by study and continent were assessed using study-specific HRs for EBV positivity. Results During median 3.0u2005years follow-up, 49% of patients died. Stage was strongly predictive of mortality, with unadjusted HRs (vs stage I) of 3.1 for stage II, 8.1 for stage III and 13.2 for stage IV. Tumour EBV positivity was 8.2% overall and inversely associated with stage (adjusted OR: 0.79 per unit change). Adjusted for stage and other confounders, EBV positivity was associated with lower mortality (HR, 0.72; 95% CI 0.61 to 0.86), with low heterogeneity among the study populations (p=0.2). The association did not significantly vary across patient or tumour characteristics. There was no significant variation among the three continent-specific HRs (p=0.4). Conclusions Our findings suggest that tumour EBV positivity is an additional prognostic indicator in gastric cancer. Further studies are warranted to identify the mechanisms underlying this protective association.


Cancer Research | 2005

Genome-Wide Association Study in Esophageal Cancer Using GeneChip Mapping 10K Array

Nan Hu; Chaoyu Wang; Ying Hu; Howard H. Yang; Carol Giffen; Ze Zhong Tang; Xiao Yu Han; Alisa M. Goldstein; Michael R. Emmert-Buck; Kenneth H. Buetow; Philip R. Taylor; Maxwell P. Lee

Whole genome association studies of complex human diseases represent a new paradigm in the postgenomic era. In this study, we report application of the Affymetrix, Inc. (Santa Clara, CA) high-density single nucleotide polymorphism (SNP) array containing 11,555 SNPs in a pilot case-control study of esophageal squamous cell carcinoma (ESCC) that included the analysis of germ line samples from 50 ESCC patients and 50 matched controls. The average genotyping call rate for the 100 samples analyzed was 96%. Using the generalized linear model (GLM) with adjustment for potential confounders and multiple comparisons, we identified 37 SNPs associated with disease, assuming a recessive mode of transmission; similarly, 48 SNPs were identified assuming a dominant mode and 53 SNPs in a continuous mode. When the 37 SNPs identified from the GLM recessive mode were used in a principal components analysis, the first principal component correctly predicted 46 of 50 cases and 47 of 50 controls. Among all the SNPs selected from GLMs for the three modes of transmission, 39 could be mapped to 1 of 33 genes. Many of these genes are involved in various cancers, including GASC1, shown previously to be amplified in ESCCs, and EPHB1 and PIK3C3. In conclusion, we have shown the feasibility of the Affymetrix 10K SNP array in genome-wide association studies of common cancers and identified new candidate loci to study in ESCC.


Gut | 2016

Genome-wide association study of gastric adenocarcinoma in Asia: a comparison of associations between cardia and non-cardia tumours

Nan Hu; Zhaoming Wang; Xin Song; Lixuan Wei; Byung Sik Kim; Neal D. Freedman; Jiwon Baek; Laurie Burdette; Jiang Chang; Charles C. Chung; Sanford M. Dawsey; Ti Ding; Yu-Tang Gao; Carol Giffen; Yaling Han; Myunghee Hong; Jia Huang; Hee Sung Kim; Woon-Puay Koh; Linda M. Liao; Yi Min Mao; You-Lin Qiao; Xiao-Ou Shu; Wen Tan; Chaoyu Wang; Chen Wu; Min-Jie Wu; Yong-Bing Xiang; Meredith Yeager; Jeong Hwan Yook

Objective Genome-wide association studies (GWAS) of gastric cancer have reported differences in single-nucleotide polymorphism (SNP) associations for tumour subtypes, particularly when divided by location into the gastric cardia versus the non-cardia. Design Here we present results for a GWAS using 2350 East Asian gastric cancer cases divided as 1189 gastric cardia and 1027 gastric non-cardia cases and 2708 controls. We also included up to 3042 cardia cases, 4359 non-cardia cases and 7548 controls for replication from two Chinese studies and one Korean study. From the GWAS, we selected 12 top SNPs for each gastric cancer subtype, 4 top SNPs for total gastric cancer and 1 SNP in MUC1 for replication testing. Results We observed genome-wide significant associations for rs10074991 in PRKAA1 at 5p13.1 for cardia (p=7.36×10−12) and non-cardia cancers (p=2.42×10−23) with per allele OR (95% CI) for the combined endpoint of 0.80 (0.77 to 0.83). At 6p21.1, rs2294693 near UNC5CL was significantly associated with gastric non-cardia cancer risk (p=2.50×10−8), with OR (95% CI) of 1.18 (1.12 to 1.26), but there was only a nominal association for cardia cancer (p=1.47×10−2). We also confirmed a previously reported association for rs4072037 in MUC1 with p=6.59×10−8 for total gastric cancer and similar estimates for cardia and non-cardia cancers. Three SNPs in PSCA previously reported to be associated with gastric non-cardia cancer showed no apparent association for cardia cancer. Conclusions Our results suggest that associations for SNPs with gastric cancer show some different results by tumour location in the stomach.


Clinical Gastroenterology and Hepatology | 2015

Association Between Circulating Levels of Sex Steroid Hormones and Barrett’s Esophagus in Men: A Case–Control Analysis

Michael B. Cook; Shannon N. Wood; Brooks D. Cash; Patrick E. Young; Ruben D. Acosta; Roni T. Falk; Ruth M. Pfeiffer; Nan Hu; Hua Su; Lemin Wang; Chaoyu Wang; Barbara Gherman; Carol Giffen; Cathy Dykes; Véronique Turcotte; Patrick Caron; Chantal Guillemette; Sanford M. Dawsey; Christian C. Abnet; Paula L. Hyland; Philip R. Taylor

BACKGROUND & AIMSnEsophageal adenocarcinoma is believed to result from the progression of gastroesophageal reflux disease to erosive esophagitis and re-epithelialization of the esophagus with a columnar cell population termed Barretts esophagus (BE). Men develop BE and esophageal adenocarcinoma more frequently than women, yet little is known about the mechanisms of this difference. We assessed whether sex steroid hormones were associated with BE in a male population.nnnMETHODSnWe analyzed data from the Barretts Esophagus Early Detection Case Control Study, based at the Walter Reed National Military Medical Center. Blood samples were collected from 174 men with BE and 213 men without BE (controls, based on endoscopic analysis); 13 sex steroid hormones were measured by mass spectrometry and sex hormone binding globulin was measured by enzyme-linked immunosorbent assay. We also calculated free estradiol, free testosterone, and free dihydrotestosterone (DHT). We used multivariable logistic regression to estimate odds ratios (ORs) and 95% confidence intervals (CIs) adjusted for age, race, smoking status, alcohol consumption, body mass index, heartburn, regurgitation, and gastroesophageal symptom score (excluding heartburn and regurgitation).nnnRESULTSnLevels of free testosterone and free DHT were associated positively with BE risk; patients in the highest quartile for these hormones were most likely to have BE (free testosterone: OR, 5.36; 95% CI, 2.21-13.03; P = .0002; free DHT: OR, 4.25; 95% CI, 1.87-9.66; P = .001). Level of estrone sulfate was associated inversely with BE risk (P for trend = .02). No other hormone was associated with BE risk. Relationships were not modified by age or BMI.nnnCONCLUSIONSnIn an analysis of men, levels of free testosterone and free DHT were significantly associated with BE.


International Journal of Cancer | 2014

Genetic variants in fas signaling pathway genes and risk of gastric cancer

Paula L. Hyland; Shih-Wen Lin; Nan Hu; Han Zhang; Lemin Wang; Hua Su; Chaoyu Wang; Ti Ding; Ze-Zhong Tang; Jin-Hu Fan; You-Lin Qiao; Xiaoqin Xiong; William Wheeler; Carol Giffen; Kai Yu; Jeff Yuenger; Laurie Burdett; Zhaoming Wang; Stephen J. Chanock; Margaret A. Tucker; Sanford M. Dawsey; Neal D. Freedman; Alisa M. Goldstein; Christian C. Abnet; Philip R. Taylor

Populations in north central China are at high risk for gastric cancers (GC), and altered FAS‐mediated cell signaling and/or apoptosis may contribute to this risk. We examined the association of 554 single nucleotide polymorphisms (SNPs) in 53 Fas signaling‐related genes using a pathway‐based approach in 1758 GC cases (1126 gastric cardia adenocarcinomas (GCA) and 632 gastric noncardia adenocarcinomas (GNCA)), and 2111 controls from a genome‐wide association study (GWAS) of GC in ethnic Chinese. SNP associations with risk of overall GC, GCA and GNCA were evaluated using unconditional logistic regressions controlling for age, sex and study. Gene‐ and pathway‐based associations were tested using the adaptive rank‐truncated product (ARTP) method. Statistical significance was evaluated empirically by permutation. Significant pathway‐based associations were observed for Fas signaling with risk of overall GC (p = 5.5E‐04) and GCA (p = 6.3E‐03), but not GNCA (p= 8.1E‐02). Among examined genes in the Fas signaling pathway, MAP2K4, FAF1, MAPK8, CASP10, CASP8, CFLAR, MAP2K1, CAP8AP2, PAK2 and IKBKB were associated with risk of GC (nominal p < 0.05), and FAF1 and MAPK8 were significantly associated with risk of both GCA and GNCA (nominal p< 0.05). Our examination of genetic variation in the Fas signaling pathway is consistent with an association of altered Fas signaling and/or apoptosis with risk of GC. As one of the first attempts to investigate a pathway‐level association, our results suggest that these genes and the Fas signaling pathway warrant further evaluation in relation to GC risk in other populations.


BMC Cancer | 2009

Biomarkers of apoptosis and survival in esophageal squamous cell carcinoma

Mikiko Takikita; Nan Hu; Jian-Zhong Shou; Quan-Hong Wang; Carol Giffen; Philip R. Taylor; Stephen M. Hewitt

BackgroundCancer of the esophagus is a deadly malignancy, and development of biomarkers that predict survival is an urgent need. The apoptotic pathways have been hypothesized as important in progression of esophageal squamous cell carcinoma (ESCC). We investigated a panel of proteins that regulate apoptosis as candidate of biomarkers of prognosis in ESCC.MethodsTissue microarray (TMA) including 313 surgically-resected cases of ESCC specimens was built for immunohistochemical interrogation. We evaluated seven genes in the FasL-Fas apoptotic pathway - FasL, Fas, FAS-associated death domain protein (FADD), phosphorylated-FADD, and caspase 8 and 10, and the antiapoptotic protein bcl-2. We studied pathway integrity and relations to risk and clinical factors, and determined the prognostic significance of each marker.ResultsFive markers showed strong inter-marker correlations (r ≥ 0.28, p < 0.001), including FasL, Fas, FADD, and caspases 8 and 10. FasL and FADD also showed modest correlations with one or more cancer risk factors, but none of the markers was significantly associated with either tumor stage or lymph node metastasis, the only two clinical factors that predicted survival in these ESCC cases. Multivariate-adjusted proportional hazard regression models showed no association between protein expression and risk of death for any of the seven markers examined.ConclusionIndividual biomarkers in the apoptosis pathway do not appear to predict survival of patients with ESCC.


Gastroenterology | 2017

Germline Mutations in PALB2, BRCA1, and RAD51C, Which Regulate DNA Recombination Repair, in Patients With Gastric Cancer

Ruta Sahasrabudhe; Paul Lott; Mabel Bohorquez; Ted Toal; Ana Estrada; John J. Suarez; Alejandro Brea-Fernández; José Cameselle-Teijeiro; Carla M. A. Pinto; Irma Ramos; Alejandra Mantilla; Rodrigo Prieto; Alejandro H. Corvalán; Enrique Norero; Carolina Alvarez; Teresa Tapia; Pilar Carvallo; Luz M. Gonzalez; Alicia Cock-Rada; Angela R. Solano; Florencia Neffa; Adriana Della Valle; Christopher Yau; Gabriela Soares; Alexander D. Borowsky; Nan Hu; Li Ji He; Xiao You Han; Magdalena Echeverry; John Suarez

Up to 10% of cases of gastric cancer are familial, but so far, only mutations in CDH1 have been associated with gastric cancer risk. To identify genetic variants that affect risk for gastric cancer, we collected blood samples from 28 patients with hereditary diffuse gastric cancer (HDGC) not associated with mutations in CDH1 and performed whole-exome sequence analysis. We then analyzed sequences of candidate genes in 333 independent HDGC and non-HDGC cases. We identified 11 cases with mutations in PALB2, BRCA1, or RAD51C genes, which regulate homologous DNA recombination. We found these mutations in 2 of 31 patients with HDGC (6.5%) and 9 of 331 patients with sporadic gastric cancer (2.8%). Most of these mutations had been previously associated with other types of tumors and partially co-segregated with gastric cancer in our study. Tumors that developed in patients with these mutations had a mutation signature associated with somatic homologous recombination deficiency. Our findings indicate that defects in homologous recombination increase risk for gastric cancer.


Cancer Causes & Control | 2009

Jasmine tea consumption and upper gastrointestinal cancer in China.

Ying Gao; Nan Hu; Xiao You Han; Carol Giffen; Ti Ding; Alisa M. Goldstein; Philip R. Taylor

IntroductionEpidemiological data on green/jasmine tea and esophageal as well as gastric cancer are limited and inconclusive.MethodsIn order to study the effect of jasmine tea in upper gastrointestinal (UGI) cancers, we evaluated 600 esophageal squamous cell carcinoma (ESCC), 598 gastric cardia cancer (GCA), and 316 gastric non-cardia cancer (GNCA) cases and 1,514 age-, gender-, and neighborhood-matched controls. Odds ratios (ORs) and 95% confidence intervals (CIs) were estimated from logistic regression adjusted for matching factors and potential confounders.ResultsAmong controls, 35% of males and 8% of females reported consumption of jasmine tea; other tea consumption was rare. Consumption of jasmine tea (ever vs. never) was not associated with risk of ESCC (ORxa0=xa01.15, 95% CI 0.92–1.44), GCA (ORxa0=xa01.14, 95% CI 0.88–1.37), or GNCA (ORxa0=xa00.85, 95% CI 0.64–1.15) in males and females combined. Among males, cumulative lifetime consumption showed a significant positive dose–response relation with ESCC risk, but not for GCA and GNCA. In exploratory analyses, occupation affected the relation between tea and ESCC such that consumption in males was associated with increased risk only in non-office workers.ConclusionOverall, we found no evidence for a protective effect of tea in esophageal or gastric cancer. Further studies of the potential effects of thermal damage, tea quality, and water quality on UGI cancers are suggested.

Collaboration


Dive into the Nan Hu's collaboration.

Top Co-Authors

Avatar

Chaoyu Wang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Philip R. Taylor

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Alisa M. Goldstein

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Christian C. Abnet

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ti Ding

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Lemin Wang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Neal D. Freedman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Sanford M. Dawsey

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Zhaoming Wang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

You-Lin Qiao

Peking Union Medical College

View shared research outputs
Researchain Logo
Decentralizing Knowledge