Network


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

Hotspot


Dive into the research topics where Liguo Geng is active.

Publication


Featured researches published by Liguo Geng.


Oncotarget | 2017

Risk assessment models for genetic risk predictors of lung cancer using two-stage replication for Asian and European populations

Yang Cheng; Tao Jiang; Meng Zhu; Zhihua Li; Jiahui Zhang; Yuzhuo Wang; Liguo Geng; Jia Liu; Wei Shen; Cheng Wang; Zhibin Hu; Guangfu Jin; Hongxia Ma; Hongbing Shen; Juncheng Dai

In the past ten years, great successes have been accumulated by taking advantage of both candidate-gene studies and genome-wide association studies. However, limited studies were available to systematically evaluate the genetic effects for lung cancer risk with large-scale and different ethnic populations. We systematically reviewed relevant literatures and filtered out 241 important genetic variants identified in 124 articles. A two-stage case-control study within specific subgroups was performed to assess the effects [Training set: 2,331 cases vs. 3,077 controls (Chinese population); testing set: 1,937 cases vs. 1,984 controls (European population)]. Variable selection and model development were used LASSO penalized regression and genetic risk score (GRS) system. Further change in area under the receiver operator characteristic curves (AUC) made by the epidemiologic model with and without GRS was used to compare predictions. It kept 38 genetic variants in our study and the ratios of lung cancer risk for subjects in the upper quartile GRS was three times higher compared to that in the low quartile (odds ratio: 4.64, 95% CI: 3.87–5.56). In addition, we found that adding genetic predictors to smoking risk factor-only model improved lung cancer predictive value greatly: AUC, 0.610 versus 0.697 (P < 0.001). Similar performance was derived in European population and the combined two data sets. Our findings suggested that genetic predictors could improve the predictive ability of risk model for lung cancer and highlighted the application among different populations, indicating that the lung cancer risk assessment model will be a promising tool for high risk population screening and prediction.


Scientific Reports | 2015

The eQTL-missense polymorphisms of APOBEC3H are associated with lung cancer risk in a Han Chinese population.

Meng Zhu; Yuzhuo Wang; Cheng Wang; Wei Shen; Jia Liu; Liguo Geng; Yang Cheng; Juncheng Dai; Guangfu Jin; Hongxia Ma; Zhibin Hu; Hongbing Shen

APOBEC (Apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like) enzymes may involve in mutagenic processes in multiple cancer types, including lung cancer. APOBEC family of cytidine deaminases induces base substitutions with a stringent TCW motif, which is widespread in multiple human cancers. We hypothesized that common missense variants in coding regions of APOBEC genes might damage the structure of proteins and modify lung cancer risk. To test this hypothesis, we systematically screened predicted deleterious polymorphisms in the exon regions of 10 APOBEC core genes (APOBEC1, APOBEC2, APOBEC3A, APOBEC3B, APOBEC3C, APOBEC3D, APOBEC3F, APOBEC3G, APOBEC3H, and APOBEC4) and evaluated them with a case-control study including 1200 cases and 1253 controls. We found that the T allele of rs139293 in exon 2 of APOBEC3H was significantly associated with decreased risk of lung cancer (odds ratio = 0.76, 95% confidence interval: 0.63–0.91). Similar inverse association of this variant was observed in subgroups. Further study showed that the T allele of rs139293 was associated with the altered expression of APOBEC3H and APOBEC3C and that the two genes were co-expressed in both tumor and adjacent normal tissues. These results indicate that genetic variants in APOBEC3H may contribute to lung cancer susceptibility in Chinese population.


Toxicology Letters | 2017

Genetic variants, PM2.5 exposure level and global DNA methylation level: A multi-center population-based study in Chinese

Jia Liu; Kaipeng Xie; Weihong Chen; Meng Zhu; Wei Shen; Jing Yuan; Yang Cheng; Liguo Geng; Yuzhuo Wang; Zhihua Li; Jiahui Zhang; Guangfu Jin; Juncheng Dai; Hongxia Ma; Jiangbo Du; Meilin Wang; Zhengdong Zhang; Zhibin Hu; Tangchun Wu; Hongbing Shen

Global DNA methylation levels can be determined by environmental and genetic factors. There are emerging evidences that methylation status can be modified as exposed to environmental factors such as PM2.5, but the genetic determinants are still largely unknown. To explore whether genetic variants contribute to global DNA methylation levels with consideration of environmental exposures, we systematically evaluated the association between genetic variants and global DNA methylation levels in 301 subjects from three cities in southern, central and northern China with different PM2.5 exposure levels (Zhuhai, Wuhan and Tianjin, respectively). Personal 24-h PM2.5 exposure levels and global DNA methylation levels for each subject were evaluated. Using Illumina Human Exome BeadChip, 241,305 SNVs was genotyped and assessed for their association with global DNA methylation levels. We found that after adjusting for age, gender, PM2.5 exposure level, pack-years of smoking and BMI, 14 SNVs were consistently associated with global DNA methylation levels with pooled P≤1.00×10-4 after meta-analysis of three cohorts, in which 8 SNVs together with age were independent factors modifying global DNA methylation levels. Joint analysis of these identified SNVs showed a significant allele-dosage association between the number of variants and global DNA methylation levels (P=1.82×10-23). In particular, we detected a significant multiplicative interaction between rs4344916 on chromosome 2p22.3 and PM2.5 exposure on global DNA methylation level (P=0.0095). Our findings indicate that genetic variants alone or in combination with PM2.5 play an important role in modifying individual global DNA methylation levels.


Gene | 2016

Genetic variants in chromatin-remodeling pathway associated with lung cancer risk in a Chinese population.

Liguo Geng; Meng Zhu; Yuzhuo Wang; Yang Cheng; Jia Liu; Wei Shen; Zhihua Li; Jiahui Zhang; Cheng Wang; Guangfu Jin; Hongxia Ma; Hongbing Shen; Zhibin Hu; Juncheng Dai

Chromatin remodeling complexes utilize the energy of ATP hydrolysis to remodel nucleosomes and have essential roles in transcriptional modulation. Increasing evidences indicate that these complexes directly interact with numerous proteins and regulate the formation of cancer. However, few studies reported the association of polymorphisms in chromatin remodeling genes and lung cancer. We hypothesized that variants in critical genes of chromatin remodeling pathway might contribute to the susceptibility of lung cancer. To validate this hypothesis, we systematically screened 40 polymorphisms in six key chromatin remodeling genes (SMARCA5, SMARCC2, SMARCD2, ARID1A, NR3C1 and SATB1) and evaluated them with a case-control study including 1341 cases and 1982 controls. Logistic regression revealed that four variants in NR3C1 and SATB1 were significantly associated with lung cancer risk after false discovery rate (FDR) correction [For NR3C1, rs9324921: odds ratio (OR)=1.23, P for FDR=0.029; rs12521436: OR=0.85, P for FDR=0.040; rs4912913: OR=1.17, P for FDR=0.040; For SATB1, rs6808523: OR=1.33, P for FDR=0.040]. Combing analysis presented a significant allele-dosage tendency for the number of risk alleles and lung cancer risk (Ptrend<0.001). Moreover, expression quantitative trait loci (eQTL) analysis revealed that these two genes were differently expressed between lung tumor and adjacent normal tissues in the database of The Cancer Genome Atlas (TCGA) (P=0.009 for rs6808523). These findings suggested that genetic variants in key chromatin remodeling genes may contribute to lung cancer risk in Chinese population. Further large and well-designed studies are warranted to validate our results.


Medicine | 2015

Genetic Variations in Key MicroRNAs are Associated With the Survival of Nonsmall Cell Lung Cancer.

Shuangshuang Wu; Wei Shen; Yun Pan; Meng Zhu; Kaipeng Xie; Liguo Geng; Yuzhuo Wang; Yan Liang; Jiali Xu; Songyu Cao; Wei Xu; Bo Chen; Zhibin Hu; Hongxia Ma; Jianqing Wu; Hongbing Shen

AbstractMicroRNAs (miRNAs) are a class of small, noncoding RNA molecules involved in carcinogenesis. It has been identified that genetic variations in miRNAs contribute to cancer risk, prognosis, and survival. In the present study, we investigated whether single nucleotide polymorphisms (SNPs) of several key miRNAs (miR-184, miR-218, and miR-124) were associated with the prognosis of nonsmall cell lung cancer (NSCLC) in a clinical cohort study including 1001 cases. Cox proportional hazards regression models were used to estimate the hazard ratios (HRs) and their 95% confidence intervals (CIs). We found that 5 SNPs were associated with NSCLC survival (rs919968, rs3775815, rs4867902, and rs6122390 in an additive model: adjusted HR = 1.15, 95% CI = 1.02–1.29; adjusted HR = 0.78, 95% CI = 0.67–0.91, adjusted HR = 1.24, 95% CI = 1.09–1.41; adjusted HR = 1.21, 95% CI = 1.07–1.36, respectively; rs298206 in a dominant model: HR = 1.25, 95% CI = 1.05–1.49). Even after the Bonferroni correction, 3 SNPs remained significant (adjusted P = 0.010, 0.010, and 0.032 for rs3775815, rs4867902, and rs6122390, respectively). Additionally, the combined analysis of these 5 SNPs showed a significant locus-dosage effect between number of unfavorable alleles (rs919968-A, rs3775815-C, rs4867902-G, rs6122390-A, and rs298206-T) and death risk of NSCLC (P for trend < 0.001). A statistically significant multiplicative interaction was found between the genotypes of rs4867902 and surgical operation status (Pint = 0.013). These findings indicated that genetic variations in miRNAs (miR-184, miR-218, and miR-124) might be prognostic markers for NSCLC patients.


International Journal of Cancer | 2017

Fine mapping of chromosome 5p15.33 identifies novel lung cancer susceptibility loci in Han Chinese.

Jing Dong; Yang Cheng; Meng Zhu; Yang Wen; Cheng Wang; Yuzhuo Wang; Liguo Geng; Wei Shen; Jia Liu; Zhihua Li; Jiahui Zhang; Hongxia Ma; Juncheng Dai; Guangfu Jin; Zhibin Hu; Hongbing Shen

Genome‐wide association studies in European and Asian populations have consistently identified chromosome 5p15.33 as a lung cancer susceptibility region. To investigate further the genetic architecture of common variants in this region, we conducted a two‐stage fine‐mapping analysis discovered by targeted resequencing of 200 cases and 300 controls individually, and validated in multiethnic lung cancer Genome wide association studies (GWASs) with 12,843 cases and 12,639 controls. Two independent variants were identified in approximate conditional analysis with GCTA and consistently validated in lung cancer GWASs in both Asian and European populations. These were rs10054203 in TERT (resequencing: OR = 1.69, p = 2.70 × 10−4; validation: OR = 1.34, p = 2.10 × 10−23 for Asian, and OR = 1.09, p = 6.00 × 10−3 for European), and rs397640 in CLPTM1L (resequencing: OR = 0.37, p = 1.19 × 10−4; validation: OR = 0.75, p = 5.89 × 10−8 for Asian, and OR = 0.90, p = 2.40 × 10−2 for European). Expression quantitative trait loci analysis showed the risk allele (C) of rs10054203 was significantly associated with lower mRNA expression of CTD‐2245Ef15.3 (p = 0.019) and Tubulin Polymerization‐Promoting Protein (TPPP, p = 0.031) in 167 lung tissues. In conclusion, in this largest and first resequencing‐based fine‐mapping analysis of 5p15.33 region in Han Chinese, we identified two novel variants associated with lung cancer susceptibility. Further validation studies and functional work is required to confirm the roles of the newly discovered variants.


Carcinogenesis | 2017

Targeted sequencing of chromosome 15q25 identified novel variants associated with risk of lung cancer and smoking behavior in Chinese

Yang Cheng; Cheng Wang; Meng Zhu; Juncheng Dai; Yuzhuo Wang; Liguo Geng; Zhihua Li; Jiahui Zhang; Hongxia Ma; Guangfu Jin; Dongxin Lin; Zhibin Hu; Hongbing Shen

Previous genome-wide association studies (GWAS) in populations of European descent identified a lung cancer susceptibility locus at 15q25 that was biologically associated with nicotine addiction. However, the allele frequency of susceptibility variants identified in this region varied dramatically across European and Asian populations, suggesting that additional risk single nucleotide polymorphism (SNPs) in Asians need to be identified. Thus, we conducted a fine-mapping study of chromosome 15q25 using targeted resequencing of 200 lung cancer cases and 300 controls of Chinese descent. An approximate conditional and joint analysis of the discovery data revealed two novel SNPs with independent effects (rs6495304: OR = 1.79, P = 9.37 × 10-4; and rs74733525: OR = 1.68, P = 8.05 × 10-3). Both variants were common in Asians but rare or monomorphic in Whites. These results were further supported by in silico validation including 8047 cases and 8898 controls from multiethnic lung cancer genome-wide association studies (GWASs) (rs6495304: OR = 1.32, P = 1.21 × 10-11; and rs74733525: OR = 1.29, P = 4.29 × 10-4); however, rs6495304 demonstrated significant effects only in ever-smokers (P = 0.004 for heterogeneity test of smoking). Mediation analysis indicated that smoking behavior may mediate the effect of rs6495304 on lung cancer risk. Furthermore, expression quantitative trait loci analysis showed the risk allele (A) of rs6495304 was significantly associated with lower mRNA expression of CHRNA3 (P = 0.029) in 81 hypothalamic tissue samples. This finding provides new insights into the association between lung cancer susceptibility and the 15q25 locus.


Mutation Research | 2016

Genetic variants in multisynthetase complex genes are associated with DNA damage levels in Chinese populations

Jia Liu; Meng Zhu; Weihong Chen; Kaipeng Xie; Wei Shen; Jing Yuan; Yang Cheng; Liguo Geng; Yuzhuo Wang; Guangfu Jin; Juncheng Dai; Hongxia Ma; Jiangbo Du; Meilin Wang; Zhengdong Zhang; Zhibin Hu; Tangchun Wu; Hongbing Shen

Aminoacyl-tRNA synthetases (ARSs) and ARS-interacting multi-functional proteins (AIMPs) form a multisynthetase complex (MSC) and play an important role in the process of DNA damage repair. We hypothesized that genetic variants in key ARSs and AIMPs might regulate the DNA damage response. Therefore, we systematically screened 23 potentially functional polymorphisms in MSC genes and evaluated the association between the genetic variants and DNA damage levels in 307 subjects from three cities in southern, central and northern China (Zhuhai, Wuhan and Tianjin, respectively). We examined personal 24-h PM2.5 exposure levels and DNA damage levels in peripheral blood lymphocytes for each subject. We found that the variant allele of rs12199241 in AIMP3 was significantly associated with DNA damage levels (β=0.343, 95%CI: 0.133-0.554, P=0.001). Meanwhile, the results of rs5030754 in EPRS and rs3784929 in KARS indicated their suggestive roles in DNA damage processes (β=0.331, 95%CI: 0.062-0.599, P=0.016 for rs5030754; β=0.192, 95%CI: 0.016-0.368, P=0.033 for rs3784929, respectively). After multiple testing, rs12199241 was still significantly associated with DNA damage levels. Combined analysis of these three polymorphisms showed a significant allele-dosage association between the number of risk alleles and higher DNA damage levels (Ptrend<0.001). These findings indicate that genetic variants in MSC genes may account for PM2.5-modulated DNA damage levels in Chinese populations.


Nature Communications | 2018

Whole-genome sequencing reveals genomic signatures associated with the inflammatory microenvironments in Chinese NSCLC patients

Cheng Wang; Rong Yin; Juncheng Dai; Yayun Gu; Shaohua Cui; Hongxia Ma; Zhihong Zhang; Jiaqi Huang; Na Qin; Tao Jiang; Liguo Geng; Meng Zhu; Zhening Pu; Fangzhi Du; Yuzhuo Wang; Jianshui Yang; Liang Chen; Qianghu Wang; Jiang Y; Lili Dong; Yihong Yao; Guangfu Jin; Zhibin Hu; Liyan Jiang; Lin Xu; Hongbing Shen

Chinese lung cancer patients have distinct epidemiologic and genomic features, highlighting the presence of specific etiologic mechanisms other than smoking. Here, we present a comprehensive genomic landscape of 149 non-small cell lung cancer (NSCLC) cases and identify 15 potential driver genes. We reveal that Chinese patients are specially characterized by not only highly clustered EGFR mutations but a mutational signature (MS3, 33.7%), that is associated with inflammatory tumor-infiltrating B lymphocytes (P = 0.001). The EGFR mutation rate is significantly increased with the proportion of the MS3 signature (P = 9.37 × 10−5). TCGA data confirm that the infiltrating B lymphocyte abundance is significantly higher in the EGFR-mutated patients (P = 0.007). Additionally, MS3-high patients carry a higher contribution of distant chromosomal rearrangements >1 Mb (P = 1.35 × 10−7), some of which result in fusions involving genes with important functions (i.e., ALK and RET). Thus, inflammatory infiltration may contribute to the accumulation of EGFR mutations, especially in never-smokers.The distinct genomic and epidemiological features of Chinese lung cancer patients suggest the presence of alternative causal mechanisms. Here, the authors present the genomic landscape of 149 Chinese NSCLC patients and reveal distinct mutational signatures associated with inflammatory microenvironments.


International Journal of Cancer | 2018

Integrating expression-related SNPs into genome-wide gene- and pathway-based analyses identified novel lung cancer susceptibility genes: Expression-related SNPs and lung cancer

Yuzhuo Wang; Weibing Wu; Meng Zhu; Cheng Wang; Wei Shen; Yang Cheng; Liguo Geng; Zhihua Li; Jiahui Zhang; Juncheng Dai; Hongxia Ma; Liang Chen; Zhibin Hu; Guangfu Jin; Hongbing Shen

Traditional pathway analysis map single nucleotide polymorphisms (SNPs) to genes according to physical position, which lacks sufficient biological bases. Here, we incorporated genetics of gene expression into gene‐ and pathway‐based analysis to identify genes and pathways associated with lung cancer risk. We identified expression‐related SNPs (eSNPs) in lung tissues and integrated these eSNPs into three lung cancer genome‐wide association studies (GWASs), including 12,843 lung cancer cases and 12,639 controls. We used SKAT‐C for gene‐based analysis, and conditional analysis to identify independent eSNPs of each gene. ARTP algorithm was used for pathway analysis. A total of 374,382 eSNPs in the GWAS datasets survived quality control, which were mapped to 5,084 genes and 2,752 pathways. In the gene‐based analysis, nine genes showed significant associations with lung cancer risk. Among them, TP63 (3q28), RP11‐650L12.2 (15q25.1) and CHRNA5 (15q25.1) were located in known lung cancer susceptibility loci. We also validated two newly identified susceptibility loci (RNASET2 and AL133458.1 in 6q27, and MPZL3 in 11q23.3). Besides, DVL3 (3q27.1), RP11‐522I20.3 (9q21.32) and CCDC116 (22q11.21) were identified as novel lung cancer susceptibility genes. Pathway analysis showed that pathways involved in protein structure, the Notch signaling pathway and the nicotinic acetylcholine receptor‐related pathways were associated with lung cancer risk. Combing eSNPs, gene‐ and pathway‐based analysis identifies novel lung cancer susceptibility genes, which serves as a powerful approach to decipher biological mechanisms underlying GWAS findings.

Collaboration


Dive into the Liguo Geng's collaboration.

Top Co-Authors

Avatar

Hongbing Shen

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Hongxia Ma

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Meng Zhu

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Yuzhuo Wang

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Zhibin Hu

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Guangfu Jin

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Juncheng Dai

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Yang Cheng

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Wei Shen

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Cheng Wang

Nanjing Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge