Network


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

Hotspot


Dive into the research topics where Lingling Cui is active.

Publication


Featured researches published by Lingling Cui.


Nature Communications | 2015

Genome-wide association analysis identifies three new risk loci for gout arthritis in Han Chinese

Changgui Li; Zhiqiang Li; Shiguo Liu; Can Wang; Lin Han; Lingling Cui; Jing-Guo Zhou; Hejian Zou; Zhen Liu; Jianhua Chen; Xiaoyu Cheng; Zhaowei Zhou; Chengcheng Ding; Meng Wang; Tong Chen; Ying Cui; Hongmei He; Keke Zhang; Congcong Yin; Yunlong Wang; Shichao Xing; Baojie Li; Jue Ji; Zhaotong Jia; Lidan Ma; Jiapeng Niu; Ying Xin; Tian Liu; Nan Chu; Qing Yu

Gout is one of the most common types of inflammatory arthritis, caused by the deposition of monosodium urate crystals in and around the joints. Previous genome-wide association studies (GWASs) have identified many genetic loci associated with raised serum urate concentrations. However, hyperuricemia alone is not sufficient for the development of gout arthritis. Here we conduct a multistage GWAS in Han Chinese using 4,275 male gout patients and 6,272 normal male controls (1,255 cases and 1,848 controls were genome-wide genotyped), with an additional 1,644 hyperuricemic controls. We discover three new risk loci, 17q23.2 (rs11653176, P=1.36 × 10−13, BCAS3), 9p24.2 (rs12236871, P=1.48 × 10−10, RFX3) and 11p15.5 (rs179785, P=1.28 × 10−8, KCNQ1), which contain inflammatory candidate genes. Our results suggest that these loci are most likely related to the progression from hyperuricemia to inflammatory gout, which will provide new insights into the pathogenesis of gout arthritis.


Journal of Gastroenterology and Hepatology | 2014

Hypothermia induced by adenosine 5′‐monophosphate attenuates injury in an L‐arginine‐induced acute pancreatitis rat model

Yunlong Wang; Weiting Guo; Yuan Li; Xinting Pan; Wenshan Lv; Lingling Cui; Changgui Li; Yangang Wang; Jidong Zhang; Bin Liu

This study sought to investigate the effects of hypothermia induced by adenosine 5′‐monophosphate (5′‐AMP) on L‐arginine (L‐Arg)‐induced acute pancreatitis in rats.


Molecular Medicine Reports | 2013

Polymorphisms in the NLRP3 gene and risk of primary gouty arthritis Corrigendum in /mmr/8/6/1888

Dong-Mei Meng; Yu-Jiao Zhou; Luan Wang; Wei Ren; Lingling Cui; Lin Han; Zheng-Hai Qu; Changgui Li; Jia-Jun Zhao

The aim of the present study was to investigate the association between genetic variants in 17 tagSNPs of the NLRP3 gene and the susceptibility to primary gouty arthritis. A genotype-phenotype analysis of 480 primary gout and 480 control patients was performed. Samples from all the patients were collected from The Affiliated Hospital of Medical College (Qingdao, China). Seventeen tagSNPs of the NLRP3 gene were amplified using polymerase chain reaction (PCR) and MassARRAY technology was used for single nucleotide polymorphism (SNP) genotyping. The genetic frequency of rs7512998 was significantly different between the gout and control patients (P<0.05), whereas no significant differences were identified for the remaining SNPs. The 17 SNPs conformed to the Hardy-Weinberg equilibrium (HWE) in the control group (P>0.05). The haplotype association among the 17 SNPs of the NLRP3 gene indicated that no individual SNP was significantly associated with primary gouty arthritis. CTATCAGCGCCCAGTGC was the most common haplotype in the case and control groups, with a frequency of 0.224 and 0.243, respectively. However, the odds ratios (ORs) of the 8 haplotypes were not identified to be significantly associated with gouty arthritis (P>0.05 for all the 8 haplotypes). To the best of our knowledge, this is the first study to investigate the association between SNPs of the NLRP3 gene and the risk of primary gouty arthritis, although no significant association was identified. Further clinical studies and functional analysis are required to explore the potential associations between NLRP3 gene polymorphisms and the risk of primary gouty arthritis.


Stem Cells International | 2016

The Establishment and Characteristics of Rat Model of Atherosclerosis Induced by Hyperuricemia.

Zhen Liu; Tong Chen; Haitao Niu; Wei Ren; Xinde Li; Lingling Cui; Changgui Li

Epidemiological studies have identified hyperuricemia as an independent risk factor for cardiovascular disease. However, the mechanism whereby hyperuricemia causes atherosclerosis remains unclear. The objective of the study was to establish a new rat model of hyperuricemia-induced atherosclerosis. Wistar-Kyoto rats were randomly allocated to either a normal diet (ND), high-fat diet (HFD), or high-adenine diet (HAD), followed by sacrifice 4, 8, or 12 weeks later. Serum uric acid and lipid levels were analyzed, pathologic changes in the aorta were observed by hematoxylin and eosin staining, and mRNA expression was evaluated by quantitative real-time polymerase chain reaction. Serum uric acid and TC were significantly increased in the HAD group at 4 weeks compared with the ND group, but there was no significant difference in serum uric acid between the ND and HFD groups. Aorta calcification occurred earlier and was more severe in the HAD group, compared with the HFD group. Proliferating cell nuclear antigen, monocyte chemotactic factor-1, intercellular adhesion molecule-1, and vascular cell adhesion molecule-1 mRNA levels were increased in the HFD and HAD groups compared with the ND group. This new animal model will be a useful tool for investigating the mechanisms responsible for hyperuricemia-induced atherosclerosis.


BMC Medical Genetics | 2015

Polymorphisms in GCKR , SLC17A1 and SLC22A12 were associated with phenotype gout in Han Chinese males: a case–control study

Zhaowei Zhou; Lingling Cui; Lin Han; Can Wang; Zhijian Song; Jiawei Shen; Zhiqiang Li; Jianhua Chen; Zujia Wen; Xiaomin Wang; Yongyong Shi; Changgui Li

BackgroundGout is a common arthritic disease resulting from elevated serum uric acid (SUA) level. A large meta-analysis including 28,141 individuals identified nine single nucleotide polymorphisms (SNPs) associated with altered SUA level in a Caucasian population. However, raised SUA level alone is not sufficient for the development of gout arthritis and most of these SNPs have not been studied in a Han Chinese population. Here, we performed a case–control association analysis to investigate the relationship between these SUA correlated SNPs and gout arthritis in Han Chinese.MethodsA total of 622 ascertained gout p9atients and 917 healthy controls were genotyped. Genome-wide significant SNPs, rs12129861, rs780094, rs734553, rs742132, rs1183201, rs12356193, rs17300741 and rs505802 in the previous SUA study, were selected for our analysis.ResultsNo deviation from the Hardy–Weinberg equilibrium was observed either in the case or control cohorts (corrected p > 0.05). Three SNPs, rs780094 (located in GCKR, corrected p = 1.78E−4, OR = 0.723), rs1183201 (located in SLC17A1, corrected p = 1.39E−7, OR = 0.572) and rs505802 (located in SLC22A12, corrected p = 0.007, OR = 0.747), were significantly associated with gout on allelic level independent of potential cofounding traits. While the remaining SNPs were not replicated. We also found significant associations of uric acid concentrations with these three SNPs (rs780094 in GCKR, corrected p = 3.94E−5; rs1183201 in SLC17A1, corrected p = 0.005; rs505802 in SLC22A12, corrected p = 0.003) and of triglycerides with rs780094 (located in GCKR, corrected p = 2.96E−4). Unfortunately, SNP-SNP interactions for these three significant SNPs were not detected (rs780094 vs rs1183201, p = 0.402; rs780094 vs rs505802, p = 0.434; rs1183201 vs rs505802, p = 0.143).ConclusionsThree SUA correlated SNPs in Caucasian population, rs780094 in GCKR, rs1183201 in SLC17A1 and rs505802 in SLC22A12 were confirmed to be associated with gout arthritis and uric acid concentrations in Han Chinese males. Considering genetic differences among populations and complicated pathogenesis of gout arthritis, more validating tests in independent populations and relevant functional experiments are suggested in future.


Kidney International | 2018

Knockout of the urate oxidase gene provides a stable mouse model of hyperuricemia associated with metabolic disorders

Jie Lu; Xu Hou; Xuan Yuan; Lingling Cui; Zhen Liu; Xinde Li; Lidan Ma; Xiaoyu Cheng; Ying Xin; Can Wang; Keke Zhang; Xuefeng Wang; Wei Ren; Ruixia Sun; Zhaotong Jia; Zibin Tian; Qing Sheng Mi; Changgui Li

The urate oxidase (Uox) gene encodes uricase that in the rodent liver degrades uric acid into allantoin, forming an obstacle for establishing stable mouse models of hyperuricemia. The loss of uricase in humans during primate evolution causes their vulnerability to hyperuricemia. Thus, we generated a Uox-knockout mouse model on a pure C57BL/6J background using the transcription activator-like effector nuclease (TALEN) technique. These Uox-knockout mice spontaneously developed hyperuricemia (over 420 μmol/l) with about 40% survival up to 62 weeks. Renal dysfunction (elevated serum creatinine and blood urea nitrogen) and glomerular/tubular lesions were observed in these Uox-knockout mice. Male Uox-knockout mice developed glycol-metabolic disorders associated with compromised insulin secretion and elevated vulnerability to streptozotocin-induced diabetes, whereas female mice developed hypertension accompanied by aberrant lipo-metabolism. Urate-lowering drugs reduced serum uric acid and improved hyperuricemia-induced disorders. Thus, uricase knockout provides a suitable mouse model to investigate hyperuricemia and associated disorders mimicking the human condition, suggesting that hyperuricemia has a causal role in the development of metabolic disorders and hypertension.


Scientific Reports | 2017

Replication of Gout/Urate Concentrations GWAS Susceptibility Loci Associated with Gout in a Han Chinese Population

Zhiqiang Li; Zhaowei Zhou; Xu Hou; Dajiang Lu; Xuan Yuan; Jie Lu; Can Wang; Lin Han; Lingling Cui; Zhen Liu; Jianhua Chen; Xiaoyu Cheng; Keke Zhang; Jue Ji; Zhaotong Jia; Lidan Ma; Ying Xin; Tian Liu; Qing Yu; Wei Ren; Xuefeng Wang; Xinde Li; Qing Sheng Mi; Yongyong Shi; Changgui Li

Gout is a chronic disease resulting from elevated serum urate (SU). Previous genome-wide association studies (GWAS) have identified dozens of susceptibility loci for SU/gout, but few have been conducted for Chinese descent. Here, we try to extensively investigate whether these loci contribute to gout risk in Han Chinese. A total of 2255 variants in linkage disequilibrium (LD) with GWAS identified SU/gout associated variants were analyzed in a Han Chinese cohort of 1255 gout patients and 1848 controls. Cumulative genetic risk score analysis was performed to assess the cumulative effect of multiple “risk” variants on gout incidence. 23 variants (41%) of LD pruned variants set (n = 56) showed nominal association with gout in our sample (p < 0.05). Some of the previously reported gout associated loci (except ALDH16A1), including ABCG2, SLC2A9, GCKR, ALDH2 and CNIH2, were replicated. Cumulative genetic risk score analyses showed that the risk of gout increased for individuals with the growing number (≥8) of the risk alleles on gout associated loci. Most of the gout associated loci identified in previous GWAS were confirmed in an independent Chinese cohort, and the SU associated loci also confer susceptibility to gout. These findings provide important information of the genetic association of gout.


Clinical and Experimental Hypertension | 2017

The ongoing role of serum uric acid in blood pressure

Hui Zhou; Yangang Wang; Lingling Cui; Ying Chen; Changgui Li; Jinjiao Zhao

ABSTRACT Positive association between blood pressure (BP) and uric acid is evident, but specific effect size of serum uric acid (SUA) at different levels on BP is unclear, and interaction effect of SUA with other metabolic factors on BP was rarely reported. A cross-sectional study was conducted by making use of data from an epidemic investigation. A total of 3658 subjects were enrolled in our data analysis. In male subjects, for each 1 mg/dL increase in the SUA level, systolic blood pressure (SBP) and diastolic blood pressure (DBP) increased by 1.339 mmHg [95% CI: 0.552–2.126] and 0.515 mmHg [95% CI: 0.013–1.016], respectively, after adjusting for age, fasting plasma glucose (FPG), triglyceride (TG), total cholesterol (TC), estimated-glomerular filtration rate (eGFR), body mass index (BMI), drinking, smoking, and waist–hip ratio (WHR). And in female subjects, for each 1 mg/dL increase in the SUA level, SBP and DBP increased by 1.180 mmHg [95% CI: 0.401–1.959] and 0.549 mmHg [95% CI: 0.086–1.011], respectively, after adjusting the same factors. In males, in subjects with SUA < 6 mg/dL, SBP increased by 0.585 mmHg [95% CI: −0.976 to 2.146] for each 1 mg/dL increase in the SUA level, while in subjects with SUA ≥ 6 mg/dL, SBP increased by 3.271 mmHg [95% CI: 1.244–5.297] (p interaction = 0.0369), after adjusting age, BMI, smoking, drinking, TG, TC, FPG, eGFR, and WHR. This difference was not observed in DBP and females. Interaction effects between SUA and BMI, TG, FPG on BP were discovered. In conclusion, higher level of SUA has a stronger effect on BP and other metabolic factors: FPG, TG, and BMI could strengthen the effect of SUA on BP.


Molecular Medicine Reports | 2013

[Corrigendum] Polymorphisms in the NLRP3 gene and risk of primary gouty arthritis

Dong-Mei Meng; Yu-Jiao Zhou; Luan Wang; Wei Ren; Lingling Cui; Lin Han; Zheng-Hai Qu; Changgui Li; Jia-Jun Zhao


Gout and Hyperuricemia | 2016

The polymorphisms of aldehyde dehydrogenase 2 gene are associated with gout disease in male Han Chinese

Can Wang; Changgui Li; Chengcheng Ding; Qing Yu; Wei Ren; Xuefeng Wang; Lingling Cui

Collaboration


Dive into the Lingling Cui'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

Jianhua Chen

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge