Network


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

Hotspot


Dive into the research topics where Huiling He is active.

Publication


Featured researches published by Huiling He.


Nature Genetics | 2009

Common variants on 9q22.33 and 14q13.3 predispose to thyroid cancer in European populations

Julius Gudmundsson; Patrick Sulem; Daniel F. Gudbjartsson; Jon G. Jonasson; Asgeir Sigurdsson; Jon Thor Bergthorsson; Huiling He; Thorarinn Blondal; Frank Geller; Margret Jakobsdottir; Droplaug N. Magnusdottir; Sigurborg Matthiasdottir; Simon N. Stacey; Oskar B Skarphedinsson; Hafdis T. Helgadottir; Wei Li; Rebecca Nagy; Esperanza Aguillo; Eduardo Faure; Enrique Prats; Berta Saez; Mariano Martinez; Gudmundur I. Eyjolfsson; Unnur S. Bjornsdottir; Hilma Holm; Kristleifur Kristjansson; Michael L. Frigge; Kristvinsson H; Jeffrey R. Gulcher; Thorvaldur Jonsson

In order to search for sequence variants conferring risk of thyroid cancer we conducted a genome-wide association study in 192 and 37,196 Icelandic cases and controls, respectively, followed by a replication study in individuals of European descent. Here we show that two common variants, located on 9q22.33 and 14q13.3, are associated with the disease. Overall, the strongest association signals were observed for rs965513 on 9q22.33 (OR = 1.75; P = 1.7 × 10−27) and rs944289 on 14q13.3 (OR = 1.37; P = 2.0 × 10−9). The gene nearest to the 9q22.33 locus is FOXE1 (TTF2) and NKX2-1 (TTF1) is among the genes located at the 14q13.3 locus. Both variants contribute to an increased risk of both papillary and follicular thyroid cancer. Approximately 3.7% of individuals are homozygous for both variants, and their estimated risk of thyroid cancer is 5.7-fold greater than that of noncarriers. In a study on a large sample set from the general population, both risk alleles are associated with low concentrations of thyroid stimulating hormone (TSH), and the 9q22.33 allele is associated with low concentration of thyroxin (T4) and high concentration of triiodothyronine (T3).


Proceedings of the National Academy of Sciences of the United States of America | 2007

Gene expression and functional evidence of epithelial-to-mesenchymal transition in papillary thyroid carcinoma invasion

Vasily Vasko; Allan V. Espinosa; William T. Scouten; Huiling He; Herbert Auer; Sandya Liyanarachchi; Alexander Larin; V Savchenko; Gary L. Francis; Albert de la Chapelle; Motoyasu Saji; Matthew D. Ringel

Papillary thyroid carcinomas (PTCs) that invade into local structures are associated with a poor prognosis, but the mechanisms for PTC invasion are incompletely defined, limiting the development of new therapies. To characterize biological processes involved in PTC invasion, we analyzed the gene expression profiles of microscopically dissected intratumoral samples from central and invasive regions of seven widely invasive PTCs and normal thyroid tissue by oligonucleotide microarray and performed confirmatory expression and functional studies. In comparison with the central regions of primary PTCs, the invasive fronts overexpressed TGF β, NFκB and integrin pathway members, and regulators of small G proteins and CDC42. Moreover, reduced levels of mRNAs encoding proteins involved in cell–cell adhesion and communication were identified, consistent with epithelial-to-mesenchymal transition (EMT). To confirm that aggressive PTCs were characterized by EMT, 34 additional PTCs were examined for expression of vimentin, a hallmark of EMT. Overexpression of vimentin was associated with PTC invasion and nodal metastasis. Functional, in vitro studies demonstrated that vimentin was required both for the development and maintenance of a mesenchymal morphology and invasiveness in thyroid cancer cells. We conclude that EMT is common in PTC invasion and that vimentin regulates thyroid cancer EMT in vitro.


Science | 2011

Mutations in U4atac snRNA, a Component of the Minor Spliceosome, in the Developmental Disorder MOPD I

Huiling He; Sandya Liyanarachchi; Keiko Akagi; Rebecca Nagy; Jingfeng Li; Rosemary C. Dietrich; Wei Li; Nikhil Sebastian; Bernard Wen; Baozhong Xin; Jarnail Singh; Pearlly S. Yan; Hansjuerg Alder; Eric Haan; Dagmar Wieczorek; Beate Albrecht; Erik G. Puffenberger; Heng Wang; Judith A. Westman; Richard A. Padgett; David E. Symer; Albert de la Chapelle

Minor RNA splicing defects can cause a major human developmental disorder. Small nuclear RNAs (snRNAs) are essential factors in messenger RNA splicing. By means of homozygosity mapping and deep sequencing, we show that a gene encoding U4atac snRNA, a component of the minor U12-dependent spliceosome, is mutated in individuals with microcephalic osteodysplastic primordial dwarfism type I (MOPD I), a severe developmental disorder characterized by extreme intrauterine growth retardation and multiple organ abnormalities. Functional assays showed that mutations (30G>A, 51G>A, 55G>A, and 111G>A) associated with MOPD I cause defective U12-dependent splicing. Endogenous U12-dependent but not U2-dependent introns were found to be poorly spliced in MOPD I patient fibroblast cells. The introduction of wild-type U4atac snRNA into MOPD I cells enhanced U12-dependent splicing. These results illustrate the critical role of minor intron splicing in human development.


Proceedings of the National Academy of Sciences of the United States of America | 2012

The polymorphism rs944289 predisposes to papillary thyroid carcinoma through a large intergenic noncoding RNA gene of tumor suppressor type

Jaroslaw Jendrzejewski; Huiling He; Hanna S. Radomska; Wei Li; Jerneja Tomsic; Sandya Liyanarachchi; Ramana V. Davuluri; Rebecca Nagy; Albert de la Chapelle

A genome-wide association study of papillary thyroid carcinoma (PTC) pinpointed two independent SNPs (rs944289 and rs965513) located in regions containing no annotated genes (14q13.3 and 9q22.33, respectively). Here, we describe a unique, long, intergenic, noncoding RNA gene (lincRNA) named Papillary Thyroid Carcinoma Susceptibility Candidate 3 (PTCSC3) located 3.2 kb downstream of rs944289 at 14q.13.3 and the expression of which is strictly thyroid specific. By quantitative PCR, PTCSC3 expression was strongly down-regulated (P = 2.84 × 10−14) in thyroid tumor tissue of 46 PTC patients and the risk allele (T) was associated with the strongest suppression (genotype [TT] (n = 21) vs. [CT] (n = 19), P = 0.004). In adjacent unaffected thyroid tissue, the genotype [TT] was associated with up-regulation of PTCSC3 ([TT] (n = 21) vs. [CT] (n = 19), P = 0.034). The SNP rs944289 was located in a binding site for the CCAAT/enhancer binding proteins (C/EBP) α and β. The risk allele destroyed the binding site in silico. Both C/EBPα and C/EBPβ activated the PTCSC3 promoter in reporter assays (P = 0.0009 and P = 0.0014, respectively) and the risk allele reduced the activation compared with the nonrisk allele (C) (P = 0.026 and P = 0.048, respectively). Restoration of PTCSC3 expression in PTC cell line cells (TPC-1 and BCPAP) inhibited cell growth (P = 0.002 and P = 0.019, respectively) and affected the expression of genes involved in DNA replication, recombination and repair, cellular movement, tumor morphology, and cell death. Our data suggest that SNP rs944289 predisposes to PTC through a previously uncharacterized, long intergenic noncoding RNA gene (PTCSC3) that has the characteristics of a tumor suppressor.


Nature Genetics | 2012

Discovery of common variants associated with low TSH levels and thyroid cancer risk

Julius Gudmundsson; Patrick Sulem; Daniel F. Gudbjartsson; Jon G. Jonasson; Gisli Masson; Huiling He; Aslaug Jonasdottir; Asgeir Sigurdsson; Simon N. Stacey; Hrefna Johannsdottir; Hafdis T. Helgadottir; Wei Li; Rebecca Nagy; Matthew D. Ringel; Richard T. Kloos; Marieke de Visser; Theo S. Plantinga; Martin den Heijer; Esperanza Aguillo; Angeles Panadero; Enrique Prats; Almudena Garcia-Castaño; Ana de Juan; Fernando Rivera; G. Bragi Walters; Hjordis Bjarnason; Laufey Tryggvadottir; Gudmundur I. Eyjolfsson; Unnur S. Bjornsdottir; Hilma Holm

To search for sequence variants conferring risk of nonmedullary thyroid cancer, we focused our analysis on 22 SNPs with a P < 5 × 10−8 in a genome-wide association study on levels of thyroid stimulating hormone (TSH) in 27,758 Icelanders. Of those, rs965513 has previously been shown to associate with thyroid cancer. The remaining 21 SNPs were genotyped in 561 Icelandic individuals with thyroid cancer (cases) and up to 40,013 controls. Variants suggestively associated with thyroid cancer (P < 0.05) were genotyped in an additional 595 non-Icelandic cases and 2,604 controls. After combining the results, three variants were shown to associate with thyroid cancer: rs966423 on 2q35 (OR = 1.34; Pcombined = 1.3 × 10−9), rs2439302 on 8p12 (OR = 1.36; Pcombined = 2.0 × 10−9) and rs116909374 on 14q13.3 (OR = 2.09; Pcombined = 4.6 × 10−11), a region previously reported to contain an uncorrelated variant conferring risk of thyroid cancer. A strong association (P = 9.1 × 10−91) was observed between rs2439302 on 8p12 and expression of NRG1, which encodes the signaling protein neuregulin 1, in blood.


Cancer Research | 2009

A Susceptibility Locus for Papillary Thyroid Carcinoma on Chromosome 8q24

Huiling He; Rebecca Nagy; Sandya Liyanarachchi; Hong Jiao; Wei Li; Saul Suster; Juha Kere; Albert de la Chapelle

Papillary thyroid carcinoma (PTC) displays higher heritability than most other cancers. To search for genes predisposing to PTC, we performed a genome-wide linkage analysis in a large family with PTC and melanoma. Among several peaks the highest was at 8q24, with a maximum nonparametric linkage (NPL) score of 7.03. Linkage analysis was then broadened to comprise 25 additional PTC families that produced a maximum NPL score of 3.2, P = 0.007 at the 8q24 locus. Fine mapping with microsatellite markers was compatible with linkage to the 8q24 locus in 10 of the 26 families. In the large family, a approximately 320 Kb haplotype was shared by individuals with PTC, melanoma, or benign thyroid disease, but not by unaffected individuals. A 12 Kb haplotype of 8 SNP markers within the larger haplotype was shared by 9 of the 10 families in which the 8q24 locus was compatible with linkage. The shared haplotype is located within 2 known overlapping protein-coding genes, thyroglobulin (TG) and Src-like adaptor (SLA). Resequencing of the coding and control regions of TG and SLA did not disclose putative mutations in PTC patients. Embedded in the TG-SLA region are three likely noncoding RNA genes, one of which (AK023948) harbors the 8-SNP haplotype. Resequencing of AK023948 and one of the other RNA genes did not reveal candidate mutations. Gene expression analysis indicated that AK023948 is significantly down-regulated in most PTC tumors. The putative noncoding RNA gene AK023948 is a candidate susceptibility gene for PTC.


International Journal of Cancer | 2007

Identification of a novel noncoding RNA gene, NAMA, that is downregulated in papillary thyroid carcinoma with BRAF mutation and associated with growth arrest

Heejei Yoon; Huiling He; Rebecca Nagy; Ramana V. Davuluri; Saul Suster; Daniel R. Schoenberg; Natalia S. Pellegata; Albert de la Chapelle

In search of tumor suppressor genes in papillary thyroid carcinoma (PTC), we previously used gene expression profiling to identify genes underexpressed in tumor compared with paired unaffected tissue. While searching for loss of heterozygosity (LOH) in genomic regions harboring candidate tumor suppressor genes, we detected LOH in a ∼20 kb region around marker D9S176. Several ESTs flanking D9S176 were underexpressed in PTC tumors, and for one of the ESTs, downregulation was highly associated with the activating BRAF mutation V600E, the most common genetic lesion in PTC. A novel gene, NAMA, (noncoding RNA associated with MAP kinase pathway and growth arrest) containing the affected EST was cloned and characterized. NAMA is weakly expressed in several human tissues, and the spliced forms are primarily detected in testis. Several characteristics of NAMA suggest that it is a nonprotein coding but functional RNA; it has no long open reading frames (ORFs); the exons exhibit low sequence identity in the evolutionarily conserved regions; it is inducible by knockdown of BRAF, inhibition of the MAP kinase pathway, growth arrest and DNA damage in cancer cell lines. We suggest that NAMA is a noncoding RNA associated with growth arrest.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Multiple functional variants in long-range enhancer elements contribute to the risk of SNP rs965513 in thyroid cancer

Huiling He; Wei Li; Sandya Liyanarachchi; Mukund Srinivas; Yanqiang Wang; Keiko Akagi; Yao Wang; Dayong Wu; Qianben Wang; Victor X. Jin; David E. Symer; Rulong Shen; John E. Phay; Rebecca Nagy; Albert de la Chapelle

Significance Papillary thyroid carcinoma (PTC) displays a strong hereditary component that is, in part, due to the additive effects of numerous low-penetrance genes or variants, but virtually no mechanistic information is available. Here, we studied a well-known low-penetrance variant (SNP rs965513) located in a region devoid of coding genes. We show that at least four variants located in the immediate vicinity of rs965513 reside in enhancer elements that bind to the promoter region shared by two adjacent thyroid-related genes, forkhead box E1 (FOXE1) and PTC susceptibility candidate 2 (PTCSC2), regulating their expression. The role of intergenic regulatory variants in cancer predisposition and carcinogenesis is growing. Further mechanistic understanding of how these variants work such as described here needs to be acquired. The [A] allele of SNP rs965513 in 9q22 has been consistently shown to be highly associated with increased papillary thyroid cancer (PTC) risk with an odds ratio of ∼1.8 as determined by genome-wide association studies, yet the molecular mechanisms remain poorly understood. Previously, we noted that the expression of two genes in the region, forkhead box E1 (FOXE1) and PTC susceptibility candidate 2 (PTCSC2), is regulated by rs965513 in unaffected thyroid tissue, but the underlying mechanisms were not elucidated. Here, we fine-mapped the 9q22 region in PTC and controls and detected an ∼33-kb linkage disequilibrium block (containing the lead SNP rs965513) that significantly associates with PTC risk. Chromatin characteristics and regulatory element signatures in this block disclosed at least three regulatory elements functioning as enhancers. These enhancers harbor at least four SNPs (rs7864322, rs12352658, rs7847449, and rs10759944) that serve as functional variants. The variant genotypes are associated with differential enhancer activities and/or transcription factor binding activities. Using the chromosome conformation capture methodology, long-range looping interactions of these elements with the promoter region shared by FOXE1 and PTCSC2 in a human papillary thyroid carcinoma cell line (KTC-1) and unaffected thyroid tissue were found. Our results suggest that multiple variants coinherited with the lead SNP and located in long-range enhancers are involved in the transcriptional regulation of FOXE1 and PTCSC2 expression. These results explain the mechanism by which the risk allele of rs965513 predisposes to thyroid cancer.


The Journal of Clinical Endocrinology and Metabolism | 2013

SRGAP1 Is a Candidate Gene for Papillary Thyroid Carcinoma Susceptibility

Huiling He; Agnieszka Bronisz; Sandya Liyanarachchi; Rebecca Nagy; Wei Li; Yungui Huang; Keiko Akagi; Motoyasu Saji; Dorota Kula; Anna Wojcicka; Nikhil Sebastian; Bernard Wen; Zbigniew Puch; Michał Kalemba; Elzbieta Stachlewska; Malgorzata Czetwertynska; Joanna Dlugosinska; Kinga Dymecka; Rafał Płoski; Marek Krawczyk; Patrick Morrison; Matthew D. Ringel; Richard T. Kloos; Krystian Jażdżewski; David E. Symer; Veronica J. Vieland; Michael C. Ostrowski; Barbara Jarząb; Albert de la Chapelle

BACKGROUND Papillary thyroid carcinoma (PTC) shows high heritability, yet efforts to find predisposing genes have been largely negative. OBJECTIVES The objective of this study was to identify susceptibility genes for PTC. METHODS A genome-wide linkage analysis was performed in 38 families. Targeted association study and screening were performed in 2 large cohorts of PTC patients and controls. Candidate DNA variants were tested in functional studies. RESULTS Linkage analysis and association studies identified the Slit-Robo Rho GTPase activating protein 1 gene (SRGAP1) in the linkage peak as a candidate gene. Two missense variants, Q149H and A275T, localized in the Fes/CIP4 homology domain segregated with the disease in 1 family each. One missense variant, R617C, located in the RhoGAP domain occurred in 1 family. Biochemical assays demonstrated that the ability to inactivate CDC42, a key function of SRGAP1, was severely impaired by the Q149H and R617C variants. CONCLUSIONS Our findings suggest that SRGAP1 is a candidate gene in PTC susceptibility. SRGAP1 is likely a low-penetrant gene, possibly of a modifier type.


Endocrine-related Cancer | 2010

Role of PTPRJ genotype in papillary thyroid carcinoma risk

Rodolfo Iuliano; Dario Palmieri; Huiling He; Angela Iervolino; Eleonora Borbone; Pierlorenzo Pallante; Alessandra Cianflone; Rebecca Nagy; Hansjuerg Alder; George A. Calin; Francesco Trapasso; Carla Giordano; Carlo M. Croce; Albert de la Chapelle; Alfredo Fusco

The strong genetic predisposition to papillary thyroid carcinoma (PTC) might be due to a combination of low-penetrance susceptibility variants. Thus, the research into gene variants involved in the increase of susceptibility to PTC is a relevant field of investigation. The gene coding for the receptor-type tyrosine phosphatase PTPRJ has been proposed as a cancer susceptibility gene, and its role as a tumor suppressor gene is well established in thyroid carcinogenesis. In this study, we want to ascertain the role of PTPRJ genotype in the risk for PTC. We performed a case-control study in which we determined the PTPRJ genotype for the non-synonymous Gln276Pro and Asp872Glu polymorphisms by PCR amplification and sequencing. We calculated allele and genotype frequencies for the considered polymorphisms of PTPRJ in a total sample of 299 cases (PTC patients) and 339 controls (healthy subjects) selected from Caucasian populations. We observed a significantly higher frequency of homozygotes for the Asp872 allele in the group of PTC patients than in the control group (odds ratio=1.61, 95% confidence interval 1.15-2.25, P=0.0053). We observed a non-significant increased frequency of homozygotes for Gln276Pro polymorphism in PTC cases in two distinct Caucasian populations. Therefore, the results reported here show that the homozygous genotype for Asp872 of PTPRJ is associated with an increased risk to develop PTC.

Collaboration


Dive into the Huiling He's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wei Li

University of Tennessee Health Science Center

View shared research outputs
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