Network


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

Hotspot


Dive into the research topics where Sherry C. Huang is active.

Publication


Featured researches published by Sherry C. Huang.


PLOS ONE | 2008

Mutation Rates of TGFBR2 and ACVR2 Coding Microsatellites in Human Cells with Defective DNA Mismatch Repair

Heekyung Chung; Dennis J. Young; Claudia G. Lopez; Thuy-Anh Le; Jeffrey K. Lee; Deena Ream-Robinson; Sherry C. Huang; John M. Carethers

Microsatellite instability promotes colonic tumorigenesis through generating frameshift mutations at coding microsatellites of tumor suppressor genes, such as TGFBR2 and ACVR2. As a consequence, signaling through these TGFβ family receptors is abrogated in DNA Mismatch repair (MMR)-deficient tumors. How these mutations occur in real time and mutational rates of these human coding sequences have not previously been studied. We utilized cell lines with different MMR deficiencies (hMLH1−/−, hMSH6−/−, hMSH3−/−, and MMR-proficient) to determine mutation rates. Plasmids were constructed in which exon 3 of TGFBR2 and exon 10 of ACVR2 were cloned +1 bp out of frame, immediately after the translation initiation codon of an enhanced GFP (EGFP) gene, allowing a −1 bp frameshift mutation to drive EGFP expression. Mutation-resistant plasmids were constructed by interrupting the coding microsatellite sequences, preventing frameshift mutation. Stable cell lines were established containing portions of TGFBR2 and ACVR2, and nonfluorescent cells were sorted, cultured for 7–35 days, and harvested for flow cytometric mutation detection and DNA sequencing at specific time points. DNA sequencing revealed a −1 bp frameshift mutation (A9 in TGFBR2 and A7 in ACVR2) in the fluorescent cells. Two distinct fluorescent populations, M1 (dim, representing heteroduplexes) and M2 (bright, representing full mutants) were identified, with the M2 fraction accumulating over time. hMLH1 deficiency revealed 11 (5.91×10−4) and 15 (2.18×10−4) times higher mutation rates for the TGFBR2 and ACVR2 microsatellites compared to hMSH6 deficiency, respectively. The mutation rate of the TGFBR2 microsatellite was ∼3 times higher in both hMLH1 and hMSH6 deficiencies than the ACVR2 microsatellite. The −1 bp frameshift mutation rates of TGFBR2 and ACVR2 microsatellite sequences are dependent upon the human MMR background.


Human Genetics | 2010

Large intron 14 rearrangement in APC results in splice defect and attenuated FAP

Thérèse M.F. Tuohy; Michelle W. Done; Michelle S. Lewandowski; Patricia Shires; Devki S. Saraiya; Sherry C. Huang; Deborah W. Neklason; Randall W. Burt

Familial adenomatous polyposis [FAP (OMIM 175100)] is an autosomal dominant colorectal cancer predisposition syndrome characterized by hundreds to thousands of colonic polyps and, if untreated by a combination of screening and/or surgical intervention, a ~99% lifetime risk of colorectal cancer. A subset of FAP patients develop an attenuated form of the condition characterized by lower numbers of colonic polyps (highly variable, but generally less than 100) and a lower lifetime risk of colorectal cancer, on the order of 70%. We report the diagnosis of three attenuated FAP families due to a 1.4-kb deletion within intron 14 of APC, originally reported clinically as a variant of unknown significance (VUS). Sequence analysis suggests that this arose through an Alu-mediated recombination event with a locus on chromosome 6q22.1. This mutation is inherited by family members who presented with an attenuated FAP phenotype, with variable age of onset and severity. Sequence analysis of mRNA revealed an increase in the level of aberrant splicing of exon 14, resulting in the generation of an exon 13–exon 15 splice-form that is predicted to lead to a frameshift and protein truncation at codon 673. The relatively mild phenotypic presentation and the intra-familial variation are consistent with the leaky nature of exon 14 splicing in normal APC. The inferred founder of these three families may account for as yet undetected affected branches of this kindred. This and similar types of intronic mutations may account for a significant proportion of FAP cases where APC clinical analysis fails because of the current limitations of testing options.


Cancer | 2011

Evidence for an hMSH3 defect in familial hamartomatous polyps

Sherry C. Huang; Jeffrey K. Lee; E. Julieta Smith; Ryan T. Doctolero; Akihiro Tajima; Stayce E. Beck; Noel Weidner; John M. Carethers

Patients with hamartomatous polyposis syndromes have increased risk for colorectal cancer (CRC). Although progression of polyps to carcinoma is observed, pathogenic mechanisms remain unknown. The authors examined whether familial hamartomatous polyps harbor defects in DNA mismatch repair (MMR), and assayed for somatic mutation of PTEN, a gene inactivated in the germline of some hamartomatous polyposis syndrome patients.


Journal of Pediatric Gastroenterology and Nutrition | 2007

Cyclooxygenase-2 expression in polyps from a patient with juvenile polyposis syndrome with mutant BMPR1A

Jayde E. Kurland; Stayce E. Beck; Carol J Solomon; Oscar S. Brann; John M. Carethers; Sherry C. Huang

Objectives: Cyclooxygenase-2 (COX-2) expression is increased in colorectal cancers and has been reported to be upregulated in Peutz-Jeghers polyps. To determine whether germline and somatic loss of BMPR1A in polyps from a patient with juvenile polyposis syndrome have altered COX-2 expression, we characterized a patient with juvenile polyposis syndrome for BMPR1A germline mutations and examined the polyps for BMPR1A expression and COX-2 expression. Patients and Methods: DNA analysis for BMPR1A was performed on a patient with juvenile polyposis syndrome. Multiple polypectomies were performed, and several polyps showed adenomatous change. Genomic DNA was extracted from polyp material for loss of heterozygosity (LOH) analyses with microsatellite markers. Immunohistochemistry was performed on sections using antibodies for BMPR1A and COX-2. Results: The kindred possessed a germline BMPR1A missense mutation. In polyp domains containing cystic and adenomatous epithelium, no LOH was observed using markers near the BMPR1A locus. Immunostaining indicated decreased expression of phospho-SMAD1 (pSMAD1), functionally downstream of the mutant BMPR1A receptor in the cystic epithelium, with further reduction in adenomatous portions within the polyp. COX-2 protein, normally not expressed in the colon, was present and increased in polyp epithelium. Conclusions: Decreased expression of pSMAD1 in the cystic epithelium with further reduction in the adenomatous area, and increase in COX-2 expression within polyps from the BMPR1A heterozygote, suggest a potential mechanism for adenomatous pathogenesis in these hamartomatous polyps. This may imply that COX-2 inhibitors could be a means for chemoprevention in this syndrome.


Journal of Pediatric Gastroenterology and Nutrition | 2014

Lynch syndrome: a pediatric perspective.

Sherry C. Huang; Carol Durno; Steven H. Erdman

ABSTRACT Colorectal cancer is a rare disease in the pediatric age group and, when present, suggests an underlying genetic predisposition. The most common hereditary colon cancer susceptibility condition, Lynch syndrome (LS), previously known as hereditary nonpolyposis colorectal cancer, is an autosomal dominant condition caused by a germline mutation in 1 of 4 DNA mismatch repair (MMR) genes: MLH1, MSH2, MSH6, or PMS2. The mutation-prone phenotype of this disorder is associated with gastrointestinal, endometrial, and other cancers and is now being identified in both symptomatic adolescents with malignancy as well in asymptomatic mutation carriers who are at risk for a spectrum of gastrointestinal and other cancers later in life. We review the DNA MMR system, our present understanding of LS in the pediatric population, and discuss the newly identified biallelic form of the disease known as constitutional mismatch repair deficiency syndrome. Both family history and tumor characteristics can help to identify patients who should undergo genetic testing for these cancer predisposition syndromes. Patients who carry either single allele (LS) or double allele (constitutional mismatch repair deficiency syndrome) mutations in the MMR genes benefit from cancer surveillance programs that target both the digestive and extraintestinal cancer risk of these diseases. Because spontaneous mutation in any one of the MMR genes is extremely rare, genetic counseling and testing are suggested for all at-risk family members.


Journal of Investigative Medicine | 2005

410 LOSS OF PTEN EXPRESSION IN MICROSATELLITE UNSTABLE COLORECTAL CANCER

Jeffrey K. Lee; Sherry C. Huang; John M. Carethers

Purpose Microsatellite unstable-high (MSI-H) colorectal cancers account for 15% of sporadic colorectal cancers and indicate genomic instability at microsatellite sequences. Many MSI mutations occur in targeted genes important for regulation of growth suppression. The tumor suppressor gene PTEN, located at chromosome sub-band 10q23.3, encodes a dual-specificity phosphatase that negatively regulates the phosphatidylinositol 3{′-kinase (PI3 K)?Akt-dependent cellular survival pathway. PTEN contains two coding hexadenine microsatellites that often change in length within cancers of the breast, endometrium, prostate, and reportedly colon cancers, and is thought to play a role in its pathogenesis. Here, we analyzed MSI-H sporadic colorectal cancers to determine if PTEN microsatellite frameshifts occur and whether somatic inactivation of PTEN occurs in the tumors with PTEN frameshift mutations. Methods 91 MSI-H sporadic colon cancers from a total cohort of 710 sporadic colon cancers were analyzed for mutations in two hexadenine tracts (exons 7 and 8) of PTEN. PTEN expression was determined by immunohistochemistry using an antibody targeting an epitope beyond the predicted truncated protein. Results 9/91 MSI-H cancers (10%) demonstrated frameshifts in exon 8 while 2/62 demonstrated frameshifts in exon 7 (3%). Of tumors with mutant PTEN, all demonstrated down regulation or complete loss of PTEN in the epithelium. Conclusions PTEN frameshifts occur at a greater rate in exon 8 than exon 7 in MSI-H sporadic colorectal cancers. PTEN frameshift mutations are associated with down-regulation or inactivation of PTEN in MSI-H sporadic colorectal cancers. Loss of this function may contribute to the pathogenesis of colon cancers.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2006

Bone morphogenetic protein signaling and growth suppression in colon cancer.

Stayce E. Beck; Barbara Jung; Antonio Fiorino; Jessica Gomez; Eunice Del Rosario; Betty L. Cabrera; Sherry C. Huang; Jimmy Y. C. Chow; John M. Carethers


Cancer Research | 2000

Genetic Heterogeneity in Familial Juvenile Polyposis

Sherry C. Huang; Christine R. Chen; Joel E. Lavine; Sharon F. Taylor; Robert O. Newbury; Thu Thao T. Pham; Luigi Ricciardiello; John M. Carethers


Current Gastroenterology Reports | 2009

Pediatric juvenile polyposis syndromes: An update

Sherry C. Huang; Steven H. Erdman


/data/revues/00223476/v138i5/S0022347601900575/ | 2011

Germline characterization of early-aged onset of hereditary non-polyposis colorectal cancer

Sherry C. Huang; Joel E. Lavine; Patricia S. Boland; Robert O. Newbury; Richard D. Kolodner; Thu-Thao T. Pham; Christian N. Arnold; C. Richard Boland; John M. Carethers

Collaboration


Dive into the Sherry C. Huang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeffrey K. Lee

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stayce E. Beck

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

C. Richard Boland

Baylor University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge