Network


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

Hotspot


Dive into the research topics where Shannon Nasser is active.

Publication


Featured researches published by Shannon Nasser.


Cancer Research | 2008

PIK3CA Mutation/PTEN Expression Status Predicts Response of Colon Cancer Cells to the Epidermal Growth Factor Receptor Inhibitor Cetuximab

Minaxi Jhawer; Sanjay Goel; Andrew J. Wilson; Cristina Montagna; Yi He Ling; Do Sun Byun; Shannon Nasser; Diego Arango; Joongho Shin; Lidija Klampfer; Leonard H. Augenlicht; Roman Perez Soler; John M. Mariadason

Cetuximab is a monoclonal antibody that targets the human epidermal growth factor receptor (EGFR). Although approved for use in EGFR-overexpressing advanced colorectal cancer, recent studies have shown a lack of association between EGFR overexpression and cetuximab response, requiring the identification of novel biomarkers predictive of response to this agent. To do so, 22 colon cancer cell lines were screened for cetuximab response in vitro and sensitive and resistant lines were identified. In sensitive cell lines, cetuximab induced a G(0)-G(1) arrest without inducing apoptosis. Notably, cetuximab-sensitive but not cetuximab-resistant cell lines were preferentially responsive to EGF-stimulated growth. Whereas neither EGFR protein/mRNA expression nor gene copy number correlated with cetuximab response, examination of the mutation status of signaling components downstream of EGFR showed that cell lines with activating PIK3CA mutations or loss of PTEN expression (PTEN null) were more resistant to cetuximab than PIK3CA wild type (WT)/PTEN-expressing cell lines (14 +/- 5.0% versus 38.5 +/- 6.4% growth inhibition, mean +/- SE; P = 0.008). Consistently, PIK3CA mutant isogenic HCT116 cells showed increased resistance to cetuximab compared with PIK3CA WT controls. Furthermore, cell lines that were PIK3CA mutant/PTEN null and Ras/BRAF mutant were highly resistant to cetuximab compared with those without dual mutations/PTEN loss (10.8 +/- 4.3% versus 38.8 +/- 5.9% growth inhibition, respectively; P = 0.002), indicating that constitutive and simultaneous activation of the Ras and PIK3CA pathways confers maximal resistance to this agent. A priori screening of colon tumors for PTEN expression status and PIK3CA and Ras/BRAF mutation status could help stratify patients likely to benefit from this therapy.


Molecular Biology of the Cell | 2008

HDAC4 Promotes Growth of Colon Cancer Cells via Repression of p21

Andrew J. Wilson; Do Sun Byun; Shannon Nasser; Lucas B. Murray; Kanyalakshmi Ayyanar; Diego Arango; Maria E. Figueroa; Ari Melnick; Gary D. Kao; Leonard H. Augenlicht; John M. Mariadason

The class II Histone deacetylase (HDAC), HDAC4, is expressed in a tissue-specific manner, and it represses differentiation of specific cell types. We demonstrate here that HDAC4 is expressed in the proliferative zone in small intestine and colon and that its expression is down-regulated during intestinal differentiation in vivo and in vitro. Subcellular localization studies demonstrated HDAC4 expression was predominantly nuclear in proliferating HCT116 cells and relocalized to the cytoplasm after cell cycle arrest. Down-regulating HDAC4 expression by small interfering RNA (siRNA) in HCT116 cells induced growth inhibition and apoptosis in vitro, reduced xenograft tumor growth, and increased p21 transcription. Conversely, overexpression of HDAC4 repressed p21 promoter activity. p21 was likely a direct target of HDAC4, because HDAC4 down-regulation increased p21 mRNA when protein synthesis was inhibited by cycloheximide. The importance of p21 repression in HDAC4-mediated growth promotion was demonstrated by the failure of HDAC4 down-regulation to induce growth arrest in HCT116 p21-null cells. HDAC4 down-regulation failed to induce p21 when Sp1 was functionally inhibited by mithramycin or siRNA-mediated down-regulation. HDAC4 expression overlapped with that of Sp1, and a physical interaction was demonstrated by coimmunoprecipitation. Chromatin immunoprecipitation (ChIP) and sequential ChIP analyses demonstrated Sp1-dependent binding of HDAC4 to the proximal p21 promoter, likely directed through the HDAC4-HDAC3-N-CoR/SMRT corepressor complex. Consistent with increased transcription, HDAC4 or SMRT down-regulation resulted in increased histone H3 acetylation at the proximal p21 promoter locus. These studies identify HDAC4 as a novel regulator of colon cell proliferation through repression of p21.


Cancer Research | 2010

Apoptotic Sensitivity of Colon Cancer Cells to Histone Deacetylase Inhibitors Is Mediated by an Sp1/Sp3-Activated Transcriptional Program Involving Immediate-Early Gene Induction

Andrew J. Wilson; Anderly C. Chueh; Lars Tögel; Georgia A. Corner; Naseem Ahmed; Sanjay Goel; Do Sun Byun; Shannon Nasser; Michele A. Houston; Minaxi Jhawer; Helena J.M. Smartt; Lucas B. Murray; Courtney Nicholas; Barbara G. Heerdt; Diego Arango; Leonard H. Augenlicht; John M. Mariadason

Histone deacetylase inhibitors (HDACi) induce growth arrest and apoptosis in colon cancer cells and are being considered for colon cancer therapy. The underlying mechanism of action of these effects is poorly defined with both transcription-dependent and -independent mechanisms implicated. We screened a panel of 30 colon cancer cell lines for sensitivity to HDACi-induced apoptosis and correlated the differences with gene expression patterns induced by HDACi in the five most sensitive and resistant lines. A robust and reproducible transcriptional response involving coordinate induction of multiple immediate-early (fos, jun, egr1, egr3, atf3, arc, nr4a1) and stress response genes (Ndrg4, Mt1B, Mt1E, Mt1F, Mt1H) was selectively induced in HDACi sensitive cells. Notably, a significant percentage of these genes were basally repressed in colon tumors. Bioinformatics analysis revealed that the promoter regions of the HDACi-induced genes were enriched for KLF4/Sp1/Sp3 transcription factor binding sites. Altering KLF4 levels failed to modulate apoptosis or transcriptional responses to HDACi treatment. In contrast, HDACi preferentially stimulated the activity of Spl/Sp3 and blocking their action attenuated both the transcriptional and apoptotic responses to HDACi treatment. Our findings link HDACi-induced apoptosis to activation of a Spl/Sp3-mediated response that involves derepression of a transcriptional network basally repressed in colon cancer.


Molecular Nutrition & Food Research | 2008

Expression of selenium-binding protein 1 characterizes intestinal cell maturation and predicts survival for patients with colorectal cancer

Tianhong Li; Wancai Yang; Maomi Li; Do Sun Byun; Chang Tong; Shannon Nasser; Min Zhuang; Diego Arango; John M. Mariadason; Leonard H. Augenlicht

To identify candidate genes involved in the development of colorectal cancer, we used cDNA microarrays to analyze gene expression differences between human colorectal tumors and paired adjacent normal mucosa. We identified approximately 3.5-fold significant downregulation of selenium-binding protein 1 (SBP1) in colorectal tumors compared to normal mucosa (p = 0.003). Importantly, stage III colorectal cancer patients with low tumor-SBP1 expression had significantly shorter disease-free and overall survival as compared with those patients with high tumor-SBP1 expression (p = 0.04 and 0.03, respectively). We further characterized the role of SBP1 in colorectal cancer in vivo and in vitro. In normal tissue, SBP1 was maximally expressed in terminally differentiated epithelial cells on the luminal surface of crypts in the large intestine. Consistent with this in vivo localization, SBP1 was upregulated during in vitro colonic cell differentiation along the absorptive (Caco-2) and secretory (HT29 Clones 16E and 19A) cell lineages. Downregulation (approximately 50%) of SBP1 expression by small interfering RNA in colonic cancer cells was associated with reduced expression of another epithelial differentiation marker, carcinoembryonic antigen (CEA), although PCNA and p21(WAF1/cip1 )expression were not altered. These data demonstrate that higher expression of SBP1 is associated with differentiation of the normal colonic epithelia and may be a positive prognostic factor for survival in stage III colorectal carcinoma.


Journal of Proteomics | 2008

Proteomic changes during intestinal cell maturation in vivo.

Jinsook Chang; Mark R. Chance; Courtney Nicholas; Naseem Ahmed; Sandra Guilmeau; Marta Flandez; Donghai Wang; Do Sun Byun; Shannon Nasser; Joseph M. Albanese; Georgia A. Corner; Barbara G. Heerdt; Andrew J. Wilson; Leonard H. Augenlicht; John M. Mariadason

Intestinal epithelial cells undergo progressive cell maturation as they migrate along the crypt-villus axis. To determine molecular signatures that define this process, proteins differentially expressed between the crypt and villus were identified by 2D-DIGE and MALDI-MS. Forty-six differentially expressed proteins were identified, several of which were validated by immunohistochemistry. Proteins upregulated in the villus were enriched for those involved in brush border assembly and lipid uptake, established features of differentiated intestinal epithelial cells. Multiple proteins involved in glycolysis were also upregulated in the villus, suggesting increased glycolysis is a feature of intestinal cell differentiation. Conversely, proteins involved in nucleotide metabolism, and protein processing and folding were increased in the crypt, consistent with functions associated with cell proliferation. Three novel paneth cell markers, AGR2, HSPA5 and RRBP1 were also identified. Notably, significant correlation was observed between overall proteomic changes and corresponding gene expression changes along the crypt-villus axis, indicating intestinal cell maturation is primarily regulated at the transcriptional level. This proteomic profiling analysis identified several novel proteins and functional processes differentially induced during intestinal cell maturation in vivo. Integration of proteomic, immunohistochemical, and parallel gene expression datasets demonstrate the coordinated manner in which intestinal cell maturation is regulated.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2011

Intestinal epithelial-specific PTEN inactivation results in tumor formation

Do Sun Byun; Naseem Ahmed; Shannon Nasser; Joongho Shin; Sheren Al-Obaidi; Sanjay Goel; Georgia A. Corner; Andrew J. Wilson; Dustin J. Flanagan; David S. Williams; Leonard H. Augenlicht; Elizabeth Vincan; John M. Mariadason

Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a negative regulator of phosphatidylinositol 3-kinase (PI3K) signaling that is frequently inactivated in colorectal cancer through mutation, loss of heterozygosity, or epigenetic mechanisms. The aim of this study was to determine the effect of intestinal-specific PTEN inactivation on intestinal epithelial homeostasis and tumorigenesis. PTEN was deleted specifically in the intestinal epithelium, by crossing PTEN(Lox/Lox) mice with villin(Cre) mice. PTEN was robustly expressed in the intestinal epithelium and maximally in the differentiated cell compartment. Targeted inactivation of PTEN in the intestinal epithelium of PTEN(Lox/Lox)/villin(Cre) mice was confirmed by genotyping, immunohistochemistry, and qPCR. While intestinal-specific PTEN deletion did not have a major effect on cell fate determination or proliferation in the small intestine, it did increase phosphorylated (p) protein kinase B (AKT) expression in the intestinal epithelium, and 19% of animals developed small intestinal adenomas and adenocarcinomas at 12 mo of age. These tumors demonstrated pAKT and nuclear β-catenin staining, indicating simultaneous activation of the PI3K/AKT and Wnt signaling pathways. These findings demonstrate that, while PTEN inactivation alone has a minimal effect on intestinal homeostasis, it can facilitate tumor promotion upon deregulation of β-catenin/TCF signaling, further establishing PTEN as a bona fide tumor suppressor gene in intestinal cancer.


Cancer Research | 2009

An A13 Repeat within the 3′-Untranslated Region of Epidermal Growth Factor Receptor (EGFR) Is Frequently Mutated in Microsatellite Instability Colon Cancers and Is Associated with Increased EGFR Expression

Ziqiang Yuan; Joongho Shin; Andrew J. Wilson; Sanjay Goel; Yi-He Ling; Naseem Ahmed; Higinio Dopeso; Minaxi Jhawer; Shannon Nasser; Cristina Montagna; Kenneth Fordyce; Leonard H. Augenlicht; Lauri A. Aaltonen; Diego Arango; Thomas K. Weber; John M. Mariadason


Gastroenterology | 2007

p27kip1 Regulates cdk2 Activity in the Proliferating Zone of the Mouse Intestinal Epithelium: Potential Role in Neoplasia

Helena J M Smartt; Sandra Guilmeau; Shannon Nasser; Courtney Nicholas; Laura Bancroft; Sharon A. Simpson; Nancy Yeh; Wancai Yang; John M. Mariadason; Andrew Koff; Leonard H. Augenlicht


Cancer Research | 2010

An A13 repeat within the 3′-untranslated region of Epidermal Growth Factor Receptor (EGFR) is frequently mutated in microsatellite instability colon cancers and is associated with increased EGFR expression (Cancer Research (2009) 69, (7811-7818))

Ziqiang Yuan; Joongho Shin; Andrew J. Wilson; Sanjay Goel; Y. H. Ling; Naseem Ahmed; Higinio Dopeso; Minaxi Jhawer; Shannon Nasser; Cristina Montagna; Kenneth Fordyce; Leonard H. Augenlicht; Lauri A. Aaltonen; Diego Arango; Thomas K. Weber; John M. Mariadason


Journal of The American College of Surgeons | 2008

A novel deletion A mutation within the 3' untranslated region of the EGFR gene contributes to EGFR overexpression in microsatellite unstable (MSI) colon cancer (CRC) that results from increased mRNA stability

Joongho Shin; Azadeh Azarbayejani; Ziqian Yuan; Minaxi Jhawer; Shannon Nasser; Kenneth Fordyce; Diego Arrango; John M. Mariadason; Thomas K. Weber

Collaboration


Dive into the Shannon Nasser's collaboration.

Top Co-Authors

Avatar

John M. Mariadason

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Minaxi Jhawer

Montefiore Medical Center

View shared research outputs
Top Co-Authors

Avatar

Diego Arango

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joongho Shin

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Sanjay Goel

Montefiore Medical Center

View shared research outputs
Top Co-Authors

Avatar

Naseem Ahmed

Montefiore Medical Center

View shared research outputs
Top Co-Authors

Avatar

Cristina Montagna

Albert Einstein College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge