Network


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

Hotspot


Dive into the research topics where Lois Myeroff is active.

Publication


Featured researches published by Lois Myeroff.


Journal of Biological Chemistry | 1995

Demonstration That Mutation of the Type II Transforming Growth Factor β Receptor Inactivates Its Tumor Suppressor Activity in Replication Error-positive Colon Carcinoma Cells

Jing Wang; LuZhe Sun; Lois Myeroff; Xiao-Fan Wang; Larry E. Gentry; Junhua Yang; Jiurong Liang; Elizabeth Zborowska; Sanford D. Markowitz; James K V Willson; Michael G. Brattain

Escape from negative growth regulation by transforming growth factor β (TGF-β) as a result of the loss of TGF-β type II receptor (RII) expression has been found to be associated with the replication error (RER) colorectal cancer genotype, which is characteristic of hereditary nonpolyposis colorectal cancers. The RER-positive HCT 116 colon carcinoma cell line was examined for RII mutations. A 1-base deletion was found within a sequence of 10 repeating adenines (nucleotides 709-718), which resulted in a frameshift mutation. Although it is reasonable to predict that the loss of RII function would be an important determinant of malignancy, the large number of potential mutations in cells of this phenotype raises the possibility that an RII mutation may not be a key event in the tumorigenic phenotype of these cells. One way to test directly the importance of RII mutations in determining the malignant phenotype would be to restore its expression. If restoration of expression leads to diminished tumorigenicity, it would indicate that RII mutation is an important determinant of malignancy in the RER phenotype. To determine whether restoration of RII would lead to reversal of malignancy in RER colon cancers, an RII expression vector was transfected into the HCT 116 cell line. RII stable clones showed mRNA and protein expression of transfected RII. The fibronectin mRNA level was increased by exogenous TGF-β treatment in a dose-dependent manner in RII-positive clones, whereas the control cells remained insensitive. The RII transfectants showed reduced clonogenicity in both monolayer culture and soft agarose. They were growth arrested at a lower saturation density than control cells. TGF-β-neutralizing antibody stimulated the proliferation of RII-transfected but not control cells, indicating that the alterations in the growth parameters of the transfected cells were due to the acquisition of autocrine-negative activity. Tumorigenicity in athymic mice was reduced and delayed in RII transfectants. These results indicate that reconstitution of TGF-β autocrine activity by reexpression of RII can reverse malignancy in RER colon cancers, thus verifying that the malignancy of hereditary nonpolyposis colorectal cancer can be directly associated with the loss of RII expression.


Proceedings of the National Academy of Sciences of the United States of America | 2003

SLC5A8, a sodium transporter, is a tumor suppressor gene silenced by methylation in human colon aberrant crypt foci and cancers

Hui Li; Lois Myeroff; Dominic J. Smiraglia; Michael F. Romero; Theresa P. Pretlow; Lakshmi Kasturi; James Lutterbaugh; Ronald M. Rerko; Graham Casey; Jean-Pierre Issa; Joseph Willis; James K V Willson; Christoph Plass; Sanford D. Markowitz

We identify a gene, SLC5A8, and show it is a candidate tumor suppressor gene whose silencing by aberrant methylation is a common and early event in human colon neoplasia. Aberrant DNA methylation has been implicated as a component of an epigenetic mechanism that silences genes in human cancers. Using restriction landmark genome scanning, we performed a global search to identify genes that would be aberrantly methylated at high frequency in human colon cancer. From among 1,231 genomic NotI sites assayed, site 3D41 was identified as methylated in 11 of 12 colon cancers profiled. Site 3D41 mapped to exon 1 of SLC5A8, a transcript that we assembled. In normal colon mucosa we found that SLC5A8 exon 1 is unmethylated and SLC5A8 transcript is expressed. In contrast, SLC5A8 exon 1 proved to be aberrantly methylated in 59% of primary colon cancers and 52% of colon cancer cell lines. SLC5A8 exon 1 methylated cells were uniformly silenced for SLC5A8 expression, but reactivated expression on treatment with a demethylating drug, 5-azacytidine. Transfection of SLC5A8 suppressed colony growth in each of three SLC5A8-deficient cell lines, but showed no suppressive effect in any of three SLC5A8-proficient cell lines. SLC5A8 exon 1 methylation is an early event, detectable in colon adenomas, and in even earlier microscopic colonic aberrant crypt foci. Structural homology and functional testing demonstrated that SLC5A8 is a member of the family of sodium solute symporters, which are now added as a class of candidate colon cancer suppressor genes.


Science | 2012

Epigenomic Enhancer Profiling Defines a Signature of Colon Cancer

Batool Akhtar-Zaidi; Richard Cowper-Sal·lari; Olivia Corradin; Alina Saiakhova; Cynthia F. Bartels; Dheepa Balasubramanian; Lois Myeroff; James Lutterbaugh; Awad Jarrar; Matthew F. Kalady; Joseph Willis; Jason H. Moore; Paul J. Tesar; Thomas LaFramboise; Sanford D. Markowitz; Mathieu Lupien; Peter C. Scacheri

Colorectal Cancer Signature The mutations and genome aberrations that characterize cancer result in often dramatically altered gene and protein expression patterns. It is these altered expression patterns that directly and indirectly drive progression of the disease. In human primary colorectal cancer cells, Akhtar-Zaidi et al. (p. 736, published online 12 April) analyzed the pattern of epigenetically modified chromatin at “enhancer” sequences that are known to be critical in the control of gene expression. An epigenetic enhancer signature was defined that was specifically associated with colorectal cancer cells. Methylation tags at long-distance gene regulatory elements provide a signature specific to cancer cells. Cancer is characterized by gene expression aberrations. Studies have largely focused on coding sequences and promoters, even though distal regulatory elements play a central role in controlling transcription patterns. We used the histone mark H3K4me1 to analyze gain and loss of enhancer activity genome-wide in primary colon cancer lines relative to normal colon crypts. We identified thousands of variant enhancer loci (VELs) that comprise a signature that is robustly predictive of the in vivo colon cancer transcriptome. Furthermore, VELs are enriched in haplotype blocks containing colon cancer genetic risk variants, implicating these genomic regions in colon cancer pathogenesis. We propose that reproducible changes in the epigenome at enhancer elements drive a specific transcriptional program to promote colon carcinogenesis.


Proceedings of the National Academy of Sciences of the United States of America | 2002

HLTF gene silencing in human colon cancer

Helen Moinova; Wei Dong Chen; Lanlan Shen; Dominic J. Smiraglia; Joseph Olechnowicz; Lakshmeswari Ravi; Lakshmi Kasturi; Lois Myeroff; Christoph Plass; Ramon Parsons; John D. Minna; James K V Willson; Sylvan B. Green; Jean-Pierre Issa; Sanford D. Markowitz

Chromatin remodeling enzymes are increasingly implicated in a variety of important cellular functions. Various components of chromatin remodeling complexes, including several members of the SWI/SNF family, have been shown to be disrupted in cancer. In this study we identified as a target for gene inactivation in colon cancer the gene for helicase-like transcription factor (HLTF), a SWI/SNF family protein. Loss of HLTF expression accompanied by HLTF promoter methylation was noted in nine of 34 colon cancer cell lines. In these cell lines HLTF expression was restored by treatment with the demethylating agent 5-azacytidine. In further studies of primary colon cancer tissues, HLTF methylation was detected in 27 of 63 cases (43%). No methylation of HLTF was detected in breast or lung cancers, suggesting selection for HLTF methylation in colonic malignancies. Transfection of HLTF suppressed 75% of colony growth in each of three different HLTF-deficient cell lines, but showed no suppressive effect in any of three HLTF-proficient cell lines. These findings show that HLTF is a common target for methylation and epigenetic gene silencing in colon cancer and suggest HLTF is a candidate colon cancer suppressor gene.


Molecular & Cellular Proteomics | 2009

Discovery and Scoring of Protein Interaction Subnetworks Discriminative of Late Stage Human Colon Cancer

Rod K. Nibbe; Sanford D. Markowitz; Lois Myeroff; Rob M. Ewing; Mark R. Chance

We used a systems biology approach to identify and score protein interaction subnetworks whose activity patterns are discriminative of late stage human colorectal cancer (CRC) versus control in colonic tissue. We conducted two gel-based proteomics experiments to identify significantly changing proteins between normal and late stage tumor tissues obtained from an adequately sized cohort of human patients. A total of 67 proteins identified by these experiments was used to seed a search for protein-protein interaction subnetworks. A scoring scheme based on mutual information, calculated using gene expression data as a proxy for subnetwork activity, was developed to score the targets in the subnetworks. Based on this scoring, the subnetwork was pruned to identify the specific protein combinations that were significantly discriminative of late stage cancer versus control. These combinations could not be discovered using only proteomics data or by merely clustering the gene expression data. We then analyzed the resultant pruned subnetwork for biological relevance to human CRC. A number of the proteins in these smaller subnetworks have been associated with the progression (CSNK2A2, PLK1, and IGFBP3) or metastatic potential (PDGFRB) of CRC. Others have been recently identified as potential markers of CRC (IFITM1), and the role of others is largely unknown in this disease (CCT3, CCT5, CCT7, and GNA12). The functional interactions represented by these signatures provide new experimental hypotheses that merit follow-on validation for biological significance in this disease. Overall the method outlines a quantitative approach for integrating proteomics data, gene expression data, and the wealth of accumulated legacy experimental data to discover significant protein subnetworks specific to disease.


Cancer Research | 2009

BCL9 Promotes Tumor Progression by Conferring Enhanced Proliferative, Metastatic, and Angiogenic Properties to Cancer Cells

Mala Mani; Daniel E. Carrasco; Yunyu Zhang; Kohichi Takada; Moshe E. Gatt; Jui Dutta-Simmons; Hiroshi Ikeda; Felipe Diaz-Griffero; Victor Pena-Cruz; Monica M. Bertagnolli; Lois Myeroff; Sanford D. Markowitz; Kenneth C. Anderson; Daniel R. Carrasco

Several components of the Wnt signaling cascade have been shown to function either as tumor suppressor proteins or as oncogenes in multiple human cancers, underscoring the relevance of this pathway in oncogenesis and the need for further investigation of Wnt signaling components as potential targets for cancer therapy. Here, using expression profiling analysis as well as in vitro and in vivo functional studies, we show that the Wnt pathway component BCL9 is a novel oncogene that is aberrantly expressed in human multiple myeloma as well as colon carcinoma. We show that BCL9 enhances beta-catenin-mediated transcriptional activity regardless of the mutational status of the Wnt signaling components and increases cell proliferation, migration, invasion, and the metastatic potential of tumor cells by promoting loss of epithelial and gain of mesenchymal-like phenotype. Most importantly, BCL9 knockdown significantly increased the survival of xenograft mouse models of cancer by reducing tumor load, metastasis, and host angiogenesis through down-regulation of c-Myc, cyclin D1, CD44, and vascular endothelial growth factor expression by tumor cells. Together, these findings suggest that deregulation of BCL9 is an important contributing factor to tumor progression. The pleiotropic roles of BCL9 reported in this study underscore its value as a drug target for therapeutic intervention in several malignancies associated with aberrant Wnt signaling.


Journal of Clinical Investigation | 1994

A benign cultured colon adenoma bears three genetically altered colon cancer oncogenes, but progresses to tumorigenicity and transforming growth factor-beta independence without inactivating the p53 tumor suppressor gene.

Sanford D. Markowitz; Lois Myeroff; Mark J. Cooper; June L. Traicoff; Mary Kochera; I James Lutterbaugh; Margaret Swiriduk; James K V Willson

We describe the spontaneous progression of a colon adenoma cell line to tumorigenicity and growth factor independence. This system allows direct comparison of biologic stages of malignant progression with alterations of colon cancer suppressor genes and oncogenes. VACO-235, a human colon adenoma cell line, is at early passages nontumorigenic in the nude mouse, unable to grow in soft agar, growth stimulated by serum and EGF, and growth inhibited by TGF-beta. VACO-235 daughter passages 93 and higher have in culture spontaneously progressed to being weakly tumorigenic, but retain all other growth characteristics of VACO-235 early passages. A mouse xenograft from late passage VACO-235 was reestablished in culture as the granddaughter cell line, VACO-411. VACO-411 is highly tumorigenic, clones in soft agar, and is unresponsive to serum, EGF, and TGF-beta. Early passage VACO-235 bears a mutant K-ras allele, bears only mutant APC alleles, expresses no DCC transcripts, and expresses only wild type p53 transcripts. VACO-411 retains the identical genotype, still expressing only wild type p53. Colonic cells after ras mutation, APC mutation, and DCC inactivation remain nontumorigenic and growth factor dependent. Malignant progression involves at least two additional steps, and in VACO-411 can proceed by a novel pathway not requiring p53 inactivation.


Genes, Chromosomes and Cancer | 2008

Mutational inactivation of TGFBR2 in microsatellite unstable colon cancer arises from the cooperation of genomic instability and the clonal outgrowth of transforming growth factor β resistant cells

Swati Biswas; Patricia Trobridge; Judith Romero-Gallo; Dean Billheimer; Lois Myeroff; James K V Willson; Sanford D. Markowitz; William M. Grady

The mutational inactivation of transforming growth factor β receptor type II (TGFBR2) occurs in ∼30% of colon cancers and promotes the formation of colon cancer by inhibiting the tumor suppressor activity of the TGFB signaling pathway. TGFBR2 mutations occur in >90% of microsatellite unstable (MSI) colon cancers and affect a polyadenine tract in exon 3 of TGFBR2, called BAT‐RII, which is vulnerable to mutation in the setting of DNA mismatch repair (MMR) system deficiency. In light of the vulnerable nature of the BAT‐RII tract in the setting of MMR inactivation and the favorable effects of TGFBR2 inactivation in colon cancer, analysis of TGFBR2 inactivation provides an opportunity to assess the roles of genomic instability vs. clonal selection in cells acquiring TGFBR2 BAT‐RII tract mutations in MSI colon cancer formation. The contribution of genomic instability and/or clonal evolution to the mutational inactivation of TGBFR2 in MSI colon cancers has not been studied in a systematic way that would allow a determination of the relative contribution of these two mechanisms in the formation of MSI colon cancer. It has not been demonstrated whether the BAT‐RII tract mutations are strictly a consequence of the BAT‐RII region being hypermutable in the setting of MMR deficiency or if the mutations are rather a consequence of clonal selection pressure against the TGFB receptor. Through the use of defined cell line systems, we show that both genomic instability and clonal selection of TGFB resistant cells contribute to the high frequency of TGFBR2 mutations in MSI colon cancer.


Genome Medicine | 2012

H3K4me3 inversely correlates with DNA methylation at a large class of non-CpG-island-containing start sites

Dheepa Balasubramanian; Batool Akhtar-Zaidi; Lingyun Song; Cynthia F. Bartels; Martina L. Veigl; Lydia Beard; Lois Myeroff; Kishore Guda; James Lutterbaugh; Joseph Willis; Gregory E. Crawford; Sanford D. Markowitz; Peter C. Scacheri

BackgroundIn addition to mutations, epigenetic silencing of genes has been recognized as a fundamental mechanism that promotes human carcinogenesis. To date, characterization of epigenetic gene silencing has largely focused on genes in which silencing is mediated by hypermethylation of promoter-associated CpG islands, associated with loss of the H3K4me3 chromatin mark. Far less is known about promoters lacking CpG-islands or genes that are repressed by alternative mechanisms.MethodsWe performed integrative ChIP-chip, DNase-seq, and global gene expression analyses in colon cancer cells and normal colon mucosa to characterize chromatin features of both CpG-rich and CpG-poor promoters of genes that undergo silencing in colon cancer.ResultsEpigenetically repressed genes in colon cancer separate into two classes based on retention or loss of H3K4me3 at transcription start sites. Quantitatively, of transcriptionally repressed genes that lose H3K4me3 in colon cancer (K4-dependent genes), a large fraction actually lacks CpG islands. Nonetheless, similar to CpG-island containing genes, cytosines located near the start sites of K4-dependent genes become DNA hypermethylated, and repressed K4-dependent genes can be reactivated with 5-azacytidine. Moreover, we also show that when the H3K4me3 mark is retained, silencing of CpG island-associated genes can proceed through an alternative mechanism in which repressive chromatin marks are recruited.ConclusionsH3K4me3 equally protects from DNA methylation at both CpG-island and non-CpG island start sites in colon cancer. Moreover, the results suggest that CpG-rich genes repressed by loss of H3K4me3 and DNA methylation represent special instances of a more general epigenetic mechanism of gene silencing, one in which gene silencing is mediated by loss of H3K4me3 and methylation of non-CpG island promoter-associated cytosines.


Oncotarget | 2015

Induction of KIAA1199/CEMIP is associated with colon cancer phenotype and poor patient survival

Stephen P. Fink; Lois Myeroff; Revital Kariv; Petra Platzer; Baozhong Xin; Debra Mikkola; Earl Lawrence; Nathan Morris; Arman Nosrati; James Willson; Joseph Willis; Martina L. Veigl; Jill S. Barnholtz-Sloan; Zhenghe Wang; Sanford D. Markowitz

Genes induced in colon cancer provide novel candidate biomarkers of tumor phenotype and aggressiveness. We originally identified KIAA1199 (now officially called CEMIP) as a transcript highly induced in colon cancer: initially designating the transcript as Colon Cancer Secreted Protein 1. We molecularly characterized CEMIP expression both at the mRNA and protein level and found it is a secreted protein induced an average of 54-fold in colon cancer. Knockout of CEMIPreduced the ability of human colon cancer cells to form xenograft tumors in athymic mice. Tumors that did grow had increased deposition of hyaluronan, linking CEMIP participation in hyaluronan degradation to the modulation of tumor phenotype. We find CEMIP mRNA overexpression correlates with poorer patient survival. In stage III only (n = 31) or in combined stage II plus stage III colon cancer cases (n = 73), 5-year overall survival was significantly better (p = 0.004 and p = 0.0003, respectively) among patients with low CEMIP expressing tumors than those with high CEMIP expressing tumors. These results demonstrate that CEMIP directly facilitates colon tumor growth, and high CEMIP expression correlates with poor outcome in stage III and in stages II+III combined cohorts. We present CEMIP as a candidate prognostic marker for colon cancer and a potential therapeutic target.

Collaboration


Dive into the Lois Myeroff's collaboration.

Top Co-Authors

Avatar

Sanford D. Markowitz

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

James K V Willson

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Joseph Willis

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

James Lutterbaugh

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Lakshmi Kasturi

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar

Ramon Parsons

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

William M. Grady

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Bert Vogelstein

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Peter C. Scacheri

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Alina Saiakhova

Case Western Reserve University

View shared research outputs
Researchain Logo
Decentralizing Knowledge