Colorectal cancer (CRC) has served as a unique platform for durable insights into and advances in molecular pathogenesis, chemoprevention, diagnosis, and therapy, not only in this venue but expanded into other disease states. The elucidation of these comprehensive aspects of CRC has been facilitated through the identification of key genes and molecular pathways. It is well accepted that the majority of sporadic colorectal cancers feature the chromosomal instability pathway, involving alterations in key tumor suppressor genes (APC and p53, but also SMAD4) and oncogenes (especially Ras, but also EGFR, c-myc, B-Raf) and their downstream effectors. A subset of sporadic colon cancers undergo microsatellite instability (MSI) pathway, typically right-sided with B-Raf mutations but without Ras mutations. Recently, it has been advocated that certain colorectal cancers feature hypermethylation. We have genetically and physically mapped a region of loss of heterozygosity (LOH) on chromosome 22q13.31 as a basis to understand the role of new genes in colorectal carcinogenesis (and other cancers). We now have discovered a new telomeric region of LOH, which harbors two microRNAs, namely Let-7a3 and Let-7b, and these microRNAs are downregulated in up to 40% of colorectal cancers. MicroRNAs have become increasingly recognized for their pivotal, diverse roles in development, differentiation, proliferation, and cancer, which is achieved the degradation or interference with translation with messenger RNAs. We hypothesize that the Let-7a3 and Let-7b microRNAs are instrumental in colorectal cancer progression, and this may be achieved by targeting the Ras oncogene, and also, the c-myc oncogene. This novel hypothesis will be pursued by the following interrelated Specific Aims: (1). To identify the roles of the Let-7a3 and Let-7b microRNAs in colorectal cancer. A. To determine the relationship between these microRNAs and clinical parameters and molecular parameters;B. To determine the function(s) of Let7a and Let7b in colorectal carcinogenesis through genetic /overexpression/knockdown studies of these microRNAs with determination of effects upon cellular proliferation, apoptosis, migration and invasion, which are critical features of the colon cancer initiation and progression. (2). To determine if the Ras and c-myc oncogenes represent targets of Let-7a3 and Let- 7b. This will be achieved through examination of Ras transcriptional and protein activities in the setting of genetic approaches, namely Let-7a3 and Let-7b overexpression/knockdown. (3) To determine the functional consequences of restoration and inhibition of Let-7a3 and Let-7b in colon cancer cells in vivo. This will be assessed by innovative three-dimensional organotypic cultures, which mimic the colon cancer microenvironment, and in athymic, irradiated nude mice through bioluminescence of tumor growth. In aggregate, our studies have the means to unravel new pathways of regulation of the Ras and c-myc oncogenes by the Let-7a3 and Let-7b microRNAs in colon cancer, especially where sporadic colorectal cancers (chromosomal instability pathway) do not harbor Ras mutations.

Public Health Relevance

Colorectal cancer (CRC) serves as a paradigm for the investigation of basic mechanisms and translation into novel chemopreventive, diagnostic and therapeutic strategies. The underlying pathogenesis of CRC may involve the Let-7a3 and Let-7b microRNAs, which in general are critical regulators of normal cellular processes to malignant transformation. Our studies hope to reveal new paradigms in CRC pathogenesis, and offer opportunities for exploiting this knowledge in novel diagnostics and therapeutics.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
5R01DK056645-11
Application #
7897807
Study Section
Special Emphasis Panel (ZRG1-DIG-C (05))
Program Officer
Hamilton, Frank A
Project Start
2000-03-01
Project End
2013-05-31
Budget Start
2010-06-01
Budget End
2011-05-31
Support Year
11
Fiscal Year
2010
Total Cost
$362,448
Indirect Cost
Name
University of Pennsylvania
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
042250712
City
Philadelphia
State
PA
Country
United States
Zip Code
19104
Chatterji, Priya; Hamilton, Kathryn E; Liang, Shun et al. (2018) The LIN28B-IMP1 post-transcriptional regulon has opposing effects on oncogenic signaling in the intestine. Genes Dev 32:1020-1034
Mizuno, Rei; Chatterji, Priya; Andres, Sarah et al. (2018) Differential Regulation of LET-7 by LIN28B Isoform-Specific Functions. Mol Cancer Res 16:403-416
Chatterji, Priya; Rustgi, Anil K (2018) RNA Binding Proteins in Intestinal Epithelial Biology and Colorectal Cancer. Trends Mol Med 24:490-506
Giroux, VĂ©ronique; Stephan, Julien; Chatterji, Priya et al. (2018) Mouse Intestinal Krt15+ Crypt Cells Are Radio-Resistant and Tumor Initiating. Stem Cell Reports 10:1947-1958
Andres, Sarah F; Williams, Kathy N; Rustgi, Anil K (2018) The Molecular Basis of Metastatic Colorectal Cancer. Curr Colorectal Cancer Rep 14:69-79
Zhou, Jin; Wu, Zhong; Wong, Gabrielle et al. (2017) CDK4/6 or MAPK blockade enhances efficacy of EGFR inhibition in oesophageal squamous cell carcinoma. Nat Commun 8:13897
Heeg, Steffen; Das, Koushik K; Reichert, Maximilian et al. (2016) ETS-Transcription Factor ETV1 Regulates Stromal Expansion and Metastasis in Pancreatic Cancer. Gastroenterology 151:540-553.e14
Liu, Qi; Zhong, Xue; Madison, Blair B et al. (2015) Assessing Computational Steps for CLIP-Seq Data Analysis. Biomed Res Int 2015:196082
Asfaha, Samuel; Hayakawa, Yoku; Muley, Ashlesha et al. (2015) Krt19(+)/Lgr5(-) Cells Are Radioresistant Cancer-Initiating Stem Cells in the Colon and Intestine. Cell Stem Cell 16:627-38
Hamilton, Kathryn E; Crissey, Mary Ann S; Lynch, John P et al. (2015) Culturing adult stem cells from mouse small intestinal crypts. Cold Spring Harb Protoc 2015:354-8

Showing the most recent 10 out of 36 publications