Functional inactivation of menin, encoded by the MEN1 gene, causes the inherited multiple endocrine neoplasia type 1 (MEN1) syndrome and some but not all sporadic pancreatic endocrine tumors. Therefore, unraveling molecular events upstream or downstream of menin could point to other causative genes and/or regulatory events responsible for such tumor types. Menin resides in a histone methylating protein complex that trimethylates histone H3 at lysine-4 (H3K4me3), an epigenetic mark for active gene expression. Therefore, we have determined a genome-wide map of menin-dependent H3K4me3 (using ChIP-Seq) and menin-dependent gene-expression program in wild-type (WT) and menin-null mouse embryonic stem cells (ESCs) and in pancreatic islet-like endocrine cells (PILECs), which we derived from WT and menin-null mouse ESCs through in vitro differentiation. We found menin-dependent H3K4me3 specifically targeting the Meg3 gene in mouse ESCs, and all four Hox loci in differentiated PILECs. Gene expression from the Meg3 locus and from all of the four Hox loci was abolished in menin-null cells. Both Meg3 and Hox loci have been implicated in MEN1-like sporadic tumors: MEG3 in pituitary tumors, and HOX in parathyroid tumors. Our data suggest that these genes with menin-dependent H3K4me3 could be relevant players in the tumorigenesis of endocrine cell types associated with MEN1. Furthermore, our work shows that menin-null mouse ESCs could also be differentiated in vitro into islet-like endocrine cells, underscoring the utility of menin-null ESC-derived specialized cell types for genome-wide analyses studies. MEG3 is a tumor suppressor long non-coding RNA. We showed that menin's tumor suppressor activity by epigenetic up-regulation of MEG3 leads to down-regulation of its target, a proto-oncogene (c-MET). We also found a reciprocal correlation of MEG3 (low) and c-MET (high) levels in human MEN1-associated and sporadic insulinomas. Our current efforts are directed towards understanding the regulation and activity of genes at the MEG3 and HOX loci. One of the tumors associated with the MEN1 syndrome is lipoma. This is a benign tumor usually only removed for cosmetic reasons. Therefore, studying this tumor of fat cells (adipocytes) is challenging due to the non-availability of tumor specimens from human MEN1 patients or from the mouse model of this disease. We used a novel approach to study lipoma cells by using in vitro differentiation to derive normal and menin-deficient adipocytes. We found a novel association of menin in the regulation of adipocyte size because menin-deficient adipocytes were larger. By gene expression microarray analysis we found novel targets of menin: differentially methylated genes including MEG3, and the mouse prolactin gene family locus. Our findings support a role for menin in the regulation of: adipocyte size, differential DNA methylation and coordinately expressed genes in gene clusters. We have shown that cyclin-dependent kinase inhibitors (CDKIs) of the INK4 family (4 genes) and the Cip/Kip family (3 genes) that negatively regulate cell cycle progression and cell proliferation have rare germline or somatic mutations in endocrine tumor states related to MEN1. Also, mouse models show an endocrine neoplasia phenotype in 'knock-in' mice homozygous for the CDK4-R24C mutation, or by the combined loss of two different CDKIs, p18 and p27. Therefore, understanding the molecular basis of CDK and CDKI regulation could provide insights into their contribution to endocrine tumorigenesis. We have investigated the contribution of cell cycle regulators in endocrine tumorigenesis, particularly mutations in CDKI genes. We are interested in investigating the molecular basis of cell cycle regulation in endocrine cells.

Project Start
Project End
Budget Start
Budget End
Support Year
7
Fiscal Year
2015
Total Cost
Indirect Cost
Name
U.S. National Inst Diabetes/Digst/Kidney
Department
Type
DUNS #
City
State
Country
Zip Code
Parekh, Vaishali I; Modali, Sita D; Welch, James et al. (2018) Frequency and consequence of the recurrent YY1 p.T372R mutation in sporadic insulinomas. Endocr Relat Cancer 25:L31-L35
Thakur, Shilpa; Daley, Brianna; Gaskins, Kelli et al. (2018) Metformin Targets Mitochondrial Glycerophosphate Dehydrogenase to Control Rate of Oxidative Phosphorylation and Growth of Thyroid Cancer In Vitro and In Vivo. Clin Cancer Res 24:4030-4043
Agarwal, Sunita K (2017) The future: genetics advances in MEN1 therapeutic approaches and management strategies. Endocr Relat Cancer 24:T119-T134
Iyer, Sucharitha; Modali, Sita D; Agarwal, Sunita K (2017) Long Noncoding RNA MEG3 Is an Epigenetic Determinant of Oncogenic Signaling in Functional Pancreatic Neuroendocrine Tumor Cells. Mol Cell Biol 37:
Keutgen, Xavier M; Nilubol, Naris; Agarwal, Sunita et al. (2016) Reoperative Surgery in Patients with Multiple Endocrine Neoplasia Type 1 Associated Primary Hyperparathyroidism. Ann Surg Oncol :
Guan, Bin; Welch, James M; Sapp, Julie C et al. (2016) GCM2-Activating Mutations in Familial Isolated Hyperparathyroidism. Am J Hum Genet 99:1034-1044
Parekh, Vaishali I; Modali, Sita D; Desai, Shruti S et al. (2015) Consequence of Menin Deficiency in Mouse Adipocytes Derived by In Vitro Differentiation. Int J Endocrinol 2015:149826
Modali, Sita D; Parekh, Vaishali I; Kebebew, Electron et al. (2015) Epigenetic regulation of the lncRNA MEG3 and its target c-MET in pancreatic neuroendocrine tumors. Mol Endocrinol 29:224-37
Desai, Shruti S; Kharade, Sampada S; Parekh, Vaishali I et al. (2015) Pro-oncogenic Roles of HLXB9 Protein in Insulinoma Cells through Interaction with Nono Protein and Down-regulation of the c-Met Inhibitor Cblb (Casitas B-lineage Lymphoma b). J Biol Chem 290:25595-608
Gara, Sudheer Kumar; Jia, Li; Merino, Maria J et al. (2015) Germline HABP2 Mutation Causing Familial Nonmedullary Thyroid Cancer. N Engl J Med 373:448-55

Showing the most recent 10 out of 15 publications