Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cancer Res ; 5(10): 1041-51, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17951404

ABSTRACT

Multiple endocrine neoplasia type 1 (MEN1) is an autosomal dominant familial cancer syndrome characterized primarily by endocrine tumors of the parathyroids, anterior pituitary, and enteropancreatic endocrine tissues. Affected individuals carry a germ-line loss-of-function mutation of the MEN1 gene, and tumors arise after loss of the second allele. Homozygous loss of Men1 in the germ line of mice results in early embryonic lethality, with defective development of neural tube, heart, liver, and craniofacial structures. We generated immortalized wild-type (WT) and menin-null mouse embryo fibroblast (MEF) cell lines and evaluated their characteristics, including global expression patterns. The WT and menin-null cell lines were aneuploid, and the nulls did not display tumorigenic characteristics in soft agar assay. Expression arrays in menin-null MEFs revealed altered expression of several extracellular matrix proteins that are critical in organogenesis. Specifically, transcripts for fibulin 2 (Fbln2), periostin (Postn), and versican [chondroitin sulfate proteoglycan (Cspg2)], genes critical for the developing heart and known to be induced by transforming growth factor-beta (TGF-beta), were decreased in their expression in menin-null MEFs. Fbln2 expression was the most affected, and the reduction in menin-null MEFs for Fbln2, Postn, and Cspg2 was 16.18-, 5.37-, and 2.15-fold, respectively. Menin-null MEFs also showed poor response to TGF-beta-induced Smad3-mediated transcription in a reporter assay, supporting a role for menin in this pathway. Postn and Cspg2 expression in WT, unlike in null MEFs, increased on TGF-beta treatment. The expression changes associated with the loss of the tumor suppressor menin provide insights into the defective organogenesis observed during early embryonic development in Men1-null mouse embryos.


Subject(s)
Embryo, Mammalian/metabolism , Extracellular Matrix Proteins/genetics , Gene Expression Regulation, Developmental , Proto-Oncogene Proteins/physiology , Tumor Suppressor Proteins/physiology , Animals , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Mice , Organogenesis/genetics , Proto-Oncogene Proteins/genetics , Transforming Growth Factor beta/pharmacology , Tumor Suppressor Proteins/genetics
2.
Ann N Y Acad Sci ; 1014: 189-98, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15153434

ABSTRACT

Multiple endocrine neoplasia type 1 (MEN1), among all syndromes, causes tumors in the highest number of tissue types. Most of the tumors are hormone producing (e.g., parathyroid, enteropancreatic endocrine, anterior pituitary) but some are not (e.g., angiofibroma). MEN1 tumors are multiple for organ type, for regions of a discontinuous organ, and for subregions of a continuous organ. Cancer contributes to late mortality; there is no effective prevention or cure for MEN1 cancers. Morbidities are more frequent from benign than malignant tumor, and both are indicators for screening. Onset age is usually earlier in a tumor type of MEN1 than of nonhereditary cases. Broad trends contrast with those in nonneoplastic excess of hormones (e.g., persistent hyperinsulinemic hypoglycemia of infancy). Most germline or somatic mutations in the MEN1 gene predict truncation or absence of encoded menin. Similarly, 11q13 loss of heterozygosity in tumors predicts inactivation of the other MEN1 copy. MEN1 somatic mutation is prevalent in nonhereditary, MEN1-like tumor types. Compiled germline and somatic mutations show almost no genotype/phenotype relation. Normal menin is 67 kDa, widespread, and mainly nuclear. It may partner with junD, NF-kB, PEM, SMAD3, RPA2, FANCD2, NM23beta, nonmuscle myosin heavy chain II-A, GFAP, and/or vimentin. These partners have not clarified menin's pathways in normal or tumor tissues. Animal models have opened approaches to menin pathways. Local overexpression of menin in Drosophila reveals its interaction with the jun-kinase pathway. The Men1+/- mouse has robust MEN1; its most important difference from human MEN1 is marked hyperplasia of pancreatic islets, a tumor precursor stage.


Subject(s)
Gene Expression Regulation, Neoplastic , Multiple Endocrine Neoplasia/genetics , Multiple Endocrine Neoplasia/physiopathology , Proto-Oncogene Proteins/genetics , Animals , Humans , Multiple Endocrine Neoplasia/pathology
3.
Proc Natl Acad Sci U S A ; 100(19): 10770-5, 2003 Sep 16.
Article in English | MEDLINE | ID: mdl-12960363

ABSTRACT

Different components of the AP1 transcription factor complex appear to have distinct effects on cell proliferation and transformation. In contrast to other AP1 components, JunD has been shown to inhibit cell proliferation. Also, in prior studies, JunD alone bound menin, product of the MEN1 tumor suppressor gene, and JunD's transcriptional activity was inhibited by menin, suggesting that JunD might achieve all or most of its unique properties through binding to menin. Analyses of JunD and menin effects on proliferation, morphology, and cyclin D1 in stable cell lines unmasked an unexpected growth promoting activity of JunD. Whereas stable overexpression of wild-type (wt) mouse JunD in JunD-/- immortalized fibroblasts inhibited their proliferation and reverted their transformed-like phenotype, overexpression of a missense mouse JunD mutant (mJunDG42E) with disabled binding to menin showed opposite or growth promoting effects. Similarly, stable overexpression of wt mouse JunD in wt immortalized fibroblasts inhibited growth. In contrast, its overexpression in Men1-/- immortalized fibroblasts enhanced their already transformed-like characteristics. To conclude, JunD changed from growth suppressor to growth promoter when its binding to menin was prevented by a JunD mutant unable to bind menin or by Men1-null genetic background.


Subject(s)
Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins , Animals , Base Sequence , Blotting, Western , Cell Division , Cell Line , Cyclin D1/metabolism , DNA Primers , Mice , Neoplasms, Experimental/metabolism
4.
Mol Cell Biol ; 23(2): 493-509, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12509449

ABSTRACT

Menin is a 70-kDa protein encoded by MEN1, the tumor suppressor gene disrupted in multiple endocrine neoplasia type 1. In a yeast two-hybrid system based on reconstitution of Ras signaling, menin was found to interact with the 32-kDa subunit (RPA2) of replication protein A (RPA), a heterotrimeric protein required for DNA replication, recombination, and repair. The menin-RPA2 interaction was confirmed in a conventional yeast two-hybrid system and by direct interaction between purified proteins. Menin-RPA2 binding was inhibited by a number of menin missense mutations found in individuals with multiple endocrine neoplasia type 1, and the interacting regions were mapped to the N-terminal portion of menin and amino acids 43 to 171 of RPA2. This region of RPA2 contains a weak single-stranded DNA-binding domain, but menin had no detectable effect on RPA-DNA binding in vitro. Menin bound preferentially in vitro to free RPA2 rather than the RPA heterotrimer or a subcomplex consisting of RPA2 bound to the 14-kDa subunit (RPA3). However, the 70-kDa subunit (RPA1) was coprecipitated from HeLa cell extracts along with RPA2 by menin-specific antibodies, suggesting that menin binds to the RPA heterotrimer or a novel RPA1-RPA2-containing complex in vivo. This finding was consistent with the extensive overlap in the nuclear localization patterns of endogenous menin, RPA2, and RPA1 observed by immunofluorescence.


Subject(s)
DNA-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins , Animals , Bacterial Proteins/metabolism , Blotting, Western , Cell Line , Cell Nucleus/metabolism , Chromatography, Gel , DNA/metabolism , DNA Damage , DNA, Complementary/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , G1 Phase , Glutathione Transferase/metabolism , HeLa Cells , Humans , Mice , Microscopy, Fluorescence , Multiple Endocrine Neoplasia/genetics , Mutation, Missense , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Replication Protein A , S Phase , Transfection , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...