Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters











Database
Language
Publication year range
1.
Mol Metab ; 3(5): 518-30, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25061557

ABSTRACT

Glucose and GLP-1 stimulate not only insulin secretion, but also the post-transcriptional induction of insulin granule biogenesis. This process involves the nucleocytoplasmic translocation of the RNA binding protein PTBP1. Binding of PTBP1 to the 3'-UTRs of mRNAs for insulin and other cargoes of beta cell granules increases their stability. Here we show that glucose enhances also the binding of PTBP1 to the 5'-UTRs of these transcripts, which display IRES activity, and their translation exclusively in a cap-independent fashion. Accordingly, glucose-induced biosynthesis of granule cargoes was unaffected by pharmacological, genetic or Coxsackievirus-mediated inhibition of cap-dependent translation. Infection with Coxsackieviruses, which also depend on PTBP1 for their own cap-independent translation, reduced instead granule stores and insulin release. These findings provide insight into the mechanism for glucose-induction of insulin granule production and on how Coxsackieviruses, which have been implicated in the pathogenesis of type 1 diabetes, can foster beta cell failure.

2.
Mol Biol Cell ; 20(11): 2785-95, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19369417

ABSTRACT

The tumor suppressor serine-threonine kinase LKB1 is mutated in Peutz-Jeghers syndrome (PJS) and in epithelial cancers, including hormone-sensitive organs such as breast, ovaries, testes, and prostate. Clinical studies in breast cancer patients show low LKB1 expression is related to poor prognosis, whereas in PJS, the risk of breast cancer is similar to the risk from germline mutations in breast cancer (BRCA) 1/BRCA2. In this study, we investigate the role of LKB1 in estrogen receptor alpha (ERalpha) signaling. We demonstrate for the first time that LKB1 binds to ERalpha in the cell nucleus in which it is recruited to the promoter of ERalpha-responsive genes. Furthermore, LKB1 catalytic activity enhances ERalpha transactivation compared with LKB1 catalytically deficient mutants. The significance of our discovery is that we demonstrate for the first time a novel functional link between LKB1 and ERalpha. Our discovery places LKB1 in a coactivator role for ERalpha signaling, broadening the scientific scope of this tumor suppressor kinase and laying the groundwork for the use of LKB1 as a target for the development of new therapies against breast cancer.


Subject(s)
Estrogen Receptor alpha/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , AMP-Activated Protein Kinase Kinases , Blotting, Western , Catalysis , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Gene Expression Regulation, Neoplastic/drug effects , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Immunoprecipitation , Luciferases/genetics , Luciferases/metabolism , Microscopy, Fluorescence , Mutation , Promoter Regions, Genetic/genetics , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
3.
Cancer Res ; 67(12): 5622-7, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17575127

ABSTRACT

Mutations in the serine-threonine tumor-suppressor kinase LKB1 are responsible for Peutz-Jeghers syndrome, characterized by hamartomatous proliferation and an increased risk of developing cancer. Mutations in lkb1 have also been identified in sporadic cancers, suggesting a wider role for LKB1 in cancer that is not limited to hamartomatous polyposis syndromes. Here, we show that LKB1 catalytically deficient mutants, when introduced into DLD1p21-/-p53-/- colorectal cancer cells, allowed for progression of cells through to S phase of cell cycle and elicited the expression of Rb, cyclin E, and cyclin A2 whereas the introduction of LKB1 lead to G1 cell cycle arrest independent of p21(WAF/CIP1) and/or p53 expression. Furthermore, we show that LKB1 catalytically deficient mutants activate the expression of cyclin D1 through recruitment to response elements within the promoter of the oncogene. In addition to compromising the tumor-suppressor function of LKB1, our findings highlight an emerging role for LKB1 catalytically deficient mutants, a gain of oncogenic properties.


Subject(s)
Adenocarcinoma/metabolism , Cell Transformation, Neoplastic/metabolism , Colorectal Neoplasms/metabolism , Cyclin D1/biosynthesis , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Adenocarcinoma/genetics , Blotting, Western , Cell Cycle/physiology , Cell Proliferation , Colorectal Neoplasms/genetics , Cyclin-Dependent Kinase Inhibitor p21/deficiency , Flow Cytometry , Humans , Immunoprecipitation , Mutation , Protein Serine-Threonine Kinases/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Suppressor Protein p53/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL