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.
Prostate ; 76(1): 97-113, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26417683

ABSTRACT

BACKGROUND: Previous results from our lab indicate a tumor suppressor role for the transmembrane protein with epidermal growth factor and two follistatin motifs 2 (TMEFF2) in prostate cancer (PCa). Here, we further characterize this role and uncover new functions for TMEFF2 in cancer and adult prostate regeneration. METHODS: The role of TMEFF2 was examined in PCa cells using Matrigel(TM) cultures and allograft models of PCa cells. In addition, we developed a transgenic mouse model that expresses TMEFF2 from a prostate specific promoter. Anatomical, histological, and metabolic characterizations of the transgenic mouse prostate were conducted. The effect of TMEFF2 in prostate regeneration was studied by analyzing branching morphogenesis in the TMEFF2-expressing mouse lobes and alterations in branching morphogenesis were correlated with the metabolomic profiles of the mouse lobes. The role of TMEFF2 in prostate tumorigenesis in whole animals was investigated by crossing the TMEFF2 transgenic mice with the TRAMP mouse model of PCa and analyzing the histopathological changes in the progeny. RESULTS: Ectopic expression of TMEFF2 impairs growth of PCa cells in Matrigel or allograft models. Surprisingly, while TMEFF2 expression in the TRAMP mouse did not have a significant effect on the glandular prostate epithelial lesions, the double TRAMP/TMEFF2 transgenic mice displayed an increased incidence of neuroendocrine type tumors. In addition, TMEFF2 promoted increased branching specifically in the dorsal lobe of the prostate suggesting a potential role in developmental processes. These results correlated with data indicating an alteration in the metabolic profile of the dorsal lobe of the transgenic TMEFF2 mice. CONCLUSIONS: Collectively, our results confirm the tumor suppressor role of TMEFF2 and suggest that ectopic expression of TMEFF2 in mouse prostate leads to additional lobe-specific effects in prostate regeneration and tumorigenesis. This points to a complex and multifunctional role for TMEFF2 during PCa progression.


Subject(s)
Adenocarcinoma , Carcinogenesis/metabolism , Membrane Proteins/metabolism , Neuroendocrine Tumors , Prostate , Prostatic Neoplasms , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Immunohistochemistry , Male , Mice , Mice, Transgenic , Neoplasm Transplantation/pathology , Neoplasm Transplantation/physiology , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Prostate/pathology , Prostate/physiology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Regeneration , Tumor Cells, Cultured
2.
PLoS One ; 8(2): e55257, 2013.
Article in English | MEDLINE | ID: mdl-23405127

ABSTRACT

The type I transmembrane protein with epidermal growth factor and two follistatin motifs 2 (TMEFF2), is expressed mainly in brain and prostate. Expression of TMEFF2 is deregulated in prostate cancer, suggesting a role in this disease, but the molecular mechanism(s) involved in this effect are not clear. Although androgens promote tmeff2 transcription, androgen delivery to castrated animals carrying CWR22 xenografts increases TMEFF2 protein levels in the absence of mRNA changes, suggesting that TMEFF2 may also be post-transcriptionally regulated. Here we show that translation of TMEFF2 is regulated by androgens. Addition of physiological concentrations of dihydrotestosterone (DHT) to prostate cancer cell lines increases translation of endogenous TMEFF2 or transfected TMEFF2-Luciferase fusions, and this effect requires the presence of upstream open reading frames (uORFs) in the 5'-untranslated region (5'-UTR) of TMEFF2. Using chemical and siRNA inhibition of the androgen receptor (AR), we show that the androgen effect on TMEFF2 translation is mediated by the AR. Importantly, DHT also promotes phosphorylation of the α subunit of the translation initiation factor 2 (eIF2α) in an AR-dependent manner, paralleling the effect on TMEFF2 translation. Moreover, endoplasmic reticulum (ER) stress conditions, which promote eIF2α phosphorylation, also stimulate TMEFF2 translation. These results indicate that androgen signaling promotes eIF2α phosphorylation and subsequent translation of TMEFF2 via a mechanism that requires uORFs in the 5'-UTR of TMEFF2.


Subject(s)
5' Untranslated Regions , Androgens/metabolism , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Prokaryotic Initiation Factor-2/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Androgens/genetics , Cell Line, Tumor , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Humans , Male , Membrane Proteins/biosynthesis , Membrane Proteins/metabolism , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/metabolism , Open Reading Frames , Phosphorylation , Prokaryotic Initiation Factor-2/metabolism , Protein Biosynthesis , Protein Subunits , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Signal Transduction
3.
J Biol Chem ; 286(18): 16091-100, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21393249

ABSTRACT

The type I transmembrane protein with epidermal growth factor and two follistatin motifs 2 (TMEFF2) is expressed in brain and prostate and overexpressed in prostate cancer, but its role in this disease is unclear. Several studies have suggested that TMEFF2 plays a role in suppressing the growth and invasive potential of human cancer cells, whereas others suggest that the shed portion of TMEFF2, which lacks the cytoplasmic region, has a growth-promoting activity. Here we show that TMEFF2 has a dual mode of action. Ectopic expression of wild-type full-length TMEFF2 inhibits soft agar colony formation, cellular invasion, and migration and increases cellular sensitivity to apoptosis. However, expression of the ectodomain portion of TMEFF2 increases cell proliferation. Using affinity chromatography and mass spectrometry, we identify sarcosine dehydrogenase (SARDH), the enzyme that converts sarcosine to glycine, as a TMEFF2-interacting protein. Co-immunoprecipitation and immunofluorescence analysis confirms the interaction of SARDH with full-length TMEFF2. The ectodomain does not bind to SARDH. Moreover, expression of the full-length TMEFF2 but not the ectodomain results in a decreased level of sarcosine in the cells. These results suggest that the tumor suppressor activity of TMEFF2 requires the cytoplasmic/transmembrane portion of the protein and correlates with its ability to bind to SARDH and to modulate the level of sarcosine.


Subject(s)
Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Sarcosine Dehydrogenase/metabolism , Sarcosine/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Line, Tumor , Glycine/genetics , Glycine/metabolism , HEK293 Cells , Humans , Male , Membrane Proteins/genetics , Mice , NIH 3T3 Cells , Neoplasm Proteins/genetics , Protein Binding , Protein Structure, Tertiary , Sarcosine/genetics , Sarcosine Dehydrogenase/genetics , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...