2013
DOI: 10.1158/1541-7786.mcr-13-0269
|View full text |Cite
|
Sign up to set email alerts
|

The Small Cell Lung Cancer-Specific Isoform of RE1-Silencing Transcription Factor (REST) Is Regulated By Neural-Specific Ser/Arg Repeat-Related Protein of 100 kDa (nSR100)

Abstract: Small cell lung cancer (SCLC) is a highly malignant form of cancer, which originates from primitive neuroendocrine cells in the lung. SCLC cells express several autocrine neurotransmitters/neuropeptides and their respective receptors. Expression of these neuronal markers is frequently regulated by RE1-silencing transcription factor (REST). In SCLC cells, an SCLC-specific isoform of REST (sREST) is highly expressed, whereas REST expression is undetectable, suggesting that the expression of sREST correlates with… Show more

Help me understand this report
View preprint versions

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
1
1
1

Citation Types

2
35
0

Year Published

2014
2014
2023
2023

Publication Types

Select...
8
1

Relationship

0
9

Authors

Journals

citations
Cited by 29 publications
(37 citation statements)
references
References 33 publications
2
35
0
Order By: Relevance
“…REST has been attributed a tumor suppressor function in breast cancer, and its expression is reduced with aggressiveness in breast cancer . Interestingly, a SCLC‐specific isoform of REST, which antagonizes REST‐mediated neuroendocrine gene expression, promoted tumor aggressiveness and neuronal phenotype in SCLCs . In colon cancer, the loss of REST expression confers a survival benefit during the progression of tumor cells .…”
Section: Discussionmentioning
confidence: 99%
“…REST has been attributed a tumor suppressor function in breast cancer, and its expression is reduced with aggressiveness in breast cancer . Interestingly, a SCLC‐specific isoform of REST, which antagonizes REST‐mediated neuroendocrine gene expression, promoted tumor aggressiveness and neuronal phenotype in SCLCs . In colon cancer, the loss of REST expression confers a survival benefit during the progression of tumor cells .…”
Section: Discussionmentioning
confidence: 99%
“…Furthermore, our studies have reported that this reprogrammed RNA splicing signature is unique to NEPC patient tumors, PDX models, and cell models, indicating a clear variance in the phenotype of NEPC and AdPC tumors . The SRRM4‐driven t‐NEPC‐specific reprogramming of the transcriptome modifies anti‐apoptotic factors (eg, Bif‐1), epigenetic modifiers (eg, MEAF6‐1), and transcriptional regulators (eg, REST, FOXA1) that are important for regulating cell survival, proliferation and tumorigenesis, and neural differentiation, respectively . Together, these studies propose a model in which SRRM4 drives t‐NEPC development through alternative splicing of downstream genes …”
Section: Introductionmentioning
confidence: 84%
“…This pathway is implicated in tumorextracellular matrix interactions that promote tumor proliferation. Preclinical studies with small-interfering RNAs have demonstrated encouraging results in suppressing sREST and restoring the REST level (81).…”
Section: Other Markersmentioning
confidence: 99%