scispace - formally typeset
Search or ask a question
Topic

Small hairpin RNA

About: Small hairpin RNA is a research topic. Over the lifetime, 9279 publications have been published within this topic receiving 285471 citations.


Papers
More filters
Journal ArticleDOI
TL;DR: The results revealed that SALL4 might serve as a functional marker for ESCC cancer stem cell, a crucial marker for prognosis and an attractive candidate for target therapy of ESCC.
Abstract: Growing evidence suggests that SALL4 plays a vital role in tumor progression and metastasis. However, the molecular mechanism of SALL4 promoting esophageal squamous cell carcinoma (ESCC) remains to be elucidated. The gene and protein expression profiles- were examined by using quantitative real-time PCR, immunohistochemistry and western blotting. Small hairpin RNA was used to evaluate the role of SALL4 both in cell lines and in animal models. Cell proliferation, apoptosis and invasion were assessed by CCK8, flow cytometry and transwell-matrigel assays. Sphere formation assay was used for cancer stem cell derivation and characterization. Our study showed that the transcription factor SALL4 was overexpressed in a majority of human ESCC tissues and closely correlated with a poor outcome. We established the lentiviral system using short hairpin RNA to knockdown SALL4 in TE7 and EC109 cells. Silencing of SALL4 inhibited the cell proliferation, induced apoptosis and the G1 phase arrest in cell cycle, decreased the ability of migration/invasion, clonogenicity and stemness in vitro. Besides, down-regulation of SALL4 enhanced the ESCC cells’ sensitivity to cisplatin. Xenograft tumor models showed that silencing of SALL4 decreased the ability to form tumors in vivo. Furthermore, our study demonstrated that SALL4 played a vital role in modulating the stemness of ESCC cells via Wnt/β-catenin signaling pathway and in epithelial-mesenchymal transition. Our results revealed that SALL4 might serve as a functional marker for ESCC cancer stem cell, a crucial marker for prognosis and an attractive candidate for target therapy of ESCC.

73 citations

Journal ArticleDOI
TL;DR: A systematic analysis of depletion of DNMT1 in colorectal (SW48), bladder (T24), and breast (T47D) cancer cells by DN MT1-specific small hairpin RNA (shRNA) targeting suggests that human cancer cells may differ in their reliance onDNMT1 for maintaining DNA methylation.
Abstract: Previous work has shown that DNA hypermethylation of tumor suppressor genes in colorectal cancer cells may be maintained in the absence of the major mammalian methyltransferase, DNA methyltransferase 1 (DNMT1). In an effort to dissect the dependency on DNMT1 to maintain such hypermethylation in different cancer types, we performed a systematic analysis of depletion of DNMT1 in colorectal (SW48), bladder (T24), and breast (T47D) cancer cells by DNMT1-specific small hairpin RNA (shRNA) targeting. We show that although DNMT1-deficient SW48 and T24 cells exhibited no observable growth defects and were able to maintain promoter hypermethylation, DNMT1-deficient T47D breast cells failed to form comparable numbers of colonies when stably selected for the incorporation of the DNMT1-specific shRNA expression vector, suggesting a growth defect with reduced levels of DNMT1. Further treatment of T47D cells with transient transfection of small interfering RNA targeting DNMT1 revealed that severely DNMT1-deficient T47D cells could not fully maintain promoter hypermethylation, and gene silencing was partially reversed at two of the three assayed loci. These observations suggest that human cancer cells may differ in their reliance on DNMT1 for maintaining DNA methylation. (Cancer Res 2006; 66(2): 729-35)

73 citations

Journal ArticleDOI
TL;DR: A novel targeted non-viral system for shRNA-mediated gene-silencing in cancer cells and results of gene silencing and apoptosis assay indicated that the targeted shRNA delivery system could efficiently down-regulate the Bcl-xL expression and induce 14% late apoptosis in target cancer cells with strong cell selectivity.

73 citations

Journal ArticleDOI
TL;DR: Findings indicate that knockdown of L-type calcium channel accessory &bgr;-subunit is capable of attenuating the hypertrophic response both in vitro and in vivo without compromising systolic performance.
Abstract: Calcium cycling figures prominently in excitation-contraction coupling and in various signaling cascades involved in the development of left ventricular hypertrophy. We hypothesized that genetic suppression of the L-type calcium channel accessory beta-subunit would modulate calcium current and suppress cardiac hypertrophy. A short hairpin RNA template sequence capable of mediating the knockdown of the L-type calcium channel accessory beta-subunit gene was incorporated into a lentiviral vector (PPT.CG.H1.beta(2)). Transduction of ventricular myocytes in vivo with the active short hairpin RNA partially inhibited the L-type calcium current. In neonatal rat cardiomyocytes, L-type calcium channel accessory beta-subunit gene knockdown reduced calcium transient amplitude. Similarly, [(3)H]leucine incorporation was attenuated in PPT.CG.H1.beta(2)-transduced neonatal rat cardiomyocytes compared with nonsilencing controls in a phenylephrine-induced hypertrophy model. In vivo gene transfer attenuated the hypertrophic response in an aortic-banded rat model of left ventricular hypertrophy, with reduced left ventricular wall thickness and heart weight/body weight ratios in PPT.CG.H1.beta(2)-injected rats at four weeks post transduction. Fractional shortening was preserved in rats treated with PPT.CG.H1.beta(2). These findings indicate that knockdown of L-type calcium channel accessory beta-subunit is capable of attenuating the hypertrophic response both in vitro and in vivo without compromising systolic performance. Suppression of the calcium channel beta subunit may represent a novel and useful therapeutic strategy for left ventricular hypertrophy.

73 citations

Journal ArticleDOI
TL;DR: Data suggest that AS3MT is the major enzyme in the oxidative methylation of inorganic arsenic (iAs) in mammalian species, although an As3MT-independent process may contribute to iAs methylation in human hepatic cells.
Abstract: Several methyltransferases have been shown to catalyze the oxidative methylation of inorganic arsenic (iAs) in mammalian species. However, the relative contributions of these enzymes to the overall capacity of cells to methylate iAs have not been characterized. Arsenic (+3 oxidation state) methyltransferase (AS3MT) that is expressed in rat and human hepatocytes catalyzes the conversion of iAs, yielding methylated metabolites that contain arsenic in +3 or +5 oxidation states. This study used short hairpin RNA (shRNA) to knock down AS3MT expression in human hepatocellular carcinoma (HepG2) cells. In a stable clonal HepG2/A cell line, AS3MT mRNA and protein levels were reduced by 83 and 88%, respectively. In comparison, the capacity to methylate iAs decreased only by 70%. These data suggest that AS3MT is the major enzyme in this pathway, although an AS3MT-independent process may contribute to iAs methylation in human hepatic cells.

73 citations


Network Information
Related Topics (5)
Cell culture
133.3K papers, 5.3M citations
91% related
Signal transduction
122.6K papers, 8.2M citations
90% related
Gene expression
113.3K papers, 5.5M citations
88% related
Regulation of gene expression
85.4K papers, 5.8M citations
86% related
Cellular differentiation
90.9K papers, 6M citations
86% related
Performance
Metrics
No. of papers in the topic in previous years
YearPapers
2023804
2022477
2021384
2020454
2019541
2018518