Downregulation of Msi1 suppresses the growth of human colon cancer by targeting p21cip1

  • Authors:
    • Chao Gao
    • Chun Han
    • Qiyao Yu
    • Yue Guan
    • Na Li
    • Jingjing Zhou
    • Yanming Tian
    • Yi Zhang
  • View Affiliations

  • Published online on: November 13, 2014     https://doi.org/10.3892/ijo.2014.2749
  • Pages: 732-740
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Musashi1 (Msi1), a member of the RNA-binding protein (RBP) family, is highly expressed in neural progenitor or stem cells for the maintenance of stemness as well as in various cancers. Emerging studies have demonstrated that it regulates cell processes by translational activation or suppresses specifically bound mRNA. In the present study, we initially reported remarkably increased expression of Msi1 in colon cancer tissues compared with adjacent non-tumor tissues. Knockdown of Msi1 significantly suppressed the proliferation, colony formation, tumorsphere formation and the progression of implanted colon cancers, and induced cell cycle attest at G0/G1 phase, along with the upregulated expression of p21cip1. Reporter assays using a chimeric mRNA that combined luciferase and the 3'-UTR of p21cip1 revealed that Msi1 decreased the reporter activity through the specific motif. Thus, the current results suggested that downregulation of Msi1 could inhibit the growth of colon cancers and Msi1 may be a promising therapeutic target molecule for human colon cancers.

Introduction

Colon cancer is one of the most common causes of cancer related mortality worldwide (1). Its overall incidence is ~5% and the 5-year survival rate ranges from 40 to 60% (2). The mechanisms of colon cancer carcinogenesis and development have drawn much attention in recent years for its high incidence and poor prognosis. However, the etiology and pathogenesis of colon cancer remain unclear, although heterogeneous genetic alterations have been reported to be responsible. The most common genetic alterations include mutations and loss of heterozygosity of tumor suppressors, such as adenomatous polyposis coli (APC) (3). Several studies have demonstrated that colon cancers were involved in the activation of Wnt, Notch, BMP and Hedgehog signaling pathways, which accompany establishment of tumorigenic state (4). Msi1 has been found to activate the Notch and Wnt signaling pathways in several types of normal and cancerous cells (5,6). Therefore, we hypothesized that Msi1 maybe have association with the progression of colon cancer.

Msi1, located at chromosome 12q24, is a member of the Musashi (MSI) family of the RNA binding proteins, which are characterized by RNA recognition motif. It can be regulated by ELAV or HuR by maintaining the stabilization of its mRNA or accumulation of mRNA translation (7,8), as well as by tumor suppressor microRNAs (9). It was first found in Drosophila as a determinant of sensory organ development (10) and highly conserved across species (11) with essential roles in the stem cell maintenance, nervous system development and tumorigenesis (12,13). In mammals, Msi1 is highly expressed in the neural progenitor and stem cells (13) for maintenance of self-renewal and differentiation. Recent studies have demonstrated that Msi1 exists in many adult cancers, including gliomas (14), gastric cancer (15), hepatoma (16) and colorectal adenoma (17). Consistently, high level of Msi1 expression is considered to be associated with many malignancies (18) and predicts poor prognosis (19,20). All these finding suggest that Msi1 function as an oncogene. This notion is further supported by the findings that ablation of Msi1 in gliomas (6) or bladder carcinoma cells (21) suppressed the cell cycle, and induced apoptosis and showed severe decline in cell numbers. However, Msi1 has also been proposed to have no significant effect on cell proliferation in human intestinal epithelial cells (22).

We performed detailed analyses on the role of Msi1 in colon carcinoma, and found that Msi1 was upregulated. Knockdown of endogenous Msi1 protein inhibited the growth and tumor formation by activating the cell cycle suppressor p21cip. Our findings support the hypothesis that Msi1 functions as an oncogene in colon cancer.

Materials and methods

Cell lines

Human colon cancer cell lines SW480, HCT116, SW620 and cervical cancer cell line HeLa were purchased from the American Type Culture Collection (ATCC), cultured in Dulbecco’s modified Eagle’s medium (DMEM; Sigma-Aldrich, St. Louis, MO, USA) supplemented with 10% fetal bovine serum (FBS; Invitrogen, Carlsbad, CA, USA) and 1% penicillin-streptomycin. All cell lines were maintained at 37°C in an atmosphere of 5% CO2.

Tissue samples

Fresh frozen specimens of matched colon cancer tissues and adjacent non-tumor tissues were from 20 patients in the Department of General Surgery of the Fourth Hospital of Hebei Medical University, obtained between March 2011 and January 2013, and stored at −80°C for further use. All of the tissues were from untreated patients who were undergoing surgery. Written informed consent was obtained from each patient before the surgery, and the study protocol was approved by the Institutional Research Ethics Committee.

Semi-quantitative real-time PCR analysis

Total RNA was isolated from colon cancer cell lines using TRIzol reagent (Invitrogen). Total cDNA was synthesized using the M-MLV Reverse Transcriptase (Fermentas, Vilnius, Lithuania). The mRNA expression levels were measured by qRT-PCR using the 7500 Real-Time PCR detection system (Applied Biosystems, Foster City, CA, USA). Amplification was performed with the SYBR Premix Ex Taq™ II kit (code: DRR081A; Takara, Dalian, China) according to a 2-step cycle procedure consisting of 45 cycles of denaturation at 95°C for 10 sec and annealing/elongation at 60°C for 30 sec. We measured mRNA levels semi-quantitatively by the Δ/Δ threshold cycle (Ct) method. GAPDH was used as internal control. Fold-changes were calculated and normalized as relative to the parental cells. The primers were used as follows: p21cip1 F, 5′-AAATCGTCCAGCGACCTTCC-3′ and p21cip1 R, 5′-GCCCTGTCCATAGCCTCTACT-3′; GAPDH F, 5′-CAAG GGCATCCTGGGCTACA-3′, GAPDH R, 5′-AAGTGGTCG TTGAGGGCAAT-3′.

Western blot analysis

Cells and clinical tissues were lysed on ice in lysis buffer (50 mM Tris-HCl, pH 7.4; 150 mM NaCl; 2 mM EDTA; 1% NP-40; and 0.1% sodium dodecyl sulfate SDS) containing freshly added protease inhibitor cocktail (Complete Mini; Roche Diagnostics, Branchburg, NJ, USA). Aliquots of samples with the same amount of protein were determined using the Bradford assay (Bio-Rad Laboratories, Hercules, CA, USA), and mixed with loading buffer (final concentrations of 62.5 mM Tris-HCl, pH 6.8, 2.3% SDS, 100 mM dithiothreitol and 0.005% bromophenol blue). Then the protein extracts were boiled and separated by SDS-PAGE and blotted to activated polyvinylidene difluoride (PVDF) membranes (Millipore, Billerica, MA, USA). Then the membranes were blocked with 5% fat-free milk and probed with first antibodies overnight. The first antibodies included the following: anti-Msi1 (1:500, sc-98845; Santa Cruz Biotechnology, Santa Cruz, CA, USA), anti-p21 (1:500, sc-397; Santa Cruz Biotechnology), and anti-β-actin (1:500, sc-47778; Santa Cruz Biotechnology). The membranes were then washed four times in PBST and incubated with a secondary antibody coupled to horseradish peroxidase (Thermo Fisher Scientific, Inc., New York, NY, USA), followed by ECL detection (Millipore) and visualization on X-ray film. Relative quantitation was determined with the AlphaView system (Cell Biosciences, Santa Clara, CA, USA) using β-actin as the loading control.

Plasmid construction

To generate plasmids that express Msi1-specific small interfering RNA (siRNA) which targeting MSI1, the oligonucleotide inserts were designed using an online siRNA design tool (Ambion, Austin, TX, USA). The double oligonucleotides were annealed and cloned into the lentivirus GV248-GFP shRNA vector (Shanghai Genechem Co., Ltd., Shanghai, China) as per the manufacturer’s introductions. Lentivirus was produced after GV248-GFP shRNA vector and two package plasmids were co-transfected to 293T cells with Lipofectamine 2000 transfection reagent (Invitrogen). Then colon cancer cells were infected by recombined lentivirus, and cell clones were selected to the amplification culture. Stably transfected clones were extracted and identified by western blotting.

A luciferase reporter vector (pMIR-REPORT; Ambion) was used to generate reporter constructs. Fragment of the 3′ UTR of human p21cip1 was amplified from HCT116 cDNA by PCR using primers p21cip1 WT F/R and cloned into the pMIR-REPORT luciferase plasmid. The mutant p21cip1 3′ UTR was generated by using the p21cip1 WT F/MU R and p21cip1 MU F/WT R primers. The following primers were used: shMsi1-1F, 5′-GATCCTGTTACATGGTGTTTCGAATTCAAGA GATTCGAAACACCATGTAACATCA-3′ and shMsi1-1R, 5′-GACAATGTACCACAAAGCTTAAGTTGAGAAAGCT TTGTGGTACATTGTAGTTCGA-3′; shMsi1-2F, 5′-GATCC TCCTGTATCATATGTAAATTTCAAGAGAATTTACATA TGATACTGGACGA-3′ and shMsi1-2R, 5′-AGGACATAGT ATACATTTAAAGTTCTCTTAAATGTATACTATGACCT GCTTTCGA-3′; p21cip1 WT F, 5′-ATTGAGCTCTAATCCGC CCACAGGAAG-3′ and p21cip1 WT R, 5′-CTCAAGCTTACA AGTAAAGTCACTAAG-3′; p21cip1 MU F, 5′-TGGGAAGCA GTGTCTTTCCTGGCACTAACGTT-3′ and p21cip1 MU R, 5′-AGACACTGCTTCCCAGCCCCATATGAGCCCAC-3′.

Cell proliferation and cell viability assays

Cells (5×104) were cultured in triplicate in 35-mm cell culture dishes. The cells were harvested longitudinally, and counted every day for one week using a hemocytometer under light microscopy.

Cell viability was assessed every other day using 3-(4,5-dimethylthiazol-yl)-2,5-diphenyl tetrazolium bromide (MTT; Sigma-Aldrich) dye according to the standard protocol. Approximately 1,000 cells/well were seeded in a 96-well plate. MTT solution (20 μl) was added to 200 μl of culture media and incubated for 4 h, and then cell growth was determined by measuring the absorbance at 490 nm.

Colony formation assay

Cells (1×103) of each type were cultured in 10-cm culture dishes and exposed to fresh media every 3 days. Colonies with diameters >0.2 mm were counted at day 14.

Tumorsphere formation assay

One hundred Msi1-KD or -control cells were seeded in 6-well plates in 2 ml of serum-free stem cell medium as described above. Fresh medium were added to each well every 3 days. The tumorspheres were analyzed on day 14 for tumorsphere forming ability.

Animal and tumor xenograft assay

Exponentially growing cells collected from stable transfectants were bilaterally inoculated into subcutaneous sites of 4- to 6-week-old Balb/c nude mice. Tumor dimensions were measured with calipers once every week, and volumes (cm3) were calculated according to the standard formula, length × width2/2. At the end of the experiment, tumors were dissected out, and the net weight per mouse was measured. The experimental protocols used herein were evaluated and approved by the Animal Care and Use Committee of the Fourth Hospital of Hebei Medical University.

Cell cycle analysis

Cells (1×106) were harvested and washed twice with PBS, followed by fixation with 75% cold ethanol at 4°C overnight. After the cells were washed twice in PBS, the cells were suspended in PBS with 50 μg/ml propidium iodide (PI; Sigma) and 10 μg/ml RNaseA (Sigma). They were then incubated at 4°C for 30 min in the dark. Cells were analyzed for DNA content by FACSCalibur (BD Biosciences) and the results of the cell cycle were analyzed by FlowJo software.

Luciferase assay

Cells were lysed in 100 μl of passive lysis buffer (Promega, Madison, WI, USA) at 48 h after transfection and assayed with a Dual-luciferase report assay kit according to the manufacturer’s instructions. For each assay, cell extract (20 μl) was used and the reaction was started by injection of 50 μl of luciferase substrate. Each reaction was measured for 10 sec in the Luminometer. Luciferase activities were expressed as the ratio of firefly to Renilla luciferase activity.

Statistical analysis

Data are presented as the mean ± standard deviation (SD). The Student’s t-test was performed using the SPSS 16.0 (SPSS, Inc., Chicago, IL, USA). P<0.05 was regarded as statistically significant.

Results

Msi1 expression in human colon cancer tissues and cell lines

To assess Msi1 expression in clinical patients, western blot analysis was conducted on 20 pairs of colon cancer tissues and matched adjacent non-tumor tissues (Fig. 1A). Msi1 expression in all 20 cancer tissues was markedly higher than that in the corresponding non-tumor tissues (Student’s t-test, P<0.01; Fig. 1B), suggesting that the activation of Msi1 may contribute to the development and progression of colon cancer.

We also evaluated the expression of Msi1 in colon cancer cell lines by western blotting. We detected a high level of Msi1 expression in SW620, HCT116 and SW480 cells (Fig. 1C).

The knockdown of Msi1 inhibits the proliferation of colon cancer cells in vitro

To determine whether Msi1 regulated the proliferation of colon cancer cell lines, the endogenous Msi1 was knocked down by short hairpin RNA (shRNA) in HCT116 and SW480 cells (Fig. 2A). The Msi1 knockdown HCT116 and SW480 cells (HCT116-KD and SW480-KD) had much lower proliferation ability than the corresponding control cells (HCT116-control and SW480-control), as measured by both cell growth curve assay (P<0.01; Fig. 2B) and cell viability assay (P<0.01; Fig. 2C), indicating that the knockdown of Msi1 suppressed the growth of colon cancer cells in vitro.

Furthermore, colony formation assays showed that the efficiency of foci formation was dramatically decreased in Msi1 knockdown clones compared with control clones (P<0.01; Fig. 2D). These findings suggested that enforced inhibition of Msi1 led to retardation of colon cancer cell growth in vitro.

Silencing of Msi1 inhibits the tumor formation of colon cancer cells in vivo and in vitro

As Msi1 modulates the proliferation in vitro, we explored whether it alters tumorigenic potential of colon cancer cells in vivo. Xenograft assays were created with nude mice, and the development and growth of solid tumors were monitored every week. The palpable tumor formation of HCT116-KD and HCT116-control cells occurred at the same time after inoculation. However, tumor development with HCT116-KD cells was significantly delayed (P<0.01; Fig. 3A). The tumor weights generated from HCT116-KD were reduced significantly (P<0.01; Fig. 3B). Similar results were observed from SW480 cells (Fig. 3A and B), suggesting that the silencing of Msi1 induces the suppression of the tumor formation and development of colon cancer, and this inhibition may have a close relationship with the decreased cell proliferation. Together, these data indicated that knockdown of Msi1 expression inhibited the growth of colon cancers.

Several studies have recently indicated the existence of CSCs (cancer stem cells) in various cancer types (2325), and CSCs are critical for the maintenance of tumor growth, progression and resistance to chemotherapy or radiotherapy, as well as recurrence and metastasis (26). It has been demonstrated that Msi1 regulates the proliferation and differentiation of CSCs. In the present study, we used a tumorsphere culture system to conduct the latent role of Msi1 on tumorsphere formation. Knockdown of Msi1 inhibited the tumorsphere formation and growth of HCT116-KD and SW480-KD cell lines, as demonstrated by the significantly reduced numbers of tumorspheres compared with the numbers in control cells (P<0.01; Fig. 3C). Our results indicate that Msi1 is a key regulator of the growth of tumorspheres.

Msi1 regulates the cell cycle of colon cancer cells in vitro

To further investigate the potential mechanism by which silence of Msi1 regulates growth of colon cancer cells, we characterized cell cycles by FACS analysis. As shown in Fig. 4A, the proportion of HCT116-KD cells in G0/G1 phase increased markedly to 69.3%, while proportion in S phase decreased to 19.0%. The ratio of G1 phase to S phase of HCT116-KD cells was much higher than that of HCT116-control cells. A similar effect was observed in SW480-KD cells (P<0.01; Fig. 4B), indicating that the knockdown of Msi1 expression blocked G1/S phase transition of cell cycle.

Msi1 negatively regulates p21cip1 expression

It has been reported that Msi1 blocked the G1/S phase transition of cell cycle by negative regulation of cyclin-dependent kinase (CDK) inhibitor p21cip1 in bladder carcinoma (21) and breast cancer (19). To test whether it also occurs in colon cancer cells, p21cip1 mRNA and protein levels were examined by qRT-PCR and western blot analysis. Significantly, increased levels of p21cip1 protein were observed in the lysates of both HCT116-KD and SW480-KD cells, but there was little change in p21cip1 mRNA levels, suggesting that Msi1 may reduce p21cip1 protein levels by inhibiting translation.

To confirm the direct interaction between Msi1 and its putative target site, the human p21cip1 3′ UTR containing the wild-type and mutant Msi1 binding sequence was cloned downstream from the luciferase report element (Fig. 5E). The relative luciferase activity of wild-type p21cip1 3′ UTR reporter was increased by 48% in HCT116-KD cells relative to controls, while the augmentation was diminished in cells transfected with the mutant reporter (P<0.01; Fig. 5F). Similarly, knockdown of Msi1 enhanced the relative luciferase activity of the wild-type in SW480 cells (P<0.01; Fig. 5F). Mutation of the Msi1 binding site abrogated the elevate effect, demonstrating the specificity of the Msi1 target sequence.

Discussion

Msi1, a multifunction RNA binding protein of MSI family, is present in many types of normal cells (2729) and is involved in CNS stem/progenitor cell fate (13,30), inner ear development (31), repair of small-intestinal and stomach injury (32,33), and maturation of photoreceptor and oocyte (34,35), intestinal metaplasia (36), atherosclerotic arteries (37) and Alzheimer disease and Pick disease (38). Its overexpression in tumor cells promotes tumor growth, invasion and metastasis, inhibits apoptosis and predicts poor prognosis. The pathophysiological functions involved have been found in several types of cancers, such as glioma (6), medulloblastoma (39) and breast cancer (19).

In the present study, we detected the expression of Msi1 in colon cancer tissues. Msi1 protein was abundant in colon cancer tissues. Low levels of Msi1 can be detected in matched adjacent non-tumor tissues. Nishimura and co-workers (40) found that Msi1 were located in the human normal colon crypt cells, indicating that it may be a possible stem cell marker of human colon epithelium. The low levels of Msi1 in normal colon tissues suggests that Msi1 may participate in the physiological function of the colon, and may be involved in the proliferation and initiation of the cells. In addition, colon cancer tissues have a significantly higher expression than normal tissues, indicating Msi1 plays important roles in colon cancers. Thus, we hypothesized that Msi1 was associated with colon cancer stem cells.

To further explore how Msi1 is involved in colon carcinogenesis, functional characterization was conducted by downregulation of Msi1 in colon cancer cell lines. We found that knockdown of Msi1 significantly inhibited cell proliferation and colony formation, induced cell cycle arrest in G0/G1 phase, demonstrating that Msi1 related signaling is a crucial regulator of the development and growth of colon cancers. Our data are in agreement with previous findings in other types of tumors (6,19,39), suggesting that Msi1 related signaling may have a similar regulatory effect on the growth of different types of human malignancies.

Several studies have recently suggested the existence of CSC in colon cancers (41,42). Cancer stem cells are a rare cell population, which have the ability for self-renewal, differentiation and tumorigenesis (43). Recent research has found that cancer stem cells are rich in tumorspheres and give rise to tumors (44). Indeed, it has been reported that Msi1 related signaling regulates the proliferation of normal and cancer stem cells (12,30). We characterized the role of Msi1 in the proliferation of colon CSC by using tumor xenograft and tumorsphere formation assays and observed that the knockdown of Msi1 expression significantly suppressed the tumor formation in vivo and remarkably decreased the number of tumorspheres formed in culture conditions in vitro that allowed the proliferation of only CSC. Therefore, it is possible that Msi1 related signaling may regulate the growth of human colon cancers by promoting the proliferation of both CSC and cancer cells.

Several lines of evidence suggest that Msi1 is a positive regulator of cell proliferation (45). Msi1 promotes cell proliferation through downregulation of the inhibitors of proliferation (6). In the present study, we also found that downregulation of Msi1 inhibited the cell cycle by blocking the G1/S transition in 2 colon cancer cell lines. Furthermore, we found that knockdown of Msi1 upregulated the expression of p21cip1. Luciferase assay revealed that Msi1 suppressed p21cip1 expression by directly binding to the consensus sequence of p21cip1 3′UTR in colon cancer cells. These results are consistent with the findings in bladder carcinoma (21) and breast cancer (19). p21cip1 is a universal cyclin dependent kinase (CDK) inhibitor that directly inhibits the activity of cyclin-CDK complexes, resulting in cell cycle arrest at G0/G1 phase. Therefore, our data suggested that cell growth retardation induced by downregulation of Msi1 in colon cancer is probably through activation of p21cip1.

In summary, we showed that Msi1 was upregulated in colon cancers, and knockdown of Msi1 inhibited the proliferation involved the coordinated direct activation of CDK inhibitor p21cip1. The present study indicates that downregulating the expression or restoring the expression of p21cip1 is a possible method to treat certain types of colon cancers.

Acknowledgements

The present study was partially supported by a grant from the National Natural Science Foundation of China (grant no. 31271223), and the Hebei Provincial Medical Foundation (grant no. ZL20140107). We gratefully thank Professor Depei Li for providing invaluable assistance.

References

1 

Weitz J, Koch M, Debus J, Hohler T, Galle PR and Buchler MW: Colorectal cancer. Lancet. 365:153–165. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Jemal A, Siegel R, Ward E, Murray T, Xu J and Thun MJ: Cancer statistics, 2007. CA Cancer J Clin. 57:43–66. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Fodde R, Smits R and Clevers H: APC, signal transduction and genetic instability in colorectal cancer. Nat Rev Cancer. 1:55–67. 2001. View Article : Google Scholar

4 

Bertrand FE, Angus CW, Partis WJ and Sigounas G: Developmental pathways in colon cancer: crosstalk between WNT, BMP, Hedgehog and Notch. Cell Cycle. 11:4344–4351. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Wang XY, Yin Y, Yuan H, Sakamaki T, Okano H and Glazer RI: Musashi1 modulates mammary progenitor cell expansion through proliferin-mediated activation of the Wnt and Notch pathways. Mol Cell Biol. 28:3589–3599. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Muto J, Imai T, Ogawa D, et al: RNA-binding protein Musashi1 modulates glioma cell growth through the post-transcriptional regulation of Notch and PI3 kinase/Akt signaling pathways. PLoS One. 7:e334312012. View Article : Google Scholar : PubMed/NCBI

7 

Ratti A, Fallini C, Cova L, et al: A role for the ELAV RNA-binding proteins in neural stem cells: stabilization of Msi1 mRNA. J Cell Sci. 119:1442–1452. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Vo DT, Abdelmohsen K, Martindale JL, et al: The oncogenic RNA-binding protein Musashi1 is regulated by HuR via mRNA translation and stability in glioblastoma cells. Mol Cancer Res. 10:143–155. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Vo DT, Qiao M, Smith AD, Burns SC, Brenner AJ and Penalva LO: The oncogenic RNA-binding protein Musashi1 is regulated by tumor suppressor miRNAs. RNA Biol. 8:817–828. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Nakamura M, Okano H, Blendy JA and Montell C: Musashi, a neural RNA-binding protein required for Drosophila adult external sensory organ development. Neuron. 13:67–81. 1994. View Article : Google Scholar : PubMed/NCBI

11 

Sakakibara S, Nakamura Y, Yoshida T, et al: RNA-binding protein Musashi family: roles for CNS stem cells and a subpopulation of ependymal cells revealed by targeted disruption and antisense ablation. Proc Natl Acad Sci USA. 99:15194–15199. 2002. View Article : Google Scholar : PubMed/NCBI

12 

Glazer RI, Vo DT and Penalva LO: Musashi1: an RBP with versatile functions in normal and cancer stem cells. Front Biosci. 17:54–64. 2012. View Article : Google Scholar

13 

Kaneko Y, Sakakibara S, Imai T, et al: Musashi1: an evolutionally conserved marker for CNS progenitor cells including neural stem cells. Dev Neurosci. 22:139–153. 2000. View Article : Google Scholar : PubMed/NCBI

14 

Toda M, Iizuka Y, Yu W, et al: Expression of the neural RNA-binding protein Musashi1 in human gliomas. Glia. 34:1–7. 2001. View Article : Google Scholar : PubMed/NCBI

15 

Nikpour P, Emadi-Baygi M, Mohhamad-Hashem F, Maracy MR and Haghjooy-Javanmard S: MSI1 overexpression in diffuse type of gastric cancer. Pathol Res Pract. 209:10–13. 2013. View Article : Google Scholar

16 

Shu HJ, Saito T, Watanabe H, et al: Expression of the Musashi1 gene encoding the RNA-binding protein in human hepatoma cell lines. Biochem Biophys Res Commun. 293:150–154. 2002. View Article : Google Scholar : PubMed/NCBI

17 

Schulenburg A, Cech P, Herbacek I, et al: CD44-positive colorectal adenoma cells express the potential stem cell markers musashi antigen (msi1) and ephrin B2 receptor (EphB2). J Pathol. 213:152–160. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Rezza A, Skah S, Roche C, Nadjar J, Samarut J and Plateroti M: The overexpression of the putative gut stem cell marker Musashi-1 induces tumorigenesis through Wnt and Notch activation. J Cell Sci. 123:3256–3265. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Wang XY, Penalva LO, Yuan H, et al: Musashi1 regulates breast tumor cell proliferation and is a prognostic indicator of poor survival. Mol Cancer. 9:2212010. View Article : Google Scholar : PubMed/NCBI

20 

Liu DC, Yang ZL and Jiang S: Identification of musashi-1 and ALDH1 as carcinogenesis, progression, and poor-prognosis related biomarkers for gallbladder adenocarcinoma. Cancer Biomark. 8:113–121. 2010.PubMed/NCBI

21 

Nikpour P, Baygi ME, Steinhoff C, et al: The RNA binding protein Musashi1 regulates apoptosis, gene expression and stress granule formation in urothelial carcinoma cells. J Cell Mol Med. 15:1210–1224. 2011. View Article : Google Scholar

22 

Murayama M, Okamoto R, Tsuchiya K, et al: Musashi-1 suppresses expression of Paneth cell-specific genes in human intestinal epithelial cells. J Gastroenterol. 44:173–182. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Margaritescu C, Pirici D, Simionescu C and Stepan A: The utility of CD44, CD117 and CD133 in identification of cancer stem cells (CSC) in oral squamous cell carcinomas (OSCC). Rom J Morphol Embryol. 52:985–993. 2011.PubMed/NCBI

24 

Janikova M, Skarda J, Dziechciarkova M, et al: Identification of CD133+/nestin+ putative cancer stem cells in non-small cell lung cancer. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 154:321–326. 2010. View Article : Google Scholar

25 

Eramo A, Lotti F, Sette G, et al: Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ. 15:504–514. 2008. View Article : Google Scholar

26 

Chinn SB, Darr OA, Peters RD and Prince ME: The role of head and neck squamous cell carcinoma cancer stem cells in tumorigenesis, metastasis, and treatment failure. Front Endocrinol (Lausanne). 3:902012.

27 

Kayahara T, Sawada M, Takaishi S, et al: Candidate markers for stem and early progenitor cells, Musashi-1 and Hes1, are expressed in crypt base columnar cells of mouse small intestine. FEBS Lett. 535:131–135. 2003. View Article : Google Scholar : PubMed/NCBI

28 

Lu X, Lin F, Fang H, Yang X and Qin L: Expression of a putative stem cell marker Musashi-1 in endometrium. Histol Histopathol. 26:1127–1133. 2011.PubMed/NCBI

29 

Raji B, Dansault A, Leemput J, et al: The RNA-binding protein Musashi-1 is produced in the developing and adult mouse eye. Mol Vis. 13:1412–1427. 2007.PubMed/NCBI

30 

Okano H, Kawahara H, Toriya M, Nakao K, Shibata S and Imai T: Function of RNA-binding protein Musashi-1 in stem cells. Exp Cell Res. 306:349–356. 2005. View Article : Google Scholar : PubMed/NCBI

31 

Sakaguchi H, Yaoi T, Suzuki T, Okano H, Hisa Y and Fushiki S: Spatiotemporal patterns of Musashi1 expression during inner ear development. Neuroreport. 15:997–1001. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Yu T, Lan SY, Wu B, et al: Musashi1 and hairy and enhancer of split 1 high expression cells derived from embryonic stem cells enhance the repair of small-intestinal injury in the mouse. Dig Dis Sci. 56:1354–1368. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Nagata H, Akiba Y, Suzuki H, Okano H and Hibi T: Expression of Musashi-1 in the rat stomach and changes during mucosal injury and restitution. FEBS Lett. 580:27–33. 2006. View Article : Google Scholar

34 

Susaki K, Kaneko J, Yamano Y, et al: Musashi-1, an RNA-binding protein, is indispensable for survival of photoreceptors. Exp Eye Res. 88:347–355. 2009. View Article : Google Scholar

35 

Arumugam K, Macnicol MC and Macnicol AM: Autoregulation of Musashi1 mRNA translation during Xenopus oocyte maturation. Mol Reprod Dev. 79:553–563. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Akasaka Y, Saikawa Y, Fujita K, et al: Expression of a candidate marker for progenitor cells, Musashi-1, in the proliferative regions of human antrum and its decreased expression in intestinal metaplasia. Histopathology. 47:348–356. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Bobryshev YV, Tran D, Botelho NK, Lord RV and Orekhov AN: Musashi-1 expression in atherosclerotic arteries and its relevance to the origin of arterial smooth muscle cells: histopathological findings and speculations. Atherosclerosis. 215:355–365. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Lovell MA and Markesbery WR: Ectopic expression of Musashi-1 in Alzheimer disease and Pick disease. J Neuropathol Exp Neurol. 64:675–680. 2005. View Article : Google Scholar : PubMed/NCBI

39 

Vo DT, Subramaniam D, Remke M, et al: The RNA-binding protein Musashi1 affects medulloblastoma growth via a network of cancer-related genes and is an indicator of poor prognosis. Am J Pathol. 181:1762–1772. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Nishimura S, Wakabayashi N, Toyoda K, Kashima K and Mitsufuji S: Expression of Musashi-1 in human normal colon crypt cells: a possible stem cell marker of human colon epithelium. Dig Dis Sci. 48:1523–1529. 2003. View Article : Google Scholar : PubMed/NCBI

41 

Ke J, Wu X, He X, et al: A subpopulation of CD24+ cells in colon cancer cell lines possess stem cell characteristics. Neoplasma. 59:282–288. 2012. View Article : Google Scholar

42 

Schneider M, Huber J, Hadaschik B, Siegers GM, Fiebig HH and Schuler J: Characterization of colon cancer cells: a functional approach characterizing CD133 as a potential stem cell marker. BMC Cancer. 12:962012. View Article : Google Scholar : PubMed/NCBI

43 

Reya T, Morrison SJ, Clarke MF and Weissman IL: Stem cells, cancer, and cancer stem cells. Nature. 414:105–111. 2001. View Article : Google Scholar : PubMed/NCBI

44 

Lin L, Fuchs J, Li C, Olson V, Bekaii-Saab T and Lin J: STAT3 signaling pathway is necessary for cell survival and tumorsphere forming capacity in ALDH+/CD133+ stem cell-like human colon cancer cells. Biochem Biophys Res Commun. 416:246–251. 2011. View Article : Google Scholar : PubMed/NCBI

45 

Kanemura Y, Mori K, Sakakibara S, et al: Musashi1, an evolutionarily conserved neural RNA-binding protein, is a versatile marker of human glioma cells in determining their cellular origin, malignancy, and proliferative activity. Differentiation. 68:141–152. 2001. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2015
Volume 46 Issue 2

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Gao C, Han C, Yu Q, Guan Y, Li N, Zhou J, Tian Y and Zhang Y: Downregulation of Msi1 suppresses the growth of human colon cancer by targeting p21cip1. Int J Oncol 46: 732-740, 2015
APA
Gao, C., Han, C., Yu, Q., Guan, Y., Li, N., Zhou, J. ... Zhang, Y. (2015). Downregulation of Msi1 suppresses the growth of human colon cancer by targeting p21cip1. International Journal of Oncology, 46, 732-740. https://doi.org/10.3892/ijo.2014.2749
MLA
Gao, C., Han, C., Yu, Q., Guan, Y., Li, N., Zhou, J., Tian, Y., Zhang, Y."Downregulation of Msi1 suppresses the growth of human colon cancer by targeting p21cip1". International Journal of Oncology 46.2 (2015): 732-740.
Chicago
Gao, C., Han, C., Yu, Q., Guan, Y., Li, N., Zhou, J., Tian, Y., Zhang, Y."Downregulation of Msi1 suppresses the growth of human colon cancer by targeting p21cip1". International Journal of Oncology 46, no. 2 (2015): 732-740. https://doi.org/10.3892/ijo.2014.2749