101
|
Götte M, Greve B, Kelsch R, Müller-Uthoff H, Weiss K, Kharabi Masouleh B, Sibrowski W, Kiesel L, Buchweitz O. The adult stem cell marker Musashi-1 modulates endometrial carcinoma cell cycle progression and apoptosis viaNotch-1 and p21 WAF1/CIP1. Int J Cancer 2011; 129:2042-2049. [DOI: 10.1002/ijc.25856] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
102
|
Changes in Musashi-1 subcellular localization correlate with cell cycle exit during postnatal retinal development. Exp Eye Res 2011; 92:344-52. [DOI: 10.1016/j.exer.2011.02.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 01/19/2011] [Accepted: 02/04/2011] [Indexed: 12/30/2022]
|
103
|
Kawase S, Imai T, Miyauchi-Hara C, Yaguchi K, Nishimoto Y, Fukami SI, Matsuzaki Y, Miyawaki A, Itohara S, Okano H. Identification of a novel intronic enhancer responsible for the transcriptional regulation of musashi1 in neural stem/progenitor cells. Mol Brain 2011; 4:14. [PMID: 21486496 PMCID: PMC3108301 DOI: 10.1186/1756-6606-4-14] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Accepted: 04/13/2011] [Indexed: 01/18/2023] Open
Abstract
Background The specific genetic regulation of neural primordial cell determination is of great interest in stem cell biology. The Musashi1 (Msi1) protein, which belongs to an evolutionarily conserved family of RNA-binding proteins, is a marker for neural stem/progenitor cells (NS/PCs) in the embryonic and post-natal central nervous system (CNS). Msi1 regulates the translation of its downstream targets, including m-Numb and p21 mRNAs. In vitro experiments using knockout mice have shown that Msi1 and its isoform Musashi2 (Msi2) keep NS/PCs in an undifferentiated and proliferative state. Msi1 is expressed not only in NS/PCs, but also in other somatic stem cells and in tumours. Based on previous findings, Msi1 is likely to be a key regulator for maintaining the characteristics of self-renewing stem cells. However, the mechanisms regulating Msi1 expression are not yet clear. Results To identify the DNA region affecting Msi1 transcription, we inserted the fusion gene ffLuc, comprised of the fluorescent Venus protein and firefly Luciferase, at the translation initiation site of the mouse Msi1 gene locus contained in a 184-kb bacterial artificial chromosome (BAC). Fluorescence and Luciferase activity, reflecting the Msi1 transcriptional activity, were observed in a stable BAC-carrying embryonic stem cell line when it was induced toward neural lineage differentiation by retinoic acid treatment. When neuronal differentiation was induced in embryoid body (EB)-derived neurosphere cells, reporter signals were detected in Msi1-positive NSCs and GFAP-positive astrocytes, but not in MAP2-positive neurons. By introducing deletions into the BAC reporter gene and conducting further reporter experiments using a minimized enhancer region, we identified a region, "D5E2," that is responsible for Msi1 transcription in NS/PCs. Conclusions A regulatory element for Msi1 transcription in NS/PCs is located in the sixth intron of the Msi1 gene. The 595-bp D5E2 intronic enhancer can transactivate Msi1 gene expression with cell-type specificity markedly similar to the endogenous Msi1 expression patterns.
Collapse
Affiliation(s)
- Satoshi Kawase
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
104
|
Sohn D, Budach W, Jänicke RU. Caspase-2 is required for DNA damage-induced expression of the CDK inhibitor p21(WAF1/CIP1). Cell Death Differ 2011; 18:1664-74. [PMID: 21475302 DOI: 10.1038/cdd.2011.34] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Although caspase-2 represents the most conserved caspase across species and was the second caspase identified, its precise function remains enigmatic. In several cell types we show that knockdown of caspase-2 specifically impaired DNA damage-induced p21 expression, whereas overexpression of a caspase-2 mutant increased p21 levels. Caspase-2 did not influence p21 mRNA transcription; moreover, various inhibitors targeting proteasomal or non-proteasomal proteases, including caspases, could not restore p21 protein levels following knockdown of caspase-2. As, however, silencing of caspase-2 impaired exogenous expression of p21 constructs containing 3'-UTR sequences, our results strongly indicate that caspase-2 regulates p21 expression at the translational level. Intriguingly, unlike depletion of caspase-2, which prevented p21 expression and thereby reverted the γ-IR-induced senescent phenotype of wild-type HCT116 colon carcinoma cells into apoptosis, knockdown of none of the caspase-2-interacting components RAIDD, RIP or DNA-PKcs was able to mimic these processes. Together, our data suggest that this novel role of caspase-2 as a translational regulator of p21 expression occurs not only independently of its enzymatic activity but also does not require known caspase-2-activating platforms.
Collapse
Affiliation(s)
- D Sohn
- Laboratory of Molecular Radiooncology, Clinic and Policlinic for Radiation Therapy and Radiooncology, University of Düsseldorf, Universitätsstrasse 1, Düsseldorf 40225, Germany
| | | | | |
Collapse
|
105
|
Li D, Peng X, Yan D, Tang H, Huang F, Yang Y, Peng Z. Msi-1 is a predictor of survival and a novel therapeutic target in colon cancer. Ann Surg Oncol 2011; 18:2074-83. [PMID: 21442350 DOI: 10.1245/s10434-011-1567-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2010] [Indexed: 01/29/2023]
Abstract
BACKGROUND Musashi1 (Msi-1), a neural RNA-binding protein, plays an important role in regulating cell differentiation in precursor cells. Recently, aberrant expression of Msi-1 has been detected in several malignancies. However, its role in the progression of colon cancer is largely unknown. MATERIALS AND METHODS We used Western blotting to examine Msi-1 protein expression in 8 cases of primary colon cancer lesions and paired normal colonic mucosa. Msi-1 expression and clinicopathological significance were determined by immunohistochemical staining in a tissue microarray (TMA) containing 203 cases of primary colon cancer paired with noncancerous tissue and 66 lymph node metastasis (LNM) tissues. RNAi was used to analyze the biological function of Msi-1 in vitro. RESULTS LNM tissue exhibited a striking increase in Msi-1 expression when compared with primary colon cancer and adjacent normal mucosa (87.9% vs. 64.5% vs. 16.7%, P < .001). Overexpression of Msi-1 was associated with higher clinical stage, T stage, lymph node metastasis, presence of distant metastasis, and Ki-67 positivity. Msi-1 served as an independent prognostic marker whose expression levels correlated with poorer metastasis-free survival (MFS) (HR 5.4; P < .001) and poorer overall survival (OS) (HR 3.8; P < .001). Msi-1 silencing significantly inhibited proliferation ability and attenuated the migration and invasion activity of colon cancer cells. CONCLUSIONS Our study provides the basis to explore the use of Msi-1 as a novel prognostic biomarker in colon cancer patients. Aberrant overexpression of Msi-1 during metastasis of colon cancer also suggests that it is a potential therapeutic target.
Collapse
Affiliation(s)
- Dawei Li
- Department of General Surgery, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
106
|
Spears E, Neufeld KL. Novel double-negative feedback loop between adenomatous polyposis coli and Musashi1 in colon epithelia. J Biol Chem 2011; 286:4946-50. [PMID: 21199875 DOI: 10.1074/jbc.c110.205922] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Loss of tumor suppressor adenomatous polyposis coli (APC) is thought to initiate the majority of all colorectal cancers. The predominant theory of colorectal carcinogenesis implicates stem cells as the initiating cells. However, relatively little is known about the function of APC in governing the homeostasis of normal intestinal stem cells. Here, we identify a novel double-negative feedback loop between APC and a translation inhibitor protein, Musashi1 (MSI1), in cultured human colonocytes. We show APC as a key factor in MSI1 regulation through Wnt signaling and identify APC mRNA as a novel target of translational inhibition by MSI1. We propose that APC/MSI1 interactions maintain homeostatic balance in the intestinal epithelium.
Collapse
Affiliation(s)
- Erick Spears
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66045, USA
| | | |
Collapse
|
107
|
MacNicol MC, Cragle CE, MacNicol AM. Context-dependent regulation of Musashi-mediated mRNA translation and cell cycle regulation. Cell Cycle 2011; 10:39-44. [PMID: 21191181 DOI: 10.4161/cc.10.1.14388] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Musashi-mediated mRNA translational control has been implicated in the promotion of physiological and pathological stem cell proliferation. During self-renewal of mammalian stem cells, Musashi has been proposed to act to repress the translation of mRNAs encoding inhibitors of cell cycle progression. By contrast, in maturing Xenopus oocytes Musashi activates translation of target mRNAs that encode proteins promoting cell cycle progression. The mechanisms directing Musashi to differentially control mRNA translation in mammalian stem cells and Xenopus oocytes is unknown. In this study, we demonstrate that the mechanisms defining Musashi function lie within the cellular context. Specifically, we show that murine Musashi acts as an activator of translation in maturing Xenopus oocytes while Xenopus Musashi functions as a repressor of target mRNA translation in mammalian cells. We further demonstrate that within the context of a primary mammalian neural stem/progenitor cell, Musashi can be converted from a repressor of mRNA translation to an activator of translation in response to extracellular stimuli. We present current models of Musashi-mediated mRNA translational control and discuss possible mechanisms for regulating Musashi function. An understanding of these mechanisms presents exciting possibilities for development of therapeutic targets to control physiological and pathological stem cell proliferation.
Collapse
Affiliation(s)
- Melanie C MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | | |
Collapse
|
108
|
Sharma P, Cline HT. Visual activity regulates neural progenitor cells in developing xenopus CNS through musashi1. Neuron 2010; 68:442-55. [PMID: 21040846 PMCID: PMC3005332 DOI: 10.1016/j.neuron.2010.09.028] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2010] [Indexed: 11/30/2022]
Abstract
Regulation of progenitor cell fate determines the numbers of neurons in the developing brain. While proliferation of neural progenitors predominates during early central nervous system (CNS) development, progenitor cell fate shifts toward differentiation as CNS circuits develop, suggesting that signals from developing circuits may regulate proliferation and differentiation. We tested whether activity regulates neurogenesis in vivo in the developing visual system of Xenopus tadpoles. Both cell proliferation and the number of musashi1-immunoreactive progenitors in the optic tectum decrease as visual system connections become stronger. Visual deprivation for 2 days increased proliferation of musashi1-immunoreactive radial glial progenitors, while visual experience increased neuronal differentiation. Morpholino-mediated knockdown and overexpression of musashi1 indicate that musashi1 is necessary and sufficient for neural progenitor proliferation in the CNS. These data demonstrate a mechanism by which increased brain activity in developing circuits decreases cell proliferation and increases neuronal differentiation through the downregulation of musashi1 in response to circuit activity.
Collapse
Affiliation(s)
- Pranav Sharma
- The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Hollis T. Cline
- The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
109
|
Rezza A, Skah S, Roche C, Nadjar J, Samarut J, Plateroti M. The overexpression of the putative gut stem cell marker Musashi-1 induces tumorigenesis through Wnt and Notch activation. J Cell Sci 2010; 123:3256-65. [PMID: 20826465 DOI: 10.1242/jcs.065284] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The RNA-binding protein Musashi-1 (Msi1) has been proposed as a marker of intestinal epithelial stem cells. These cells are responsible for the continuous renewal of the intestinal epithelium. Although the function of Msi1 has been studied in several organs from different species and in mammalian cell lines, its function and molecular regulation in mouse intestinal epithelium progenitor cells are still undefined. We describe here that, in these cells, the expression of Msi1 is regulated by the canonical Wnt pathway, through a mechanism involving a functional Tcf/Lef binding site on its promoter. An in vitro study in intestinal epithelium primary cultures showed that Msi1 overexpression promotes progenitor proliferation and activates Wnt and Notch pathways. Moreover, Msi1-overexpressing cells exhibit tumorigenic properties in xenograft experiments. These data point to a positive feedback loop between Msi1 and Wnt in intestinal epithelial progenitors. They also suggest that Msi1 has oncogenic properties in these cells, probably through induction of both the Wnt and Notch pathways.
Collapse
Affiliation(s)
- Amelie Rezza
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, INRA, Ecole Normale Supérieure de Lyon, 46 Allée d'Italie, 69364 Lyon Cedex 07, France
| | | | | | | | | | | |
Collapse
|
110
|
Scoumanne A, Cho SJ, Zhang J, Chen X. The cyclin-dependent kinase inhibitor p21 is regulated by RNA-binding protein PCBP4 via mRNA stability. Nucleic Acids Res 2010; 39:213-24. [PMID: 20817677 PMCID: PMC3017617 DOI: 10.1093/nar/gkq778] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
RNA-binding proteins (RBPs) play a major role in many post-transcriptional processes, including mRNA stability, alternative splicing and translation. PCBP4, also called MCG10, is an RBP belonging to the poly(C)-binding protein family and a target of p53 tumor suppressor. Ectopic expression of PCBP4 induces cell-cycle arrest in G2 and apoptosis. To identify RNA targets regulated by PCBP4 and further decipher its function, we generated multiple cell lines in which PCBP4 is either inducibly over-expressed or knocked down. We found that PCBP4 expression decreases cyclin-dependent kinase inhibitor p21 induction in response to DNA damage. We also provided evidence that PCBP4 regulates p21 expression independently of p53. In addition, we showed that a deficiency in PCBP4 enhances p21 induction upon DNA damage. To validate PCBP4 regulation of p21, we made PCBP4-deficient mice and showed that p21 expression is markedly increased in PCBP4-deficient primary mouse embryo fibroblasts compared to that in wild-type counterparts. Finally, we uncovered that PCBP4 binds to the 3′-UTR of p21 transcript in vitro and in vivo to regulate p21 mRNA stability. Taken together, we revealed that PCBP4 regulates both basal and stress-induced p21 expression through binding p21 3′-UTR and modulating p21 mRNA stability.
Collapse
Affiliation(s)
- Ariane Scoumanne
- The Center for Comparative Oncology, University of California at Davis, Davis, CA 95616, USA
| | | | | | | |
Collapse
|
111
|
Kuwako KI, Kakumoto K, Imai T, Igarashi M, Hamakubo T, Sakakibara SI, Tessier-Lavigne M, Okano HJ, Okano H. Neural RNA-binding protein Musashi1 controls midline crossing of precerebellar neurons through posttranscriptional regulation of Robo3/Rig-1 expression. Neuron 2010; 67:407-21. [PMID: 20696379 DOI: 10.1016/j.neuron.2010.07.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2010] [Indexed: 12/11/2022]
Abstract
Precisely regulated spatiotemporal gene expression is essential for the establishment of neural circuits. In contrast to the increasing evidence for transcriptional regulation of axon guidance cues and receptors, the role of posttranscriptional regulation in axon guidance, especially in vivo, remains poorly characterized. Here, we demonstrate that the expression of Slit receptor Robo3/Rig-1, which plays crucial roles in axonal midline crossing, is regulated by a neural RNA-binding protein Musashi1 (Msi1). Msi1 binds to Robo3 mRNA through RNA recognition motifs and increases the protein level of Robo3 without affecting its mRNA level. In Msi1-deficient precerebellar neurons, Robo3 protein, but not its mRNA, is dramatically reduced. Moreover, similar to defects in Robo3-deficient mice, axonal midline crossing and neuronal migration of precerebellar neurons are severely impaired in Msi1-deficient mice. Together, these findings indicate that Msi1-mediated posttranscriptional regulation of Robo3 controls midline crossing of precerebellar neurons.
Collapse
Affiliation(s)
- Ken-ichiro Kuwako
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
112
|
Wang XY, Penalva LO, Yuan H, Linnoila RI, Lu J, Okano H, Glazer RI. Musashi1 regulates breast tumor cell proliferation and is a prognostic indicator of poor survival. Mol Cancer 2010; 9:221. [PMID: 20727204 PMCID: PMC2939568 DOI: 10.1186/1476-4598-9-221] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2010] [Accepted: 08/21/2010] [Indexed: 12/23/2022] Open
Abstract
Background Musashi1 (Msi1) is a conserved RNA-binding protein that regulates the Notch and Wnt pathways, and serves as a stem cell marker in the breast and other tissues. It is unknown how Msi1 relates to other breast cancer markers, whether it denotes tumor initiating cells (TICs), and how it affects gene expression and tumor cell survival in breast cancer cells. Results Msi1 expression was analyzed in 20 breast cancer cell lines and in 140 primary breast tumors by western blotting and immunohistochemistry, respectively. Lentivirus RNA interference was used to reduce Msi1 expression in breast cancer cell lines MCF-7 and T47D grown as spheroid cultures and to assess stem cell gene expression and the growth of these cell lines as xenografts. In normal human breast tissue, Msi1 was expressed in 10.6% of myoepithelum and 1.2% of ductal epithelium in the terminal ductal lobular unit (TDLU), whereas, less than 0.05% of ductal epithelium and myoepithelium in large ducts outside the TDLU expressed Msi1. Msi1 was expressed in 55% of the breast cancer cell lines and correlated with ErbB2 expression in 50% of the cell lines. Msi1 was expressed in 68% of primary tumors and in 100% of lymph node metastases, and correlated with 5 year survival. Msi1 was enriched in CD133+ MCF-7 and T47D cells and in spheroid cultures of these cells, and Msi1 'knockdown' (KD) with a lentivirus-expressed shRNA decreased the number and size of spheroid colonies. Msi1 KD reduced Notch1, c-Myc, ErbB2 and pERK1/2 expression, and increased p21CIP1 expression, which is consistent with known Msi1 target mRNAs. Msi1 KD also reduced the expression of the somatic and embryonic stem cell markers, CD133, Bmi1, Sox2, Nanog and Oct4. Xenografts of MCF-7 and T47D Msi1 KD cells resulted in a marked reduction of tumor growth, reduced Msi1 and Notch1 expression and increased p21CIP1 expression. Conclusion Msi1 is a negative prognostic indicator of breast cancer patient survival, and is indicative of tumor cells with stem cell-like characteristics. Msi1 KD reduces tumor cell survival and tumor xenograft growth, suggesting that it may represent a novel target for drug discovery.
Collapse
Affiliation(s)
- Xiao-Yang Wang
- Department of Oncology, Georgetown University, and Lombardi Comprehensive Cancer Center, Washington, DC 20007, USA.
| | | | | | | | | | | | | |
Collapse
|
113
|
Ito T, Kwon HY, Zimdahl B, Congdon KL, Blum J, Lento WE, Zhao C, Lagoo A, Gerrard G, Foroni L, Goldman J, Goh H, Kim SH, Kim DW, Chuah C, Oehler VG, Radich JP, Jordan CT, Reya T. Regulation of myeloid leukaemia by the cell-fate determinant Musashi. Nature 2010; 466:765-8. [PMID: 20639863 PMCID: PMC2918284 DOI: 10.1038/nature09171] [Citation(s) in RCA: 286] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2008] [Accepted: 05/13/2010] [Indexed: 12/25/2022]
Abstract
Chronic myelogenous leukaemia (CML) can progress from a slow growing chronic phase to an aggressive blast crisis phase, but the molecular basis of this transition remains poorly understood. Here we have used mouse models of CML to show that disease progression is regulated by the Musashi-Numb signalling axis. Specifically, we find that the chronic phase is marked by high levels of Numb expression whereas the blast crisis phase has low levels of Numb expression, and that ectopic expression of Numb promotes differentiation and impairs advanced-phase disease in vivo. As a possible explanation for the decreased levels of Numb in the blast crisis phase, we show that NUP98-HOXA9, an oncogene associated with blast crisis CML, can trigger expression of the RNA-binding protein Musashi2 (Msi2), which in turn represses Numb. Notably, loss of Msi2 restores Numb expression and significantly impairs the development and propagation of blast crisis CML in vitro and in vivo. Finally we show that Msi2 expression is not only highly upregulated during human CML progression but is also an early indicator of poorer prognosis. These data show that the Musashi-Numb pathway can control the differentiation of CML cells, and raise the possibility that targeting this pathway may provide a new strategy for the therapy of aggressive leukaemias.
Collapse
MESH Headings
- Animals
- Blast Crisis/genetics
- Blast Crisis/metabolism
- Blast Crisis/pathology
- Cell Differentiation/genetics
- Disease Progression
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Gene Expression Regulation, Neoplastic
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Membrane Proteins/biosynthesis
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred C57BL
- Nerve Tissue Proteins/biosynthesis
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Nuclear Pore Complex Proteins/genetics
- Nuclear Pore Complex Proteins/metabolism
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Prognosis
- RNA-Binding Proteins/biosynthesis
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Receptor, Notch1/metabolism
- Signal Transduction
- Tumor Suppressor Protein p53/metabolism
- Up-Regulation
Collapse
Affiliation(s)
- Takahiro Ito
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27710
| | - Hyog Young Kwon
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27710
| | - Bryan Zimdahl
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27710
| | - Kendra L. Congdon
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27710
| | - Jordan Blum
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27710
| | - William E. Lento
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27710
| | - Chen Zhao
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27710
| | - Anand Lagoo
- Department of Pathology Duke University Medical Center, Durham, NC, 27710
| | - Gareth Gerrard
- Department of Haematology, Imperial College London, Hammersmith Hospital, London, W12 0NN
| | - Letizia Foroni
- Department of Haematology, Imperial College London, Hammersmith Hospital, London, W12 0NN
| | - John Goldman
- Department of Haematology, Imperial College London, Hammersmith Hospital, London, W12 0NN
| | - Harriet Goh
- Division of Hematology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Soo-Hyun Kim
- Division of Hematology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Dong-Wook Kim
- Division of Hematology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Charles Chuah
- Department of Haematology, Singapore General Hospital, Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, Singapore
| | - Vivian G. Oehler
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109
| | - Jerald P. Radich
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109
| | - Craig T. Jordan
- James P. Wilmot Cancer Center, University of Rochester School of Medicine, Rochester, NY 14642
| | - Tannishtha Reya
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27710
| |
Collapse
|
114
|
MACNICOL MELANIEC, MACNICOL ANGUSM. Developmental timing of mRNA translation--integration of distinct regulatory elements. Mol Reprod Dev 2010; 77:662-9. [PMID: 20652998 PMCID: PMC2910371 DOI: 10.1002/mrd.21191] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Targeted mRNA translation is emerging as a critical mechanism to control gene expression during developmental processes. Exciting new findings have revealed a critical role for regulatory elements within the mRNA untranslated regions to direct the timing of mRNA translation. Regulatory elements can be targeted by sequence-specific binding proteins to direct either repression or activation of mRNA translation in response to developmental signals. As new regulatory elements continue to be identified it has become clear that targeted mRNAs can contain multiple regulatory elements, directing apparently contradictory translational patterns. How is this complex regulatory input integrated? In this review, we focus on a new challenge area-how sequence-specific RNA binding proteins respond to developmental signals and functionally integrate to regulate the extent and timing of target mRNA translation. We discuss current understanding with a particular emphasis on the control of cell cycle progression that is mediated through a complex interplay of distinct mRNA regulatory elements during Xenopus oocyte maturation.
Collapse
Affiliation(s)
- MELANIE C. MACNICOL
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - ANGUS M. MACNICOL
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
115
|
Jung YS, Qian Y, Chen X. Examination of the expanding pathways for the regulation of p21 expression and activity. Cell Signal 2010; 22:1003-12. [PMID: 20100570 PMCID: PMC2860671 DOI: 10.1016/j.cellsig.2010.01.013] [Citation(s) in RCA: 333] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Accepted: 01/16/2010] [Indexed: 02/06/2023]
Abstract
p21(Waf1/Cip1/Sdi1) was originally identified as an inhibitor of cyclin-dependent kinases, a mediator of p53 in growth suppression and a marker of cellular senescence. p21 is required for proper cell cycle progression and plays a role in cell death, DNA repair, senescence and aging, and induced pluripotent stem cell reprogramming. Although transcriptional regulation is considered to be the initial control point for p21 expression, there is growing evidence that post-transcriptional and post-translational regulations play a critical role in p21 expression and activity. This review will briefly discuss the activity of p21 and focus on current knowledge of the determinants that control p21 transcription, mRNA stability and translation, and protein stability and activity.
Collapse
Affiliation(s)
- Yong-Sam Jung
- Center for Comparative Oncology, University of California, Davis, California 95616, USA
| | - Yingjuan Qian
- Center for Comparative Oncology, University of California, Davis, California 95616, USA
| | - Xinbin Chen
- Center for Comparative Oncology, University of California, Davis, California 95616, USA
| |
Collapse
|
116
|
Abstract
Carcinomas may arise as a disorder of regeneration, so that a malignant cell may represent a failure to fully attain the characteristics of differentiated tissue. We hypothesized that there is a differential distribution of progenitor cell markers among different histological types of lung cancers, with poorly differentiated tumors being more likely to express progenitor stem cell markers. The study was limited to paraffin-embedded archival material of resected untreated pulmonary carcinomas, including adenocarcinoma, squamous cell carcinoma, large cell carcinoma, and small cell carcinoma. The sections were stained for putative stem cells markers (Musashi-1, Musashi-2, CD34, CD21, KIT, CD133, p63, and OCT-4). Positivity was read as isolated, focal, or diffuse staining. Stem cell markers were detected in all histological types of pulmonary carcinomas. There was a difference in the expression of markers among the histological types. Small cell carcinoma showed diffuse positivity for most of the markers; in contrast to focal or negative staining in other histological groups. An inverse relationship between CD21 and Musashi-1 was observed. No staining for OCT-4 and CD34 was seen in any of the tumor types. Hierarchical clustering based on marker expression separated tumors into two groups, with one group marked by high expression of Musashi-1 and KIT, contained most of the poorly differentiated adenocarcinomas and small cell carcinomas. Therefore, stem cell markers are expressed in lung cancers with different patterns seen for different histological types and degrees of differentiation.
Collapse
|
117
|
Todaro M, Francipane MG, Medema JP, Stassi G. Colon cancer stem cells: promise of targeted therapy. Gastroenterology 2010; 138:2151-62. [PMID: 20420952 DOI: 10.1053/j.gastro.2009.12.063] [Citation(s) in RCA: 354] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Revised: 11/28/2009] [Accepted: 12/15/2009] [Indexed: 12/12/2022]
Abstract
First developed for hematologic disorders, the concept of cancer stem cells (CSCs) was expanded to solid tumors, including colorectal cancer (CRC). The traditional model of colon carcinogenesis includes several steps that occur via mutational activation of oncogenes and inactivation of tumor suppressor genes. Intestinal epithelial cells exist for a shorter amount of time than that required to accumulate tumor-inducing genetic changes, so researchers have investigated the concept that CRC arises from the long-lived stem cells, rather than from the differentiated epithelial cells. Colon CSCs were originally identified through the expression of the CD133 glycoprotein using an antibody directed to its epitope AC133. It is not clear if CD133 is a marker of colon CSCs-other cell surface markers, such as epithelial-specific antigen, CD44, CD166, Musashi-1, CD29, CD24, leucine-rich repeat-containing G-protein-coupled receptor 5, and aldehyde dehydrogenase 1, have been proposed. In addition to initiating and sustaining tumor growth, CSCs are believed to mediate cancer relapse after chemotherapy. How can we identify and analyze colon CSCs and what agents are being designed to kill this chemotherapy-refractory population?
Collapse
Affiliation(s)
- Matilde Todaro
- Department of Surgical and Oncological Sciences, Cellular and Molecular Pathophysiology Laboratory, Palermo, Italy
| | | | | | | |
Collapse
|
118
|
Nikpour P, Baygi ME, Steinhoff C, Hader C, Luca AC, Mowla SJ, Schulz WA. The RNA binding protein Musashi1 regulates apoptosis, gene expression and stress granule formation in urothelial carcinoma cells. J Cell Mol Med 2010; 15:1210-24. [PMID: 20477901 PMCID: PMC3822633 DOI: 10.1111/j.1582-4934.2010.01090.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The RNA-binding protein Musashi1 (MSI1) is a marker of progenitor cells in the nervous system functioning as a translational repressor. We detected MSI1 mRNA in several bladder carcinoma cell lines, but not in cultured normal uroepithelial cells, whereas the paralogous MSI2 gene was broadly expressed. Knockdown of MSI1 expression by siRNA induced apoptosis and a severe decline in cell numbers in 5637 bladder carcinoma cells. Microarray analysis of gene expression changes after MSI1 knockdown significantly up-regulated 735 genes, but down-regulated only 31. Up-regulated mRNAs contained a highly significantly greater number and density of Musashi binding sites. Therefore, a much larger set of mRNAs may be regulated by Musashi1, which may affect not only their translation, but also their turnover. The study confirmed p21CIP1 and Numb proteins as targets of Musashi1, suggesting additionally p27KIP1 in cell-cycle regulation and Jagged-1 in Notch signalling. A significant number of up-regulated genes encoded components of stress granules (SGs), an organelle involved in translational regulation and mRNA turnover, and impacting on apoptosis. Accordingly, heat shock induced SG formation was augmented by Musashi1 down-regulation. Our data show that ectopic MSI1 expression may contribute to tumorigenesis in selected bladder cancers through multiple mechanisms and reveal a previously unrecognized function of Musashi1 in the regulation of SG formation.
Collapse
Affiliation(s)
- Parvaneh Nikpour
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | | | | | | | | | | |
Collapse
|
119
|
Morgan SC, Yasin S, Uwanogho D, Jeffries A, Price J. Positional Specification in a Neural Stem Cell Line Involves Modulation of Musashi1 Expression. Stem Cells Dev 2010; 19:579-92. [DOI: 10.1089/scd.2009.0108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Sarah C. Morgan
- Centre for the Cellular Basis of Behaviour, King’s College London, Institute of Psychiatry, Denmark Hill, London, United Kingdom
| | - Shireena Yasin
- Centre for the Cellular Basis of Behaviour, King’s College London, Institute of Psychiatry, Denmark Hill, London, United Kingdom
| | - Dafe Uwanogho
- Centre for the Cellular Basis of Behaviour, King’s College London, Institute of Psychiatry, Denmark Hill, London, United Kingdom
| | - Aaron Jeffries
- Centre for the Cellular Basis of Behaviour, King’s College London, Institute of Psychiatry, Denmark Hill, London, United Kingdom
| | - Jack Price
- Centre for the Cellular Basis of Behaviour, King’s College London, Institute of Psychiatry, Denmark Hill, London, United Kingdom
| |
Collapse
|
120
|
Anant S, Houchen CW, Pawar V, Ramalingam S. Role of RNA-Binding Proteins in Colorectal Carcinogenesis. CURRENT COLORECTAL CANCER REPORTS 2010; 6:68-73. [PMID: 20401169 PMCID: PMC2853959 DOI: 10.1007/s11888-010-0048-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
RNA-binding proteins (RBPs) play key roles in the posttranscriptional regulation of gene expression. RBPs control various posttranscriptional events, including splicing, polyadenylation, mRNA stability, transport, and translation. It is becoming apparent that RBPs play a significant role in pathophysiologic conditions such as inflammation and cancer. More importantly, we and others have begun dissecting the role of mRNA stability and translation in regulating gene expression, dysregulation of which has serious consequences for the fate of the cell. In this article, we discuss this emerging area of posttranscriptional gene regulation and the role of RBPs in the aberrant expression of proteins in tumorigenesis.
Collapse
Affiliation(s)
- Shrikant Anant
- Section of Digestive Diseases and Nutrition, Department of Internal Medicine, University of Oklahoma Health Sciences Center, 920 Stanton L. Young Boulevard WP1345, Oklahoma City, OK 73104, USA; Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | | | | |
Collapse
|
121
|
Horisawa K, Imai T, Okano H, Yanagawa H. 3'-Untranslated region of doublecortin mRNA is a binding target of the Musashi1 RNA-binding protein. FEBS Lett 2009; 583:2429-34. [PMID: 19573529 DOI: 10.1016/j.febslet.2009.06.045] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Accepted: 06/25/2009] [Indexed: 11/17/2022]
Abstract
Musashi1 (Msi1) is an RNA-binding protein that is highly expressed in neural stem cells, and is considered to be a stemness factor. A known function of Msi1 is translational repression of specifically bound mRNAs. Although the basic mechanism and some target RNAs have been reported, further survey of interactors is necessary to understand the integrated function of Msi1. By screening using an mRNA display technique, we found that doublecortin (dcx) mRNA is a specific binding target of Msi1 in vitro. We confirmed that Msil repressed translation of a luciferase reporter gene linked to the selected 3'-untranslated region fragment of dcx in Neuro2A cells.
Collapse
Affiliation(s)
- Kenichi Horisawa
- Department of Biosciences and Informatics, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama 223-8522, Japan
| | | | | | | |
Collapse
|
122
|
Murayama M, Okamoto R, Tsuchiya K, Akiyama J, Nakamura T, Sakamoto N, Kanai T, Watanabe M. Musashi-1 suppresses expression of Paneth cell-specific genes in human intestinal epithelial cells. J Gastroenterol 2009; 44:173-82. [PMID: 19214660 DOI: 10.1007/s00535-008-2284-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2008] [Accepted: 08/16/2008] [Indexed: 02/04/2023]
Abstract
BACKGROUND Musashi-1 (Msi-1) is a RNA-binding protein, known as a putative marker of intestinal stem cells (ISCs). However, little is known about the function of Msi-1 within human intestinal epithelial cells (IECs). Thus, the present study aimed to clarify the role of Msi-1 in differentiation and proliferation of IECs. METHODS A human intestinal epithelial cell line stably expressing Msi-1 was established. Proliferation of the established cell lines was measured by bromodeoxyuridine incorporation, whereas differentiation were assessed by reverse transcriptase-polymerase chain reaction (RT-PCR) analysis of lineage-specific genes. Activities of the Notch and Wnt pathways were examined either by reporter assays or expression of downstream target genes. The distribution of Msi-1 and PLA2G2A expression in vivo was determined by immunohistochemistry. RESULTS Constitutive expression of Msi-1 in IECs had no significant effect on cell proliferation, but suppressed expression of Paneth cell-specific genes, including PLA2G2A. Msi-1 appeared to suppress expression of the PLA2G2A gene at the mRNA level. Analysis of Notch and Wnt pathway activity, however, revealed no significant change upon Msi-1 expression. The expression of Msi-1 and PLA2G2A in vivo was restricted to IECs residing at the lowest part of the human intestinal crypt, but was clearly separated to within basal columnar cells or mature Paneth cells, respectively. CONCLUSIONS Msi-1 suppresses expression of Paneth cell-specific genes in IECs, presumably through a pathway independent from Notch or Wnt. These findings suggest Msi-1 is a negative regulator of Paneth cell differentiation, an may contribute to maintain the undifferentiated phenotype of ISCs.
Collapse
Affiliation(s)
- Minekazu Murayama
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | | | | | | | | | | | | | | |
Collapse
|
123
|
Waggoner SA, Johannes GJ, Liebhaber SA. Depletion of the poly(C)-binding proteins alphaCP1 and alphaCP2 from K562 cells leads to p53-independent induction of cyclin-dependent kinase inhibitor (CDKN1A) and G1 arrest. J Biol Chem 2009; 284:9039-49. [PMID: 19211566 PMCID: PMC2666552 DOI: 10.1074/jbc.m806986200] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2008] [Revised: 02/03/2009] [Indexed: 12/23/2022] Open
Abstract
The alpha-globin poly(C)-binding proteins (alphaCPs) comprise an abundant and widely expressed set of K-homolog domain RNA-binding proteins. alphaCPs regulate the expression of a number of cellular and viral mRNAs at the levels of splicing, stability, and translation. Previous surveys have identified 160 mRNAs that are bound by alphaCP in the human hematopoietic cell line, K562. To explore the functions of these alphaCP/mRNA interactions, we identified mRNAs whose levels are altered in K562 cells acutely depleted of the two major alphaCP proteins, alphaCP1 and alphaCP2. Microarray analysis identified 27 mRNAs that are down-regulated and 14 mRNAs that are up-regulated in the alphaCP1/2-co-depleted cells. This alphaCP1/2 co-depletion was also noted to inhibit cell proliferation and trigger a G(1) cell cycle arrest. Targeted analysis of genes involved in cell cycle control revealed a marked increase in p21(WAF) mRNA and protein. Analysis of mRNP complexes in K562 cells demonstrates in vivo association of p21(WAF) mRNA with alphaCP1 and alphaCP2. In vitro binding assays indicate that a 127-nucleotide region of the 3'-untranslated region of p21(WAF) interacts with both alphaCP1 and alphaCP2, and co-depletion of alphaCP1/2 results in a marked increase in p21(WAF) mRNA half-life. p21(WAF) induction and G(1) arrest in the alphaCP1/2-co-depleted cells occur in the absence of p53 and are not observed in cells depleted of the individual alphaCP isoforms. The apparent redundancy in the actions of alphaCP1 and alphaCP2 upon p21(WAF) expression correlates with a parallel redundancy in their effects on cell cycle control. These data reveal a pivotal role for alphaCP1 and alphaCP2 in a p53-independent pathway of p21(WAF) control and cell cycle progression.
Collapse
Affiliation(s)
- Shelly A Waggoner
- Department of Genetics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | | | |
Collapse
|
124
|
de Sousa Abreu R, Sanchez-Diaz PC, Vogel C, Burns SC, Ko D, Burton TL, Vo DT, Chennasamudaram S, Le SY, Shapiro BA, Penalva LOF. Genomic analyses of musashi1 downstream targets show a strong association with cancer-related processes. J Biol Chem 2009; 284:12125-35. [PMID: 19258308 DOI: 10.1074/jbc.m809605200] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Musashi1 (Msi1) is a highly conserved RNA-binding protein with pivotal functions in stem cell maintenance, nervous system development, and tumorigenesis. Despite its importance, only three direct mRNA targets have been characterized so far: m-numb, CDKN1A, and c-mos. Msi1 has been shown to affect their translation by binding to short elements located in the 3'-untranslated region. To better understand Msi1 functions, we initially performed an RIP-Chip analysis in HEK293T cells; this method consists of isolation of specific RNA-protein complexes followed by identification of the RNA component via microarrays. A group of 64 mRNAs was found to be enriched in the Msi1-associated population compared with controls. These genes belong to two main functional categories pertinent to tumorigenesis: 1) cell cycle, cell proliferation, cell differentiation, and apoptosis and 2) protein modification (including ubiquitination and ubiquitin cycle). To corroborate our findings, we examined the impact of Msi1 expression on both mRNA (transcriptomic) and protein (proteomic) expression levels. Genes whose mRNA levels were affected by Msi1 expression have a Gene Ontology distribution similar to RIP-Chip results, reinforcing Msi1 participation in cancer-related processes. The proteomics study revealed that Msi1 can have either positive or negative effects on gene expression of its direct targets. In summary, our results indicate that Msi1 affects a network of genes and could function as a master regulator during development and tumor formation.
Collapse
Affiliation(s)
- Raquel de Sousa Abreu
- Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, Department of Cellular and Structural Biology, San Antonio, TX 78229-390, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
125
|
Abstract
p53 tumor suppressor plays a pivotal role in maintaining genomic integrity and preventing cancer development. The importance of p53 in tumor suppression is illustrated by the observation that about 50% human tumor cells have a dysfunctional p53 pathway. Although it has been well accepted that the activity of p53 is mainly controlled through post-translational modifications, recent studies have revealed that posttranscriptional regulations of p53 by various RNA-binding proteins also play a crucial role in modulating p53 activity and its downstream targets.
Collapse
Affiliation(s)
- Jin Zhang
- Center for Comparative Oncology, University of California, Davis, CA 95616, USA
| | | |
Collapse
|
126
|
Kaneko J, Chiba C. Immunohistochemical analysis of Musashi-1 expression during retinal regeneration of adult newt. Neurosci Lett 2008; 450:252-7. [PMID: 19028551 DOI: 10.1016/j.neulet.2008.11.031] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2008] [Revised: 11/05/2008] [Accepted: 11/12/2008] [Indexed: 11/29/2022]
Abstract
The adult newt retinal regeneration is an ideal model for studying retinal regeneration by transdifferentiation of the retinal pigment epithelium (RPE) cells. Accumulated evidence suggests that the RNA-binding protein Musashi-1 (Msi1) is expressed in mature photoreceptors and RPE cells as well as in retinal stem/progenitor cells, being essential for vision. We have been investigating whether Msi1 is also essential for retinal regeneration. In the last paper [K. Susaki, J. Kaneko, Y. Yamano, K. Nakamura, W. Inami, T. Yoshikawa, Y. Ozawa, S. Shibata, O. Matsuzaki, H. Okano, C. Chiba, Musashi-1, an RNA-binding protein, is indispensable for survival of photoreceptors. Exp. Eye Res. (in press)], we showed that the expression profiles of Msi1 transcripts and protein isoforms change during retinal regeneration. In the current report, we show by immunohistochemistry that Msi1 is expressed in transdifferentiating cells or cells of regenerating retinal tissues. Upon retinectomy, Msi1 protein, which is expressed in the nuclei of intact (stage E-0) RPE cells, changed its subcellular localization, being expressed in both the nucleus and cytoplasm of the RPE-derived stem-like cells at stage E-1. As the retinal rudiment/regenerating retina (rR) and renewing RPE (rRPE) are specified from the stem-like cell population (stage E-2), Msi1 expression was maintained or up-regulated in the rR, while down-regulated in the rRPE. During further retinal regeneration, Msi1 expression was decreased in association with cell differentiation, except for photoreceptors and RPE cells whose Msi1 expression increased as they differentiate. Thus, Msi1 is likely to be regulated at various cellular events during retinal regeneration, implying that Msi1 may have multi-functions in retinal regeneration. All together, it is probable that Msi1 is one of the essential factors that need to be regulated in retinal regeneration.
Collapse
Affiliation(s)
- Jun Kaneko
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki 305-8572, Japan
| | | |
Collapse
|
127
|
Sanchez-Diaz PC, Burton TL, Burns SC, Hung JY, Penalva LOF. Musashi1 modulates cell proliferation genes in the medulloblastoma cell line Daoy. BMC Cancer 2008; 8:280. [PMID: 18826648 PMCID: PMC2572071 DOI: 10.1186/1471-2407-8-280] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2008] [Accepted: 09/30/2008] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Musashi1 (Msi1) is an RNA binding protein with a central role during nervous system development and stem cell maintenance. High levels of Msi1 have been reported in several malignancies including brain tumors thereby associating Msi1 and cancer. METHODS We used the human medulloblastoma cell line Daoy as model system in this study to knock down the expression of Msi1 and determine the effects upon soft agar growth and neurophere formation. Quantitative RT-PCR was conducted to evaluate the expression of cell proliferation, differentiation and survival genes in Msi1 depleted Daoy cells. RESULTS We observed that MSI1 expression was elevated in Daoy cells cultured as neurospheres compared to those grown as monolayer. These data indicated that Msi1 might be involved in regulating proliferation in cancer cells. Here we show that shRNA mediated Msi1 depletion in Daoy cells notably impaired their ability to form colonies in soft agar and to grow as neurospheres in culture. Moreover, differential expression of a group of Notch, Hedgehog and Wnt pathway related genes including MYCN, FOS, NOTCH2, SMO, CDKN1A, CCND2, CCND1, and DKK1, was also found in the Msi1 knockdown, demonstrating that Msi1 modulated the expression of a subset of cell proliferation, differentiation and survival genes in Daoy. CONCLUSION Our data suggested that Msi1 may promote cancer cell proliferation and survival as its loss seems to have a detrimental effect in the maintenance of medulloblastoma cancer cells. In this regard, Msi1 might be a positive regulator of tumor progression and a potential target for therapy.
Collapse
Affiliation(s)
- Patricia C Sanchez-Diaz
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, TX, USA.
| | | | | | | | | |
Collapse
|
128
|
Abstract
One of the earliest genes identified with stem and early progenitor cells is the RNA-binding protein, Musashi1 (Msi1). Through gene profiling of mammary epithelial cells transduced with Msi1, a unique autocrine signaling pathway was identified that activates both the Wnt and Notch pathways. This process was associated with increased secretion of the growth factor, PLF1 and inhibition of the secreted Wnt pathway inhibitor, DKK3. Identification of PLF1 as an effector of these pathways in the absence of the DKK3 tumor suppressor provides a new avenue for investigating differences between normal and malignant tissues, and potentially targeting tumor stem cells.
Collapse
Affiliation(s)
- Robert I Glazer
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA.
| | | | | | | |
Collapse
|
129
|
Expression and functional analysis of musashi-like genes in planarian CNS regeneration. Mech Dev 2008; 125:631-45. [DOI: 10.1016/j.mod.2008.03.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2007] [Revised: 03/01/2008] [Accepted: 03/11/2008] [Indexed: 01/01/2023]
|
130
|
Post-transcriptional regulation of myelin formation. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2008; 1779:486-94. [PMID: 18590840 DOI: 10.1016/j.bbagrm.2008.06.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2008] [Revised: 05/15/2008] [Accepted: 06/03/2008] [Indexed: 12/21/2022]
Abstract
Myelin is a specialized structure of the nervous system that both enhances electrical conductance and protects neurons from degeneration. In the central nervous system, extensively polarized oligodendrocytes form myelin by wrapping cellular processes in a spiral pattern around neuronal axons. Myelin formation requires the oligodendrocyte to regulate gene expression in response to changes in its extracellular environment. Because these changes occur at a distance from the cell body, post-transcriptional control of gene expression allows the cell to fine-tune its response. Here, we review the RNA-binding proteins that control myelin formation in the brain, highlighting the molecular mechanisms by which they control gene expression and drawing parallels from studies in other cell types.
Collapse
|
131
|
Kawahara H, Imai T, Imataka H, Tsujimoto M, Matsumoto K, Okano H. Neural RNA-binding protein Musashi1 inhibits translation initiation by competing with eIF4G for PABP. ACTA ACUST UNITED AC 2008; 181:639-53. [PMID: 18490513 PMCID: PMC2386104 DOI: 10.1083/jcb.200708004] [Citation(s) in RCA: 159] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Musashi1 (Msi1) is an RNA-binding protein that is highly expressed in neural stem cells. We previously reported that Msi1 contributes to the maintenance of the immature state and self-renewal activity of neural stem cells through translational repression of m-Numb. However, its translation repression mechanism has remained unclear. Here, we identify poly(A) binding protein (PABP) as an Msi1-binding protein, and find Msi1 competes with eIF4G for PABP binding. This competition inhibits translation initiation of Msi1's target mRNA. Indeed, deletion of the PABP-interacting domain in Msi1 abolishes its function. We demonstrate that Msi1 inhibits the assembly of the 80S, but not the 48S, ribosome complex. Consistent with these conclusions, Msi1 colocalizes with PABP and is recruited into stress granules, which contain the stalled preinitiation complex. However, Msi1 with mutations in two RNA recognition motifs fails to accumulate into stress granules. These results provide insight into the mechanism by which sequence-specific translational repression occurs in stem cells through the control of translation initiation.
Collapse
Affiliation(s)
- Hironori Kawahara
- Department of Physiology, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan
| | | | | | | | | | | |
Collapse
|
132
|
MacNicol AM, Wilczynska A, MacNicol MC. Function and regulation of the mammalian Musashi mRNA translational regulator. Biochem Soc Trans 2008; 36:528-30. [PMID: 18481998 PMCID: PMC2562719 DOI: 10.1042/bst0360528] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The evolutionarily conserved RNA-binding protein, Musashi, regulates neural stem cell self-renewal. Musashi expression is also indicative of stem cell populations in breast and intestinal tissues and is linked to cell overproliferation in cancers of these tissues. Musashi has been primarily implicated as a repressor of target mRNAs in stem cell populations. However, little is known about the mechanism by which Musashi exerts mRNA translational control or how Musashi function is regulated. Recent findings in oocytes of the frog, Xenopus, indicate an unexpected role for Musashi as an activator of a number of maternal mRNAs during meiotic cell cycle progression. Given the importance of Musashi function in stem cell biology and the implications of aberrant Musashi expression in cancer, it is critical that we understand the molecular processes that regulate Musashi function.
Collapse
Affiliation(s)
- Angus M MacNicol
- Department of Neurobiology and Developmental Sciences, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W. Markham, Little Rock, AR 72205, USA.
| | | | | |
Collapse
|
133
|
Wang XY, Yin Y, Yuan H, Sakamaki T, Okano H, Glazer RI. Musashi1 modulates mammary progenitor cell expansion through proliferin-mediated activation of the Wnt and Notch pathways. Mol Cell Biol 2008; 28:3589-99. [PMID: 18362162 PMCID: PMC2423292 DOI: 10.1128/mcb.00040-08] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2008] [Revised: 02/06/2008] [Accepted: 03/13/2008] [Indexed: 01/17/2023] Open
Abstract
The RNA-binding protein Musashi1 (Msi1) is a positive regulator of Notch-mediated transcription in Drosophila melanogaster and neural progenitor cells and has been identified as a putative human breast stem cell marker. Here we describe a novel functional role for Msi1: its ability to drive progenitor cell expansion along the luminal and myoepithelial lineages. Expression of Msi1 in mammary epithelial cells increases the abundance of CD24(hi) Sca-1(+), CD24(hi) CD29(+), CK19, CK6, and double-positive CK14/CK18 progenitor cells. Proliferation is associated with increased proliferin-1 (PLF1) and reduced Dickkopf-3 (DKK3) secretion into the conditioned medium from Msi-expressing cells, which is associated with increased colony formation and extracellular signal-regulated kinase (ERK) phosphorylation. Treatment with the MEK inhibitor U0126 inhibits ERK activation and decreases Notch and beta-catenin/T-cell factor (TCF) reporter activity resulting from Msi1 expression. Reduction of DKK3 in control cells with a short hairpin RNA (shRNA) increases Notch and beta-catenin/TCF activation, whereas reduction of PLF1 with a shRNA in Msi1-expressing cells inhibits these pathways. These results identify Msi1 as a key determinant of the mammary lineage through its ability to coordinate cell cycle entry and activate the Notch and Wnt pathways by a novel autocrine process involving PLF1 and DKK3.
Collapse
Affiliation(s)
- Xiao-Yang Wang
- Department of Oncology, Georgetown University, and Lombardi Comprehensive Cancer Center, Washington, DC 20007, USA
| | | | | | | | | | | |
Collapse
|
134
|
Sureban SM, May R, George RJ, Dieckgraefe BK, McLeod HL, Ramalingam S, Bishnupuri KS, Natarajan G, Anant S, Houchen CW. Knockdown of RNA binding protein musashi-1 leads to tumor regression in vivo. Gastroenterology 2008; 134:1448-58. [PMID: 18471519 DOI: 10.1053/j.gastro.2008.02.057] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2007] [Accepted: 01/31/2008] [Indexed: 01/06/2023]
Abstract
BACKGROUND & AIMS In the gut, tumorigenesis is thought to arise from the stem cell population located near the base of intestinal and colonic crypts. The RNA binding protein musashi-1 (Msi-1) is a putative intestinal and progenitor/stem cell marker. Msi-1 expression is increased during rat brain development and in APC(min/+) mice tumors. This study examined a potential role of Msi-1 in tumorigenesis. METHODS Msi-1 small interfering RNA (siRNA) was administered as a liposomal preparation to HCT116 colon adenocarcinoma xenografts in athymic nude mice and tumor volume was measured. Cell proliferation was assessed by hexosaminidase and 3-(4,5-dimethylthiazol 2-yl)-2,5-diphenyltetrazolium bromide MTT assays. siRNA-transfected cells were subjected to 12 Gy gamma-irradiation. Apoptosis was assessed by immunoreactive activated caspase-3 and mitosis was assessed by phosphorylated histone H3 staining. The tumor xenografts were stained similarly for phosphorylated histone H3, activated caspase-3, terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling, Notch-1, and p21(WAF1). Furthermore, siRNA-transfected cells were subjected to cell-cycle analysis and Western blot analyses for Notch-1 and p21(WAF1). RESULTS Knockdown of Msi-1 resulted in tumor growth arrest in xenografts, reduced cancer cell proliferation, and increased apoptosis alone and in combination with radiation injury. siRNA-mediated reduction of Msi-1 lead to mitotic catastrophe in tumor cells. Moreover, there was inhibition of Notch-1 and up-regulation of p21(WAF1) after knockdown of Msi-1. CONCLUSIONS Our results show the involvement of Msi-1 in cancer cell proliferation, inhibition of apoptosis, and mitotic catastrophe, suggesting an important potential mechanism for its role in tumorigenesis.
Collapse
Affiliation(s)
- Sripathi M Sureban
- Department of Medicine, Division of Digestive Diseases and Nutrition, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
135
|
Dobson NR, Zhou Y, Flint NC, Armstrong RC. Musashi1 RNA-binding protein regulates oligodendrocyte lineage cell differentiation and survival. Glia 2008; 56:318-30. [PMID: 18098125 PMCID: PMC2663423 DOI: 10.1002/glia.20615] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2007] [Revised: 10/30/2007] [Accepted: 11/14/2007] [Indexed: 11/10/2022]
Abstract
Expression of Musashi1 (Msi1), an evolutionarily conserved RNA-binding protein, in neural stem cells of the subventricular zone in the postnatal and adult CNS indicates a potential role in the generation of oligodendrocytes. We now show Msi1 expression in a subset of oligodendrocyte progenitor (OP) cells in white matter areas temporally and spatially associated with oligodendrogenesis in the postnatal CNS. Msi1 function was evaluated by infection of OP cells with retroviral transduction of Msi1 or knockdown of endogenous Msi1. Retroviral expression of Msi1 significantly reduced the proportion of mature oligodendrocytes generated from OP cells in vitro and in vivo during myelination. Msi1 transduction also promoted OP survival, particularly under conditions of challenge from oxidative stress, while Msi1 siRNA knockdown resulted in dramatic OP cell death. Furthermore, in experimental demyelination Msi1 expression was increased among cells associated with lesions, including OP cells, indicating a potential role in the generation of remyelinating oligodendrocytes.
Collapse
Affiliation(s)
- Nicole R. Dobson
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Department of Pediatrics, Walter Reed Army Medical Center, Washington, DC
| | - Yong‐Xing Zhou
- Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Nicole C. Flint
- Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Regina C. Armstrong
- Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
136
|
Bani-Yaghoub M, Kubu CJ, Cowling R, Rochira J, Nikopoulos GN, Bellum S, Verdi JM. A switch in numb isoforms is a critical step in cortical development. Dev Dyn 2007; 236:696-705. [PMID: 17253625 DOI: 10.1002/dvdy.21072] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Loss of numb function suggests that numb maintains progenitors in an undifferentiated state. Herein, we demonstrate that numb1 and numb3 are expressed in undifferentiated cortical progenitors, whereas numb2 and numb4 become prominent throughout differentiation. To further assess the role of different numb isoforms in cortical neural development, we first created a Numb-null state with antisense morpholino, followed by the re-expression of specific numb isoforms. The re-expression of numb1 or numb3 resulted in a significant reduction of neural differentiation, correlating with an expansion of the cortical progenitor pool. In contrast, the expression of numb2 or numb4 resulted in a reduction of proliferating progenitors and a corresponding increase in mammalian achete-scute homologue (MASH1) expression, concurrent with the appearance of the microtubule[corrected]-associated [corrected] protein-2-positive neurons. Of interest, the effect of numb isoforms on neural differentiation could not be directly related to Notch, because classic canonical Notch signaling assays failed to uncover any differences in the four isoforms to inhibit the Notch downstream events. This finding suggests that numb may have other signaling properties during neuronal differentiation in addition to augmenting notch signal strength.
Collapse
Affiliation(s)
- Mahmud Bani-Yaghoub
- Laboratory of Neural Stem Cell Biology, Robarts Research Institute, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
137
|
Abstract
The axolotl mutant strain, short toes (s/s), can regenerate spinal cord and tail, but not limbs. This makes s/s potentially very useful for limb regeneration studies. This mutant merits a new examination that integrates the original description of the mutant, existing experimental studies, new data and current thinking about stem cells and regeneration. There are still major gaps in information about this mutant; the gene(s) causing the defects has not yet been discovered, and even the histological description is incomplete, especially regarding muscle abnormalities. In the short toes limb, MyHC (myosin heavy chain)-1, MyHC-2b and pax7 are down-regulated. In particular, all three MyHC genes and pax7 are highly expressed in the normal limb, but almost lost in the s/s limb. MyHC genes are one of the main components of skeletal muscle, and Pax7 is the skeletal muscle satellite cell marker. Histological experiments confirm that severe s/s has lost most skeletal muscle and myosin. These results suggest that skeletal muscle, which includes satellite cells, could play an important role in axolotl limb regeneration.
Collapse
Affiliation(s)
- Kazuna Sato
- The Indiana University Center for Regenerative Biology and Medicine, Department of Biology, Indiana University-Purdue University Indianapolis (IUPUI), Indianapolis, Indiana 46202-5132, USA
| | | |
Collapse
|
138
|
Watanabe K, Kondo K, Takeuchi N, Okano H, Yamasoba T. Musashi-1 expression in postnatal mouse olfactory epithelium. Neuroreport 2007; 18:641-4. [PMID: 17426590 DOI: 10.1097/wnr.0b013e3280bef7e2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We investigated the age-related change in the distribution of a molecular marker for neural stem and precursor cells, Musashi-1, in the olfactory epithelium of mice from 1 day up to 16 months of age using immunohistochemistry. We also compared the distribution pattern of Musashi-1 with that of growth-associated protein 43, the olfactory marker protein, and Notch-1. Musashi-1 was expressed in the globose basal cell layer and the lower part of the growth-associated protein 43-positive layer, with immunoreactivity decreasing with aging. Notch-1 was observed only in the early postnatal period. These findings are consistent with the fact that globose basal cells are proliferating olfactory precursor cells and that their ability to generate new neurons decreases with aging.
Collapse
Affiliation(s)
- Kenta Watanabe
- Department of Otolaryngology, Faculty of Medicine, University of Tokyo, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
139
|
Wilson JM, Sato K, Chernoff EAG, Belecky-Adams TL. Expression patterns of chick Musashi-1 in the developing nervous system. Gene Expr Patterns 2007; 7:817-25. [PMID: 17544341 DOI: 10.1016/j.modgep.2007.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2006] [Revised: 03/26/2007] [Accepted: 04/15/2007] [Indexed: 10/23/2022]
Abstract
Vertebrate homologues of musashi have recently been referred to as neural stem cell markers because of their expression patterns and RNA-binding interactions. In the context of the notch signaling pathway, Musashi-1 (Msi-1) is a regulator of neural cell generation, cooperating with notch to maintain mitosis. In an effort to identify definitive stem cell markers of the neural retina, a portion of the Msi-1 cDNA was cloned, and the expression of Msi-1 in the chick eye was analyzed. Using an Msi-1-specific antibody and RNA probe, we show that expression of Msi-1 in the early neural tube is consistent with neural stem identity. In the neural retina, expression starts shortly before embryonic day 3 (E3) and continues up to and including E18. A BrdU incorporation assay shows Msi-1 to be found in both proliferating and differentiating cells of E5 neural retina. At E8 (when proliferation is complete in the fundus of the retina) and E18 (mature retina) Msi-1 expression was found in the ciliary marginal zone (CMZ) as well as in a subpopulation of differentiated cells, including photoreceptors and ganglion cells.
Collapse
Affiliation(s)
- Jonathan M Wilson
- Department of Biology and Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | | | | | | |
Collapse
|
140
|
Shu L, Yan W, Chen X. RNPC1, an RNA-binding protein and a target of the p53 family, is required for maintaining the stability of the basal and stress-induced p21 transcript. Genes Dev 2006; 20:2961-72. [PMID: 17050675 PMCID: PMC1620019 DOI: 10.1101/gad.1463306] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
p21, a cyclin-dependent kinase inhibitor, is transcriptionally regulated by the p53 family to induce cell cycle arrest. p21 is also regulated post-transcriptionally upon DNA damage in a p53-dependent manner, but the mechanism is uncertain. Here, we found that RNPC1, an RNA-binding protein and a target of the p53 family, is required for maintaining the stability of the basal and stress-induced p21 transcript. Specifically, we showed that RNPC1 is induced by the p53 family and DNA damage in a p53-dependent manner. The RNPC1 gene encodes at least two alternative spliced isoforms, RNPC1a and RNPC1b, both of which contain an intact RNA recognition motif. Interestingly, we found that RNPC1a, but not RNPC1b, induces cell cycle arrest in G1, although both isoforms are expressed in the nucleus and cytoplasm. In addition, we found that while both isoforms directly bind to the 3' untranslated region in p21 transcript, only RNPC1a is able to stabilize both the basal and stress-induced p21 transcripts. Conversely, RNPC1a knockdown destabilizes p21 transcript. Finally, we found that RNPC1a is required to maintain the stability of p21 transcript induced by p53.
Collapse
Affiliation(s)
- Limin Shu
- Department of Cell Biology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | |
Collapse
|
141
|
Charlesworth A, Wilczynska A, Thampi P, Cox LL, MacNicol AM. Musashi regulates the temporal order of mRNA translation during Xenopus oocyte maturation. EMBO J 2006; 25:2792-801. [PMID: 16763568 PMCID: PMC1500856 DOI: 10.1038/sj.emboj.7601159] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2005] [Accepted: 04/28/2006] [Indexed: 11/08/2022] Open
Abstract
A strict temporal order of maternal mRNA translation is essential for meiotic cell cycle progression in oocytes of the frog Xenopus laevis. The molecular mechanisms controlling the ordered pattern of mRNA translational activation have not been elucidated. We report a novel role for the neural stem cell regulatory protein, Musashi, in controlling the translational activation of the mRNA encoding the Mos proto-oncogene during meiotic cell cycle progression. We demonstrate that Musashi interacts specifically with the polyadenylation response element in the 3' untranslated region of the Mos mRNA and that this interaction is necessary for early Mos mRNA translational activation. A dominant inhibitory form of Musashi blocks maternal mRNA cytoplasmic polyadenylation and meiotic cell cycle progression. Our data suggest that Musashi is a target of the initiating progesterone signaling pathway and reveal that late cytoplasmic polyadenylation element-directed mRNA translation requires early, Musashi-dependent mRNA translation. These findings indicate that Musashi function is necessary to establish the temporal order of maternal mRNA translation during Xenopus meiotic cell cycle progression.
Collapse
Affiliation(s)
- Amanda Charlesworth
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Anna Wilczynska
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Prajitha Thampi
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Linda L Cox
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Angus M MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- The Arkansas Cancer Research Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Neurobiology and Developmental Sciences, The Arkansas Cancer Research Center, Slot 814, University of Arkansas for Medical Sciences, 4301 West Markham, Little Rock, AR 72205, USA. Tel.: +1 501 686 8164; Fax: +1 501 686 6517; E-mail:
| |
Collapse
|
142
|
Ratti A, Fallini C, Cova L, Fantozzi R, Calzarossa C, Zennaro E, Pascale A, Quattrone A, Silani V. A role for the ELAV RNA-binding proteins in neural stem cells: stabilization of Msi1 mRNA. J Cell Sci 2006; 119:1442-52. [PMID: 16554442 DOI: 10.1242/jcs.02852] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Post-transcriptional regulation exerted by neural-specific RNA-binding proteins plays a pivotal role in the development and maintenance of the nervous system. Neural ELAV proteins are key inducers of neuronal differentiation through the stabilization and/or translational enhancement of target transcripts bearing the AU-rich elements (AREs), whereas Musashi-1 maintains the stem cell proliferation state by acting as a translational repressor. Since the gene encoding Musashi-1 (Msi1) contains a conserved ARE in its 3' untranslated region, we focused on the possibility of a mechanistic relationship between ELAV proteins and Musashi-1 in cell fate commitment. Colocalization of neural ELAV proteins with Musashi-1 clearly shows that ELAV proteins are expressed at early stages of neural commitment, whereas interaction studies demonstrate that neural ELAV proteins exert an ARE-dependent binding activity on the Msi1 mRNA. This binding activity has functional effects, since the ELAV protein family member HuD is able to stabilize the Msi1 ARE-containing mRNA in a sequence-dependent way in a deadenylation/degradation assay. Furthermore activation of the neural ELAV proteins by phorbol esters in human SH-SY5Y cells is associated with an increase of Musashi-1 protein content in the cytoskeleton. We propose that ELAV RNA-binding proteins exert an important post-transcriptional control on Musashi-1 expression in the transition from proliferation to neural differentiation of stem/progenitor cells.
Collapse
Affiliation(s)
- Antonia Ratti
- Department of Neuroscience, Dino Ferrari Centre, University of Milan-IRCCS Istituto Auxologico Italiano, Via Zucchi 18, 20095 Cusano Milanino, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Siddall NA, McLaughlin EA, Marriner NL, Hime GR. The RNA-binding protein Musashi is required intrinsically to maintain stem cell identity. Proc Natl Acad Sci U S A 2006; 103:8402-7. [PMID: 16717192 PMCID: PMC1570104 DOI: 10.1073/pnas.0600906103] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
A key goal of regenerative medicine is an understanding of the genetic factors that define the properties of stem cells. However, stem cell research in mammalian tissue has been hampered by a paucity of stem cell-specific markers. Although increasing evidence suggests that members of the Musashi (Msi) family of RNA-binding proteins play important functions in progenitor cells, it remains unclear whether there is a stem cell-autonomous requirement for Msi because of an inability to distinguish stem cells from early-lineage cells in mammalian tissues. Here, using the Drosophila testis as a model system for the study of stem cell regulation, we show specific evidence for a cell-autonomous requirement for Msi family proteins in regulating stem cell differentiation, leading to the identification of an RNA-binding protein required for spermatogonial stem cell maintenance. We found that loss of Msi function disrupts the balance between germ-line stem cell renewal and differentiation, resulting in the premature differentiation of germ-line stem cells. Moreover, we found that, although Msi is expressed in both somatic and germ cells, Msi function is required intrinsically in stem cells for maintenance of stem cell identity. We also discovered a requirement for Msi function in male meiosis, revealing that Msi has distinct roles at different stages of germ cell differentiation. We describe the complementary expression patterns of the murine Msi paralogues Msi1 and Msi2 during spermatogenesis, which support the idea of distinct, evolutionarily conserved roles of Msi.
Collapse
Affiliation(s)
- Nicole A. Siddall
- *Department of Anatomy and Cell Biology, University of Melbourne, Melbourne VIC 3010, Australia
- School of Environmental and Life Sciences, University of Newcastle, Callaghan NSW 2308, Australia; and
| | - Eileen A. McLaughlin
- School of Environmental and Life Sciences, University of Newcastle, Callaghan NSW 2308, Australia; and
- Australian Research Council Centre of Excellence in Biotechnology and Development, Callaghan NSW 2308, Australia
| | - Neisha L. Marriner
- *Department of Anatomy and Cell Biology, University of Melbourne, Melbourne VIC 3010, Australia
- School of Environmental and Life Sciences, University of Newcastle, Callaghan NSW 2308, Australia; and
| | - Gary R. Hime
- *Department of Anatomy and Cell Biology, University of Melbourne, Melbourne VIC 3010, Australia
- School of Environmental and Life Sciences, University of Newcastle, Callaghan NSW 2308, Australia; and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|