51
|
Vizzini A, Parisi MG, Cardinale L, Testasecca L, Cammarata M. Evolution of Ciona intestinalis Tumor necrosis factor alpha (CiTNFα): Polymorphism, tissues expression, and 3D modeling. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 67:107-116. [PMID: 27829139 DOI: 10.1016/j.dci.2016.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 11/04/2016] [Accepted: 11/05/2016] [Indexed: 06/06/2023]
Abstract
Although the Tumor necrosis factor gene superfamily seems to be very conserved in vertebrates, phylogeny, tissue expression, genomic and gene organization, protein domains and polymorphism analyses showed that a strong change has happened mostly in invertebrates in which protochordates were a constraint during the immune-molecules history and evolution. RT PCR was used to investigate differential gene expression in different tissues. The expression shown was greater in the pharynx. Single-nucleotide polymorphism has been investigated in Ciona intestinalis Tumor necrosis factor alpha (CiTNFα) mRNA isolated from the pharynx of 30 ascidians collected from Licata, Sicily (Italy), by denaturing gradient gel electrophoresis (DGGE). For this analysis, CiTNFα nucleotide sequence was separated into two fragments, TNF-1 and -2, respectively, of 630 and 540 bp. We defined 23 individual DGGE patterns (named 1 to 10 for TNF-1 and 1 to 13 for TNF-2). Five patterns for TNF-1 accounted for <10% of the individuals, whereas the pattern 13 of TNF-2 accounted for >20% of the individuals. All the patterns were verified by direct sequencing. Single base-pair mutations were observed mainly within COOH-terminus, leading to 30 nucleotide sequence variants and 30 different coding sequences segregating in two main different clusters. Although most of the base mutations were silent, four propeptide variants were detected and six amino acid replacements occurred within COOH-terminus. Statistical tests for neutrality indicated negative selection pressure on signal and mature peptide domains, but possible positive selection pressure on COOH-terminus domain. Lastly we displayed the in silico 3D structure analysis including the CiTNFα variable region.
Collapse
Affiliation(s)
- Aiti Vizzini
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Via Archirafi 18, Palermo, Italy
| | - Maria Giovanna Parisi
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Via Archirafi 18, Palermo, Italy
| | - Laura Cardinale
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Via Archirafi 18, Palermo, Italy
| | - Lelia Testasecca
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Via Archirafi 18, Palermo, Italy
| | - Matteo Cammarata
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Via Archirafi 18, Palermo, Italy.
| |
Collapse
|
52
|
Kudinov AE, Karanicolas J, Golemis EA, Boumber Y. Musashi RNA-Binding Proteins as Cancer Drivers and Novel Therapeutic Targets. Clin Cancer Res 2017; 23:2143-2153. [PMID: 28143872 DOI: 10.1158/1078-0432.ccr-16-2728] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 12/16/2016] [Accepted: 12/19/2016] [Indexed: 12/12/2022]
Abstract
Aberrant gene expression that drives human cancer can arise from epigenetic dysregulation. Although much attention has focused on altered activity of transcription factors and chromatin-modulating proteins, proteins that act posttranscriptionally can potently affect expression of oncogenic signaling proteins. The RNA-binding proteins (RBP) Musashi-1 (MSI1) and Musashi-2 (MSI2) are emerging as regulators of multiple critical biological processes relevant to cancer initiation, progression, and drug resistance. Following identification of Musashi as a regulator of progenitor cell identity in Drosophila, the human Musashi proteins were initially linked to control of maintenance of hematopoietic stem cells, then stem cell compartments for additional cell types. More recently, the Musashi proteins were found to be overexpressed and prognostic of outcome in numerous cancer types, including colorectal, lung, and pancreatic cancers; glioblastoma; and several leukemias. MSI1 and MSI2 bind and regulate the mRNA stability and translation of proteins operating in essential oncogenic signaling pathways, including NUMB/Notch, PTEN/mTOR, TGFβ/SMAD3, MYC, cMET, and others. On the basis of these activities, MSI proteins maintain cancer stem cell populations and regulate cancer invasion, metastasis, and development of more aggressive cancer phenotypes, including drug resistance. Although RBPs are viewed as difficult therapeutic targets, initial efforts to develop MSI-specific inhibitors are promising, and RNA interference-based approaches to inhibiting these proteins have had promising outcomes in preclinical studies. In the interim, understanding the function of these translational regulators may yield insight into the relationship between mRNA expression and protein expression in tumors, guiding tumor-profiling analysis. This review provides a current overview of Musashi as a cancer driver and novel therapeutic target. Clin Cancer Res; 23(9); 2143-53. ©2017 AACR.
Collapse
Affiliation(s)
- Alexander E Kudinov
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - John Karanicolas
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Erica A Golemis
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Yanis Boumber
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania. .,Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| |
Collapse
|
53
|
Formation of mos RNA granules in the zebrafish oocyte that differ from cyclin B1 RNA granules in distribution, density and regulation. Eur J Cell Biol 2016; 95:563-573. [PMID: 27756483 DOI: 10.1016/j.ejcb.2016.10.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 09/07/2016] [Accepted: 10/03/2016] [Indexed: 12/28/2022] Open
Abstract
Many translationally repressed mRNAs are deposited in the oocyte cytoplasm for progression of the meiotic cell cycle and early development. mos and cyclin B1 mRNAs encode proteins promoting oocyte meiosis, and translational control of these mRNAs is important for normal progression of meiotic cell division. We previously demonstrated that cyclin B1 mRNA forms RNA granules in the zebrafish and mouse oocyte cytoplasm and that the formation of RNA granules is crucial for regulating the timing of translational activation of the mRNA. However, whether the granule formation is specific to cyclin B1 mRNA remains unknown. In this study, we found that zebrafish mos mRNA forms granules distinct from those of cyclin B1 mRNA. Fluorescent in situ hybridization analysis showed that cyclin B1 RNA granules were assembled in dense clusters, while mos RNA granules were distributed diffusely in the animal polar cytoplasm. Sucrose density gradient ultracentrifugation analysis showed that the density of mos RNA granules was partly lower than that of cyclin B1 mRNA. Similar to cyclin B1 RNA granules, mos RNA granules were disassembled after initiation of oocyte maturation at the timing at which the poly(A) tail was elongated. However, while almost all of the granules of cyclin B1 were disassembled simultaneously, a fraction of mos RNA granules firstly disappeared and then a large part of them was disassembled. In addition, while cyclin B1 RNA granules were disassembled in a manner dependent on actin filament depolymerization, certain fractions of mos RNA granules were disassembled independently of actin filaments. These results suggest that cytoplasmic regulation of translationally repressed mRNAs by formation of different RNA granules is a key mechanism for translational control of distinct mRNAs in the oocyte.
Collapse
|
54
|
de Araujo PR, Gorthi A, da Silva AE, Tonapi SS, Vo DT, Burns SC, Qiao M, Uren PJ, Yuan ZM, Bishop AJR, Penalva LOF. Musashi1 Impacts Radio-Resistance in Glioblastoma by Controlling DNA-Protein Kinase Catalytic Subunit. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2271-8. [PMID: 27470713 PMCID: PMC5012509 DOI: 10.1016/j.ajpath.2016.05.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 05/13/2016] [Indexed: 12/30/2022]
Abstract
The conserved RNA-binding protein Musashi1 (MSI1) has been characterized as a stem cell marker, controlling the balance between self-renewal and differentiation and as a key oncogenic factor in numerous solid tumors, including glioblastoma. To explore the potential use of MSI1 targeting in therapy, we studied MSI1 in the context of radiation sensitivity. Knockdown of MSI1 led to a decrease in cell survival and an increase in DNA damage compared to control in cells treated with ionizing radiation. We subsequently examined mechanisms of double-strand break repair and found that loss of MSI1 reduces the frequency of nonhomologous end-joining. This phenomenon could be attributed to the decreased expression of DNA-protein kinase catalytic subunit, which we have previously identified as a target of MSI1. Collectively, our results suggest a role for MSI1 in double-strand break repair and that its inhibition may enhance the effect of radiotherapy.
Collapse
Affiliation(s)
- Patricia Rosa de Araujo
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, Texas; Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas
| | - Aparna Gorthi
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, Texas; Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas
| | - Acarizia E da Silva
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, Texas; Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas
| | - Sonal S Tonapi
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, Texas; Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas
| | - Dat T Vo
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, Texas; Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas
| | - Suzanne C Burns
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, Texas
| | - Mei Qiao
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, Texas
| | - Philip J Uren
- Molecular and Computational Biology Section, Division of Biological Sciences, University of Southern California, Los Angeles, California
| | - Zhi-Min Yuan
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts
| | - Alexander J R Bishop
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, Texas; Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas.
| | - Luiz O F Penalva
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, Texas; Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas.
| |
Collapse
|
55
|
Bertolin AP, Katz MJ, Yano M, Pozzi B, Acevedo JM, Blanco-Obregón D, Gándara L, Sorianello E, Kanda H, Okano H, Srebrow A, Wappner P. Musashi mediates translational repression of the Drosophila hypoxia inducible factor. Nucleic Acids Res 2016; 44:7555-67. [PMID: 27141964 PMCID: PMC5027473 DOI: 10.1093/nar/gkw372] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 04/23/2016] [Indexed: 12/22/2022] Open
Abstract
Adaptation to hypoxia depends on a conserved α/β heterodimeric transcription factor called Hypoxia Inducible Factor (HIF), whose α-subunit is regulated by oxygen through different concurrent mechanisms. In this study, we have identified the RNA binding protein dMusashi, as a negative regulator of the fly HIF homologue Sima. Genetic interaction assays suggested that dMusashi participates of the HIF pathway, and molecular studies carried out in Drosophila cell cultures showed that dMusashi recognizes a Musashi Binding Element in the 3' UTR of the HIFα transcript, thereby mediating its translational repression in normoxia. In hypoxic conditions dMusashi is downregulated, lifting HIFα repression and contributing to trigger HIF-dependent gene expression. Analysis performed in mouse brains revealed that murine Msi1 protein physically interacts with HIF-1α transcript, suggesting that the regulation of HIF by Msi might be conserved in mammalian systems. Thus, Musashi is a novel regulator of HIF that inhibits responses to hypoxia specifically when oxygen is available.
Collapse
Affiliation(s)
| | - Maximiliano J Katz
- Instituto Leloir, Patricias Argentinas 435, Buenos Aires (1405), Argentina
| | - Masato Yano
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, 1-757, Asahimachidori, Chuo-ku, Niigata, Niigata 951-8510, Japan
| | - Berta Pozzi
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE, UBA-CONICET), Ciudad Universitaria, Pabellón 2, Buenos Aires (C1428EHA), Argentina Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, Buenos Aires (C1428EHA), Argentina
| | - Julieta M Acevedo
- Instituto Leloir, Patricias Argentinas 435, Buenos Aires (1405), Argentina
| | | | - Lautaro Gándara
- Instituto Leloir, Patricias Argentinas 435, Buenos Aires (1405), Argentina
| | | | - Hiroshi Kanda
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Anabella Srebrow
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE, UBA-CONICET), Ciudad Universitaria, Pabellón 2, Buenos Aires (C1428EHA), Argentina Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, Buenos Aires (C1428EHA), Argentina
| | - Pablo Wappner
- Instituto Leloir, Patricias Argentinas 435, Buenos Aires (1405), Argentina Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, Buenos Aires (C1428EHA), Argentina
| |
Collapse
|
56
|
Sutherland JM, Siddall NA, Hime GR, McLaughlin EA. RNA binding proteins in spermatogenesis: an in depth focus on the Musashi family. Asian J Androl 2016; 17:529-36. [PMID: 25851660 PMCID: PMC4492041 DOI: 10.4103/1008-682x.151397] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Controlled gene regulation during gamete development is vital for maintaining reproductive potential. During the complex process of mammalian spermatogenesis, male germ cells experience extended periods of the inactive transcription despite heavy translational requirements for continued growth and differentiation. Hence, spermatogenesis is highly reliant on mechanisms of posttranscriptional regulation of gene expression, facilitated by RNA binding proteins (RBPs), which remain abundantly expressed throughout this process. One such group of proteins is the Musashi family, previously identified as critical regulators of testis germ cell development and meiosis in Drosophila, and also shown to be vital to sperm development and reproductive potential in the mouse. This review describes the role and function of RBPs within the scope of male germ cell development, focusing on our recent knowledge of the Musashi proteins in spermatogenesis. The functional mechanisms utilized by RBPs within the cell are outlined in depth, and the significance of sub-cellular localization and stage-specific expression in relation to the mode and impact of posttranscriptional regulation is also highlighted. We emphasize the historical role of the Musashi family of RBPs in stem cell function and cell fate determination, as originally characterized in Drosophila and Xenopus, and conclude with our current understanding of the differential roles and functions of the mammalian Musashi proteins, Musashi-1 and Musashi-2, with a primary focus on our findings in spermatogenesis. This review highlights both the essential contribution of RBPs to posttranscriptional regulation and the importance of the Musashi family as master regulators of male gamete development.
Collapse
Affiliation(s)
| | | | | | - Eileen A McLaughlin
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| |
Collapse
|
57
|
Yamagishi R, Tsusaka T, Mitsunaga H, Maehata T, Hoshino SI. The STAR protein QKI-7 recruits PAPD4 to regulate post-transcriptional polyadenylation of target mRNAs. Nucleic Acids Res 2016; 44:2475-90. [PMID: 26926106 PMCID: PMC4824116 DOI: 10.1093/nar/gkw118] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/16/2016] [Indexed: 12/20/2022] Open
Abstract
Emerging evidence has demonstrated that regulating the length of the poly(A) tail on an mRNA is an efficient means of controlling gene expression at the post-transcriptional level. In early development, transcription is silenced and gene expression is primarily regulated by cytoplasmic polyadenylation. In somatic cells, considerable progress has been made toward understanding the mechanisms of negative regulation by deadenylation. However, positive regulation through elongation of the poly(A) tail has not been widely studied due to the difficulty in distinguishing whether any observed increase in length is due to the synthesis of new mRNA, reduced deadenylation or cytoplasmic polyadenylation. Here, we overcame this barrier by developing a method for transcriptional pulse-chase analysis under conditions where deadenylases are suppressed. This strategy was used to show that a member of the Star family of RNA binding proteins, QKI, promotes polyadenylation when tethered to a reporter mRNA. Although multiple RNA binding proteins have been implicated in cytoplasmic polyadenylation during early development, previously only CPEB was known to function in this capacity in somatic cells. Importantly, we show that only the cytoplasmic isoform QKI-7 promotes poly(A) tail extension, and that it does so by recruiting the non-canonical poly(A) polymerase PAPD4 through its unique carboxyl-terminal region. We further show that QKI-7 specifically promotes polyadenylation and translation of three natural target mRNAs (hnRNPA1, p27kip1 and β-catenin) in a manner that is dependent on the QKI response element. An anti-mitogenic signal that induces cell cycle arrest at G1 phase elicits polyadenylation and translation of p27kip1 mRNA via QKI and PAPD4. Taken together, our findings provide significant new insight into a general mechanism for positive regulation of gene expression by post-transcriptional polyadenylation in somatic cells.
Collapse
Affiliation(s)
- Ryota Yamagishi
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Takeshi Tsusaka
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Hiroko Mitsunaga
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Takaharu Maehata
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Shin-ichi Hoshino
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| |
Collapse
|
58
|
CPEB and miR-15/16 Co-Regulate Translation of Cyclin E1 mRNA during Xenopus Oocyte Maturation. PLoS One 2016; 11:e0146792. [PMID: 26829217 PMCID: PMC4734764 DOI: 10.1371/journal.pone.0146792] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 12/22/2015] [Indexed: 01/07/2023] Open
Abstract
Cell cycle transitions spanning meiotic maturation of the Xenopus oocyte and early embryogenesis are tightly regulated at the level of stored inactive maternal mRNA. We investigated here the translational control of cyclin E1, required for metaphase II arrest of the unfertilised egg and the initiation of S phase in the early embryo. We show that the cyclin E1 mRNA is regulated by both cytoplasmic polyadenylation elements (CPEs) and two miR-15/16 target sites within its 3’UTR. Moreover, we provide evidence that maternal miR-15/16 microRNAs co-immunoprecipitate with CPE-binding protein (CPEB), and that CPEB interacts with the RISC component Ago2. Experiments using competitor RNA and mutated cyclin E1 3’UTRs suggest cooperation of the regulatory elements to sustain repression of the cyclin E1 mRNA during early stages of maturation when CPEB becomes limiting and cytoplasmic polyadenylation of repressed mRNAs begins. Importantly, injection of anti-miR-15/16 LNA results in the early polyadenylation of endogenous cyclin E1 mRNA during meiotic maturation, and an acceleration of GVBD, altogether strongly suggesting that the proximal CPEB and miRNP complexes act to mutually stabilise each other. We conclude that miR-15/16 and CPEB co-regulate cyclin E1 mRNA. This is the first demonstration of the co-operation of these two pathways.
Collapse
|
59
|
Malhotra S, Vinod PK, Mansfeld J, Stemmann O, Mayer TU. RETRACTED: The Anaphase-Promoting Complex/Cyclosome Is Essential for Entry into Meiotic M-Phase. Dev Cell 2016; 36:94-102. [DOI: 10.1016/j.devcel.2015.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 11/12/2015] [Accepted: 12/03/2015] [Indexed: 11/16/2022]
|
60
|
Reyes JM, Ross PJ. Cytoplasmic polyadenylation in mammalian oocyte maturation. WILEY INTERDISCIPLINARY REVIEWS-RNA 2015; 7:71-89. [PMID: 26596258 DOI: 10.1002/wrna.1316] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 10/02/2015] [Accepted: 10/07/2015] [Indexed: 12/21/2022]
Abstract
Oocyte developmental competence is the ability of the mature oocyte to be fertilized and subsequently drive early embryo development. Developmental competence is acquired by completion of oocyte maturation, a process that includes nuclear (meiotic) and cytoplasmic (molecular) changes. Given that maturing oocytes are transcriptionally quiescent (as are early embryos), they depend on post-transcriptional regulation of stored transcripts for protein synthesis, which is largely mediated by translational repression and deadenylation of transcripts within the cytoplasm, followed by recruitment of specific transcripts in a spatiotemporal manner for translation during oocyte maturation and early development. Motifs within the 3' untranslated region (UTR) of messenger RNA (mRNA) are thought to mediate repression and downstream activation by their association with binding partners that form dynamic protein complexes that elicit differing effects on translation depending on cell stage and interacting proteins. The cytoplasmic polyadenylation (CP) element, Pumilio binding element, and hexanucleotide polyadenylation signal are among the best understood motifs involved in CP, and translational regulation of stored transcripts as their binding partners have been relatively well-characterized. Knowledge of CP in mammalian oocytes is discussed as well as novel approaches that can be used to enhance our understanding of the functional and contributing features to transcript CP and translational regulation during mammalian oocyte maturation. WIREs RNA 2016, 7:71-89. doi: 10.1002/wrna.1316 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Juan M Reyes
- Department of Animal Science, University of California, Davis, CA, USA
| | - Pablo J Ross
- Department of Animal Science, University of California, Davis, CA, USA
| |
Collapse
|
61
|
Fox RG, Park FD, Koechlein CS, Kritzik M, Reya T. Musashi Signaling in Stem Cells and Cancer. Annu Rev Cell Dev Biol 2015; 31:249-67. [DOI: 10.1146/annurev-cellbio-100814-125446] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Raymond G. Fox
- Department of Pharmacology,
- Moores Cancer Center, and
- Sanford Consortium for Regenerative Medicine, La Jolla, California 92037
| | - Frederick D. Park
- Department of Pharmacology,
- Moores Cancer Center, and
- Division of Gastroenterology, University of California San Diego School of Medicine, La Jolla, California 92093;
- Sanford Consortium for Regenerative Medicine, La Jolla, California 92037
| | - Claire S. Koechlein
- Department of Pharmacology,
- Moores Cancer Center, and
- Sanford Consortium for Regenerative Medicine, La Jolla, California 92037
| | - Marcie Kritzik
- Department of Pharmacology,
- Moores Cancer Center, and
- Sanford Consortium for Regenerative Medicine, La Jolla, California 92037
| | - Tannishtha Reya
- Department of Pharmacology,
- Moores Cancer Center, and
- Sanford Consortium for Regenerative Medicine, La Jolla, California 92037
| |
Collapse
|
62
|
Mardakheh FK, Paul A, Kümper S, Sadok A, Paterson H, Mccarthy A, Yuan Y, Marshall CJ. Global Analysis of mRNA, Translation, and Protein Localization: Local Translation Is a Key Regulator of Cell Protrusions. Dev Cell 2015; 35:344-57. [PMID: 26555054 PMCID: PMC4643311 DOI: 10.1016/j.devcel.2015.10.005] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 08/23/2015] [Accepted: 10/08/2015] [Indexed: 12/12/2022]
Abstract
Polarization of cells into a protrusive front and a retracting cell body is the hallmark of mesenchymal-like cell migration. Many mRNAs are localized to protrusions, but it is unclear to what degree mRNA localization contributes toward protrusion formation. We performed global quantitative analysis of the distributions of mRNAs, proteins, and translation rates between protrusions and the cell body by RNA sequencing (RNA-seq) and quantitative proteomics. Our results reveal local translation as a key determinant of protein localization to protrusions. Accordingly, inhibition of local translation destabilizes protrusions and inhibits mesenchymal-like morphology. Interestingly, many mRNAs localized to protrusions are translationally repressed. Specific cis-regulatory elements within mRNA UTRs define whether mRNAs are locally translated or repressed. Finally, RNAi screening of RNA-binding proteins (RBPs) enriched in protrusions revealed trans-regulators of localized translation that are functionally important for protrusions. We propose that by deciphering the localized mRNA UTR code, these proteins regulate protrusion stability and mesenchymal-like morphology.
Collapse
Affiliation(s)
- Faraz K Mardakheh
- Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| | - Angela Paul
- Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Sandra Kümper
- Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Amine Sadok
- Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Hugh Paterson
- Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Afshan Mccarthy
- Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Yinyin Yuan
- Division of Molecular Pathology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Christopher J Marshall
- Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| |
Collapse
|
63
|
Miyazaki S, Sato Y, Asano T, Nagamura Y, Nonomura KI. Rice MEL2, the RNA recognition motif (RRM) protein, binds in vitro to meiosis-expressed genes containing U-rich RNA consensus sequences in the 3'-UTR. PLANT MOLECULAR BIOLOGY 2015; 89:293-307. [PMID: 26319516 DOI: 10.1007/s11103-015-0369-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 08/22/2015] [Indexed: 06/04/2023]
Abstract
Post-transcriptional gene regulation by RNA recognition motif (RRM) proteins through binding to cis-elements in the 3'-untranslated region (3'-UTR) is widely used in eukaryotes to complete various biological processes. Rice MEIOSIS ARRESTED AT LEPTOTENE2 (MEL2) is the RRM protein that functions in the transition to meiosis in proper timing. The MEL2 RRM preferentially associated with the U-rich RNA consensus, UUAGUU[U/A][U/G][A/U/G]U, dependently on sequences and proportionally to MEL2 protein amounts in vitro. The consensus sequences were located in the putative looped structures of the RNA ligand. A genome-wide survey revealed a tendency of MEL2-binding consensus appearing in 3'-UTR of rice genes. Of 249 genes that conserved the consensus in their 3'-UTR, 13 genes spatiotemporally co-expressed with MEL2 in meiotic flowers, and included several genes whose function was supposed in meiosis; such as Replication protein A and OsMADS3. The proteome analysis revealed that the amounts of small ubiquitin-related modifier-like protein and eukaryotic translation initiation factor3-like protein were dramatically altered in mel2 mutant anthers. Taken together with transcriptome and gene ontology results, we propose that the rice MEL2 is involved in the translational regulation of key meiotic genes on 3'-UTRs to achieve the faithful transition of germ cells to meiosis.
Collapse
Affiliation(s)
- Saori Miyazaki
- Experimental Farm, National Institute of Genetics, Mishima, Shizuoka, 411-8540, Japan.
- Department of Genetics, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Mishima, Shizuoka, 411-8540, Japan.
- Office for the Promotion of Global Education Programs, Shizuoka University, Jyouhoku, Nakaku, Hamamatsu, Shizuoka, 432-8561, Japan.
| | - Yutaka Sato
- Genome Resource Unit, Agrogenomics Research Center, National Institute of Agrobiological Sciences, Kannondai 2-1-2, Tsukuba, Ibaraki, 305-8602, Japan.
| | - Tomoya Asano
- Division of Functional Genomics, Advanced Science Research Center, Kanazawa University, Takaramachi, Kanazawa, 920-0934, Japan.
- Wakasa Seikatsu Co. Ltd, 22 Naginataboko-cho, Shijo-Karasuma, Shimogyo-ku, Kyoto, 600-8008, Japan.
| | - Yoshiaki Nagamura
- Genome Resource Unit, Agrogenomics Research Center, National Institute of Agrobiological Sciences, Kannondai 2-1-2, Tsukuba, Ibaraki, 305-8602, Japan.
| | - Ken-Ichi Nonomura
- Experimental Farm, National Institute of Genetics, Mishima, Shizuoka, 411-8540, Japan.
- Department of Genetics, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Mishima, Shizuoka, 411-8540, Japan.
| |
Collapse
|
64
|
Nahas GR, Murthy RG, Patel SA, Ganta T, Greco SJ, Rameshwar P. The RNA-binding protein Musashi 1 stabilizes the oncotachykinin 1 mRNA in breast cancer cells to promote cell growth. FASEB J 2015; 30:149-59. [PMID: 26373800 DOI: 10.1096/fj.15-278770] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 08/17/2015] [Indexed: 12/20/2022]
Abstract
Substance P and its truncated receptor exert oncogenic effects. The high production of substance P in breast cancer cells (BCCs) is caused by the enhancement of tachykinin (TAC)1 translation by cytosolic factor. In vitro translational studies and mRNA stabilization analyses indicate that BCCs contain the factor needed to increase TAC1 translation and to stabilize the mRNA. Prediction of protein folding, RNA-shift analysis, and proteomic analysis identified a 40 kDa molecule that interacts with the noncoding exon 7. Western blot analysis and RNA supershift identified Musashi 1 (Msi1) as the binding protein. Ectopic expression of TAC1 in nontumorigenic breast cells (BCs) indicates that TAC1 regulates its stability by increasing Msi1. Using a reporter gene system, we showed that Msi1 competes with microRNA (miR)130a and -206 for the 3' UTR of exon 7/TAC1. In the absence of Msi1 and miR130a and -206, reporter gene activity decreased, indicating that Msi1 expression limits TAC1 expression. Tumor growth was significantly decreased when nude BALB/c mice were injected with Msi1-knockdown BCCs. In summary, the RNA-binding protein Msi1 competes with miR130a and -206 for interaction with TAC1 mRNA, to stabilize and increase its translation. Consequently, these interactions increase tumor growth.
Collapse
Affiliation(s)
- George R Nahas
- Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Raghav G Murthy
- Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Shyam A Patel
- Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Teja Ganta
- Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Steven J Greco
- Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Pranela Rameshwar
- Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| |
Collapse
|
65
|
MacNicol MC, Cragle CE, Arumugam K, Fosso B, Pesole G, MacNicol AM. Functional Integration of mRNA Translational Control Programs. Biomolecules 2015. [PMID: 26197342 PMCID: PMC4598765 DOI: 10.3390/biom5031580] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Regulated mRNA translation plays a key role in control of cell cycle progression in a variety of physiological and pathological processes, including in the self-renewal and survival of stem cells and cancer stem cells. While targeting mRNA translation presents an attractive strategy for control of aberrant cell cycle progression, mRNA translation is an underdeveloped therapeutic target. Regulated mRNAs are typically controlled through interaction with multiple RNA binding proteins (RBPs) but the mechanisms by which the functions of distinct RBPs bound to a common target mRNA are coordinated are poorly understood. The challenge now is to gain insight into these mechanisms of coordination and to identify the molecular mediators that integrate multiple, often conflicting, inputs. A first step includes the identification of altered mRNA ribonucleoprotein complex components that assemble on mRNAs bound by multiple, distinct RBPs compared to those recruited by individual RBPs. This review builds upon our knowledge of combinatorial control of mRNA translation during the maturation of oocytes from Xenopus laevis, to address molecular strategies that may mediate RBP diplomacy and conflict resolution for coordinated control of mRNA translational output. Continued study of regulated ribonucleoprotein complex dynamics promises valuable new insights into mRNA translational control and may suggest novel therapeutic strategies for the treatment of disease.
Collapse
Affiliation(s)
- Melanie C MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
- Center for Translational Neuroscience, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Chad E Cragle
- Interdisciplinary BioSciences Graduate Program, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Karthik Arumugam
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Bruno Fosso
- Institute of Biomembranes and Bioenergetics, National Research Council, Bari 70126, Italy.
| | - Graziano Pesole
- Institute of Biomembranes and Bioenergetics, National Research Council, Bari 70126, Italy.
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Bari 70125, Italy.
| | - Angus M MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
- Center for Translational Neuroscience, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|
66
|
Knockout of RNA Binding Protein MSI2 Impairs Follicle Development in the Mouse Ovary: Characterization of MSI1 and MSI2 during Folliculogenesis. Biomolecules 2015; 5:1228-44. [PMID: 26131972 PMCID: PMC4598749 DOI: 10.3390/biom5031228] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 05/29/2015] [Accepted: 06/09/2015] [Indexed: 01/20/2023] Open
Abstract
Characterizing the mechanisms underlying follicle development in the ovary is crucial to understanding female fertility and is an area of increasing research interest. The RNA binding protein Musashi is essential for post-transcriptional regulation of oocyte maturation in Xenopus and is expressed during ovarian development in Drosophila. In mammals Musashi is important for spermatogenesis and male fertility, but its role in the ovary has yet to be characterized. In this study we determined the expression of mammalian Musashi proteins MSI1 and MSI2 during mouse folliculogenesis, and through the use of a MSI2-specific knockout mouse model we identified that MSI2 is essential for normal follicle development. Time-course characterization of MSI1 and MSI2 revealed distinct differences in steady-state mRNA levels and protein expression/localization at important developmental time-points during folliculogenesis. Using a gene-trap mouse model that inactivates Msi2, we observed a significant decrease in ovarian mass, and change in follicle-stage composition due to developmental blocking of antral stage follicles and pre-antral follicle loss through atresia. We also confirmed that hormonally stimulated Msi2-deficient mice produce significantly fewer MII oocytes (60.9% less than controls, p < 0.05). Furthermore, the majority of these oocytes are of poor viability (62.2% non-viable/apoptotic, p < 0.05), which causes a reduction in female fertility evidenced by decreased litter size in Msi2-deficient animals (33.1% reduction to controls, p < 0.05). Our findings indicate that MSI1 and MSI2 display distinct expression profiles during mammalian folliculogenesis and that MSI2 is required for pre-antral follicle development.
Collapse
|
67
|
Horisawa K, Imai T, Okano H, Yanagawa H. The Musashi family RNA-binding proteins in stem cells. Biomol Concepts 2015; 1:59-66. [PMID: 25961986 DOI: 10.1515/bmc.2010.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The Musashi family is an evolutionarily conserved group of RNA-binding proteins. In mammal, two members of the group, Msi1 and Msi2, have been identified to date. Msi1 is considered to play roles in maintaining the stem cell status (stemness) of neural stem/progenitor cells in adults and in the development of central nervous system through translational regulation of its target mRNAs, which encode regulators of signal transduction and the cell cycle. Recently, strong expression of Msi1 in various somatic stem/progenitor cells of adult tissues, such as eye, gut, stomach, breast, and hair follicle, has been reported. The protein is also expressed in various cancer cells, and ectopically emerging cells have been found in neural tissues of patients with diseases involving neural disorder, including epilepsy. Many novel target mRNAs and regulatory pathways of Msi1 have been reported in recent years. Here, we present a review of the functions and action mechanisms of Msi1 protein and discuss possible directions for further study.
Collapse
|
68
|
Lan L, Appelman C, Smith AR, Yu J, Larsen S, Marquez RT, Liu H, Wu X, Gao P, Roy A, Anbanandam A, Gowthaman R, Karanicolas J, De Guzman RN, Rogers S, Aubé J, Ji M, Cohen RS, Neufeld KL, Xu L. Natural product (-)-gossypol inhibits colon cancer cell growth by targeting RNA-binding protein Musashi-1. Mol Oncol 2015; 9:1406-20. [PMID: 25933687 DOI: 10.1016/j.molonc.2015.03.014] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 03/09/2015] [Accepted: 03/27/2015] [Indexed: 02/07/2023] Open
Abstract
Musashi-1 (MSI1) is an RNA-binding protein that acts as a translation activator or repressor of target mRNAs. The best-characterized MSI1 target is Numb mRNA, whose encoded protein negatively regulates Notch signaling. Additional MSI1 targets include the mRNAs for the tumor suppressor protein APC that regulates Wnt signaling and the cyclin-dependent kinase inhibitor P21(WAF-1). We hypothesized that increased expression of NUMB, P21 and APC, through inhibition of MSI1 RNA-binding activity might be an effective way to simultaneously downregulate Wnt and Notch signaling, thus blocking the growth of a broad range of cancer cells. We used a fluorescence polarization assay to screen for small molecules that disrupt the binding of MSI1 to its consensus RNA binding site. One of the top hits was (-)-gossypol (Ki = 476 ± 273 nM), a natural product from cottonseed, known to have potent anti-tumor activity and which has recently completed Phase IIb clinical trials for prostate cancer. Surface plasmon resonance and nuclear magnetic resonance studies demonstrate a direct interaction of (-)-gossypol with the RNA binding pocket of MSI1. We further showed that (-)-gossypol reduces Notch/Wnt signaling in several colon cancer cell lines having high levels of MSI1, with reduced SURVIVIN expression and increased apoptosis/autophagy. Finally, we showed that orally administered (-)-gossypol inhibits colon cancer growth in a mouse xenograft model. Our study identifies (-)-gossypol as a potential small molecule inhibitor of MSI1-RNA interaction, and suggests that inhibition of MSI1's RNA binding activity may be an effective anti-cancer strategy.
Collapse
Affiliation(s)
- Lan Lan
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS, USA
| | - Carl Appelman
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS, USA
| | - Amber R Smith
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS, USA
| | - Jia Yu
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS, USA; School of Chemistry and Chemical Engineering, Southeast University, Nanjing, China
| | - Sarah Larsen
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS, USA
| | - Rebecca T Marquez
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS, USA
| | - Hao Liu
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS, USA
| | - Xiaoqing Wu
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS, USA
| | - Philip Gao
- COBRE Protein Production Group, The University of Kansas, Lawrence, KS, USA
| | - Anuradha Roy
- High Throughput Screening Laboratory, The University of Kansas, Lawrence, KS, USA
| | | | - Ragul Gowthaman
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS, USA; Center for Bioinformatics, The University of Kansas, Lawrence, KS, USA
| | - John Karanicolas
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS, USA; Center for Bioinformatics, The University of Kansas, Lawrence, KS, USA
| | - Roberto N De Guzman
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS, USA
| | - Steven Rogers
- Center of Biomedical Research Excellence, Center for Cancer Experimental Therapeutics, The University of Kansas, Lawrence, KS, USA
| | - Jeffrey Aubé
- Center of Biomedical Research Excellence, Center for Cancer Experimental Therapeutics, The University of Kansas, Lawrence, KS, USA; Specialized Chemistry Center, The University of Kansas, Lawrence, KS, USA; Center for Chemical Methodologies and Library Development, The University of Kansas, Lawrence, KS, USA; Department of Medicinal Chemistry, The University of Kansas, Lawrence, KS, USA
| | - Min Ji
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, China
| | - Robert S Cohen
- Department of Biological Sciences, Clemson University, Clemson, SC, USA
| | - Kristi L Neufeld
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS, USA
| | - Liang Xu
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS, USA; Department of Radiation Oncology, The University of Kansas Cancer Center, Kansas City, KS, USA.
| |
Collapse
|
69
|
MacNicol AM, Hardy LL, Spencer HJ, MacNicol MC. Neural stem and progenitor cell fate transition requires regulation of Musashi1 function. BMC DEVELOPMENTAL BIOLOGY 2015; 15:15. [PMID: 25888190 PMCID: PMC4369890 DOI: 10.1186/s12861-015-0064-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 02/26/2015] [Indexed: 12/28/2022]
Abstract
Background There is increasing evidence of a pivotal role for regulated mRNA translation in control of developmental cell fate transitions. Physiological and pathological stem and progenitor cell self-renewal is maintained by the mRNA-binding protein, Musashi1 through repression of translation of key mRNAs encoding cell cycle inhibitory proteins. The mechanism by which Musashi1 function is modified to allow translation of these target mRNAs under conditions that require inhibition of cell cycle progression, is unknown. Results In this study, we demonstrate that differentiation of primary embryonic rat neural stem/progenitor cells (NSPCs) or human neuroblastoma SH-SY5Y cells results in the rapid phosphorylation of Musashi1 on the evolutionarily conserved site serine 337 (S337). Phosphorylation of this site has been shown to be required for cell cycle control during the maturation of Xenopus oocytes. S337 phosphorylation in mammalian NSPCs and human SH-SY5Y cells correlates with the de-repression and translation of a Musashi reporter mRNA and with accumulation of protein from the endogenous Musashi target mRNA, p21WAF1/CIP1. Inhibition of Musashi regulatory phosphorylation, through expression of a phospho-inhibitory mutant Musashi1 S337A or over-expression of the wild-type Musashi, blocked differentiation of both NSPCs and SH-SY5Y cells. Musashi1 was similarly phosphorylated in NSPCs and SH-SY5Y cells under conditions of nutrient deprivation-induced cell cycle arrest. Expression of the Musashi1 S337A mutant protein attenuated nutrient deprivation-induced NSPC and SH-SY5Y cell death. Conclusions Our data suggest that in response to environmental cues that oppose cell cycle progression, regulation of Musashi function is required to promote target mRNA translation and cell fate transition. Forced modulation of Musashi1 function may present a novel therapeutic strategy to oppose pathological stem cell self-renewal.
Collapse
Affiliation(s)
- Angus M MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W. Markham, Slot 814, Little Rock, AR, 72205, USA. .,Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W. Markham, Little Rock, AR, 72205, USA.
| | - Linda L Hardy
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W. Markham, Slot 814, Little Rock, AR, 72205, USA.
| | - Horace J Spencer
- Department of Biostatistics, University of Arkansas for Medical Sciences, 4301 W. Markham, Little Rock, AR, 72205, USA.
| | - Melanie C MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W. Markham, Slot 814, Little Rock, AR, 72205, USA. .,Center for Translational Neuroscience, University of Arkansas for Medical Sciences, 4301 W. Markham, Little Rock, AR, 72205, USA.
| |
Collapse
|
70
|
Sutherland JM, Sobinoff AP, Fraser BA, Redgrove KA, Davidson TL, Siddall NA, Koopman P, Hime GR, McLaughlin EA. RNA binding protein Musashi-1 directly targets Msi2 and Erh during early testis germ cell development and interacts with IPO5 upon translocation to the nucleus. FASEB J 2015; 29:2759-68. [PMID: 25782991 DOI: 10.1096/fj.14-265868] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 02/26/2015] [Indexed: 12/19/2022]
Abstract
Controlled gene regulation during gamete development is vital for maintaining reproductive potential. During the process of gamete development, male germ cells experience extended periods of inactive transcription despite requirements for continued growth and differentiation. Spermatogenesis therefore provides an ideal model to study the effects of posttranscriptional control on gene regulation. During spermatogenesis posttranscriptional regulation is orchestrated by abundantly expressed RNA-binding proteins. One such group of RNA-binding proteins is the Musashi family, previously identified as a critical regulator of testis germ cell development and meiosis in Drosophila and also shown to be vital to sperm development and reproductive potential in the mouse. We focus in depth on the role and function of the vertebrate Musashi ortholog Musashi-1 (MSI1). Through detailed expression studies and utilizing our novel transgenic Msi1 testis-specific overexpression model, we have identified 2 unique RNA-binding targets of MSI1 in spermatogonia, Msi2 and Erh, and have demonstrated a role for MSI1 in translational regulation. We have also provided evidence to suggest that nuclear import protein, IPO5, facilitates the nuclear translocation of MSI1 to the transcriptionally silenced XY chromatin domain in meiotic pachytene spermatocytes, resulting in the release of MSI1 RNA-binding targets. This firmly establishes MSI1 as a master regulator of posttranscriptional control during early spermatogenesis and highlights the significance of the subcellular localization of RNA binding proteins in relation to their function.
Collapse
Affiliation(s)
- Jessie M Sutherland
- *School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia; and Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Alexander P Sobinoff
- *School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia; and Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Barbara A Fraser
- *School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia; and Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Kate A Redgrove
- *School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia; and Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Tara-Lynne Davidson
- *School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia; and Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Nicole A Siddall
- *School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia; and Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Peter Koopman
- *School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia; and Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Gary R Hime
- *School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia; and Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Eileen A McLaughlin
- *School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia; and Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
71
|
Zearfoss NR, Deveau LM, Clingman CC, Schmidt E, Johnson ES, Massi F, Ryder SP. A conserved three-nucleotide core motif defines Musashi RNA binding specificity. J Biol Chem 2014; 289:35530-41. [PMID: 25368328 DOI: 10.1074/jbc.m114.597112] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Musashi (MSI) family proteins control cell proliferation and differentiation in many biological systems. They are overexpressed in tumors of several origins, and their expression level correlates with poor prognosis. MSI proteins control gene expression by binding RNA and regulating its translation. They contain two RNA recognition motif (RRM) domains, which recognize a defined sequence element. The relative contribution of each nucleotide to the binding affinity and specificity is unknown. We analyzed the binding specificity of three MSI family RRM domains using a quantitative fluorescence anisotropy assay. We found that the core element driving recognition is the sequence UAG. Nucleotides outside of this motif have a limited contribution to binding free energy. For mouse MSI1, recognition is determined by the first of the two RRM domains. The second RRM adds affinity but does not contribute to binding specificity. In contrast, the recognition element for Drosophila MSI is more extensive than the mouse homolog, suggesting functional divergence. The short nature of the binding determinant suggests that protein-RNA affinity alone is insufficient to drive target selection by MSI family proteins.
Collapse
Affiliation(s)
- N Ruth Zearfoss
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Laura M Deveau
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Carina C Clingman
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Eric Schmidt
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Emily S Johnson
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Francesca Massi
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Sean P Ryder
- From the Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| |
Collapse
|
72
|
Clingman CC, Deveau LM, Hay SA, Genga RM, Shandilya SMD, Massi F, Ryder SP. Allosteric inhibition of a stem cell RNA-binding protein by an intermediary metabolite. eLife 2014; 3. [PMID: 24935936 PMCID: PMC4094780 DOI: 10.7554/elife.02848] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 06/15/2014] [Indexed: 01/22/2023] Open
Abstract
Gene expression and metabolism are coupled at numerous levels. Cells must sense and respond to nutrients in their environment, and specialized cells must synthesize metabolic products required for their function. Pluripotent stem cells have the ability to differentiate into a wide variety of specialized cells. How metabolic state contributes to stem cell differentiation is not understood. In this study, we show that RNA-binding by the stem cell translation regulator Musashi-1 (MSI1) is allosterically inhibited by 18-22 carbon ω-9 monounsaturated fatty acids. The fatty acid binds to the N-terminal RNA Recognition Motif (RRM) and induces a conformational change that prevents RNA association. Musashi proteins are critical for development of the brain, blood, and epithelium. We identify stearoyl-CoA desaturase-1 as a MSI1 target, revealing a feedback loop between ω-9 fatty acid biosynthesis and MSI1 activity. We propose that other RRM proteins could act as metabolite sensors to couple gene expression changes to physiological state.
Collapse
Affiliation(s)
- Carina C Clingman
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, United States
| | - Laura M Deveau
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, United States
| | - Samantha A Hay
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, United States
| | - Ryan M Genga
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, United States
| | - Shivender M D Shandilya
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, United States
| | - Francesca Massi
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, United States
| | - Sean P Ryder
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, United States
| |
Collapse
|
73
|
Hahnel S, Quack T, Parker-Manuel SJ, Lu Z, Vanderstraete M, Morel M, Dissous C, Cailliau K, Grevelding CG. Gonad RNA-specific qRT-PCR analyses identify genes with potential functions in schistosome reproduction such as SmFz1 and SmFGFRs. Front Genet 2014; 5:170. [PMID: 24959172 PMCID: PMC4050651 DOI: 10.3389/fgene.2014.00170] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 05/21/2014] [Indexed: 01/15/2023] Open
Abstract
In the search for new strategies to fight schistosomiasis, the unique reproductive biology of Schistosoma mansoni has come into the focus of research. The development of the gonads and the ability of egg production are fundamental not only for continuing the life cycle but also for pathogenicity. Previous studies of schistosome biology demonstrated an influence of pairing on gonad development of the female and on gene expression profiles in both genders. Due to the limited access to specific tissues, however, most of these studies were done at the level of whole worms neglecting individual tissues that may be targets of pairing-dependent processes. Recently, we established a protocol allowing the isolation of testes and ovaries from adult S. mansoni. Here, we describe tissue-specific qRT-PCR analyses comparing transcript levels of selected genes on the basis of RNA from gonads and whole worms. Gene expression in ovary and testes was in some cases found to be significantly influenced by pairing, which was not traceable in whole worms. Among the candidate genes identified as regulated by pairing in gonads were the frizzled homolog SmFz1 and the two fibroblast growth factor receptor homologs SmFGFR-A and SmFGFR-B. First functional characterizations were done, including comparative qRT-PCR analyses, in situ-localization experiments, heterologous expression in Xenopus oocytes (SmFGFR-A/B), and inhibitor studies using the Fz/Dvl-pathway inhibitor 3289-8625, or BIBF1120 blocking FGFR-signaling. Besides confirming gonad localization and receptor functions, inhibitor-induced phenotypes were observed in vitro such as decreased egg production as well as drastic effects on gonad differentiation, morphology, embryogenesis, and survival of adult worms. In summary, these results emphasise the usefulness of tissue-specific qRT-PCRs for selection of candidate genes with important roles in reproduction, allowing subsequent studies to determine their suitability as drug targets.
Collapse
Affiliation(s)
- Steffen Hahnel
- Biologisch-Medizinisches Forschungszentrum Seltersberg, Institute of Parasitology, Justus-Liebig-University Giessen, Germany
| | - Thomas Quack
- Biologisch-Medizinisches Forschungszentrum Seltersberg, Institute of Parasitology, Justus-Liebig-University Giessen, Germany
| | - Sophia J Parker-Manuel
- Biologisch-Medizinisches Forschungszentrum Seltersberg, Institute of Parasitology, Justus-Liebig-University Giessen, Germany
| | - Zhigang Lu
- Biologisch-Medizinisches Forschungszentrum Seltersberg, Institute of Parasitology, Justus-Liebig-University Giessen, Germany
| | - Mathieu Vanderstraete
- CIIL - Center of Infection and Immunity of Lille, CNRS-UMR 8204, INSERM U1019, Institut Pasteur de Lille, Université Lille Nord de France Lille Cedex, France
| | - Marion Morel
- CIIL - Center of Infection and Immunity of Lille, CNRS-UMR 8204, INSERM U1019, Institut Pasteur de Lille, Université Lille Nord de France Lille Cedex, France
| | - Colette Dissous
- CIIL - Center of Infection and Immunity of Lille, CNRS-UMR 8204, INSERM U1019, Institut Pasteur de Lille, Université Lille Nord de France Lille Cedex, France
| | - Katia Cailliau
- Laboratoire de Régulation des Signaux de Division, EA 4479, IFR 147, Université Lille 1 Sciences et Technology, Villeneuve d'Ascq Cedex France
| | - Christoph G Grevelding
- Biologisch-Medizinisches Forschungszentrum Seltersberg, Institute of Parasitology, Justus-Liebig-University Giessen, Germany
| |
Collapse
|
74
|
Sutherland JM, Fraser BA, Sobinoff AP, Pye VJ, Davidson TL, Siddall NA, Koopman P, Hime GR, McLaughlin EA. Developmental Expression of Musashi-1 and Musashi-2 RNA-Binding Proteins During Spermatogenesis: Analysis of the Deleterious Effects of Dysregulated Expression1. Biol Reprod 2014; 90:92. [DOI: 10.1095/biolreprod.113.115261] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
75
|
Takahashi K, Kotani T, Katsu Y, Yamashita M. Possible involvement of insulin-like growth factor 2 mRNA-binding protein 3 in zebrafish oocyte maturation as a novel cyclin B1 mRNA-binding protein that represses the translation in immature oocytes. Biochem Biophys Res Commun 2014; 448:22-7. [DOI: 10.1016/j.bbrc.2014.04.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 04/06/2014] [Indexed: 01/10/2023]
|
76
|
Cragle C, MacNicol AM. Musashi protein-directed translational activation of target mRNAs is mediated by the poly(A) polymerase, germ line development defective-2. J Biol Chem 2014; 289:14239-51. [PMID: 24644291 DOI: 10.1074/jbc.m114.548271] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The mRNA-binding protein, Musashi, has been shown to regulate translation of select mRNAs and to control cellular identity in both stem cells and cancer cells. Within the mammalian cells, Musashi has traditionally been characterized as a repressor of translation. However, we have demonstrated that Musashi is an activator of translation in progesterone-stimulated oocytes of the frog Xenopus laevis, and recent evidence has revealed Musashi's capability to function as an activator of translation in mammalian systems. The molecular mechanism by which Musashi directs activation of target mRNAs has not been elucidated. Here, we report a specific association of Musashi with the noncanonical poly(A) polymerase germ line development defective-2 (GLD2) and map the association domain to 31 amino acids within the C-terminal domain of Musashi. We show that loss of GLD2 interaction through deletion of the binding domain or treatment with antisense oligonucleotides compromises Musashi function. Additionally, we demonstrate that overexpression of both Musashi and GLD2 significantly enhances Musashi function. Finally, we report a similar co-association also occurs between murine Musashi and GLD2 orthologs, suggesting that coupling of Musashi to the polyadenylation apparatus is a conserved mechanism to promote target mRNA translation.
Collapse
Affiliation(s)
- Chad Cragle
- From the Interdiciplinary Biomedical Sciences, Departments of Neurobiology and Developmental Sciences
| | - Angus M MacNicol
- Departments of Neurobiology and Developmental Sciences, Physiology and Biophysics, and Genetics, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas 722205
| |
Collapse
|
77
|
Hadziselimovic N, Vukojevic V, Peter F, Milnik A, Fastenrath M, Fenyves B, Hieber P, Demougin P, Vogler C, de Quervain DF, Papassotiropoulos A, Stetak A. Forgetting Is Regulated via Musashi-Mediated Translational Control of the Arp2/3 Complex. Cell 2014; 156:1153-1166. [DOI: 10.1016/j.cell.2014.01.054] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 11/27/2013] [Accepted: 01/17/2014] [Indexed: 01/21/2023]
|
78
|
Gene structure and spatio-temporal expression of chicken LPIN2. Mol Biol Rep 2014; 41:4081-91. [PMID: 24562627 DOI: 10.1007/s11033-014-3278-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Accepted: 02/13/2014] [Indexed: 10/25/2022]
Abstract
LPIN2 is one of the members of the Lipin family, which acts as a phosphatidate phosphatase enzyme. In this study, we identified the cDNA sequence and exonic variants of chicken LPIN2, and evaluated its spatio-temporal expression patterns. It indicated that chicken LPIN2 cDNA contained a 2,664-bp open reading frame flanked by a 176-bp 5' untranslated region and a 429-bp 3' untranslated region, predicted encoding one protein of 886 amino acids. Fourteen variants (three missense mutations) were detected from the coding region of chicken LPIN2. W265L was predicted to affect the gene function (p < 0.01) and eight synonymous mutations were predicted to affect the binding sites of SR proteins, which suggested the important functions of these variants. Real-time quantitative PCR revealed that LPIN2 in two genotypic chickens (LD and HB chickens, with difference in growth rate) presented similar tissue expression patterns, which was liver and ovary enriched with low abundance in skeleton muscles. Chicken LPIN2 exhibited tissue-specific temporal-expression patterns during postnatal development (0-16 weeks). Chicken cutaneous LPIN2 was in steady-state mRNA levels during postnatal development; chicken LPIN2 mRNA in pectoralis major had a prominent level at 0 week-old, then dropped dramatically at 4 week-old and maintained a relatively low level through 4-16 weeks; while chicken hepatic LPIN2 had a relatively high expression at 0 week-old, with a relatively low level through 4-12 weeks and a slight increase at 16 week-old. The studies about the basic gene features of chicken LPIN2 would lay the foundation for further exploring its biological function.
Collapse
|
79
|
Rutledge CE, Lau HT, Mangan H, Hardy LL, Sunnotel O, Guo F, MacNicol AM, Walsh CP, Lees-Murdock DJ. Efficient translation of Dnmt1 requires cytoplasmic polyadenylation and Musashi binding elements. PLoS One 2014; 9:e88385. [PMID: 24586322 PMCID: PMC3930535 DOI: 10.1371/journal.pone.0088385] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 01/06/2014] [Indexed: 12/14/2022] Open
Abstract
Regulation of DNMT1 is critical for epigenetic control of many genes and for genome stability. Using phylogenetic analysis we characterized a block of 27 nucleotides in the 3′UTR of Dnmt1 mRNA identical between humans and Xenopus and investigated the role of the individual elements contained within it. This region contains a cytoplasmic polyadenylation element (CPE) and a Musashi binding element (MBE), with CPE binding protein 1 (CPEB1) known to bind to the former in mouse oocytes. The presence of these elements usually indicates translational control by elongation and shortening of the poly(A) tail in the cytoplasm of the oocyte and in some somatic cell types. We demonstrate for the first time cytoplasmic polyadenylation of Dnmt1 during periods of oocyte growth in mouse and during oocyte activation in Xenopus. Furthermore we show by RNA immunoprecipitation that Musashi1 (MSI1) binds to the MBE and that this element is required for polyadenylation in oocytes. As well as a role in oocytes, site-directed mutagenesis and reporter assays confirm that mutation of either the MBE or CPE reduce DNMT1 translation in somatic cells, but likely act in the same pathway: deletion of the whole conserved region has more severe effects on translation in both ES and differentiated cells. In adult cells lacking MSI1 there is a greater dependency on the CPE, with depletion of CPEB1 or CPEB4 by RNAi resulting in substantially reduced levels of endogenous DNMT1 protein and concurrent upregulation of the well characterised CPEB target mRNA cyclin B1. Our findings demonstrate that CPE- and MBE-mediated translation regulate DNMT1 expression, representing a novel mechanism of post-transcriptional control for this gene.
Collapse
Affiliation(s)
- Charlotte E. Rutledge
- Transcriptional Regulation and Epigenetics Research Group, School of Biomedical Sciences, University of Ulster, Coleraine, North Ireland, United Kingdom
| | - Ho-Tak Lau
- Transcriptional Regulation and Epigenetics Research Group, School of Biomedical Sciences, University of Ulster, Coleraine, North Ireland, United Kingdom
| | - Hazel Mangan
- Transcriptional Regulation and Epigenetics Research Group, School of Biomedical Sciences, University of Ulster, Coleraine, North Ireland, United Kingdom
| | - Linda L. Hardy
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Olaf Sunnotel
- Transcriptional Regulation and Epigenetics Research Group, School of Biomedical Sciences, University of Ulster, Coleraine, North Ireland, United Kingdom
| | - Fan Guo
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Angus M. MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Colum P. Walsh
- Transcriptional Regulation and Epigenetics Research Group, School of Biomedical Sciences, University of Ulster, Coleraine, North Ireland, United Kingdom
| | - Diane J. Lees-Murdock
- Transcriptional Regulation and Epigenetics Research Group, School of Biomedical Sciences, University of Ulster, Coleraine, North Ireland, United Kingdom
- * E-mail:
| |
Collapse
|
80
|
Park SM, Deering RP, Lu Y, Tivnan P, Lianoglou S, Al-Shahrour F, Ebert BL, Hacohen N, Leslie C, Daley GQ, Lengner CJ, Kharas MG. Musashi-2 controls cell fate, lineage bias, and TGF-β signaling in HSCs. ACTA ACUST UNITED AC 2014; 211:71-87. [PMID: 24395885 PMCID: PMC3892968 DOI: 10.1084/jem.20130736] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Musashi-2 is an important regulator of the hematopoietic stem cell translatome and balances HSC homeostasis and lineage bias. Hematopoietic stem cells (HSCs) are maintained through the regulation of symmetric and asymmetric cell division. We report that conditional ablation of the RNA-binding protein Msi2 results in a failure of HSC maintenance and engraftment caused by a loss of quiescence and increased commitment divisions. Contrary to previous studies, we found that these phenotypes were independent of Numb. Global transcriptome profiling and RNA target analysis uncovered Msi2 interactions at multiple nodes within pathways that govern RNA translation, stem cell function, and TGF-β signaling. Msi2-null HSCs are insensitive to TGF-β–mediated expansion and have decreased signaling output, resulting in a loss of myeloid-restricted HSCs and myeloid reconstitution. Thus, Msi2 is an important regulator of the HSC translatome and balances HSC homeostasis and lineage bias.
Collapse
Affiliation(s)
- Sun-Mi Park
- Molecular Pharmacology and Chemistry Program, 2 Center for Cell Engineering, and 3 Computational Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Charlesworth A, Meijer HA, de Moor CH. Specificity factors in cytoplasmic polyadenylation. WILEY INTERDISCIPLINARY REVIEWS-RNA 2014; 4:437-61. [PMID: 23776146 PMCID: PMC3736149 DOI: 10.1002/wrna.1171] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Revised: 04/08/2013] [Accepted: 04/09/2013] [Indexed: 12/12/2022]
Abstract
Poly(A) tail elongation after export of an messenger RNA (mRNA) to the cytoplasm is called cytoplasmic polyadenylation. It was first discovered in oocytes and embryos, where it has roles in meiosis and development. In recent years, however, has been implicated in many other processes, including synaptic plasticity and mitosis. This review aims to introduce cytoplasmic polyadenylation with an emphasis on the factors and elements mediating this process for different mRNAs and in different animal species. We will discuss the RNA sequence elements mediating cytoplasmic polyadenylation in the 3' untranslated regions of mRNAs, including the CPE, MBE, TCS, eCPE, and C-CPE. In addition to describing the role of general polyadenylation factors, we discuss the specific RNA binding protein families associated with cytoplasmic polyadenylation elements, including CPEB (CPEB1, CPEB2, CPEB3, and CPEB4), Pumilio (PUM2), Musashi (MSI1, MSI2), zygote arrest (ZAR2), ELAV like proteins (ELAVL1, HuR), poly(C) binding proteins (PCBP2, αCP2, hnRNP-E2), and Bicaudal C (BICC1). Some emerging themes in cytoplasmic polyadenylation will be highlighted. To facilitate understanding for those working in different organisms and fields, particularly those who are analyzing high throughput data, HUGO gene nomenclature for the human orthologs is used throughout. Where human orthologs have not been clearly identified, reference is made to protein families identified in man.
Collapse
Affiliation(s)
- Amanda Charlesworth
- Department of Integrative Biology, University of Colorado Denver, Denver, CO, USA
| | | | | |
Collapse
|
82
|
Mohindra V, Tripathi RK, Singh RK, Lal KK. Molecular characterization and expression analysis of three hypoxia-inducible factor alpha subunits, HIF-1α, -2α and -3α in hypoxia-tolerant Indian catfish, Clarias batrachus [Linnaeus, 1758]. Mol Biol Rep 2013; 40:5805-15. [PMID: 24065526 DOI: 10.1007/s11033-013-2685-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 09/14/2013] [Indexed: 01/01/2023]
Abstract
The present study aimed at characterization of three HIF-α subunits, HIF-1α -2α and -3α from hypoxia-tolerant Clarias batrachus, as well as to elucidate their expression pattern under short and long-term hypoxic conditions and identification of biomarker candidate. The complete cDNAs of HIF-1α, -2α and -3α were 2,833, 4,270 and 3,256 bp in length, encoding 774, 818 and 628 amino acid residues, respectively. In C. batrachus, HIF-α subunits were structurally similar in DNA binding, dimerization, degradation and transcriptional activation domains, but differed in their oxygen-dependent degradation domains. Presence of c-Jun N-terminal kinase binding domain in HIF-α subunits was reported here for the first time in fish. In adult C. batrachus, three HIF-α mRNAs were detected in different tissues under normoxic conditions, however HIF-1α was highly expressed in all the tissues studied, in comparison to HIF-2α and -3α. Short-term hypoxia exposure caused significant increase in three HIF-α transcripts in brain, liver and head kidney, while after long-term hypoxia exposure, significant up-regulation of HIF-1α in spleen and -2α in muscle was observed and HIF-3α significantly down-regulated in head kidney. These observations suggest that the differential expression of HIF-α subunits in C. batrachus was hypoxic time period dependent and may play specialized roles in adaptive response to hypoxia. HIF-2α, with its highly elevated expression in muscle tissues, can be a robust biomarker candidate for exposure to hypoxic environment.
Collapse
Affiliation(s)
- Vindhya Mohindra
- National Bureau of Fish Genetic Resources (ICAR), Canal Ring Road, P.O. Dilkusha, Lucknow, 226002, UP, India,
| | | | | | | |
Collapse
|
83
|
Sutherland JM, McLaughlin EA, Hime GR, Siddall NA. The Musashi family of RNA binding proteins: master regulators of multiple stem cell populations. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 786:233-45. [PMID: 23696360 DOI: 10.1007/978-94-007-6621-1_13] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
In order to maintain their unlimited capacity to divide, stem cells require controlled temporal and spatial protein expression. The Musashi family of RNA-binding proteins have been shown to exhibit this necessary translational control through both repression and activation in order to regulate multiple stem cell populations. This chapter looks in depth at the initial discovery and characterisation of Musashi in the model organism Drosophila, and its subsequent emergence as a master regulator in a number of stem cell populations. Furthermore the unique roles for mammalian Musashi-1 and Musashi-2 in different stem cell types are correlated with the perceived diagnostic power of Musashi expression in specific stem cell derived oncologies. In particular the potential role for Musashi in the identification and treatment of human cancer is considered, with a focus on the role of Musashi-2 in leukaemia. Finally, the manipulation of Musashi expression is proposed as a potential avenue towards the targeted treatment of specific aggressive stem cell cancers.
Collapse
Affiliation(s)
- Jessie M Sutherland
- Priority Research Centre in Reproductive Science, School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, 2308, Australia
| | | | | | | |
Collapse
|
84
|
Zar1 represses translation in Xenopus oocytes and binds to the TCS in maternal mRNAs with different characteristics than Zar2. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2013; 1829:1034-46. [PMID: 23827238 DOI: 10.1016/j.bbagrm.2013.06.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 06/20/2013] [Accepted: 06/24/2013] [Indexed: 12/23/2022]
Abstract
Maternal mRNAs are translationally regulated during early development. Zar1 and its closely related homolog, Zar2, are both crucial in early development. Xenopus laevis Zygote arrest 2 (Zar2) binds to the Translational Control Sequence (TCS) in maternal mRNAs and regulates translation. The molecular mechanism of Zar1 has not been described. Here we report similarities and differences between Xenopus Zar1 and Zar2. Analysis of Zar sequences in vertebrates revealed two Zar family members with conserved, characteristic amino acid differences in the C-terminal domain. The presence of only two vertebrate Zar proteins was supported by analyzing Zar1 synteny. We propose that the criteria for naming Zar sequences are based on the characteristic amino acids and the chromosomal context. We also propose reclassification of some Zar sequences. We found that Zar1 is expressed throughout oogenesis and is stable during oocyte maturation. The N-terminal domain of Zar1 repressed translation of a reporter construct in immature oocytes. Both Zar1 and Zar2 bound to the TCS in the Wee1 and Mos 3' UTRs using a zinc finger in the C-terminal domain. However, Zar1 had much higher affinity for RNA than Zar2. To show the functional significance of the conserved amino acid substitutions, these residues in Zar2 were mutated to those found in Zar1. We show that these residues contributed to the different RNA binding characteristics of Zar1 compared to Zar2. Our study shows that Zar proteins have generally similar molecular functions in the translational regulation of maternal mRNAs, but they may have different roles in early development.
Collapse
|
85
|
Phillips CD, Butler B, Fondon JW, Mantilla-Meluk H, Baker RJ. Contrasting evolutionary dynamics of the developmental regulator PAX9, among bats, with evidence for a novel post-transcriptional regulatory mechanism. PLoS One 2013; 8:e57649. [PMID: 23469040 PMCID: PMC3585407 DOI: 10.1371/journal.pone.0057649] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 01/23/2013] [Indexed: 11/18/2022] Open
Abstract
Morphological evolution can be the result of natural selection favoring modification of developmental signaling pathways. However, little is known about the genetic basis of such phenotypic diversity. Understanding these mechanisms is difficult for numerous reasons, yet studies in model organisms often provide clues about the major developmental pathways involved. The paired-domain gene, PAX9, is known to be a key regulator of development, particularly of the face and teeth. In this study, using a comparative genetics approach, we investigate PAX9 molecular evolution among mammals, focusing on craniofacially diversified (Phyllostomidae) and conserved (Vespertilionidae) bat families, and extend our comparison to other orders of mammal. Open-reading frame analysis disclosed signatures of selection, in which a small percentage of residues vary, and lineages acquire different combinations of variation through recurrent substitution and lineage specific changes. A few instances of convergence for specific residues were observed between morphologically convergent bat lineages. Bioinformatic analysis for unknown PAX9 regulatory motifs indicated a novel post-transcriptional regulatory mechanism involving a Musashi protein. This regulation was assessed through fluorescent reporter assays and gene knockdowns. Results are compatible with the hypothesis that the number of Musashi binding-elements in PAX9 mRNA proportionally regulates protein translation rate. Although a connection between morphology and binding element frequency was not apparent, results indicate this regulation would vary among craniofacially divergent bat species, but be static among conserved species. Under this model, Musashi's regulatory control of alternative human PAX9 isoforms would also vary. The presence of Musashi-binding elements within PAX9 of all mammals examined, chicken, zebrafish, and the fly homolog of PAX9, indicates this regulatory mechanism is ancient, originating basal to much of the animal phylogeny.
Collapse
Affiliation(s)
- Caleb D Phillips
- Department of Biological Sciences, Texas Tech University, Lubbock, Texas, United States of America.
| | | | | | | | | |
Collapse
|
86
|
Mohindra V, Tripathi RK, Singh A, Singh B. Molecular characterization and expression analysis of a novel cystatin-like gene in a hypoxia-tolerant Indian catfish, Clarias batrachus [Linnaeus, 1758]. FISH & SHELLFISH IMMUNOLOGY 2013; 34:683-687. [PMID: 23246809 DOI: 10.1016/j.fsi.2012.11.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 11/08/2012] [Accepted: 11/09/2012] [Indexed: 06/01/2023]
Abstract
A novel member of Cystatin superfamily was identified from Indian catfish, Clarias batrachus, in response to oxidation stress induced by environmental hypoxia. Integrated genomic approaches, expression profiling and computational techniques showed that CbCystatin had putative cystatin/monelin like domain and might be a transmembrane and/or intermediate protein in signaling pathways. CbCystatin was found to be clustered into family 2 Cystatins. At transcriptional level, its expression was significantly up-regulated in response to short as well as long periods (more than 20 fold) of hypoxia, suggesting its positive association with oxygen concentrations lower than physiological concentrations.
Collapse
Affiliation(s)
- Vindhya Mohindra
- National Bureau of Fish Genetic Resources (ICAR), Lucknow, UP, India.
| | | | | | | |
Collapse
|
87
|
Takahashi T, Suzuki H, Imai T, Shibata S, Tabuchi Y, Tsuchimoto K, Okano H, Hibi T. Musashi-1 post-transcriptionally enhances phosphotyrosine-binding domain-containing m-Numb protein expression in regenerating gastric mucosa. PLoS One 2013; 8:e53540. [PMID: 23308249 PMCID: PMC3537613 DOI: 10.1371/journal.pone.0053540] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 11/30/2012] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVE Upregulation of the RNA-binding protein Musashi-1 (Msi1) has been shown to occur in rat gastric corpus mucosa after ethanol-induced mucosal injury. However, there is no direct evidence linking Msi1 with gastric regeneration. We examined the process of tissue repair after acute gastric mucosal injury with Msi1-knock-out (KO) mice to clarify the role of Msi1 and Msi1-dependent regulation of m-Numb expression in regenerating gastric mucosa. METHODS Acute gastric injury was induced in Msi1-KO and wild-type ICR mice by administering absolute ethanol. Expression of the splicing variants of m-Numb mRNA and protein in the gastric mucosa were analyzed by quantitative RT-PCR and western blotting, respectively. RESULTS We demonstrated that phosphotyrosine-binding domain-containing m-Numb expression was significantly upregulated at both the mRNA and protein levels in wild-type mice at 3 h after ethanol-induced acute gastric injury. In contrast, in Msi1-KO mice, the m-Numb protein was expressed weakly, and was associated with delayed regeneration of the injured gastric mucosal epithelium. In the Msi1-KO mouse, the ratio of m-Numb mRNA to total m-Numb mRNA in the heavy polysome fractions was lower than that in the wild-type mouse. Further, we showed that m-Numb-enhancement in gastric mucous cells induced the expression of prostate stem cell antigen and metallothionein-2. Under the m-Numb enhancing condition, the gastric cells exhibited enhanced cell proliferation and were significantly more resistant to H(2)O(2)-induced cell death than control cells. CONCLUSIONS Msi1-dependent post-transcriptional enhancement of m-Numb is crucial in gastric epithelial regeneration.
Collapse
Affiliation(s)
- Tetsufumi Takahashi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
- Laboratory of Pathophysiology, Division of Clinical Medicine, School of Pharmacy, Kitasato University, Tokyo, Japan
| | - Hidekazu Suzuki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
- * E-mail:
| | - Takao Imai
- Department of Physiology, School of Medicine, Keio University, Tokyo, Japan
| | - Shinsuke Shibata
- Department of Physiology, School of Medicine, Keio University, Tokyo, Japan
| | - Yoshiaki Tabuchi
- Division of Molecular Genetics, Life Science Research Center, University of Toyama, Toyama, Japan
| | - Kanji Tsuchimoto
- Laboratory of Pathophysiology, Division of Clinical Medicine, School of Pharmacy, Kitasato University, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, School of Medicine, Keio University, Tokyo, Japan
| | - Toshifumi Hibi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| |
Collapse
|
88
|
Plateroti M, de Araujo PR, da Silva AE, Penalva LOF. The RNA-Binding Protein Musashi1: A Major Player in Intestinal Epithelium Renewal and Colon Cancer Development. CURRENT COLORECTAL CANCER REPORTS 2012; 8:290-297. [PMID: 23914149 DOI: 10.1007/s11888-012-0141-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aberrant gene expression is the cause and the consequence of tumorigenesis. A major component of gene expression is translation regulation; a process whose main players are RNA-binding-proteins (RBPs). More than 800 RBPs have been identified in the human genome and several of them have been shown to control gene networks associated with relevant cancer processes. A more systematic characterization of RBPs starts to reveal that similar to transcription factors, they can function as tumor suppressors or oncogenes. A relevant example is Musashi1 (Msi1), which is emerging as a critical regulator of tumorigenesis in multiple cancer types, including colon cancer. Msi1 is a stem marker in several tissues and is critical in maintaining the balance between self-renewal and differentiation. However, a boost in Msi1 expression can most likely lead cells towards an oncogenic pathway. In this article, we discuss the parallels between Msi1 function in normal renewal of intestinal epithelium and in colon cancer.
Collapse
Affiliation(s)
- Michelina Plateroti
- Centre de Génétique et de Physiologie Moléculaire et Cellulaire, Université Claude Bernard Lyon 1, France. 16 Rue Raphael Dubois, 69622 Villeurbanne, Cedex France
| | | | | | | |
Collapse
|
89
|
Abstract
PURPOSE OF REVIEW Recent work has shown that the Musashi 2 (Msi2) gene plays important roles in normal and malignant hematopoiesis. Here, we give an overview on the role of Msi2 in the regulation and function of primitive hematopoietic cells as well as in leukaemic progression. We also discuss the molecular pathways in which Msi2 acts in both normal and leukaemic blood cells. RECENT FINDINGS Msi2 gain and loss of function experiments have shown that it plays an important role in regulating the heamatopoietic stem cell pool. Msi2 has also been found to be overexpressed in human myeloid leukaemias correlating with poor prognosis, therefore Msi2 may be considered as a prognostic marker for acute myeloid leukaemia. SUMMARY Further studies into the molecular pathways through which Msi2 modulates primitive progenitor function will provide insight into the regulation of normal haematopoiesis and a better understanding of the mechanisms governing the leukaemic transformation process. This will be crucial for the development of effective therapies.
Collapse
|
90
|
Balmaceda-Aguilera C, Martos-Sitcha J, Mancera J, Martínez‐Rodríguez G. Cloning and expression pattern of facilitative glucose transporter 1 (GLUT1) in gilthead sea bream Sparus aurata in response to salinity acclimation. Comp Biochem Physiol A Mol Integr Physiol 2012; 163:38-46. [DOI: 10.1016/j.cbpa.2012.04.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 04/03/2012] [Accepted: 04/25/2012] [Indexed: 01/05/2023]
|
91
|
Arumugam K, Macnicol MC, Macnicol AM. Autoregulation of Musashi1 mRNA translation during Xenopus oocyte maturation. Mol Reprod Dev 2012; 79:553-63. [PMID: 22730340 DOI: 10.1002/mrd.22060] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 06/04/2012] [Indexed: 11/06/2022]
Abstract
The mRNA translational control protein, Musashi, plays a critical role in cell fate determination through sequence-specific interactions with select target mRNAs. In proliferating stem cells, Musashi exerts repression of target mRNAs to promote cell cycle progression. During stem cell differentiation, Musashi target mRNAs are de-repressed and translated. Recently, we have reported an obligatory requirement for Musashi to direct translational activation of target mRNAs during Xenopus oocyte meiotic cell cycle progression. Despite the importance of Musashi in cell cycle regulation, only a few target mRNAs have been fully characterized. In this study, we report the identification and characterization of a new Musashi target mRNA in Xenopus oocytes. We demonstrate that progesterone-stimulated translational activation of the Xenopus Musashi1 mRNA is regulated through a functional Musashi binding element (MBE) in the Musashi1 mRNA 3' untranslated region (3' UTR). Mutational disruption of the MBE prevented translational activation of Musashi1 mRNA and its interaction with Musashi protein. Further, elimination of Musashi function through microinjection of inhibitory antisense oligonucleotides prevented progesterone-induced polyadenylation and translation of the endogenous Musashi1 mRNA. Thus, Xenopus Musashi proteins regulate translation of the Musashi1 mRNA during oocyte maturation. Our results indicate that the hierarchy of sequential and dependent mRNA translational control programs involved in directing progression through meiosis are reinforced by an intricate series of nested, positive feedback loops, including Musashi mRNA translational autoregulation. These autoregulatory positive feedback loops serve to amplify a weak initiating signal into a robust commitment for the oocyte to progress through the cell cycle and become competent for fertilization.
Collapse
Affiliation(s)
- Karthik Arumugam
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, 4301W Markham, Little Rock, AR 72205, USA
| | | | | |
Collapse
|
92
|
Xenopus laevis zygote arrest 2 (zar2) encodes a zinc finger RNA-binding protein that binds to the translational control sequence in the maternal Wee1 mRNA and regulates translation. Dev Biol 2012; 369:177-90. [PMID: 22732570 DOI: 10.1016/j.ydbio.2012.06.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Revised: 06/12/2012] [Accepted: 06/17/2012] [Indexed: 11/22/2022]
Abstract
Zygote arrest (Zar) proteins are crucial for early embryonic development, but their molecular mechanism of action is unknown. The Translational Control Sequence (TCS) in the 3' untranslated region (UTR) of the maternal mRNA, Wee1, mediates translational repression in immature Xenopus oocytes and translational activation in mature oocytes, but the protein that binds to the TCS and mediates translational control is not known. Here we show that Xenopus laevis Zar2 (encoded by zar2) binds to the TCS in maternal Wee1 mRNA and represses translation in immature oocytes. Using yeast 3 hybrid assays and electrophoretic mobility shift assays, Zar2 was shown to bind specifically to the TCS in the Wee1 3'UTR. RNA binding required the presence of Zn(2+) and conserved cysteines in the C-terminal domain, suggesting that Zar2 contains a zinc finger. Consistent with regulating maternal mRNAs, Zar2 was present throughout oogenesis, and endogenous Zar2 co-immunoprecipitated endogenous Wee1 mRNA from immature oocytes, demonstrating the physiological significance of the protein-RNA interaction. Interestingly, Zar2 levels decreased during oocyte maturation. Dual luciferase reporter tethered assays showed that Zar2 repressed translation in immature oocytes. Translational repression was relieved during oocyte maturation and this coincided with degradation of Zar2 during maturation. This is the first report of a molecular function of zygote arrest proteins. These data show that Zar2 contains a zinc finger and is a trans-acting factor for the TCS in maternal mRNAs in immature Xenopus oocytes.
Collapse
|
93
|
Saravanaperumal SA, Pediconi D, Renieri C, La Terza A. Skipping of exons by premature termination of transcription and alternative splicing within intron-5 of the sheep SCF gene: a novel splice variant. PLoS One 2012; 7:e38657. [PMID: 22719917 PMCID: PMC3376141 DOI: 10.1371/journal.pone.0038657] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 05/08/2012] [Indexed: 11/23/2022] Open
Abstract
Stem cell factor (SCF) is a growth factor, essential for haemopoiesis, mast cell development and melanogenesis. In the hematopoietic microenvironment (HM), SCF is produced either as a membrane-bound (-) or soluble (+) forms. Skin expression of SCF stimulates melanocyte migration, proliferation, differentiation, and survival. We report for the first time, a novel mRNA splice variant of SCF from the skin of white merino sheep via cloning and sequencing. Reverse transcriptase (RT)-PCR and molecular prediction revealed two different cDNA products of SCF. Full-length cDNA libraries were enriched by the method of rapid amplification of cDNA ends (RACE-PCR). Nucleotide sequencing and molecular prediction revealed that the primary 1519 base pair (bp) cDNA encodes a precursor protein of 274 amino acids (aa), commonly known as 'soluble' isoform. In contrast, the shorter (835 and/or 725 bp) cDNA was found to be a 'novel' mRNA splice variant. It contains an open reading frame (ORF) corresponding to a truncated protein of 181 aa (vs 245 aa) with an unique C-terminus lacking the primary proteolytic segment (28 aa) right after the D(175)G site which is necessary to produce 'soluble' form of SCF. This alternative splice (AS) variant was explained by the complete nucleotide sequencing of splice junction covering exon 5-intron (5)-exon 6 (948 bp) with a premature termination codon (PTC) whereby exons 6 to 9/10 are skipped (Cassette Exon, CE 6-9/10). We also demonstrated that the Northern blot analysis at transcript level is mediated via an intron-5 splicing event. Our data refine the structure of SCF gene; clarify the presence (+) and/or absence (-) of primary proteolytic-cleavage site specific SCF splice variants. This work provides a basis for understanding the functional role and regulation of SCF in hair follicle melanogenesis in sheep beyond what was known in mice, humans and other mammals.
Collapse
Affiliation(s)
| | - Dario Pediconi
- School of Environmental Sciences, University of Camerino, via Gentile III da Varano, Camerino (MC), Italy
| | - Carlo Renieri
- School of Environmental Sciences, University of Camerino, via Gentile III da Varano, Camerino (MC), Italy
| | - Antonietta La Terza
- School of Environmental Sciences, University of Camerino, via Gentile III da Varano, Camerino (MC), Italy
| |
Collapse
|
94
|
Han D, Liu XY, Jiao GZ, Liang B, He N, Gao WQ, Tan JH. Cyclin B1 turnover and the mechanism causing insensitivity of fully grown mouse oocytes to cycloheximide inhibition of meiotic resumption. Theriogenology 2012; 77:1900-10. [PMID: 22444557 DOI: 10.1016/j.theriogenology.2012.01.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 12/16/2011] [Accepted: 01/13/2012] [Indexed: 11/18/2022]
Abstract
Cyclin B1 turnover and the insensitivity of fully-grown mouse oocytes to cycloheximide (CHX) inhibition of germinal vesicle breakdown (GVBD) were examined by assaying GVBD and cyclin B1 levels after treatment of oocytes with various combinations of eCG and CHX. Whereas over 95% of oocytes underwent GVBD after culture for 24 h with CHX alone, only 10% did so after culture with CHX + eCG (P < 0.05). In addition, preculture with eCG alone had no effect, but preculture with eCG + CHX prevented GVBD during a second culture with CHX alone. Therefore, we inferred that eCG delayed GVBD long enough for CHX inhibition of protein synthesis to allow cyclin B1 to decrease below a threshold where GVBD became dependent upon its de novo synthesis. However, western blot revealed no cyclin B1 synthesis, but cyclin B1 degradation, as long as GVs were maintained intact with eCG. Regarding the function of CHX in preculture without protein synthesis to block subsequent GVBD, whereas eCG delayed GVBD for only 3 h, CHX had an ongoing effect that further postponed GVBD, thus allowing cyclin B1 to decrease below the threshold. When oocytes precultured with eCG + CHX were further cultured without eCG and CHX, cyclin B1 first decreased but then, because of the ongoing effects of CHX, increased to a level sufficient to induce GVBD. The content of P34Cdc2 was not altered under any of the culture conditions (P > 0.05). We concluded that insensitivity of mouse germinal vesicle (GV) oocytes to CHX was due to the presence of sufficient cyclin B1, and that cyclin B1 level in such oocytes was maintained by an equilibrium between synthesis and degradation.
Collapse
Affiliation(s)
- Dong Han
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai-an City, China
| | | | | | | | | | | | | |
Collapse
|
95
|
Muto J, Imai T, Ogawa D, Nishimoto Y, Okada Y, Mabuchi Y, Kawase T, Iwanami A, Mischel PS, Saya H, Yoshida K, Matsuzaki Y, Okano H. RNA-binding protein Musashi1 modulates glioma cell growth through the post-transcriptional regulation of Notch and PI3 kinase/Akt signaling pathways. PLoS One 2012; 7:e33431. [PMID: 22428049 PMCID: PMC3299785 DOI: 10.1371/journal.pone.0033431] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 02/08/2012] [Indexed: 12/11/2022] Open
Abstract
Musashi1 (MSI1) is an RNA-binding protein that plays critical roles in nervous-system development and stem-cell self-renewal. Here, we examined its role in the progression of glioma. Short hairpin RNA (shRNA)-based MSI1-knock down (KD) in glioblastoma and medulloblastoma cells resulted in a significantly lower number of self renewing colony on day 30 (a 65% reduction), compared with non-silencing shRNA-treated control cells, indicative of an inhibitory effect of MSI1-KD on tumor cell growth and survival. Immunocytochemical staining of the MSI1-KD glioblastoma cells indicated that they ectopically expressed metaphase markers. In addition, a 2.2-fold increase in the number of MSI1-KD cells in the G2/M phase was observed. Thus, MSI1-KD caused the prolongation of mitosis and reduced the cell survival, although the expression of activated Caspase-3 was unaltered. We further showed that MSI1-KD glioblastoma cells xenografted into the brains of NOD/SCID mice formed tumors that were 96.6% smaller, as measured by a bioluminescence imaging system (BLI), than non-KD cells, and the host survival was longer (49.3±6.1 days vs. 33.6±3.6 days; P<0.01). These findings and other cell biological analyses suggested that the reduction of MSI1 in glioma cells prolonged the cell cycle by inducing the accumulation of Cyclin B1. Furthermore, MSI1-KD reduced the activities of the Notch and PI3 kinase-Akt signaling pathways, through the up-regulation of Numb and PTEN, respectively. Exposure of glioma cells to chemical inhibitors of these pathways reduced the number of spheres and living cells, as did MSI1-KD. These results suggest that MSI1 increases the growth and/or survival of certain types of glioma cells by promoting the activation of both Notch and PI3 kinase/Akt signaling.
Collapse
Affiliation(s)
- Jun Muto
- Department of Physiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
- Division of Neurosurgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Takao Imai
- Department of Physiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Daisuke Ogawa
- Department of Physiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Yoshinori Nishimoto
- Department of Physiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Yohei Okada
- Department of Physiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Yo Mabuchi
- Department of Physiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Takeshi Kawase
- Division of Neurosurgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Akio Iwanami
- Pathology and Laboratory Medicine, David Geffen UCLA School of Medicine, Los Angeles, California, United States of America
| | - Paul S. Mischel
- Pathology and Laboratory Medicine, David Geffen UCLA School of Medicine, Los Angeles, California, United States of America
| | - Hideyuki Saya
- Division of Gene regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Kazunari Yoshida
- Division of Neurosurgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Yumi Matsuzaki
- Department of Physiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
96
|
Kronja I, Orr-Weaver TL. Translational regulation of the cell cycle: when, where, how and why? Philos Trans R Soc Lond B Biol Sci 2012; 366:3638-52. [PMID: 22084390 DOI: 10.1098/rstb.2011.0084] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Translational regulation contributes to the control of archetypal and specialized cell cycles, such as the meiotic and early embryonic cycles. Late meiosis and early embryogenesis unfold in the absence of transcription, so they particularly rely on translational repression and activation of stored maternal mRNAs. Here, we present examples of cell cycle regulators that are translationally controlled during different cell cycle and developmental transitions in model organisms ranging from yeast to mouse. Our focus also is on the RNA-binding proteins that affect cell cycle progression by recognizing special features in untranslated regions of mRNAs. Recent research highlights the significance of the cytoplasmic polyadenylation element-binding protein (CPEB). CPEB determines polyadenylation status, and consequently translational efficiency, of its target mRNAs in both transcriptionally active somatic cells as well as in transcriptionally silent mature Xenopus oocytes and early embryos. We discuss the role of CPEB in mediating the translational timing and in some cases spindle-localized translation of critical regulators of Xenopus oogenesis and early embryogenesis. We conclude by outlining potential directions and approaches that may provide further insights into the translational control of the cell cycle.
Collapse
Affiliation(s)
- Iva Kronja
- Whitehead Institute and Department of Biology, Massachusetts Institute of Technology, Nine Cambridge Center, Cambridge, MA 02142, USA
| | | |
Collapse
|
97
|
Arumugam K, MacNicol MC, Wang Y, Cragle CE, Tackett AJ, Hardy LL, MacNicol AM. Ringo/cyclin-dependent kinase and mitogen-activated protein kinase signaling pathways regulate the activity of the cell fate determinant Musashi to promote cell cycle re-entry in Xenopus oocytes. J Biol Chem 2012; 287:10639-10649. [PMID: 22215682 DOI: 10.1074/jbc.m111.300681] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Cell cycle re-entry during vertebrate oocyte maturation is mediated through translational activation of select target mRNAs, culminating in the activation of mitogen-activated protein kinase and cyclin B/cyclin-dependent kinase (CDK) signaling. The temporal order of targeted mRNA translation is crucial for cell cycle progression and is determined by the timing of activation of distinct mRNA-binding proteins. We have previously shown in oocytes from Xenopus laevis that the mRNA-binding protein Musashi targets translational activation of early class mRNAs including the mRNA encoding the Mos proto-oncogene. However, the molecular mechanism by which Musashi function is activated is unknown. We report here that activation of Musashi1 is mediated by Ringo/CDK signaling, revealing a novel role for early Ringo/CDK function. Interestingly, Musashi1 activation is subsequently sustained through mitogen-activated protein kinase signaling, the downstream effector of Mos mRNA translation, thus establishing a positive feedback loop to amplify Musashi function. The identified regulatory sites are present in mammalian Musashi proteins, and our data suggest that phosphorylation may represent an evolutionarily conserved mechanism to control Musashi-dependent target mRNA translation.
Collapse
Affiliation(s)
- Karthik Arumugam
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas 722205
| | - Melanie C MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas 722205; Center for Translational Neuroscience, University of Arkansas for Medical Sciences, Little Rock, Arkansas 722205
| | - Yiying Wang
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas 722205
| | - Chad E Cragle
- Interdisciplinary BioSciences Graduate Program, University of Arkansas for Medical Sciences, Little Rock, Arkansas 722205
| | - Alan J Tackett
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 722205
| | - Linda L Hardy
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas 722205
| | - Angus M MacNicol
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas 722205; Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas 722205; Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas 722205.
| |
Collapse
|
98
|
Clarke HJ. Post-transcriptional control of gene expression during mouse oogenesis. Results Probl Cell Differ 2012; 55:1-21. [PMID: 22918798 DOI: 10.1007/978-3-642-30406-4_1] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Post-transcriptional mechanisms play a central role in regulating gene expression during oogenesis and early embryogenesis. Growing oocytes accumulate an enormous quantity of messenger RNAs (mRNAs), but transcription decreases dramatically near the end of growth and is undetectable during meiotic maturation. Following fertilization, the embryo is initially transcriptionally inactive and then becomes active at a species-specific stage of early cleavage. Meanwhile, beginning during maturation and continuing after fertilization, the oocyte mRNAs are eliminated, allowing the embryonic genome to assume control of development. How the mammalian oocyte manages the storage, translation, and degradation of the huge quantity and diversity of mRNAs that it harbours has been the focus of enormous research effort and is the subject of this review. We discuss the roles of sequences within the 3'-untranslated region of certain mRNAs and the proteins that bind to them, sequence-non-specific RNA-binding proteins, and recent studies implicating ribonucleoprotein processing (P-) bodies and cytoplasmic lattices. We also discuss mechanisms that may control the temporally regulated translational activation of different mRNAs during meiotic maturation, as well as the signals that trigger silencing and degradation of the oocyte mRNAs. We close by highlighting areas for future research including the potential key role of small RNAs in regulating gene expression in oocytes.
Collapse
Affiliation(s)
- Hugh J Clarke
- Department of Obstetrics and Gynecology, McGill University Health Centre, Montréal, QC, Canada.
| |
Collapse
|
99
|
Mai J, Virtue A, Maley E, Tran T, Yin Y, Meng S, Pansuria M, Jiang X, Wang H, Yang XF. MicroRNAs and other mechanisms regulate interleukin-17 cytokines and receptors. Front Biosci (Elite Ed) 2012. [PMID: 22201969 DOI: 10.2741/474] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Interleukin-17 cytokines are a family of pro-inflammatory cytokines. Our current studies found: i) IL-17 cytokines are not ubiquitously expressed, but several receptors and TRAF3IP2 are ubiquitously expressed in tissues with a few exceptions; ii) heart and vascular tissue are in the second tier of readiness to respond to IL-17 cytokine stimulation; iii) alternative transcription starting sites and alternative spliced isoforms are found in IL-17 cytokine and receptor transcripts; iv) higher hypomethylation status is associated with higher expressions of IL-17 receptors; v) the binding sites of several RNA binding proteins are found in the 3'UTRs of the mRNAs of IL-17 cytokines and receptors; and vi) numerous microRNA binding sites are statistically equivalent to that of experimentally verified microRNAs-mRNA interactions in the 3'UTRs of IL-17 cytokine and receptor mRNAs. These results suggest that mechanisms including alternative promoters, alternative splicing, RNA binding proteins, and microRNAs regulate the structures and expressions of IL-17 cytokines and receptors. These results provide an insight into the roles of IL-17 in mediating inflammation and immunity.
Collapse
Affiliation(s)
- Jietang Mai
- Department of Pharmacology and Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Mai J, Virtue A, Maley E, Tran T, Yin Y, Meng S, Pansuria M, Jiang X, Wang H, Yang XF. MicroRNAs and other mechanisms regulate interleukin-17 cytokines and receptors. Front Biosci (Elite Ed) 2012; 4:1478-95. [PMID: 22201969 PMCID: PMC3289104 DOI: 10.2741/e474] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Interleukin-17 cytokines are a family of pro-inflammatory cytokines. Our current studies found: i) IL-17 cytokines are not ubiquitously expressed, but several receptors and TRAF3IP2 are ubiquitously expressed in tissues with a few exceptions; ii) heart and vascular tissue are in the second tier of readiness to respond to IL-17 cytokine stimulation; iii) alternative transcription starting sites and alternative spliced isoforms are found in IL-17 cytokine and receptor transcripts; iv) higher hypomethylation status is associated with higher expressions of IL-17 receptors; v) the binding sites of several RNA binding proteins are found in the 3'UTRs of the mRNAs of IL-17 cytokines and receptors; and vi) numerous microRNA binding sites are statistically equivalent to that of experimentally verified microRNAs-mRNA interactions in the 3'UTRs of IL-17 cytokine and receptor mRNAs. These results suggest that mechanisms including alternative promoters, alternative splicing, RNA binding proteins, and microRNAs regulate the structures and expressions of IL-17 cytokines and receptors. These results provide an insight into the roles of IL-17 in mediating inflammation and immunity.
Collapse
Affiliation(s)
- Jietang Mai
- Department of Pharmacology and Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Anthony Virtue
- Department of Pharmacology and Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Erin Maley
- Department of Pharmacology and Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Tran Tran
- Department of Pharmacology and Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Ying Yin
- Department of Pharmacology and Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Shu Meng
- Department of Pharmacology and Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Meghana Pansuria
- Department of Pharmacology and Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Xiaohua Jiang
- Department of Pharmacology and Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Hong Wang
- Department of Pharmacology and Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Xiao-Feng Yang
- Department of Pharmacology and Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| |
Collapse
|