1
|
Ghosh N, Chatterjee D, Datta A. Tumor heterogeneity and resistance in glioblastoma: the role of stem cells. Apoptosis 2025:10.1007/s10495-025-02123-y. [PMID: 40375039 DOI: 10.1007/s10495-025-02123-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2025] [Indexed: 05/18/2025]
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive and treatment-resistant brain tumor, characterized by its heterogeneity and the presence of glioblastoma stem cells (GSCs). GSCs are a subpopulation of cells within the tumor that possess self-renewal and differentiation capabilities, contributing to tumor initiation, progression, and recurrence. This review explores the unique biological properties of GSCs, including their molecular markers, signalling pathways, and interactions with the tumor microenvironment. We discuss the mechanisms by which GSCs evade conventional therapies, such as enhanced DNA repair and metabolic plasticity, which complicate treatment outcomes. Furthermore, we highlight recent advancements in identifying novel biomarkers and therapeutic targets that may improve the efficacy of treatments aimed at GSCs. The potential of targeted therapies, including immunotherapy and combination strategies, is also examined to overcome the challenges posed by GSCs. Ultimately, a deeper understanding of GSC biology is essential for developing personalized treatment approaches that can enhance patient outcomes in glioblastoma.
Collapse
Affiliation(s)
- Nikita Ghosh
- Department of Neuroscience Technology, School of Allied Health Sciences, Yenepoya, Mangalore, Karnataka, India
| | | | - Aparna Datta
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata, India.
| |
Collapse
|
2
|
Banik J, Moreira ARS, Lim J, Tomlinson S, Hardy LL, Lagasse A, Haney A, Crimmins MR, Boehm U, Odle AK, MacNicol MC, Childs GV, MacNicol AM. The Musashi RNA binding proteins direct the translational activation of key pituitary mRNAs. Sci Rep 2024; 14:5918. [PMID: 38467682 PMCID: PMC10928108 DOI: 10.1038/s41598-024-56002-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 02/29/2024] [Indexed: 03/13/2024] Open
Abstract
The pituitary functions as a master endocrine gland that secretes hormones critical for regulation of a wide variety of physiological processes including reproduction, growth, metabolism and stress responses. The distinct hormone-producing cell lineages within the pituitary display remarkable levels of cell plasticity that allow remodeling of the relative proportions of each hormone-producing cell population to meet organismal demands. The molecular mechanisms governing pituitary cell plasticity have not been fully elucidated. Our recent studies have implicated a role for the Musashi family of sequence-specific mRNA binding proteins in the control of pituitary hormone production, pituitary responses to hypothalamic stimulation and modulation of pituitary transcription factor expression in response to leptin signaling. To date, these actions of Musashi in the pituitary appear to be mediated through translational repression of the target mRNAs. Here, we report Musashi1 directs the translational activation, rather than repression, of the Prop1, Gata2 and Nr5a1 mRNAs which encode key pituitary lineage specification factors. We observe that Musashi1 further directs the translational activation of the mRNA encoding the glycolipid Neuronatin (Nnat) as determined both in mRNA reporter assays as well as in vivo. Our findings suggest a complex bifunctional role for Musashi1 in the control of pituitary cell function.
Collapse
Affiliation(s)
- Jewel Banik
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Ana Rita Silva Moreira
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Juchan Lim
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Sophia Tomlinson
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, 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
| | - Alex Lagasse
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Anessa Haney
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - Meghan R Crimmins
- Arkansas Children's Nutrition Center, Arkansas Children's Hospital, Little Rock, AR, USA
| | - Ulrich Boehm
- Department of Experimental Pharmacology, Center for Molecular Signaling, Saarland University School of Medicine, Homburg, Germany
| | - Angela K Odle
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, 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
| | - Gwen V Childs
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W Markham, Slot 814, Little Rock, AR, 72205, USA
| | - 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.
| |
Collapse
|
3
|
Haiduk TS, Sicking M, Brücksken KA, Espinoza-Sánchez NA, Eder KM, Kemper B, Eich HT, Götte M, Greve B, Troschel FM. Dysregulated Stem Cell Markers Musashi-1 and Musashi-2 are Associated with Therapy Resistance in Inflammatory Breast Cancer. Arch Med Res 2023; 54:102855. [PMID: 37481823 DOI: 10.1016/j.arcmed.2023.102855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 06/21/2023] [Accepted: 07/11/2023] [Indexed: 07/25/2023]
Abstract
BACKGROUND AND AIM While preliminary evidence points to pro-tumorigenic roles for the Musashi (MSI) RNA-binding proteins Musashi-1 (MSI1) and Musashi-2 (MSI2) in some breast cancer subtypes, no data exist for inflammatory breast cancer (IBC). METHODS MSI gene expression was quantified in IBC SUM149PT cells. We then used small interfering RNA-based MSI1 and MSI2 double knockdown (DKD) to understand gene expression and functional changes upon MSI depletion. We characterized cancer stem cell characteristics, cell apoptosis and cell cycle progression via flow cytometry, mammospheres via spheroid assays, migration and proliferation via digital holographic microscopy, and cell viability using BrdU assays. Chemoresistance was determined for paclitaxel and cisplatin with MTT assays and radioresistance was assessed with clonogenic analyses. In parallel, we supported our in vitro data by analyzing publicly available patient IBC gene expression datasets. RESULTS MSI1 and MSI2 are upregulated in breast cancer generally and IBC specifically. MSI2 is more commonly expressed compared to MSI1. MSI DKD attenuated proliferation, cell cycle progression, migration, and cell viability while increasing apoptosis. Stem cell characteristics CD44(+)/CD24(-), TERT and Oct4 were associated with MSI expression in vivo and were decreased in vitro after MSI DKD as was ALDH expression and mammosphere formation. In vivo, chemoresistant tumors were characterized by MSI upregulation upon chemotherapy application. In vitro, MSI DKD was able to alleviate chemo- and radioresistance. CONCLUSIONS The Musashi RNA binding proteins are dysregulated in IBC and associated with tumor proliferation, cancer stem cell phenotype, chemo- and radioresistance. MSI downregulation alleviates therapy resistance and attenuates tumor proliferation in vitro.
Collapse
Affiliation(s)
- Tiffany S Haiduk
- Department of Radiation Oncology, University Hospital Münster, Münster, Germany
| | - Mark Sicking
- Department of Radiation Oncology, University Hospital Münster, Münster, Germany
| | - Kathrin A Brücksken
- Department of Radiation Oncology, University Hospital Münster, Münster, Germany
| | - Nancy A Espinoza-Sánchez
- Department of Radiation Oncology, University Hospital Münster, Münster, Germany; Department of Gynecology and Obstetrics, University Hospital Münster, Münster, Germany
| | - Kai Moritz Eder
- Biomedical Technology Center, Medical Faculty, University of Münster, Münster, Germany
| | - Björn Kemper
- Biomedical Technology Center, Medical Faculty, University of Münster, Münster, Germany
| | - Hans Theodor Eich
- Department of Radiation Oncology, University Hospital Münster, Münster, Germany
| | - Martin Götte
- Department of Gynecology and Obstetrics, University Hospital Münster, Münster, Germany
| | - Burkhard Greve
- Department of Radiation Oncology, University Hospital Münster, Münster, Germany
| | - Fabian M Troschel
- Department of Radiation Oncology, University Hospital Münster, Münster, Germany.
| |
Collapse
|
4
|
Moreira ARS, Lim J, Urbaniak A, Banik J, Bronson K, Lagasse A, Hardy L, Haney A, Allensworth M, Miles TK, Gies A, Byrum SD, Wilczynska A, Boehm U, Kharas M, Lengner C, MacNicol MC, Childs GV, MacNicol AM, Odle AK. Musashi Exerts Control of Gonadotrope Target mRNA Translation During the Mouse Estrous Cycle. Endocrinology 2023; 164:bqad113. [PMID: 37477898 PMCID: PMC10402870 DOI: 10.1210/endocr/bqad113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/30/2023] [Accepted: 07/20/2023] [Indexed: 07/22/2023]
Abstract
The anterior pituitary controls key biological processes, including growth, metabolism, reproduction, and stress responses through distinct cell types that each secrete specific hormones. The anterior pituitary cells show a remarkable level of cell type plasticity that mediates the shifts in hormone-producing cell populations that are required to meet organismal needs. The molecular mechanisms underlying pituitary cell plasticity are not well understood. Recent work has implicated the pituitary stem cell populations and specifically, the mRNA binding proteins of the Musashi family in control of pituitary cell type identity. In this study we have identified the target mRNAs that mediate Musashi function in the adult mouse pituitary and demonstrate the requirement for Musashi function in vivo. Using Musashi RNA immunoprecipitation, we identify a cohort of 1184 mRNAs that show specific Musashi binding. Identified Musashi targets include the Gnrhr mRNA, which encodes the gonadotropin-releasing hormone receptor (GnRHR), and the Fshb mRNA, encoding follicle-stimulating hormone (FSH). Reporter assays reveal that Musashi functions to exert repression of translation of the Fshb mRNA, in addition to the previously observed repression of the Gnrhr mRNA. Importantly, mice engineered to lack Musashi in gonadotropes demonstrate a failure to repress translation of the endogenous Gnrhr and Fshb mRNAs during the estrous cycle and display a significant heterogeneity in litter sizes. The range of identified target mRNAs suggests that, in addition to these key gonadotrope proteins, Musashi may exert broad regulatory control over the pituitary proteome in a cell type-specific manner.
Collapse
Affiliation(s)
- Ana Rita Silva Moreira
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Juchan Lim
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Alicja Urbaniak
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jewel Banik
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Katherine Bronson
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Alex Lagasse
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Linda Hardy
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Anessa Haney
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Melody Allensworth
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Tiffany K Miles
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Allen Gies
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Arkansas Children's Research Institute, Arkansas Children's Hospital, Little Rock, AR 72202, USA
| | - Ania Wilczynska
- Bit.bio, The Dorothy Hodgkin Building, Babraham Research Campus, Cambridge CB22 3FH, UK
| | - Ulrich Boehm
- Department of Experimental Pharmacology, Center for Molecular Signaling, Saarland University School of Medicine, Homburg 66421, Germany
| | - Michael Kharas
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Christopher Lengner
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19146, USA
| | - Melanie C MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Gwen V Childs
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Angus M MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Angela K Odle
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
5
|
Chen X, Wang Y, Xu Z, Cheng ML, Ma QQ, Li RT, Wang ZJ, Zhao H, Zuo X, Li XF, Fang X, Qin CF. Zika virus RNA structure controls its unique neurotropism by bipartite binding to Musashi-1. Nat Commun 2023; 14:1134. [PMID: 36854751 PMCID: PMC9972320 DOI: 10.1038/s41467-023-36838-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 02/17/2023] [Indexed: 03/02/2023] Open
Abstract
Human RNA binding protein Musashi-1 (MSI1) plays a critical role in neural progenitor cells (NPCs) by binding to various host RNA transcripts. The canonical MSI1 binding site (MBS), A/GU(1-3)AG single-strand motif, is present in many RNA virus genomes, but only Zika virus (ZIKV) genome has been demonstrated to bind MSI1. Herein, we identified the AUAG motif and the AGAA tetraloop in the Xrn1-resistant RNA 2 (xrRNA2) as the canonical and non-canonical MBS, respectively, and both are crucial for ZIKV neurotropism. More importantly, the unique AGNN-type tetraloop is evolutionally conserved, and distinguishes ZIKV from other known viruses with putative MBSs. Integrated structural analysis showed that MSI1 binds to the AUAG motif and AGAA tetraloop of ZIKV in a bipartite fashion. Thus, our results not only identified an unusual viral RNA structure responsible for MSI recognition, but also revealed a role for the highly structured xrRNA in controlling viral neurotropism.
Collapse
Affiliation(s)
- Xiang Chen
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing, 100071, China
| | - Yan Wang
- Beijing Advanced Innovation Center for Structural Biology and Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Zhonghe Xu
- Beijing Advanced Innovation Center for Structural Biology and Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Meng-Li Cheng
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing, 100071, China
| | - Qing-Qing Ma
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing, 100071, China
| | - Rui-Ting Li
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing, 100071, China
| | - Zheng-Jian Wang
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing, 100071, China
| | - Hui Zhao
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing, 100071, China
| | - Xiaobing Zuo
- X-ray Science Division, Argonne National Laboratory, Lemont, IL, 60439, USA
| | - Xiao-Feng Li
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing, 100071, China
| | - Xianyang Fang
- Beijing Advanced Innovation Center for Structural Biology and Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Cheng-Feng Qin
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing, 100071, China.
- Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing, 100071, China.
| |
Collapse
|
6
|
Splicing factor SRSF3 represses translation of p21 cip1/waf1 mRNA. Cell Death Dis 2022; 13:933. [PMID: 36344491 PMCID: PMC9640673 DOI: 10.1038/s41419-022-05371-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 11/09/2022]
Abstract
Serine/arginine-rich splicing factor 3 (SRSF3) is an RNA binding protein that most often regulates gene expression at the splicing level. Although the role of SRSF3 in mRNA splicing in the nucleus is well known, its splicing-independent role outside of the nucleus is poorly understood. Here, we found that SRSF3 exerts a translational control of p21 mRNA. Depletion of SRSF3 induces cellular senescence and increases the expression of p21 independent of p53. Consistent with the expression patterns of SRSF3 and p21 mRNA in the TCGA database, SRSF3 knockdown increases the p21 mRNA level and its translation efficiency as well. SRSF3 physically associates with the 3'UTR region of p21 mRNA and the translational initiation factor, eIF4A1. Our study proposes a model in which SRSF3 regulates translation by interacting with eIF4A1 at the 3'UTR region of p21 mRNA. We also found that SRSF3 localizes to the cytoplasmic RNA granule along with eIF4A1, which may assist in translational repression therein. Thus, our results provide a new mode of regulation for p21 expression, a crucial regulator of the cell cycle and senescence, which occurs at the translational level and involves SRSF3.
Collapse
|
7
|
Matalkah F, Jeong B, Sheridan M, Horstick E, Ramamurthy V, Stoilov P. The Musashi proteins direct post-transcriptional control of protein expression and alternate exon splicing in vertebrate photoreceptors. Commun Biol 2022; 5:1011. [PMID: 36153373 PMCID: PMC9509328 DOI: 10.1038/s42003-022-03990-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 09/13/2022] [Indexed: 11/23/2022] Open
Abstract
The Musashi proteins, MSI1 and MSI2, are conserved RNA binding proteins with a role in the maintenance and renewal of stem cells. Contrasting with this role, terminally differentiated photoreceptor cells express high levels of MSI1 and MSI2, pointing to a role for the two proteins in vision. Combined knockout of Msi1 and Msi2 in mature photoreceptor cells abrogated the retinal response to light and caused photoreceptor cell death. In photoreceptor cells the Musashi proteins perform distinct nuclear and cytoplasmic functions. In the nucleus, the Musashi proteins promote splicing of photoreceptor-specific alternative exons. Surprisingly, conserved photoreceptor-specific alternative exons in genes critical for vision proved to be dispensable, raising questions about the selective pressures that lead to their conservation. In the cytoplasm MSI1 and MSI2 activate protein expression. Loss of Msi1 and Msi2 lead to reduction in the levels of multiple proteins including proteins required for vision and photoreceptor survival. The requirement for MSI1 and MSI2 in terminally differentiated photoreceptors alongside their role in stem cells shows that, depending on cellular context, these two proteins can control processes ranging from cell proliferation to sensory perception.
Collapse
Affiliation(s)
- Fatimah Matalkah
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV, USA
| | - Bohye Jeong
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV, USA
| | - Macie Sheridan
- Undergraduate Program in Biochemistry, West Virginia University, Morgantown, WV, USA
| | - Eric Horstick
- Department of Biology, West Virginia University, Morgantown, WV, USA
- Department of Neuroscience, West Virginia University, Morgantown, WV, USA
| | - Visvanathan Ramamurthy
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV, USA
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV, USA
| | - Peter Stoilov
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
8
|
Knockdown of the stem cell marker Musashi-1 inhibits endometrial cancer growth and sensitizes cells to radiation. Stem Cell Res Ther 2022; 13:212. [PMID: 35619161 PMCID: PMC9137084 DOI: 10.1186/s13287-022-02891-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/02/2022] [Indexed: 11/22/2022] Open
Abstract
Background Endometrial carcinoma is the most common gynecological cancer in Europe. Musashi-1 is known to be a key regulator of endometrial cancer stem cells and a negative prognostic marker. In the present study, we aimed to understand growth and gene expression patterns in endometrial carcinoma after Musashi-1 knockdown in vitro and in vivo. Changes in therapeutic resistance were also assessed.
Methods First, we performed analyses to understand Musashi-1 expression patterns using The Cancer Genome Atlas database. We then proceeded to assess effects of small interfering RNA-based Musashi-1 targeting in two endometrial carcinoma cell lines, Ishikawa and KLE. After quantifying baseline changes in cell metabolism, we used MTT tests to assess chemotherapy effects and colony formation assays to understand changes in radioresistance. For mechanistic study, we used quantitative polymerase chain reaction (qPCR) and western blotting of key Musashi-1 target genes and compared results to primary tissue database studies. Finally, xenograft experiments in a mouse model helped understand in vivo effects of Musashi-1 knockdown. Results Musashi-1 is aberrantly expressed in primary tumor tissues. In vitro, silencing of Musashi-1 resulted in a strong decline in cell proliferation and radioresistance, while chemoresistance remained unchanged. Loss of Musashi-1 led to downregulation of telomerase, DNA-dependent protein kinase, the Notch pathway and overexpression of cyclin-dependent kinase inhibitor p21, the latter of which we identified as a key mediator of Msi-1 knockdown-related anti-proliferative signaling. In vivo, the anti-proliferative effect was confirmed, with Msi-1 knockdown tumors being about 40% reduced in size. Conclusions Musashi-1 knockdown resulted in a strong decrease in endometrial cancer proliferation and a loss of radioresistance, suggesting therapeutic potential. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02891-3.
Collapse
|
9
|
Nishanth MJ, Jha S. Global Exploration of RNA-Binding Proteins in Exercise-Induced Adult Hippocampal Neurogenesis: A Transcriptome Meta-analysis and Computational Study. Biochem Genet 2022; 60:2471-2488. [PMID: 35546218 DOI: 10.1007/s10528-022-10230-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 04/18/2022] [Indexed: 11/02/2022]
Abstract
Voluntary physical exercise is a robust enhancer of adult hippocampal neurogenesis (AHN). A complete understanding of the molecular regulation of AHN is important in order to exploit the benefits of the process toward therapeutic approaches. Several factors such as epigenetic modifiers, non-coding RNAs, and transcription factors have been reported to regulate AHN. However, there is a limited understanding of the impact of RNA-binding proteins (RBPs) on exercise-mediated AHN, in spite of their well-documented significance in embryonic neurogenesis. The present study is the first global analysis to catalog the potential RBPs influencing exercise-mediated AHN. Here, a transcriptome meta-analysis was conducted to study exercise-mediated gene expression modulation in hippocampi of adult mice. Next, potential RBPs influencing transcriptome-wide expression changes via untranslated regions (UTRs) were identified. Among other RBPs, MATR3, Musashi, TIA1, and FXR2 (known critical modulators of neurogenesis) were found to potentially regulate gene expression patterns. Subsequently, binding sites of known neurogenesis-regulating RBPs were identified in the UTRs of AHN-associated genes modulated by exercise. Finally, a number of RBPs including RBFOX1, RBFOX3, and QKI (known regulators of neurogenesis) were found to be highly expressed in mouse hippocampal formation and also potentially interact with other RBPs, suggesting their combinatorial functioning in exercise-induced AHN. Thus, the present meta-analysis-based computational study identified several RBPs potentially important in exercise-induced AHN, which could form a foundation for further experiments to unravel RBP-mediated regulation of AHN.
Collapse
Affiliation(s)
- M J Nishanth
- School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, 613401, India
| | - Shanker Jha
- School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, 613401, India.
| |
Collapse
|
10
|
Karmakar S, Ramirez O, Paul KV, Gupta AK, Kumari V, Botti V, de Los Mozos IR, Neuenkirchen N, Ross RJ, Karanicolas J, Neugebauer KM, Pillai MM. Integrative genome-wide analysis reveals EIF3A as a key downstream regulator of translational repressor protein Musashi 2 (MSI2). NAR Cancer 2022; 4:zcac015. [PMID: 35528200 PMCID: PMC9070473 DOI: 10.1093/narcan/zcac015] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/04/2022] [Accepted: 04/19/2022] [Indexed: 01/29/2023] Open
Abstract
Musashi 2 (MSI2) is an RNA binding protein (RBP) that regulates asymmetric cell division and cell fate decisions in normal and cancer stem cells. MSI2 appears to repress translation by binding to 3′ untranslated regions (3′UTRs) of mRNA, but the identity of functional targets remains unknown. Here, we used individual nucleotide resolution cross-linking and immunoprecipitation (iCLIP) to identify direct RNA binding partners of MSI2 and integrated these data with polysome profiling to obtain insights into MSI2 function. iCLIP revealed specific MSI2 binding to thousands of mRNAs largely in 3′UTRs, but translational differences were restricted to a small fraction of these transcripts, indicating that MSI2 regulation is not triggered by simple binding. Instead, the functional targets identified here were bound at higher density and contain more ‘UAG’ motifs compared to targets bound nonproductively. To further distinguish direct and indirect targets, MSI2 was acutely depleted. Surprisingly, only 50 transcripts were found to undergo translational induction on acute loss. Using complementary approaches, we determined eukaryotic translation initiation factor 3A (EIF3A) to be an immediate, direct target. We propose that MSI2 downregulation of EIF3A amplifies these effects on translation. Our results also underscore the challenges in defining functional targets of RBPs since mere binding does not imply a discernible functional interaction.
Collapse
Affiliation(s)
| | - Oscar Ramirez
- Section of Hematology, Yale Cancer Center, New Haven, CT 06511, USA
| | - Kiran V Paul
- Section of Hematology, Yale Cancer Center, New Haven, CT 06511, USA
| | - Abhishek K Gupta
- Section of Hematology, Yale Cancer Center, New Haven, CT 06511, USA
| | - Vandana Kumari
- Section of Hematology, Yale Cancer Center, New Haven, CT 06511, USA
| | - Valentina Botti
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Igor Ruiz de Los Mozos
- Institute of Neurology, University College London and The Francis Crick Institute, London NW1 1AT, UK
| | - Nils Neuenkirchen
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Robert J Ross
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - John Karanicolas
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Karla M Neugebauer
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Manoj M Pillai
- Section of Hematology, Yale Cancer Center, New Haven, CT 06511, USA
| |
Collapse
|
11
|
Zhang W, Liu L, Zhao S, Chen L, Wei Y, Chen W, Ge F. Research progress on RNA‑binding proteins in breast cancer (Review). Oncol Lett 2022; 23:121. [PMID: 35261635 PMCID: PMC8867207 DOI: 10.3892/ol.2022.13241] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 02/03/2022] [Indexed: 11/28/2022] Open
Abstract
Breast cancer is the most common malignancy among women, and the abnormal regulation of gene expression serves an important role in its occurrence and development. However, the molecular mechanisms underlying gene expression are highly complex and heterogeneous, and RNA-binding proteins (RBPs) are among the key regulatory factors. RBPs bind targets in an environment-dependent or environment-independent manner to influence mRNA stability and the translation of genes involved in the formation, progression, metastasis and treatment of breast cancer. Due to the growing interest in these regulators, the present review summarizes the most influential studies concerning RBPs associated with breast cancer to elucidate the role of RBPs in breast cancer and to assess how they interact with other key pathways to provide new molecular targets for the diagnosis and treatment of breast cancer.
Collapse
Affiliation(s)
- Wenzhu Zhang
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Linlin Liu
- School of Forensic Medicine, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Shengdi Zhao
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Liang Chen
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Yuxian Wei
- Department of Endocrine Breast Surgery, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Wenlin Chen
- Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650118, P.R. China
| | - Fei Ge
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
12
|
Landínez-Macías M, Urwyler O. The Fine Art of Writing a Message: RNA Metabolism in the Shaping and Remodeling of the Nervous System. Front Mol Neurosci 2021; 14:755686. [PMID: 34916907 PMCID: PMC8670310 DOI: 10.3389/fnmol.2021.755686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/18/2021] [Indexed: 01/25/2023] Open
Abstract
Neuronal morphogenesis, integration into circuits, and remodeling of synaptic connections occur in temporally and spatially defined steps. Accordingly, the expression of proteins and specific protein isoforms that contribute to these processes must be controlled quantitatively in time and space. A wide variety of post-transcriptional regulatory mechanisms, which act on pre-mRNA and mRNA molecules contribute to this control. They are thereby critically involved in physiological and pathophysiological nervous system development, function, and maintenance. Here, we review recent findings on how mRNA metabolism contributes to neuronal development, from neural stem cell maintenance to synapse specification, with a particular focus on axon growth, guidance, branching, and synapse formation. We emphasize the role of RNA-binding proteins, and highlight their emerging roles in the poorly understood molecular processes of RNA editing, alternative polyadenylation, and temporal control of splicing, while also discussing alternative splicing, RNA localization, and local translation. We illustrate with the example of the evolutionary conserved Musashi protein family how individual RNA-binding proteins are, on the one hand, acting in different processes of RNA metabolism, and, on the other hand, impacting multiple steps in neuronal development and circuit formation. Finally, we provide links to diseases that have been associated with the malfunction of RNA-binding proteins and disrupted post-transcriptional regulation.
Collapse
Affiliation(s)
- María Landínez-Macías
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.,Molecular Life Sciences Program, Life Science Zurich Graduate School, University of Zurich and Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Olivier Urwyler
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.,Molecular Life Sciences Program, Life Science Zurich Graduate School, University of Zurich and Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland.,Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Double-Stranded RNA Structural Elements Holding the Key to Translational Regulation in Cancer: The Case of Editing in RNA-Binding Motif Protein 8A. Cells 2021; 10:cells10123543. [PMID: 34944051 PMCID: PMC8699885 DOI: 10.3390/cells10123543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/08/2021] [Accepted: 12/13/2021] [Indexed: 12/30/2022] Open
Abstract
Mesothelioma is an aggressive cancer associated with asbestos exposure. RNA-binding motif protein 8a (RBM8A) mRNA editing increases in mouse tissues upon asbestos exposure. The aim of this study was to further characterize the role of RBM8A in mesothelioma and the consequences of its mRNA editing. RBM8A protein expression was higher in mesothelioma compared to mesothelial cells. Silencing RBM8A changed splicing patterns in mesothelial and mesothelioma cells but drastically reduced viability only in mesothelioma cells. In the tissues of asbestos-exposed mice, editing of Rbm8a mRNA was associated with increased protein immunoreactivity, with no change in mRNA levels. Increased adenosine deaminase acting on dsRNA (ADAR)-dependent editing of Alu elements in the RBM8A 3′UTR was observed in mesothelioma cells compared to mesothelial cells. Editing stabilized protein expression. The unedited RBM8A 3′UTR had a stronger interaction with Musashi (MSI) compared to the edited form. The silencing of MSI2 in mesothelioma or overexpression of Adar2 in mesothelial cells resulted in increased RBM8A protein levels. Therefore, ADAR-dependent editing contributes to maintaining elevated RBM8A protein levels in mesothelioma by counteracting MSI2-driven downregulation. A wider implication of this mechanism for the translational control of protein expression is suggested by the editing of similarly structured Alu elements in several other transcripts.
Collapse
|
14
|
Dual Knockdown of Musashi RNA-Binding Proteins MSI-1 and MSI-2 Attenuates Putative Cancer Stem Cell Characteristics and Therapy Resistance in Ovarian Cancer Cells. Int J Mol Sci 2021; 22:ijms222111502. [PMID: 34768932 PMCID: PMC8584030 DOI: 10.3390/ijms222111502] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/14/2021] [Accepted: 10/21/2021] [Indexed: 01/06/2023] Open
Abstract
In ovarian cancer, therapy resistance mechanisms complicate cancer cell eradication. Targeting Musashi RNA-binding proteins (MSI) may increase therapeutic efficacy. Database analyses were performed to identify gene expression associations between MSI proteins and key therapy resistance and cancer stem cell (CSC) genes. Then, ovarian cancer cells were subjected to siRNA-based dual knockdown of MSI-1 and MSI-2. CSC and cell cycle gene expression was investigated using quantitative polymerase chain reaction (qPCR), western blots, and flow cytometry. Metabolic activity and chemoresistance were assessed by MTT assay. Clonogenic assays were used to quantify cell survival post-irradiation. Database analyses demonstrated positive associations between MSI proteins and putative CSC markers NOTCH, MYC, and ALDH4A1 and negative associations with NOTCH inhibitor NUMB. MSI-2 expression was negatively associated with the apoptosis regulator p21. MSI-1 and MSI-2 were positively correlated, informing subsequent dual knockdown experiments. After MSI silencing, CSC genes were downregulated, while cell cycle progression was reduced. Metabolic activity was decreased in some cancer cells. Both chemo- and radioresistance were reduced after dual knockdown, suggesting therapeutic potential. Dual knockdown of MSI proteins is a promising venue to impede tumor growth and sensitize ovarian cancer cells to irradiation and chemotherapy.
Collapse
|
15
|
Troschel FM, Palenta H, Borrmann K, Heshe K, Hua SH, Yip GW, Kiesel L, Eich HT, Götte M, Greve B. Knockdown of the prognostic cancer stem cell marker Musashi-1 decreases radio-resistance while enhancing apoptosis in hormone receptor-positive breast cancer cells via p21 WAF1/CIP1. J Cancer Res Clin Oncol 2021; 147:3299-3312. [PMID: 34291358 PMCID: PMC8484224 DOI: 10.1007/s00432-021-03743-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/13/2021] [Indexed: 02/02/2023]
Abstract
Purpose While the stem cell marker Musashi-1 (MSI-1) has been identified as a key player in a wide array of malignancies, few findings exist on its prognostic relevance and relevance for cancer cell death and therapy resistance in breast cancer. Methods First, we determined prognostic relevance of MSI-1 in database analyses regarding multiple survival outcomes. To substantiate findings, MSI-1 was artificially downregulated in MCF-7 breast cancer cells and implications for cancer stem cell markers, cell apoptosis and apoptosis regulator p21, proliferation and radiation response were analyzed via flow cytometry and colony formation. Radiation-induced p21 expression changes were investigated using a dataset containing patient samples obtained before and after irradiation and own in vitro experiments. Results MSI-1 is a negative prognostic marker for disease-free and distant metastasis-free survival in breast cancer and tends to negatively influence overall survival. MSI-1 knockdown downregulated stem cell gene expression and proliferation, but increased p21 levels and apoptosis. Similar to the MSI-1 knockdown effect, p21 expression was strongly increased after irradiation and was expressed at even higher levels in MSI-1 knockdown cells after irradiation. Finally, combined use of MSI-1 silencing and irradiation reduced cancer cell survival. Conclusion MSI-1 is a prognostic marker in breast cancer. MSI-1 silencing downregulates proliferation while increasing apoptosis. The anti-proliferation mediator p21 was upregulated independently after both MSI-1 knockdown and irradiation and even more after both treatments combined, suggesting synergistic potential. Radio-sensitization effects after combining radiation and MSI-1 knockdown underline the potential of MSI-1 as a therapeutic target. Supplementary Information The online version contains supplementary material available at 10.1007/s00432-021-03743-y.
Collapse
Affiliation(s)
- Fabian M Troschel
- Department of Radiation Oncology, University Hospital Münster, 48149, Münster, Germany.
| | - Heike Palenta
- Department of Gynecology and Obstetrics, University Hospital Münster, 48149, Münster, Germany
| | - Katrin Borrmann
- Department of Radiation Oncology, University Hospital Münster, 48149, Münster, Germany
| | - Kristin Heshe
- Department of Radiation Oncology, University Hospital Münster, 48149, Münster, Germany
| | - San Hue Hua
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117594, Singapore
| | - George W Yip
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117594, Singapore
| | - Ludwig Kiesel
- Department of Gynecology and Obstetrics, University Hospital Münster, 48149, Münster, Germany
| | - Hans Theodor Eich
- Department of Radiation Oncology, University Hospital Münster, 48149, Münster, Germany
| | - Martin Götte
- Department of Gynecology and Obstetrics, University Hospital Münster, 48149, Münster, Germany
| | - Burkhard Greve
- Department of Radiation Oncology, University Hospital Münster, 48149, Münster, Germany
| |
Collapse
|
16
|
Getmantseva L, Kolosova M, Bakoev F, Zimina A, Bakoev S. Genomic Regions and Candidate Genes Linked to Capped Hock in Pig. Life (Basel) 2021; 11:life11060510. [PMID: 34073088 PMCID: PMC8228005 DOI: 10.3390/life11060510] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 01/13/2023] Open
Abstract
Capped hock affects the exterior of pedigree pigs, making them unsalable and resulting in a negative impact on the efficiency of pig-breeding centers. The purpose of this paper was to carry out pilot studies aimed at finding genomic regions and genes linked to the capped hock in pigs. The studies were carried out on Landrace pigs (n = 75) and Duroc pigs (n = 70). To identify genomic regions linked to capped hock in pigs, we used smoothing FST statistics. Genotyping was performed with GeneSeek® GGP Porcine HD Genomic Profiler v1 (Illumina Inc, San Diego, CA, USA). The research results showed 70 SNPs linked to capped hock in Landrace (38 SNPs) and Duroc (32 SNPs). The identified regions overlapped with QTLs related with health traits (blood parameters) and meat and carcass traits (fatness). In total, 31 genes were identified (i.e., 17 genes in Landrace, 14 genes in Durocs). Three genes appeared in both the Landrace and Duroc groups, including A2ML1 (SSC5), ROBO2 (SSC13), and MSI1 (SSC14). We identified genomic regions directly or indirectly linked to capped hock, which thus might contribute to identifying genetic variants and using them as genetic markers in pig breeding.
Collapse
Affiliation(s)
- Lyubov Getmantseva
- Federal Research Center for Animal Husbandry Named after Academy Member L.K. Ernst, 142132 Dubrovitsy, Russia; (M.K.); (F.B.); (S.B.)
- Correspondence: (L.G.); (A.Z.); Tel.: +7-(4967)-65-11-01 (L.G. & A.Z.)
| | - Maria Kolosova
- Federal Research Center for Animal Husbandry Named after Academy Member L.K. Ernst, 142132 Dubrovitsy, Russia; (M.K.); (F.B.); (S.B.)
- Department of Biotechnology, Don State Agrarian University, 346493 Persianovski, Russia
| | - Faridun Bakoev
- Federal Research Center for Animal Husbandry Named after Academy Member L.K. Ernst, 142132 Dubrovitsy, Russia; (M.K.); (F.B.); (S.B.)
| | - Anna Zimina
- Federal Research Center for Animal Husbandry Named after Academy Member L.K. Ernst, 142132 Dubrovitsy, Russia; (M.K.); (F.B.); (S.B.)
- Correspondence: (L.G.); (A.Z.); Tel.: +7-(4967)-65-11-01 (L.G. & A.Z.)
| | - Siroj Bakoev
- Federal Research Center for Animal Husbandry Named after Academy Member L.K. Ernst, 142132 Dubrovitsy, Russia; (M.K.); (F.B.); (S.B.)
- Centre for Strategic Planning and Management of Biomedical Health Risks, 123182 Moscow, Russia
| |
Collapse
|
17
|
Bley N, Hmedat A, Müller S, Rolnik R, Rausch A, Lederer M, Hüttelmaier S. Musashi-1-A Stemness RBP for Cancer Therapy? BIOLOGY 2021; 10:407. [PMID: 34062997 PMCID: PMC8148009 DOI: 10.3390/biology10050407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 04/29/2021] [Accepted: 05/01/2021] [Indexed: 12/12/2022]
Abstract
The RNA-binding protein Musashi-1 (MSI1) promotes stemness during development and cancer. By controlling target mRNA turnover and translation, MSI1 is implicated in the regulation of cancer hallmarks such as cell cycle or Notch signaling. Thereby, the protein enhanced cancer growth and therapy resistance to standard regimes. Due to its specific expression pattern and diverse functions, MSI1 represents an interesting target for cancer therapy in the future. In this review we summarize previous findings on MSI1's implications in developmental processes of other organisms. We revisit MSI1's expression in a set of solid cancers, describe mechanistic details and implications in MSI1 associated cancer hallmark pathways and highlight current research in drug development identifying the first MSI1-directed inhibitors with anti-tumor activity.
Collapse
Affiliation(s)
- Nadine Bley
- Department for Molecular Cell Biology, Institute for Molecular Medicine, Martin Luther University Halle/Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany; (A.H.); (S.M.); (R.R.); (A.R.); (M.L.); (S.H.)
- Core Facility Imaging, Institute for Molecular Medicine, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany
| | - Ali Hmedat
- Department for Molecular Cell Biology, Institute for Molecular Medicine, Martin Luther University Halle/Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany; (A.H.); (S.M.); (R.R.); (A.R.); (M.L.); (S.H.)
| | - Simon Müller
- Department for Molecular Cell Biology, Institute for Molecular Medicine, Martin Luther University Halle/Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany; (A.H.); (S.M.); (R.R.); (A.R.); (M.L.); (S.H.)
| | - Robin Rolnik
- Department for Molecular Cell Biology, Institute for Molecular Medicine, Martin Luther University Halle/Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany; (A.H.); (S.M.); (R.R.); (A.R.); (M.L.); (S.H.)
| | - Alexander Rausch
- Department for Molecular Cell Biology, Institute for Molecular Medicine, Martin Luther University Halle/Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany; (A.H.); (S.M.); (R.R.); (A.R.); (M.L.); (S.H.)
- Core Facility Imaging, Institute for Molecular Medicine, Martin Luther University Halle-Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany
| | - Marcell Lederer
- Department for Molecular Cell Biology, Institute for Molecular Medicine, Martin Luther University Halle/Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany; (A.H.); (S.M.); (R.R.); (A.R.); (M.L.); (S.H.)
| | - Stefan Hüttelmaier
- Department for Molecular Cell Biology, Institute for Molecular Medicine, Martin Luther University Halle/Wittenberg, Charles Tanford Protein Center, Kurt–Mothes–Str. 3A, 06120 Halle, Germany; (A.H.); (S.M.); (R.R.); (A.R.); (M.L.); (S.H.)
| |
Collapse
|
18
|
Padial-Molina M, Crespo-Lora V, Candido-Corral C, Martin-Morales N, Abril-Garcia D, Galindo-Moreno P, Hernandez-Cortes P, O’Valle F. Expression of Musashi-1 Increases in Bone Healing. Int J Mol Sci 2021; 22:ijms22073395. [PMID: 33810326 PMCID: PMC8037090 DOI: 10.3390/ijms22073395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/16/2022] Open
Abstract
Musashi-1 (MSI1) is an RNA-binding protein that regulates progenitor cells in adult and developing organisms to maintain self-renewal capacities. The role of musashi-1 in the bone healing environment and its relation with other osteogenic factors is unknown. In the current study, we analyze the expression of MSI1 in an experimental model of rat femoral bone fractures. We also analyze the relation between MSI1 expression and the expression of two osteogenic markers: periostin (POSTN) and runt-related transcription factor 2 (RUNX2). We use histological, immunohistochemical, and qPCR techniques to evaluate bone healing and the expression of MSI1, POSTN, and RUNX2 over time (4, 7, and 14 days). We compare our findings with non-fractured controls. We find that in bone calluses, the number of cells expressing MSI1 and RUNX2 increase over time and the intensity of POSTN expression decreases over time. Within bone calluses, we find the presence of MSI1 expression in mesenchymal stromal cells, osteoblasts, and osteocytes but not in hypertrophic chondrocytes. After 14 days, the expression of MSI1, POSTN, and RUNX2 was significantly correlated. Thus, we conclude that musashi-1 potentially serves in the osteogenic differentiation of mesenchymal stromal cells and bone healing. Therefore, further studies are needed to determine the possibility of musashi-1′s role as a clinical biomarker of bone healing and therapeutic agent for bone regeneration.
Collapse
Affiliation(s)
- Miguel Padial-Molina
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, and Centre for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (M.P.-M.); (N.M.-M.); (D.A.-G.)
| | - Vicente Crespo-Lora
- Department of Pathology, University of Granada, 18071 Granada, Spain; (V.C.-L.); (C.C.-C.)
| | - Clara Candido-Corral
- Department of Pathology, University of Granada, 18071 Granada, Spain; (V.C.-L.); (C.C.-C.)
| | - Nati Martin-Morales
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, and Centre for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (M.P.-M.); (N.M.-M.); (D.A.-G.)
- Department of Pathology, University of Granada, 18071 Granada, Spain; (V.C.-L.); (C.C.-C.)
| | - Dario Abril-Garcia
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, and Centre for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (M.P.-M.); (N.M.-M.); (D.A.-G.)
| | - Pablo Galindo-Moreno
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, and Centre for Biomedical Research (CIBM), University of Granada, 18071 Granada, Spain; (M.P.-M.); (N.M.-M.); (D.A.-G.)
- Correspondence:
| | - Pedro Hernandez-Cortes
- Department of Orthopedic Surgery, San Cecilio University Hospital, 18071 Granada, Spain;
| | - Francisco O’Valle
- Department of Pathology, Institute of Biopathology and Regenerative Medicine (IBIMER, CIBM), and Institute of Biosanitary (ibs-Granada), University of Granada, 18071 Granada, Spain;
| |
Collapse
|
19
|
Baroni M, Yi C, Choudhary S, Lei X, Kosti A, Grieshober D, Velasco M, Qiao M, Burns SS, Araujo PR, DeLambre T, Son MY, Plateroti M, Ferreira MAR, Hasty EP, Penalva LOF. Musashi1 Contribution to Glioblastoma Development via Regulation of a Network of DNA Replication, Cell Cycle and Division Genes. Cancers (Basel) 2021; 13:1494. [PMID: 33804958 PMCID: PMC8036803 DOI: 10.3390/cancers13071494] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/17/2021] [Accepted: 03/21/2021] [Indexed: 11/21/2022] Open
Abstract
RNA-binding proteins (RBPs) function as master regulators of gene expression. Alterations in their levels are often observed in tumors with numerous oncogenic RBPs identified in recent years. Musashi1 (Msi1) is an RBP and stem cell gene that controls the balance between self-renewal and differentiation. High Msi1 levels have been observed in multiple tumors including glioblastoma and are often associated with poor patient outcomes and tumor growth. A comprehensive genomic analysis identified a network of cell cycle/division and DNA replication genes and established these processes as Msi1's core regulatory functions in glioblastoma. Msi1 controls this gene network via two mechanisms: direct interaction and indirect regulation mediated by the transcription factors E2F2 and E2F8. Moreover, glioblastoma lines with Msi1 knockout (KO) displayed increased sensitivity to cell cycle and DNA replication inhibitors. Our results suggest that a drug combination strategy (Msi1 + cell cycle/DNA replication inhibitors) could be a viable route to treat glioblastoma.
Collapse
Affiliation(s)
- Mirella Baroni
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA; (M.B.); (C.Y.); (X.L.); (A.K.); (D.G.); (M.V.); (M.Q.); (P.R.A.); (T.D.)
| | - Caihong Yi
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA; (M.B.); (C.Y.); (X.L.); (A.K.); (D.G.); (M.V.); (M.Q.); (P.R.A.); (T.D.)
- Third Xiangya Hospital, Central South University, Changsha 410000, China
| | - Saket Choudhary
- Computational Biology and Bioinformatics, University of Southern California, Los Angeles, CA 90089, USA;
| | - Xiufen Lei
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA; (M.B.); (C.Y.); (X.L.); (A.K.); (D.G.); (M.V.); (M.Q.); (P.R.A.); (T.D.)
| | - Adam Kosti
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA; (M.B.); (C.Y.); (X.L.); (A.K.); (D.G.); (M.V.); (M.Q.); (P.R.A.); (T.D.)
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Denise Grieshober
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA; (M.B.); (C.Y.); (X.L.); (A.K.); (D.G.); (M.V.); (M.Q.); (P.R.A.); (T.D.)
| | - Mitzli Velasco
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA; (M.B.); (C.Y.); (X.L.); (A.K.); (D.G.); (M.V.); (M.Q.); (P.R.A.); (T.D.)
| | - Mei Qiao
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA; (M.B.); (C.Y.); (X.L.); (A.K.); (D.G.); (M.V.); (M.Q.); (P.R.A.); (T.D.)
| | - Suzanne S. Burns
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA; (M.B.); (C.Y.); (X.L.); (A.K.); (D.G.); (M.V.); (M.Q.); (P.R.A.); (T.D.)
| | - Patricia R. Araujo
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA; (M.B.); (C.Y.); (X.L.); (A.K.); (D.G.); (M.V.); (M.Q.); (P.R.A.); (T.D.)
| | - Talia DeLambre
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA; (M.B.); (C.Y.); (X.L.); (A.K.); (D.G.); (M.V.); (M.Q.); (P.R.A.); (T.D.)
| | - Mi Young Son
- Department of Molecular Medicine, Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX 78229, USA; (M.Y.S.); (E.P.H.)
| | - Michelina Plateroti
- Team: Development, Cancer and Stem Cells, Université de Strasbourg, Inserm, IRFAC/UMR-S1113, FMTS, 67200 Strasbourg, France;
| | | | - E. Paul Hasty
- Department of Molecular Medicine, Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX 78229, USA; (M.Y.S.); (E.P.H.)
| | - Luiz O. F. Penalva
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA; (M.B.); (C.Y.); (X.L.); (A.K.); (D.G.); (M.V.); (M.Q.); (P.R.A.); (T.D.)
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
20
|
Sundar J, Matalkah F, Jeong B, Stoilov P, Ramamurthy V. The Musashi proteins MSI1 and MSI2 are required for photoreceptor morphogenesis and vision in mice. J Biol Chem 2021; 296:100048. [PMID: 33168629 PMCID: PMC7948980 DOI: 10.1074/jbc.ra120.015714] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/30/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022] Open
Abstract
The Musashi family of RNA-binding proteins is known for its role in stem-cell renewal and is a negative regulator of cell differentiation. Interestingly, in the retina, the Musashi proteins MSI1 and MSI2 are differentially expressed throughout the cycle of retinal development, with MSI2 protein displaying robust expression in the adult retinal tissue. In this study, we investigated the importance of Musashi proteins in the development and function of photoreceptor neurons in the retina. We generated a pan-retinal and rod photoreceptor neuron-specific conditional KO mouse lacking MSI1 and MSI2. Independent of the sex, photoreceptor neurons with simultaneous deletion of Msi1 and Msi2 were unable to respond to light and displayed severely disrupted photoreceptor outer segment morphology and ciliary defects. Mice lacking MSI1 and MSI2 in the retina exhibited neuronal degeneration, with complete loss of photoreceptors within 6 months. In concordance with our earlier studies that proposed a role for Musashi proteins in regulating alternative splicing, the loss of MSI1 and MSI2 prevented the use of photoreceptor-specific exons in transcripts critical for outer segment morphogenesis, ciliogenesis, and synaptic transmission. Overall, we demonstrate a critical role for Musashi proteins in the morphogenesis of terminally differentiated photoreceptor neurons. This role is in stark contrast with the canonical function of these two proteins in the maintenance and renewal of stem cells.
Collapse
Affiliation(s)
- Jesse Sundar
- Department of Biochemistry, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, West Virginia, USA
| | - Fatimah Matalkah
- Department of Biochemistry, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, West Virginia, USA
| | - Bohye Jeong
- Department of Biochemistry, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, West Virginia, USA
| | - Peter Stoilov
- Department of Biochemistry, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, West Virginia, USA.
| | - Visvanathan Ramamurthy
- Department of Biochemistry, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, West Virginia, USA; Department of Ophthalmology and Visual Sciences, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, West Virginia, USA; Department of Neuroscience, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, West Virginia, USA.
| |
Collapse
|
21
|
Yarmishyn AA, Yang YP, Lu KH, Chen YC, Chien Y, Chou SJ, Tsai PH, Ma HI, Chien CS, Chen MT, Wang ML. Musashi-1 promotes cancer stem cell properties of glioblastoma cells via upregulation of YTHDF1. Cancer Cell Int 2020; 20:597. [PMID: 33317545 PMCID: PMC7734781 DOI: 10.1186/s12935-020-01696-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022] Open
Abstract
Background Glioblastoma (GBM) is the most lethal brain tumor characterized by high morbidity and limited treatment options. Tumor malignancy is usually associated with the epigenetic marks, which coordinate gene expression to ascertain relevant phenotypes. One of such marks is m6A modification of RNA, whose functional effects are dependent on the YTH family m6A reader proteins. Methods and results In this study, we investigated the expression of five
YTH family proteins in different GBM microarray datasets from the Oncomine
database, and identified YTHDF1 as the most highly overexpressed member of this
family in GBM. By performing the knockdown of YTHDF1 in a GBM cell line, we
found that it positively regulates proliferation, chemoresistance and cancer
stem cell-like properties. Musashi-1 (MSI1) is a postranscriptional gene
expression regulator associated with high oncogenicity in GBM. By knocking down
and overexpressing MSI1, we found that it positively regulates YTHDF1
expression. The inhibitory effects
imposed on the processes of proliferation and migration by YTHDF1 knockdown
were shown to be partially rescued by concomitant overexpression of MSI1. MSI1
and YTHDF1 were shown to be positively correlated in clinical glioma samples,
and their concomitant upregulation was associated with decreased survival of
glioma patients. We identified the direct regulation of YTHDF1 by MSI1. Conclusions Given the fact that both proteins are master
regulators of gene expression, and both of them are unfavorable factors in GBM,
we suggest that in any future studies aimed to uncover the prognostic value and
therapy potential, these two proteins should be considered together.
Collapse
Affiliation(s)
- Aliaksandr A Yarmishyn
- Division of Basic Research, Department of Medical Research, Taipei Veterans General Hospital, 112, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, 112, Taipei, Taiwan
| | - Yi-Ping Yang
- Division of Basic Research, Department of Medical Research, Taipei Veterans General Hospital, 112, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, 112, Taipei, Taiwan.,School of Pharmaceutical Sciences, National Yang-Ming University, 112, Taipei, Taiwan
| | - Kai-Hsi Lu
- Department of Medical Research and Education, Cheng-Hsin General Hospital, 112, Taipei, Taiwan
| | - Yi-Chen Chen
- Division of Basic Research, Department of Medical Research, Taipei Veterans General Hospital, 112, Taipei, Taiwan
| | - Yueh Chien
- Division of Basic Research, Department of Medical Research, Taipei Veterans General Hospital, 112, Taipei, Taiwan
| | - Shih-Jie Chou
- Division of Basic Research, Department of Medical Research, Taipei Veterans General Hospital, 112, Taipei, Taiwan
| | - Ping-Hsing Tsai
- Division of Basic Research, Department of Medical Research, Taipei Veterans General Hospital, 112, Taipei, Taiwan
| | - Hsin-I Ma
- Department of Neurological Surgery, Tri-Service General Hospital and National Defense Medical Center, 114, Taipei, Taiwan
| | - Chian-Shiu Chien
- Division of Basic Research, Department of Medical Research, Taipei Veterans General Hospital, 112, Taipei, Taiwan.,Institute of Pharmacology, National Yang-Ming University, 112, Taipei, Taiwan
| | - Ming-Teh Chen
- School of Medicine, National Yang-Ming University, 112, Taipei, Taiwan.,Institute of Pharmacology, National Yang-Ming University, 112, Taipei, Taiwan.,Department of Neurosurgery, Taipei Veterans General Hospital, 112, Taipei, Taiwan
| | - Mong-Lien Wang
- Division of Basic Research, Department of Medical Research, Taipei Veterans General Hospital, 112, Taipei, Taiwan. .,School of Medicine, National Yang-Ming University, 112, Taipei, Taiwan. .,Institute of Food Safety and Health Risk Assessment, National Yang Ming University, 112, Taipei, Taiwan.
| |
Collapse
|
22
|
AMD1 is required for the maintenance of leukemic stem cells and promotes chronic myeloid leukemic growth. Oncogene 2020; 40:603-617. [PMID: 33203990 DOI: 10.1038/s41388-020-01547-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/15/2020] [Accepted: 10/28/2020] [Indexed: 12/18/2022]
Abstract
Polyamines are critical elements in mammals, but it remains unknown whether adenosyl methionine decarboxylase (AMD1), a rate-limiting enzyme in polyamine synthesis, is required for myeloid leukemia. Here, we found that leukemic stem cells (LSCs) were highly differentiated, and leukemia progression was severely impaired in the absence of AMD1 in vivo. AMD1 was highly upregulated as chronic myeloid leukemia (CML) progressed from the chronic phase to the blast crisis phase, and was associated with the poor prognosis of CML patients. In addition, the pharmacological inhibition of AMD1 by AO476 treatment resulted in a robust reduction of the progression of leukemic cells both in vitro and in vivo. Mechanistically, AMD1 depletion induced loss of mitochondrial membrane potential and accumulation of reactive oxygen species (ROS), resulting in the differentiation of LSCs via oxidative stress and aberrant activation of unfolded protein response (UPR) pathway, which was partially rescued by the addition of polyamine. These results indicate that AMD1 is an essential element in the progression of myeloid leukemia and could be an attractive target for the treatment of the disease.
Collapse
|
23
|
Chiremba TT, Neufeld KL. Constitutive Musashi1 expression impairs mouse postnatal development and intestinal homeostasis. Mol Biol Cell 2020; 32:28-44. [PMID: 33175598 PMCID: PMC8098822 DOI: 10.1091/mbc.e20-03-0206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Evolutionarily conserved RNA-binding protein Musashi1 (Msi1) can regulate developmentally relevant genes. Here we report the generation and characterization of a mouse model that allows inducible Msi1 overexpression in a temporal and tissue-specific manner. We show that ubiquitous Msi1 induction in ∼5-wk-old mice delays overall growth, alters organ-to-body proportions, and causes premature death. Msi1-overexpressing mice had shortened intestines, diminished intestinal epithelial cell (IEC) proliferation, and decreased growth of small intestine villi and colon crypts. Although Lgr5-positive intestinal stem cell numbers remained constant in Msi1-overexpressing tissue, an observed reduction in Cdc20 expression provided a potential mechanism underlying the intestinal growth defects. We further demonstrated that Msi1 overexpression affects IEC differentiation in a region-specific manner, with ileum tissue being influenced the most. Ilea of mutant mice displayed increased expression of enterocyte markers, but reduced expression of the goblet cell marker Mucin2 and fewer Paneth cells. A higher hairy and enhancer of split 1:mouse atonal homolog 1 ratio in ilea from Msi1-overexpressing mice implicated Notch signaling in inducing enterocyte differentiation. Together, this work implicates Msi1 in mouse postnatal development of multiple organs, with Notch signaling alterations contributing to intestinal defects. This new mouse model will be a useful tool to further elucidate the role of Msi1 in other tissue settings.
Collapse
Affiliation(s)
- Thelma T Chiremba
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045
| | - Kristi L Neufeld
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045
| |
Collapse
|
24
|
Allensworth-James ML, Odle AK, Lim J, LaGasse AN, Miles TK, Hardy LL, Haney AC, MacNicol MC, MacNicol AM, Childs GV. Metabolic signalling to somatotrophs: Transcriptional and post-transcriptional mediators. J Neuroendocrinol 2020; 32:e12883. [PMID: 32657474 PMCID: PMC8086172 DOI: 10.1111/jne.12883] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/17/2020] [Accepted: 06/10/2020] [Indexed: 12/19/2022]
Abstract
In normal individuals, pituitary somatotrophs optimise body composition by responding to metabolic signals from leptin. To identify mechanisms behind the regulation of somatotrophs by leptin, we used Cre-LoxP technology to delete leptin receptors (LEPR) selectively in somatotrophs and developed populations purified by fluorescence-activated cell sorting (FACS) that contained 99% somatotrophs. FACS-purified, Lepr-null somatotrophs showed reduced levels of growth hormone (GH), growth hormone-releasing hormone receptor (GHRHR), and Pou1f1 proteins and Gh (females) and Ghrhr (both sexes) mRNAs. Pure somatotrophs also expressed thyroid-stimulating hormone (TSH) and prolactin (PRL), both of which were reduced in pure somatotrophs lacking LEPR. This introduced five gene products that were targets of leptin. In the present study, we tested the hypothesis that leptin is both a transcriptional and a post-transcriptional regulator of these gene products. Our tests showed that Pou1f1 and/or the Janus kinase/signal transducer and activator of transcription 3 transcriptional regulatory pathways are implicated in the leptin regulation of Gh or Ghrhr mRNAs. We then focused on potential actions by candidate microRNAs (miRNAs) with consensus binding sites on the 3' UTR of Gh or Ghrhr mRNAs. Somatotroph Lepr-null deletion mutants expressed elevated levels of miRNAs including miR1197-3p (in females), miR103-3p and miR590-3p (both sexes), which bind Gh mRNA, or miRNA-325-3p (elevated in both sexes), which binds Ghrhr mRNA. This elevation indicates repression of translation in the absence of LEPR. In addition, after detecting binding sites for Musashi on Tshb and Prl 3' UTR, we determined that Musashi1 repressed translation of both mRNAs in in vitro fluc assays and that Prl mRNA was enriched in Musashi immunoprecipitation assays. Finally, we tested ghrelin actions to determine whether its nitric oxide-mediated signalling pathways would restore somatotroph functions in deletion mutants. Ghrelin did not restore either GHRH binding or GH secretion in vitro. These studies show an unexpectedly broad role for leptin with respect to maintaining somatotroph functions, including the regulation of PRL and TSH in subsets of somatotrophs that may be progenitor cells.
Collapse
Affiliation(s)
- Melody L Allensworth-James
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Angela K Odle
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Juchan Lim
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Alex N LaGasse
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Tiffany K Miles
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Linda L Hardy
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Anessa C Haney
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Melanie C MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Angus M MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Gwen V Childs
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
25
|
Orzechowska EJ, Katano T, Bialkowska AB, Yang VW. Interplay among p21 Waf1/Cip1, MUSASHI-1 and Krüppel-like factor 4 in activation of Bmi1-Cre ER reserve intestinal stem cells after gamma radiation-induced injury. Sci Rep 2020; 10:18300. [PMID: 33110120 PMCID: PMC7591575 DOI: 10.1038/s41598-020-75171-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 10/12/2020] [Indexed: 12/23/2022] Open
Abstract
Gamma radiation is a commonly used adjuvant treatment for abdominally localized cancer. Since its therapeutic potential is limited due to gastrointestinal (GI) syndrome, elucidation of the regenerative response following radiation-induced gut injury is needed to develop a preventive treatment. Previously, we showed that Krüppel-like factor 4 (KLF4) activates certain quiescent intestinal stem cells (ISCs) marked by Bmi1-CreER to give rise to regenerating crypts following γ irradiation. In the current study, we showed that γ radiation-induced expression of p21Waf1/Cip1 in Bmi1-CreER cells is likely mitigated by MUSASHI-1 (MSI1) acting as a negative regulator of p21Waf1/Cip1 mRNA translation, which promotes exit of the Bmi1-CreER cells from a quiescent state. Additionally, Bmi1-specific Klf4 deletion resulted in decreased numbers of MSI1+ cells in regenerating crypts compared to those of control mice. We showed that KLF4 binds to the Msi1 promoter and activates its expression in vitro. Since MSI1 has been shown to be crucial for crypt regeneration, this finding elucidates a pro-proliferative role of KLF4 during the postirradiation regenerative response. Taken together, our data suggest that the interplay among p21Waf1/Cip1, MSI1 and KLF4 regulates Bmi1-CreER cell survival, exit from quiescence and regenerative potential upon γ radiation-induced injury.
Collapse
Affiliation(s)
- Emilia J Orzechowska
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA.,Department of Molecular Biology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Takahito Katano
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA.,Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Agnieszka B Bialkowska
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA.
| | - Vincent W Yang
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA. .,Department of Physiology and Biophysics, Renaissance School of Medicine at Stony, Brook University, Stony Brook, NY, USA.
| |
Collapse
|
26
|
Lan L, Liu J, Xing M, Smith AR, Wang J, Wu X, Appelman C, Li K, Roy A, Gowthaman R, Karanicolas J, Somoza AD, Wang CCC, Miao Y, De Guzman R, Oakley BR, Neufeld KL, Xu L. Identification and Validation of an Aspergillus nidulans Secondary Metabolite Derivative as an Inhibitor of the Musashi-RNA Interaction. Cancers (Basel) 2020; 12:cancers12082221. [PMID: 32784494 PMCID: PMC7463734 DOI: 10.3390/cancers12082221] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 08/01/2020] [Accepted: 08/06/2020] [Indexed: 12/16/2022] Open
Abstract
RNA-binding protein Musashi-1 (MSI1) is a key regulator of several stem cell populations. MSI1 is involved in tumor proliferation and maintenance, and it regulates target mRNAs at the translational level. The known mRNA targets of MSI1 include Numb, APC, and P21WAF-1, key regulators of Notch/Wnt signaling and cell cycle progression, respectively. In this study, we aim to identify small molecule inhibitors of MSI1-mRNA interactions, which could block the growth of cancer cells with high levels of MSI1. Using a fluorescence polarization (FP) assay, we screened small molecules from several chemical libraries for those that disrupt the binding of MSI1 to its consensus RNA. One cluster of hit compounds is the derivatives of secondary metabolites from Aspergillus nidulans. One of the top hits, Aza-9, from this cluster was further validated by surface plasmon resonance and nuclear magnetic resonance spectroscopy, which demonstrated that Aza-9 binds directly to MSI1, and the binding is at the RNA binding pocket. We also show that Aza-9 binds to Musashi-2 (MSI2) as well. To test whether Aza-9 has anti-cancer potential, we used liposomes to facilitate Aza-9 cellular uptake. Aza-9-liposome inhibits proliferation, induces apoptosis and autophagy, and down-regulates Notch and Wnt signaling in colon cancer cell lines. In conclusion, we identified a series of potential lead compounds for inhibiting MSI1/2 function, while establishing a framework for identifying small molecule inhibitors of RNA binding proteins using FP-based screening methodology.
Collapse
Affiliation(s)
- Lan Lan
- Departments of Molecular Biosciences, the University of Kansas, Lawrence, KS 66045, USA; (L.L.); (J.L.); (A.R.S.); (X.W.); (C.A.); (K.L.); (R.D.G.); (B.R.O.); (K.L.N.)
| | - Jiajun Liu
- Departments of Molecular Biosciences, the University of Kansas, Lawrence, KS 66045, USA; (L.L.); (J.L.); (A.R.S.); (X.W.); (C.A.); (K.L.); (R.D.G.); (B.R.O.); (K.L.N.)
| | - Minli Xing
- Bio-NMR Core Facility, the University of Kansas, Lawrence, KS 66045, USA;
| | - Amber R. Smith
- Departments of Molecular Biosciences, the University of Kansas, Lawrence, KS 66045, USA; (L.L.); (J.L.); (A.R.S.); (X.W.); (C.A.); (K.L.); (R.D.G.); (B.R.O.); (K.L.N.)
| | - Jinan Wang
- Center for Computational Biology, the University of Kansas, Lawrence, KS 66045, USA; (J.W.); (R.G.); (Y.M.)
| | - Xiaoqing Wu
- Departments of Molecular Biosciences, the University of Kansas, Lawrence, KS 66045, USA; (L.L.); (J.L.); (A.R.S.); (X.W.); (C.A.); (K.L.); (R.D.G.); (B.R.O.); (K.L.N.)
| | - Carl Appelman
- Departments of Molecular Biosciences, the University of Kansas, Lawrence, KS 66045, USA; (L.L.); (J.L.); (A.R.S.); (X.W.); (C.A.); (K.L.); (R.D.G.); (B.R.O.); (K.L.N.)
| | - Ke Li
- Departments of Molecular Biosciences, the University of Kansas, Lawrence, KS 66045, USA; (L.L.); (J.L.); (A.R.S.); (X.W.); (C.A.); (K.L.); (R.D.G.); (B.R.O.); (K.L.N.)
| | - Anuradha Roy
- High Throughput Screening Laboratory, the University of Kansas, Lawrence, KS 66045, USA;
| | - Ragul Gowthaman
- Center for Computational Biology, the University of Kansas, Lawrence, KS 66045, USA; (J.W.); (R.G.); (Y.M.)
| | - John Karanicolas
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA;
| | - Amber D. Somoza
- Department of Chemistry, University of Southern California, Los Angeles, CA 90007, USA; (A.D.S.); (C.C.C.W.)
| | - Clay C. C. Wang
- Department of Chemistry, University of Southern California, Los Angeles, CA 90007, USA; (A.D.S.); (C.C.C.W.)
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90007, USA
| | - Yinglong Miao
- Center for Computational Biology, the University of Kansas, Lawrence, KS 66045, USA; (J.W.); (R.G.); (Y.M.)
| | - Roberto De Guzman
- Departments of Molecular Biosciences, the University of Kansas, Lawrence, KS 66045, USA; (L.L.); (J.L.); (A.R.S.); (X.W.); (C.A.); (K.L.); (R.D.G.); (B.R.O.); (K.L.N.)
| | - Berl R. Oakley
- Departments of Molecular Biosciences, the University of Kansas, Lawrence, KS 66045, USA; (L.L.); (J.L.); (A.R.S.); (X.W.); (C.A.); (K.L.); (R.D.G.); (B.R.O.); (K.L.N.)
| | - Kristi L. Neufeld
- Departments of Molecular Biosciences, the University of Kansas, Lawrence, KS 66045, USA; (L.L.); (J.L.); (A.R.S.); (X.W.); (C.A.); (K.L.); (R.D.G.); (B.R.O.); (K.L.N.)
- Department of Cancer Biology, the University of Kansas Cancer Center, Kansas City, KS 66160, USA
| | - Liang Xu
- Departments of Molecular Biosciences, the University of Kansas, Lawrence, KS 66045, USA; (L.L.); (J.L.); (A.R.S.); (X.W.); (C.A.); (K.L.); (R.D.G.); (B.R.O.); (K.L.N.)
- Department of Radiation Oncology, the University of Kansas Cancer Center, Kansas City, KS 66160, USA
- Correspondence:
| |
Collapse
|
27
|
Mendonça LS, Nóbrega C, Tavino S, Brinkhaus M, Matos C, Tomé S, Moreira R, Henriques D, Kaspar BK, Pereira de Almeida L. Ibuprofen enhances synaptic function and neural progenitors proliferation markers and improves neuropathology and motor coordination in Machado-Joseph disease models. Hum Mol Genet 2020; 28:3691-3703. [PMID: 31127937 DOI: 10.1093/hmg/ddz097] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 04/22/2019] [Accepted: 05/07/2019] [Indexed: 01/09/2023] Open
Abstract
Machado-Joseph disease or spinocerebellar ataxia type 3 is an inherited neurodegenerative disease associated with an abnormal glutamine over-repetition within the ataxin-3 protein. This mutant ataxin-3 protein affects several cellular pathways, leading to neuroinflammation and neuronal death in specific brain regions resulting in severe clinical manifestations. Presently, there is no therapy able to modify the disease progression. Nevertheless, anti-inflammatory pharmacological intervention has been associated with positive outcomes in other neurodegenerative diseases. Thus, the present work aimed at investigating whether ibuprofen treatment would alleviate Machado-Joseph disease. We found that ibuprofen-treated mouse models presented a significant reduction in the neuroinflammation markers, namely Il1b and TNFa mRNA and IKB-α protein phosphorylation levels. Moreover, these mice exhibited neuronal preservation, cerebellar atrophy reduction, smaller mutant ataxin-3 inclusions and motor performance improvement. Additionally, neural cultures of Machado-Joseph disease patients' induced pluripotent stem cells-derived neural stem cells incubated with ibuprofen showed increased levels of neural progenitors proliferation and synaptic markers such as MSI1, NOTCH1 and SYP. These findings were further confirmed in ibuprofen-treated mice that display increased neural progenitor numbers (Ki67 positive) in the subventricular zone. Furthermore, interestingly, ibuprofen treatment enhanced neurite total length and synaptic function of human neurons. Therefore, our results indicate that ibuprofen reduces neuroinflammation and induces neuroprotection, alleviating Machado-Joseph disease-associated neuropathology and motor impairments. Thus, our findings demonstrate that ibuprofen treatment has the potential to be used as a neuroprotective therapeutic approach in Machado-Joseph disease.
Collapse
Affiliation(s)
- Liliana S Mendonça
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Clévio Nóbrega
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Silvia Tavino
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Maximilian Brinkhaus
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Carlos Matos
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Sandra Tomé
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Ricardo Moreira
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Daniel Henriques
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Brian K Kaspar
- The Research Institute at Nationwide Children's Hospital, Ohio State University School of Medicine, Columbus, Ohio 43205, USA
| | - Luís Pereira de Almeida
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
28
|
Pötschke R, Gielen G, Pietsch T, Kramm C, Klusmann JH, Hüttelmaier S, Kühnöl CD. Musashi1 enhances chemotherapy resistance of pediatric glioblastoma cells in vitro. Pediatr Res 2020; 87:669-676. [PMID: 31756732 DOI: 10.1038/s41390-019-0628-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 09/25/2019] [Indexed: 11/09/2022]
Abstract
BACKGROUND Glioblastoma (GBM) is the most aggressive form of glioma in adults and children and is associated with very poor prognosis. Pediatric tumors are biologically distinct from adult GBM and differ in response to current GBM treatment protocols. Regarding pediatric GBM, new drug combinations and the molecular background of chemotherapy effects need to be investigated, in order to increase patient survival outcome. METHODS The expression of the RNA-binding protein Musashi1 (MSI1) in pediatric glioma samples of different WHO tumor grades was investigated on the protein (immunohistochemistry) and on the RNA level (publicly accessible RNA sequencing dataset). The impact of the chemotherapeutic temozolomide (TMZ) in combination with valproic acid (VPA) was tested in two pediatric glioblastoma-derived cell lines. The supportive effect of MSI1 expression against this treatment was investigated via transient knockdown and protein overexpression. RESULTS MSI1 expression correlates with pediatric high-grade glioma (HGG). The combination of TMZ with VPA significantly increases the impact of drug treatment on cell viability in vitro. MSI1 was found to promote drug resistance to the combined treatment with TMZ and VPA. CONCLUSION MSI1 expression is a potential marker for pediatric HGG and increases chemoresistance. Inhibition of MSI1 might lead to an improved patient outcome and therapy response.
Collapse
Affiliation(s)
- Rebecca Pötschke
- Molecular Cell Biology, Institute of Molecular Medicine, Martin-Luther-University, Halle (Saale), Germany.,Department of Pediatric Hematology/Oncology, University Hospital, Halle (Saale), Germany
| | - Gerrit Gielen
- Institute of Neuropathology, University Hospital, Bonn, Germany
| | - Torsten Pietsch
- Institute of Neuropathology, University Hospital, Bonn, Germany
| | - Christof Kramm
- Division of Pediatric Hematology and Oncology, University Medical Center, Göttingen, Germany
| | - Jan-Henning Klusmann
- Department of Pediatric Hematology/Oncology, University Hospital, Halle (Saale), Germany
| | - Stefan Hüttelmaier
- Molecular Cell Biology, Institute of Molecular Medicine, Martin-Luther-University, Halle (Saale), Germany.
| | - Caspar D Kühnöl
- Department of Pediatric Hematology/Oncology, University Hospital, Halle (Saale), Germany.
| |
Collapse
|
29
|
Montalbano M, McAllen S, Sengupta U, Puangmalai N, Bhatt N, Ellsworth A, Kayed R. Tau oligomers mediate aggregation of RNA-binding proteins Musashi1 and Musashi2 inducing Lamin alteration. Aging Cell 2019; 18:e13035. [PMID: 31532069 PMCID: PMC6826126 DOI: 10.1111/acel.13035] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 08/07/2019] [Accepted: 08/17/2019] [Indexed: 01/01/2023] Open
Abstract
The exact mechanisms leading to neurodegeneration in Alzheimer's disease (AD) and other tauopathies are not yet entirely understood. However, it is known that several RNA-binding proteins (RBPs) form toxic aggregates and also interact with tau in such granules in tauopathies, including AD. The Musashi (MSI) family of RBPs, consisting of two homologues: Musashi1 and Musashi2, have not been extensively investigated in neurodegenerative diseases. Here, using a tau inducible HEK (iHEK) model we investigate whether MSI proteins contribute to the aggregation of toxic tau oligomers (TauO). Wild-type and mutant P301L tau iHEK cells are used to study the effect of different tau variants on the cellular localization of MSI proteins. Interestingly, we observe that tau co-localizes with MSI in the cytoplasm and nuclei, altering the nuclear transport of MSI. Furthermore, incremental changes in the size and density of nuclear MSI/tau foci are observed. We also report here that TauO interact with MSI to cause the formation of distinct nuclear aggregates. Moreover, tau/MSI aggregates induce structural changes to LaminB1, leading to nuclear instability. These results illustrate a possible mechanism of neurodegeneration mediated by the aggregation of MSI proteins and TauO, suggesting that MSI plays a critical role in cellular dysfunction.
Collapse
Affiliation(s)
- Mauro Montalbano
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTXUSA
- Department of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTXUSA
| | - Salome McAllen
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTXUSA
- Department of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTXUSA
| | - Urmi Sengupta
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTXUSA
- Department of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTXUSA
| | - Nicha Puangmalai
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTXUSA
- Department of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTXUSA
| | - Nemil Bhatt
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTXUSA
- Department of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTXUSA
| | - Anna Ellsworth
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTXUSA
- Department of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTXUSA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTXUSA
- Department of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTXUSA
| |
Collapse
|
30
|
Abbaszadegan MR, Keyvani V, Moghbeli M. Genetic and molecular bases of esophageal Cancer among Iranians: an update. Diagn Pathol 2019; 14:97. [PMID: 31470870 PMCID: PMC6717340 DOI: 10.1186/s13000-019-0875-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 08/22/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Esophageal cancer is one of the leading causes of cancer related deaths among the Iranians. There is still a high ratio of mortality and low 5 years survival which are related to the late onset and diagnosis. Majority of patients refer for the treatment in advanced stages of tumor progression. MAIN BODY It is required to define an efficient local panel of diagnostic and prognostic markers for the Iranians. Indeed such efficient specific panel of markers will pave the way to decrease the mortality rate and increase the 5 years survival among the Iranian patients via the early diagnosis and targeted therapy. CONCLUSION in present review we have reported all of the molecular markers in different signaling pathways and cellular processes which have been assessed among the Iranian esophageal cancer patients until now.
Collapse
Affiliation(s)
| | - Vahideh Keyvani
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Meysam Moghbeli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
31
|
Cragle CE, MacNicol MC, Byrum SD, Hardy LL, Mackintosh SG, Richardson WA, Gray NK, Childs GV, Tackett AJ, MacNicol AM. Musashi interaction with poly(A)-binding protein is required for activation of target mRNA translation. J Biol Chem 2019; 294:10969-10986. [PMID: 31152063 PMCID: PMC6635449 DOI: 10.1074/jbc.ra119.007220] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 05/09/2019] [Indexed: 12/21/2022] Open
Abstract
The Musashi family of mRNA translational regulators controls both physiological and pathological stem cell self-renewal primarily by repressing target mRNAs that promote differentiation. In response to differentiation cues, Musashi can switch from a repressor to an activator of target mRNA translation. However, the molecular events that distinguish Musashi-mediated translational activation from repression are not understood. We have previously reported that Musashi function is required for the maturation of Xenopus oocytes and specifically for translational activation of specific dormant maternal mRNAs. Here, we employed MS to identify cellular factors necessary for Musashi-dependent mRNA translational activation. We report that Musashi1 needs to associate with the embryonic poly(A)-binding protein (ePABP) or the canonical somatic cell poly(A)-binding protein PABPC1 for activation of Musashi target mRNA translation. Co-immunoprecipitation studies demonstrated an increased Musashi1 interaction with ePABP during oocyte maturation. Attenuation of endogenous ePABP activity severely compromised Musashi function, preventing downstream signaling and blocking oocyte maturation. Ectopic expression of either ePABP or PABPC1 restored Musashi-dependent mRNA translational activation and maturation of ePABP-attenuated oocytes. Consistent with these Xenopus findings, PABPC1 remained associated with Musashi under conditions of Musashi target mRNA de-repression and translation during mammalian stem cell differentiation. Because association of Musashi1 with poly(A)-binding proteins has previously been implicated only in repression of Musashi target mRNAs, our findings reveal novel context-dependent roles for the interaction of Musashi with poly(A)-binding protein family members in response to extracellular cues that control cell fate.
Collapse
Affiliation(s)
- Chad E Cragle
- Department of Neurobiology and Developmental Sciences
| | - Melanie C MacNicol
- Department of Neurobiology and Developmental Sciences,; Center for Translational Neuroscience
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology,; Arkansas Children's Research Institute
| | - Linda L Hardy
- Department of Neurobiology and Developmental Sciences
| | | | - William A Richardson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom
| | - Nicola K Gray
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom
| | - Gwen V Childs
- Department of Neurobiology and Developmental Sciences,; Center for Translational Neuroscience
| | - Alan J Tackett
- Department of Biochemistry and Molecular Biology,; Arkansas Children's Research Institute
| | - Angus M MacNicol
- Department of Neurobiology and Developmental Sciences,; Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205 and.
| |
Collapse
|
32
|
Sengupta U, Montalbano M, McAllen S, Minuesa G, Kharas M, Kayed R. Formation of Toxic Oligomeric Assemblies of RNA-binding Protein: Musashi in Alzheimer's disease. Acta Neuropathol Commun 2018; 6:113. [PMID: 30367664 PMCID: PMC6203984 DOI: 10.1186/s40478-018-0615-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 10/10/2018] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder associated with structural and functional alterations of brain cells causing progressive deterioration of memory and other cognitive functions. Recent studies demonstrate that several neurodegenerative diseases, including AD exhibit RNA-binding proteins (RBPs) pathologies, including TAR DNA -binding protein (TDP-43), fused in sarcoma (FUS), superoxide dismutase (SOD1) and T-interacting antigen-1 (TIA-1), highlighting the role of RBPs in neurodegeneration. One such group of RBPs, Musashi proteins comprised of MSI1 and MSI2, has been long studied in neurogenesis and cancer biology. Herein, we have investigated the aggregation properties of MSI1 and MSI2 by in vitro assays, their expression and accumulation as well as their possible interactions with other cellular proteins, such as tau in AD pathology. We have performed atomic force microscopy, Western blot, and immunoprecipitation to demonstrate the aggregation properties of recombinant Musashi proteins. Furthermore, we have studied cortical brain sections from AD (N = 4) and age-matched non-demented subjects (N = 4) by Western blot and immunofluorescence microscopy to investigate MSI1 and MSI2 levels and their localization in human brain tissues. Musashi proteins showed in vitro aggregation properties by forming oligomers. We have observed an increase in Musashi proteins levels in AD brain tissues as compared with age-matched non-demented subjects. Moreover, Musashi proteins are observed to form oligomers in the diseased brain tissues. Interestingly, the co-immunofluorescence study has revealed a change in fluorescence pattern of oligomeric Musashi proteins and tau with a high association in the perinuclear area of the cells suggesting changes in function of Musashi proteins. Our data have demonstrated for the first time that MSI1 and MSI2 are present in an oligomeric state in AD brains compared to the age-matched non-demented subjects and that these large assemblies co-localize with tau contributing to the neurodegenerative pathogenesis.
Collapse
|
33
|
O'Valle F, de Buitrago JG, Hernández-Cortés P, Padial-Molina M, Crespo-Lora V, Cobo M, Aguilar D, Galindo-Moreno P. Increased Expression of Musashi-1 Evidences Mesenchymal Repair in Maxillary Sinus Floor Elevation. Sci Rep 2018; 8:12243. [PMID: 30116022 PMCID: PMC6095840 DOI: 10.1038/s41598-018-29908-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/17/2018] [Indexed: 11/21/2022] Open
Abstract
This study aimed to analyze the expression of Musashi-1 (MSI1) in maxillary native bone and grafted bone after maxillary sinus floor elevation. To do so, fifty-seven bone biopsies from 45 participants were studied. Eighteen samples were collected from native bone while 39 were obtained 6 months after maxillary sinus grafting procedures. Musashi-1 was analyzed by immunohistochemistry and RT-PCR. MSI1 was detected in osteoblasts and osteocytes in 97.4% (38/39) of grafted areas. In native bone, MSI1 was detected in only 66.6% (12/18) of the biopsies, mainly in osteocytes. Detection of MSI1 was significantly higher in osteoprogenitor mesenchymal cells of grafted biopsies (p < 0.001) but minor in smooth muscle and endothelial cells; no expression was detected in adipocytes. The mesenchymal cells of the non-mineralized tissue of native bone showed very low nuclear expression of MSI1, in comparison to fusiform cells in grafted areas (0.28(0.13) vs. 2.10(0.14), respectively; p < 0.001). Additionally, the detection of MSI1 mRNA was significantly higher in biopsies from grafted areas than those from native bone (1.00(0.51) vs. 60.34(35.2), respectively; p = 0.029). Thus, our results regardig the significantly higher detection of Musashi-1 in grafted sites than in native bone reflects its importance in the remodeling/repair events that occur after maxillary sinus floor elevation in humans.
Collapse
Affiliation(s)
- Francisco O'Valle
- Department of Pathology & Biopathology and Medicine Regenerative Institute (IBIMER, CIBM), University of Granada, Granada, Spain.,Institute of Biosanitary (Ibs-Granada), University of Granada, Granada, Spain
| | - Juan G de Buitrago
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, Granada, Spain
| | - Pedro Hernández-Cortés
- Department of Orthopedic Surgery, San Cecilio University Hospital of Granada, Granada, Spain
| | - Miguel Padial-Molina
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, Granada, Spain
| | - Vicente Crespo-Lora
- Department of Pathology & Biopathology and Medicine Regenerative Institute (IBIMER, CIBM), University of Granada, Granada, Spain
| | - Marien Cobo
- Department of Genomic Medicine & GENYO, Centre for Genomics and Oncological Research, Pfizer - University of Granada - Andalusian Regional Government, Granada, Spain
| | - David Aguilar
- Department of Pathology & Biopathology and Medicine Regenerative Institute (IBIMER, CIBM), University of Granada, Granada, Spain
| | - Pablo Galindo-Moreno
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, Granada, Spain.
| |
Collapse
|
34
|
Ma L, Shan Y, Ma H, Elshoura I, Nafees M, Yang K, Yin W. Identification of a novel splice variant of the human musashi-1 gene. Oncol Lett 2018; 16:5441-5448. [PMID: 30250616 DOI: 10.3892/ol.2018.9300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 02/28/2018] [Indexed: 11/06/2022] Open
Abstract
Musashi-1 (Msi1) is an evolutionarily conserved RNA-binding protein that has been reported to be the key regulator in malignancies and with involvement in cancer stemness. In the present study, a novel Msi1 transcript variant generated by alternative splicing was identified and termed Msi1 variant 2. This variant was observed to be ubiquitously expressed in cancerous and non-cancerous cells compared with its wild-type variant, which is preferentially expressed in cancer cells. Notably, the expression levels of Msi1 variant 2 were inversely associated with the protein expression levels of Msi1 in various cancer cells. This naturally truncated variant contains 899 nucleotides and a skipping event of exons 3 and 4, which leads to the emergence of a premature TGA stop codon in exon 5. The present results also demonstrated that hypoxia increased the resistance of H460 cells to cisplatin by suppressing the exon 3 and 4 skipping event of Msi1. In summary, the present study identified a novel splice variant of Msi1 lacking two complete RNA recognition motifs, and revealed the role of exon 3 and 4 skipping of Msi1 pre-mRNA in regulating cisplatin resistance under hypoxia. These observations indicate that targeting Msi1 alternative splicing could represent a valuable strategy to repress Msi1 signaling in tumors overexpressing this RNA-binding protein.
Collapse
Affiliation(s)
- Lin Ma
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, Jiangsu 210046, P.R. China
| | - Yating Shan
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, Jiangsu 210046, P.R. China
| | - Heliang Ma
- Department of Radiology, Jinan Central Hospital, Jinan, Shandong 250013, P.R. China
| | - Ihab Elshoura
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, Jiangsu 210046, P.R. China
| | - Muhammad Nafees
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210046, P.R. China
| | - Kaiyong Yang
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, Jiangsu 210046, P.R. China
| | - Wu Yin
- State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, Jiangsu 210046, P.R. China
| |
Collapse
|
35
|
Lan L, Liu H, Smith AR, Appelman C, Yu J, Larsen S, Marquez RT, Wu X, Liu FY, Gao P, Gowthaman R, Karanicolas J, De Guzman RN, Rogers S, Aubé J, Neufeld KL, Xu L. Natural product derivative Gossypolone inhibits Musashi family of RNA-binding proteins. BMC Cancer 2018; 18:809. [PMID: 30097032 PMCID: PMC6086024 DOI: 10.1186/s12885-018-4704-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 07/30/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The Musashi (MSI) family of RNA-binding proteins is best known for the role in post-transcriptional regulation of target mRNAs. Elevated MSI1 levels in a variety of human cancer are associated with up-regulation of Notch/Wnt signaling. MSI1 binds to and negatively regulates translation of Numb and APC (adenomatous polyposis coli), negative regulators of Notch and Wnt signaling respectively. METHODS Previously, we have shown that the natural product (-)-gossypol as the first known small molecule inhibitor of MSI1 that down-regulates Notch/Wnt signaling and inhibits tumor xenograft growth in vivo. Using a fluorescence polarization (FP) competition assay, we identified gossypolone (Gn) with a > 20-fold increase in Ki value compared to (-)-gossypol. We validated Gn binding to MSI1 using surface plasmon resonance, nuclear magnetic resonance, and cellular thermal shift assay, and tested the effects of Gn on colon cancer cells and colon cancer DLD-1 xenografts in nude mice. RESULTS In colon cancer cells, Gn reduced Notch/Wnt signaling and induced apoptosis. Compared to (-)-gossypol, the same concentration of Gn is less active in all the cell assays tested. To increase Gn bioavailability, we used PEGylated liposomes in our in vivo studies. Gn-lip via tail vein injection inhibited the growth of human colon cancer DLD-1 xenografts in nude mice, as compared to the untreated control (P < 0.01, n = 10). CONCLUSION Our data suggest that PEGylation improved the bioavailability of Gn as well as achieved tumor-targeted delivery and controlled release of Gn, which enhanced its overall biocompatibility and drug efficacy in vivo. This provides proof of concept for the development of Gn-lip as a molecular therapy for colon cancer with MSI1/MSI2 overexpression.
Collapse
Affiliation(s)
- Lan Lan
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA
| | - Hao Liu
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA
- Current address: School of Pharmacy, Southwest Medical University, Luzhou City, China
| | - Amber R Smith
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA
| | - Carl Appelman
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA
| | - Jia Yu
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, China
| | - Sarah Larsen
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA
| | - Rebecca T Marquez
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA
| | - Xiaoqing Wu
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA
| | - Frank Y Liu
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA
| | - Philip Gao
- Protein Production Group, NIH COBRE in Protein Structure and Function, Lawrence, USA
| | - Ragul Gowthaman
- Center for Computational Biology, University of Kansas, Lawrence, Kansas, USA
| | - John Karanicolas
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Roberto N De Guzman
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA
| | - Steven Rogers
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Jeffrey Aubé
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Kristi L Neufeld
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA
| | - Liang Xu
- Departments of Molecular Biosciences, University of Kansas, 4002 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS, 66045-7534, USA.
- Department of Radiation Oncology, University of Kansas Cancer Center, Kansas City, Kansas, USA.
| |
Collapse
|
36
|
Liu L, Qiu F, Chen J, Wu D, Nong Q, Zhou Y, Lu J. Functional Polymorphism in the MSI1 Gene Promoter Confers a Decreased Risk of Lung Cancer in Chinese by Reducing MSI1 Expression. Curr Genomics 2018; 19:375-383. [PMID: 30065613 PMCID: PMC6030856 DOI: 10.2174/1389202919666171128151544] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 12/12/2016] [Accepted: 01/29/2017] [Indexed: 12/15/2022] Open
Abstract
Background: Musashi1 (MSI1) is a characteristic stem cell marker that regulates the balance between cell self-renewal and differentiation. Evidence has identified MSI1 as a pivotal oncogenic regulator in diverse malignancies. However, little evidence uncovers the role of genetic variations of MSI1 gene in cancer etiology. Objective: The aim of this study was to investigate the association between genetic variants in the MSI1 gene and lung cancer risk. Methods: Based on a two-stage retrospective study with a total of 1559 patients with lung cancer and 1667 healthy controls, we evaluated the relevance between three putative functional SNPs in the MSI1 promoter (i.e., -2696T>C[rs7959801], -2297T>C[rs3742038] and -1081C>T[rs34570155]) and lung cancer risk. Results: We found that the SNP rs7959801T>C was significantly associated with lung cancer susceptibility. Compared to those with rs7959801TT wild-genotype, individuals with CT/CC variant genotypes exerted consistently beneficial roles in lung cancer risk in the discovery set (adjusted odd ratios [OR] = 0.67; 95% confidence interval [CI] = 0.57-0.80), and in the validation set (OR=0.69; 95%CI=0.54-0.88). Functional assays indicated that the allele transformation from T to C in rs7959801 of MSI1 gene arrestingly decreased its transcription activity in vitro. Furthermore, the expression levels of MSI1 were significantly lower in the patients with CT/CC variants than in those who were with TT genotype. Conclusion: Our findings suggested that the rs7959801T>C polymorphism in the MSI1 promoter conferred a decreased risk to lung cancer by reducing the expression of MSI1 and it may be a promising indicator for lung cancer predisposition.
Collapse
Affiliation(s)
- Lin Liu
- The State Key Laboratory of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, 151 Yanjiangxi Road, Guangzhou, 510120, China.,The School of Public Health, The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, Guangzhou Medical University, 195 Dongfengxi Road, Guangzhou, 510182, China
| | - Fuman Qiu
- The State Key Laboratory of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, 151 Yanjiangxi Road, Guangzhou, 510120, China.,The School of Public Health, The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, Guangzhou Medical University, 195 Dongfengxi Road, Guangzhou, 510182, China
| | - Jiansong Chen
- The State Key Laboratory of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, 151 Yanjiangxi Road, Guangzhou, 510120, China.,The School of Public Health, The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, Guangzhou Medical University, 195 Dongfengxi Road, Guangzhou, 510182, China
| | - Di Wu
- The State Key Laboratory of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, 151 Yanjiangxi Road, Guangzhou, 510120, China.,The School of Public Health, The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, Guangzhou Medical University, 195 Dongfengxi Road, Guangzhou, 510182, China
| | - Qingqing Nong
- Department of Environmental Health, Guangxi Medical University, 22 Shuangyong road, Nanning530021, China
| | - Yifeng Zhou
- Department of Genetics, Medical College of Soochow University, 199 Renai road, Suzhou215123, China
| | - Jiachun Lu
- The State Key Laboratory of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, 151 Yanjiangxi Road, Guangzhou, 510120, China.,The School of Public Health, The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, Guangzhou Medical University, 195 Dongfengxi Road, Guangzhou, 510182, China
| |
Collapse
|
37
|
Chernoff EAG, Sato K, Salfity HVN, Sarria DA, Belecky-Adams T. Musashi and Plasticity of Xenopus and Axolotl Spinal Cord Ependymal Cells. Front Cell Neurosci 2018. [PMID: 29535610 PMCID: PMC5835034 DOI: 10.3389/fncel.2018.00045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The differentiated state of spinal cord ependymal cells in regeneration-competent amphibians varies between a constitutively active state in what is essentially a developing organism, the tadpole of the frog Xenopus laevis, and a quiescent, activatable state in a slowly growing adult salamander Ambystoma mexicanum, the Axolotl. Ependymal cells are epithelial in intact spinal cord of all vertebrates. After transection, body region ependymal epithelium in both Xenopus and the Axolotl disorganizes for regenerative outgrowth (gap replacement). Injury-reactive ependymal cells serve as a stem/progenitor cell population in regeneration and reconstruct the central canal. Expression patterns of mRNA and protein for the stem/progenitor cell-maintenance Notch signaling pathway mRNA-binding protein Musashi (msi) change with life stage and regeneration competence. Msi-1 is missing (immunohistochemistry), or at very low levels (polymerase chain reaction, PCR), in both intact regeneration-competent adult Axolotl cord and intact non-regeneration-competent Xenopus tadpole (Nieuwkoop and Faber stage 62+, NF 62+). The critical correlation for successful regeneration is msi-1 expression/upregulation after injury in the ependymal outgrowth and stump-region ependymal cells. msi-1 and msi-2 isoforms were cloned for the Axolotl as well as previously unknown isoforms of Xenopus msi-2. Intact Xenopus spinal cord ependymal cells show a loss of msi-1 expression between regeneration-competent (NF 50-53) and non-regenerating stages (NF 62+) and in post-metamorphosis froglets, while msi-2 displays a lower molecular weight isoform in non-regenerating cord. In the Axolotl, embryos and juveniles maintain Msi-1 expression in the intact cord. In the adult Axolotl, Msi-1 is absent, but upregulates after injury. Msi-2 levels are more variable among Axolotl life stages: rising between late tailbud embryos and juveniles and decreasing in adult cord. Cultures of regeneration-competent Xenopus tadpole cord and injury-responsive adult Axolotl cord ependymal cells showed an identical growth factor response. Epidermal growth factor (EGF) maintains mesenchymal outgrowth in vitro, the cells are proliferative and maintain msi-1 expression. Non-regeneration competent Xenopus ependymal cells, NF 62+, failed to attach or grow well in EGF+ medium. Ependymal Msi-1 expression in vivo and in vitro is a strong indicator of regeneration competence in the amphibian spinal cord.
Collapse
Affiliation(s)
- Ellen A G Chernoff
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Kazuna Sato
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Hai V N Salfity
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Deborah A Sarria
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Teri Belecky-Adams
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| |
Collapse
|
38
|
Yu X, Mi L, Dong J, Zou J. Long intergenic non-protein-coding RNA 1567 (LINC01567) acts as a "sponge" against microRNA-93 in regulating the proliferation and tumorigenesis of human colon cancer stem cells. BMC Cancer 2017; 17:716. [PMID: 29110645 PMCID: PMC5674857 DOI: 10.1186/s12885-017-3731-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 10/30/2017] [Indexed: 02/08/2023] Open
Abstract
Background Cancer stem cells (CSCs) are considered to be the major factor in tumor initiation, progression, metastasis, recurrence and chemoresistance. Maintaining the stemness and promoting differentiation of these cells involve various factors. Recently, long non-coding RNAs (lncRNAs) have been identified as new regulatory factors in human cancer cells. However, the function of lncRNAs in colon CSCs is still unknown. Methods Primary colon cancer cells were maintained in serum-free medium to form spheres and CD133+/CD166+/CD44+ spheroid cells were selected using FACS technique. Then we detected growth curve, colony formation, invasion and migration ability, and tumorigenicity of CD133+/CD166+/CD44+ cells. LOCCS-siRNA and pcDNA-LOCCS plasmid vectors were constructed and transfected to evaluate impact of the lncRNA. We also performed dual luciferase reporter assay to verify the interaction of LOCCS and miR-93. Results The research explored lncRNA expression and the regulatory role of novel lncRNAs in colon CSCs. Using the stem cell markers CD133, CD166 and CD44, we found a subpopulation of highly tumorigenic human colon cancer cells. They displayed some characteristics of stem cells, including the ability to proliferate and form colonies, to resist chemotherapeutic drugs, and to produce xenografts in nude mice. We also found an lncRNA, LOCCS, with obviously upregulated expression in colon CSCs. Knockdown of LOCCS reduced cell proliferation, invasion, migration, and generation of tumor xenografts. Furthermore, microRNA-93 (miR-93) and Musashi-1 mediated the tumor suppression of LOCCS knockdown. Conclusions There was reciprocal repression between LOCCS and miR-93. Research on mechanisms suggested direct binding, as a predicted miR-93 binding site was identified in LOCCS. This comprehensive analysis of LOCCS in colon CSCs provides insight for elucidating important roles of the lncRNA–microRNA functional network in human colon cancer. Electronic supplementary material The online version of this article (10.1186/s12885-017-3731-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaofeng Yu
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, West Yan'an Road 221, Shanghai, 200040, China
| | - Lin Mi
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, West Yan'an Road 221, Shanghai, 200040, China
| | - Jie Dong
- Drug Clinical Trial Organization Office, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, China
| | - Jian Zou
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, West Yan'an Road 221, Shanghai, 200040, China.
| |
Collapse
|
39
|
Mukohyama J, Shimono Y, Minami H, Kakeji Y, Suzuki A. Roles of microRNAs and RNA-Binding Proteins in the Regulation of Colorectal Cancer Stem Cells. Cancers (Basel) 2017; 9:cancers9100143. [PMID: 29064439 PMCID: PMC5664082 DOI: 10.3390/cancers9100143] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 10/17/2017] [Accepted: 10/17/2017] [Indexed: 12/28/2022] Open
Abstract
Colorectal cancer stem cells (CSCs) are responsible for the initiation, progression and metastasis of human colorectal cancers, and have been characterized by the expression of cell surface markers, such as CD44, CD133, CD166 and LGR5. MicroRNAs (miRNAs) are differentially expressed between CSCs and non-tumorigenic cancer cells, and play important roles in the maintenance and regulation of stem cell properties of CSCs. RNA binding proteins (RBPs) are emerging epigenetic regulators of various RNA processing events, such as splicing, localization, stabilization and translation, and can regulate various types of stem cells. In this review, we summarize current evidences on the roles of miRNA and RBPs in the regulation of colorectal CSCs. Understanding the epigenetic regulation of human colorectal CSCs will help to develop biomarkers for colorectal cancers and to identify targets for CSC-targeting therapies.
Collapse
Affiliation(s)
- Junko Mukohyama
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan.
- Division of Gastrointestinal Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan.
- Department of Pathology and Cell Biology, Department of Medicine (Division of Digestive and Liver Diseases) and Herbert Irving Comprehensive Cancer Center (HICCC), Columbia University, New York, NY 10032, USA.
| | - Yohei Shimono
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan.
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine, Kobe, Hyogo 6500017, Japan.
| | - Hironobu Minami
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine, Kobe, Hyogo 6500017, Japan.
| | - Yoshihiro Kakeji
- Division of Gastrointestinal Surgery, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan.
| | - Akira Suzuki
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan.
| |
Collapse
|
40
|
MacNicol MC, Cragle CE, McDaniel FK, Hardy LL, Wang Y, Arumugam K, Rahmatallah Y, Glazko GV, Wilczynska A, Childs GV, Zhou D, MacNicol AM. Evasion of regulatory phosphorylation by an alternatively spliced isoform of Musashi2. Sci Rep 2017; 7:11503. [PMID: 28912529 PMCID: PMC5599597 DOI: 10.1038/s41598-017-11917-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 09/01/2017] [Indexed: 01/06/2023] Open
Abstract
The Musashi family of RNA binding proteins act to promote stem cell self-renewal and oppose cell differentiation predominantly through translational repression of mRNAs encoding pro-differentiation factors and inhibitors of cell cycle progression. During tissue development and repair however, Musashi repressor function must be dynamically regulated to allow cell cycle exit and differentiation. The mechanism by which Musashi repressor function is attenuated has not been fully established. Our prior work indicated that the Musashi1 isoform undergoes site-specific regulatory phosphorylation. Here, we demonstrate that the canonical Musashi2 isoform is subject to similar regulated site-specific phosphorylation, converting Musashi2 from a repressor to an activator of target mRNA translation. We have also characterized a novel alternatively spliced, truncated isoform of human Musashi2 (variant 2) that lacks the sites of regulatory phosphorylation and fails to promote translation of target mRNAs. Consistent with a role in opposing cell cycle exit and differentiation, upregulation of Musashi2 variant 2 was observed in a number of cancers and overexpression of the Musashi2 variant 2 isoform promoted cell transformation. These findings indicate that alternately spliced isoforms of the Musashi protein family possess distinct functional and regulatory properties and suggest that differential expression of Musashi isoforms may influence cell fate decisions.
Collapse
Affiliation(s)
- Melanie C MacNicol
- University of Arkansas for Medical Sciences, Department of Neurobiology and Developmental Sciences, 4301 W. Markham, Little Rock, 72205, AR, USA.,University of Arkansas for Medical Science, Center for Translational Neuroscience, 4301 W. Markham, Little Rock, 72205, AR, USA
| | - Chad E Cragle
- University of Arkansas for Medical Sciences, Department of Neurobiology and Developmental Sciences, 4301 W. Markham, Little Rock, 72205, AR, USA
| | - F Kennedy McDaniel
- University of Arkansas for Medical Sciences, Department of Neurobiology and Developmental Sciences, 4301 W. Markham, Little Rock, 72205, AR, USA
| | - Linda L Hardy
- University of Arkansas for Medical Sciences, Department of Neurobiology and Developmental Sciences, 4301 W. Markham, Little Rock, 72205, AR, USA
| | - Yan Wang
- University of Arkansas for Medical Sciences, Department of Pharmaceutical Sciences, 4301 W. Markham, Little Rock, 72205, AR, USA.,Department of Orthopedics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510182, PR China
| | - Karthik Arumugam
- University of Arkansas for Medical Sciences, Department of Physiology and Biophysics, 4301 W. Markham, Little Rock, 72205, AR, USA.,Center for Genomic Regulation, Department of Gene Regulation, Stem Cells and Cancer, C/Dr. Aiguader 88, 08003, Barcelona, Spain
| | - Yasir Rahmatallah
- University of Arkansas for Medical Sciences, Department of Biomedical Informatics, 4301 W. Markham, Little Rock, 72205, AR, USA
| | - Galina V Glazko
- University of Arkansas for Medical Sciences, Department of Biomedical Informatics, 4301 W. Markham, Little Rock, 72205, AR, USA
| | | | - Gwen V Childs
- University of Arkansas for Medical Sciences, Department of Neurobiology and Developmental Sciences, 4301 W. Markham, Little Rock, 72205, AR, USA.,University of Arkansas for Medical Science, Center for Translational Neuroscience, 4301 W. Markham, Little Rock, 72205, AR, USA
| | - Daohong Zhou
- University of Arkansas for Medical Sciences, Department of Pharmaceutical Sciences, 4301 W. Markham, Little Rock, 72205, AR, USA
| | - Angus M MacNicol
- University of Arkansas for Medical Sciences, Department of Neurobiology and Developmental Sciences, 4301 W. Markham, Little Rock, 72205, AR, USA. .,Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W. Markham, Little Rock, AR, 72205, United States.
| |
Collapse
|
41
|
Structural Insight into the Recognition of r(UAG) by Musashi-1 RBD2, and Construction of a Model of Musashi-1 RBD1-2 Bound to the Minimum Target RNA. Molecules 2017; 22:molecules22071207. [PMID: 28753936 PMCID: PMC6152312 DOI: 10.3390/molecules22071207] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 07/14/2017] [Accepted: 07/14/2017] [Indexed: 12/23/2022] Open
Abstract
Musashi-1 (Msi1) controls the maintenance of stem cells and tumorigenesis through binding to its target mRNAs and subsequent translational regulation. Msi1 has two RNA-binding domains (RBDs), RBD1 and RBD2, which recognize r(GUAG) and r(UAG), respectively. These minimal recognition sequences are connected by variable linkers in the Msi1 target mRNAs, however, the molecular mechanism by which Msi1 recognizes its targets is not yet understood. We previously determined the solution structure of the Msi1 RBD1:r(GUAGU) complex. Here, we determined the first structure of the RBD2:r(GUAGU) complex. The structure revealed that the central trinucleotide, r(UAG), is specifically recognized by the intermolecular hydrogen-bonding and aromatic stacking interactions. Importantly, the C-terminal region, which is disordered in the free form, took a certain conformation, resembling a helix. The observation of chemical shift perturbation and intermolecular NOEs, together with increases in the heteronuclear steady-state {1H}-15N NOE values on complex formation, indicated the involvement of the C-terminal region in RNA binding. On the basis of the two complex structures, we built a structural model of consecutive RBDs with r(UAGGUAG) containing both minimal recognition sequences, which resulted in no steric hindrance. The model suggests recognition of variable lengths (n) of the linker up to n = 50 may be possible.
Collapse
|
42
|
Weill L, Belloc E, Castellazzi CL, Méndez R. Musashi 1 regulates the timing and extent of meiotic mRNA translational activation by promoting the use of specific CPEs. Nat Struct Mol Biol 2017; 24:672-681. [DOI: 10.1038/nsmb.3434] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 06/14/2017] [Indexed: 12/31/2022]
|
43
|
Chavali PL, Stojic L, Meredith LW, Joseph N, Nahorski MS, Sanford TJ, Sweeney TR, Krishna BA, Hosmillo M, Firth AE, Bayliss R, Marcelis CL, Lindsay S, Goodfellow I, Woods CG, Gergely F. Neurodevelopmental protein Musashi-1 interacts with the Zika genome and promotes viral replication. Science 2017; 357:83-88. [PMID: 28572454 PMCID: PMC5798584 DOI: 10.1126/science.aam9243] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 05/16/2017] [Indexed: 12/21/2022]
Abstract
A recent outbreak of Zika virus in Brazil has led to a simultaneous increase in reports of neonatal microcephaly. Zika targets cerebral neural precursors, a cell population essential for cortical development, but the cause of this neurotropism remains obscure. Here we report that the neural RNA-binding protein Musashi-1 (MSI1) interacts with the Zika genome and enables viral replication. Zika infection disrupts the binding of MSI1 to its endogenous targets, thereby deregulating expression of factors implicated in neural stem cell function. We further show that MSI1 is highly expressed in neural progenitors of the human embryonic brain and is mutated in individuals with autosomal recessive primary microcephaly. Selective MSI1 expression in neural precursors could therefore explain the exceptional vulnerability of these cells to Zika infection.
Collapse
Affiliation(s)
- Pavithra L Chavali
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UK
| | - Lovorka Stojic
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UK
| | - Luke W Meredith
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 2QQ, UK
| | - Nimesh Joseph
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UK
| | - Michael S Nahorski
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK
| | - Thomas J Sanford
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 2QQ, UK
| | - Trevor R Sweeney
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 2QQ, UK
| | - Ben A Krishna
- Department of Medicine, University of Cambridge, Hills Road, Cambridge CB2 2QQ, UK
| | - Myra Hosmillo
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 2QQ, UK
| | - Andrew E Firth
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 2QQ, UK
| | - Richard Bayliss
- Faculty of Biological Sciences, Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Carlo L Marcelis
- Department of Human Genetics, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Susan Lindsay
- Institute of Genetic Medicine, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Ian Goodfellow
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 2QQ, UK
| | - C Geoffrey Woods
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK
| | - Fanni Gergely
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UK.
| |
Collapse
|
44
|
Kharas MG, Lengner CJ. Stem Cells, Cancer, and MUSASHI in Blood and Guts. Trends Cancer 2017; 3:347-356. [PMID: 28718412 DOI: 10.1016/j.trecan.2017.03.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 03/17/2017] [Accepted: 03/20/2017] [Indexed: 01/08/2023]
Abstract
The mammalian MSI family of RNA-binding proteins (RBPs) have important roles as oncoproteins in an array of tumor types, including leukemias, glioblastomas, and pancreatic, breast, lung, and colorectal cancers. The mammalian Msi genes, Msi1 and Msi2, have been most thoroughly investigated in two highly proliferative tissues prone to oncogenic transformation: the hematopoietic lineage and the intestinal epithelium. Despite their vast phenotypic differences, MSI proteins appear to have an analogous role in governing the stem cell compartment in both of these tissues, potentially providing a paradigm for a broader understanding of MSI function and oncogenic activities. In this review, we focus on the function of MSI in the blood and the intestine, and discuss therapeutic strategies for targeting this pathway.
Collapse
Affiliation(s)
- Michael G Kharas
- Molecular Pharmacology Program and Center for Cell Engineering, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA.
| | - Christopher J Lengner
- Department of Biomedical Sciences, School of Veterinary Medicine, and Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19146, USA.
| |
Collapse
|
45
|
Peters D, Radine C, Reese A, Budach W, Sohn D, Jänicke RU. The DEAD-box RNA helicase DDX41 is a novel repressor of p21 WAF1/CIP1 mRNA translation. J Biol Chem 2017; 292:8331-8341. [PMID: 28348086 DOI: 10.1074/jbc.m116.772327] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/17/2017] [Indexed: 12/31/2022] Open
Abstract
The cyclin-dependent kinase inhibitor p21 is an important player in stress pathways exhibiting both tumor-suppressive and oncogenic functions. Thus, expression of p21 has to be tightly controlled, which is achieved by numerous mechanisms at the transcriptional, translational, and posttranslational level. Performing immunoprecipitation of bromouridine-labeled p21 mRNAs that had been incubated before with cytoplasmic extracts of untreated HCT116 colon carcinoma cells, we identified the DEAD-box RNA helicase DDX41 as a novel regulator of p21 expression. DDX41 specifically precipitates with the 3'UTR, but not with the 5'UTR, of p21 mRNA. Knockdown of DDX41 increases basal and γ irradiation-induced p21 protein levels without affecting p21 mRNA expression. Conversely, overexpression of DDX41 strongly inhibits expression of a FLAG-p21 and a luciferase construct, but only in the presence of the p21 3'UTR. Together, these data suggest that this helicase regulates p21 expression at the translational level independent of the transcriptional activity of p53. However, knockdown of DDX41 completely fails to increase p21 protein levels in p53-deficient HCT116 cells. Moreover, posttranslational up-regulation of p21 achieved in both p53+/+ and p53-/- HCT116 cells in response to pharmaceutical inhibition of the proteasome (by MG-132) or p90 ribosomal S6 kinases (by BI-D1870) is further increased by knockdown of DDX41 only in p53-proficient but not in p53-deficient cells. Although our data demonstrate that DDX41 suppresses p21 translation without disturbing the function of p53 to directly induce p21 mRNA expression, this process indirectly requires p53, perhaps in the form of another p53 target gene or as a still undefined posttranscriptional function of p53.
Collapse
Affiliation(s)
- Dominik Peters
- Laboratory of Molecular Radiooncology, Clinic for Radiation Therapy and Radiooncology, Medical Faculty of the Heinrich Heine University, 40255 Düsseldorf, Germany
| | - Claudia Radine
- Laboratory of Molecular Radiooncology, Clinic for Radiation Therapy and Radiooncology, Medical Faculty of the Heinrich Heine University, 40255 Düsseldorf, Germany
| | - Alina Reese
- Laboratory of Molecular Radiooncology, Clinic for Radiation Therapy and Radiooncology, Medical Faculty of the Heinrich Heine University, 40255 Düsseldorf, Germany
| | - Wilfried Budach
- Laboratory of Molecular Radiooncology, Clinic for Radiation Therapy and Radiooncology, Medical Faculty of the Heinrich Heine University, 40255 Düsseldorf, Germany
| | - Dennis Sohn
- Laboratory of Molecular Radiooncology, Clinic for Radiation Therapy and Radiooncology, Medical Faculty of the Heinrich Heine University, 40255 Düsseldorf, Germany
| | - Reiner U Jänicke
- Laboratory of Molecular Radiooncology, Clinic for Radiation Therapy and Radiooncology, Medical Faculty of the Heinrich Heine University, 40255 Düsseldorf, Germany.
| |
Collapse
|
46
|
Shou Z, Jin X, He X, Zhao Z, Chen Y, Ye M, Yao J. Overexpression of Musashi-1 protein is associated with progression and poor prognosis of gastric cancer. Oncol Lett 2017; 13:3556-3566. [PMID: 28521458 PMCID: PMC5431268 DOI: 10.3892/ol.2017.5879] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 01/12/2017] [Indexed: 12/13/2022] Open
Abstract
Musashi-1, an evolutionally conserved RNA-binding protein, has been implicated in the promotion of pathological stem cell proliferation, including tumorigenesis. The objective of the present study was to evaluate the expression of Musashi-1 protein and its implications in the progression and prognosis of gastric cancer. The expression level of Musashi-1 protein in gastric cancer was determined by western blotting and immunohistochemistry, and compared with the clinicopathological parameters. The present study revealed that the expression level of Musashi-1 protein in gastric cancer was significantly upregulated and correlated with the tumor size, tumor-node-metastasis (TNM) stage, Lauren classification, depth of invasion, vessel invasion, lymph node metastasis and distant metastasis. The mean survival time for patients with low expression levels of Musashi-1 was significantly longer compared with patients with high expression levels of Musashi-1. For each TNM stage, the mean survival time for patients with a low Musashi-1 expression levels was also significantly longer compared with patients with a high Musashi-1 expression level. Notably, TNM stage II patients with a low Musashi-1 expression level demonstrated a longer mean survival time compared with TNM stage I patients with high Musashi-1 expression level (56.8 vs. 42.3 months; P=0.001), and TNM stage III patients with low Musashi-1 expression level exhibited a longer mean survival time compared with TNM stage II patients with a high Musashi-1 expression level (44.0 vs. 33.8 months; P=0.034). Multivariate Cox's regression test demonstrated that Musashi-1 protein expression level was an independent prognostic indicator for the survival rate of the patients with gastric cancer. The results of the present study highlighted an important role for Musashi-1 protein in the progression of gastric cancer. The detection of the Musashi-1 protein expression level alone or in combination with TNM staging may aid the prediction of the prognosis of patients with gastric cancer.
Collapse
Affiliation(s)
- Zhangxuan Shou
- Department of Pharmaceutical Sciences, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Xue Jin
- Department of Pharmaceutical Sciences, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Xujun He
- Key Laboratory of Gastroenterology of Zhejiang, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Zhongsheng Zhao
- Department of Pathology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Yuan Chen
- Department of Pathology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Meihua Ye
- Department of Pathology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Jiong Yao
- Department of Medical Records and Statistics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
47
|
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: 196] [Impact Index Per Article: 24.5] [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
|
48
|
Lee J, An S, Choi YM, Lee J, Ahn KJ, Lee JH, Kim TJ, An IS, Bae S. Musashi-2 is a novel regulator of paclitaxel sensitivity in ovarian cancer cells. Int J Oncol 2016; 49:1945-1952. [PMID: 27600258 DOI: 10.3892/ijo.2016.3683] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 08/22/2016] [Indexed: 11/06/2022] Open
Abstract
As few prognostic markers and symptoms have been identified, ovarian cancer is typically diagnosed at an advanced stage, and a majority of patients will relapse and develop resistance to anticancer drugs such as paclitaxel. Musashi-2 (MSI2) is a regulator of gene translation and functions as an oncogenic protein and a marker of poor prognosis in various types of cancer. However, the biological and clinical significance of MSI2 in ovarian cancer remains unclear. Using a tissue microarray-based assay, we demonstrated that MSI2 was highly expressed in advanced, serous ovarian cancer tissues. In addition, MSI2-overexpressing ovarian cancer cells exhibited increased viability, proliferation and growth. We found that MSI2 was overexpressed in paclitaxel-resistant ovarian cancer SKOV3-TR cells but not in paclitaxel-sensitive cell lines. The loss of MSI2 expression in lentivirus-mediated stable MSI2 knockdown SKOV3-TR cells impaired paclitaxel resistance as determined using cell viability and apoptosis assays. In contrast, lentivirus-mediated MSI2 overexpression promoted the development of paclitaxel resistance in paclitaxel-sensitive ovarian cancer cells. The results of the present study are the first to demonstrate that MSI2 is a valuable marker of advanced, serous ovarian cancer and that MSI2 plays an important role in paclitaxel resistance.
Collapse
Affiliation(s)
- Jeongsuk Lee
- Research Institute for Molecular-Targeted Drugs, Department of Biological Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Sungkwan An
- Research Institute for Molecular-Targeted Drugs, Department of Biological Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Yeong Min Choi
- Research Institute for Molecular-Targeted Drugs, Department of Biological Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Junwoo Lee
- Research Institute for Molecular-Targeted Drugs, Department of Biological Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Kyu Joong Ahn
- Department of Dermatology, Konkuk University School of Medicine, Seoul 05030, Republic of Korea
| | - Jae Ho Lee
- Laboratory of Molecular Oncology, Cheil General Hospital and Women's Healthcare Center, Dankook University College of Medicine, Seoul 04619, Republic of Korea
| | - Tae Jin Kim
- Department of Obstetrics and Gynecology, Cheil General Hospital and Women's Healthcare Center, Dankook University College of Medicine, Seoul 04619, Republic of Korea
| | - In-Sook An
- Gene Cell Pharm Corporation, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea
| | - Seunghee Bae
- Research Institute for Molecular-Targeted Drugs, Department of Biological Engineering, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
49
|
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: 40] [Impact Index Per Article: 4.4] [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
|
50
|
Moghbeli M, Rad A, Farshchian M, Taghehchian N, Gholamin M, Abbaszadegan MR. Correlation Between Meis1 and Msi1 in Esophageal Squamous Cell Carcinoma. J Gastrointest Cancer 2016; 47:273-277. [PMID: 27142513 DOI: 10.1007/s12029-016-9824-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
PURPOSE Homeobox (HOX) transcription factors are critical regulators of cell fate, stem cell functions, and gastrointestinal development. They require three-amino acid loop extension (TALE) homeodomain proteins such as Meis1 to enhance their transcriptional efficiencies. There are complicated associations between different signaling pathways such as the Wnt and NOTCH and tumor progression. It has been investigated that GSK-3 as an important component of the Wnt pathway facilitates the expression of HOX target genes. Therefore, in the present study, we assessed the probable correlation between Wnt, NOTCH, and HOX genes in esophageal squamous cell carcinoma (ESCC) progression and metastasis through the correlational study between the Msi1 as an important activator for both of the NOTCH and Wnt pathways and Meis1. METHODS Levels of Meis1 and Msi1 messenger RNA (mRNA) expression in 51 ESCC patients were compared to the normal tissues using real-time polymerase chain reaction. RESULTS Only 3 out of 51 (5.9 %) cases had Meis1/Msi1 overexpression and also 3/51 (5.9 %) cases had Meis1/Msi1 underexpression. There was a significant correlation between the Msi1 and Mesi1 mRNA expression (p = 0.037). All of the Msi1/Meis1 underexpressed tumors were poorly differentiated (p = 0.003). Meis1 under/Msi1 overexpressed cases also were in T3 tumor depth of invasion (p = 0.019). And there was a significant correlation between the Msi1/Meis1 underexpression and gender (p = 0.045). CONCLUSIONS Our results show that Meis1 may have a positive feedback with Msi1 during the ESCC progression.
Collapse
Affiliation(s)
- Meysam Moghbeli
- Division of Human Genetics, Immunology Research Center, Avicenna Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abolfazl Rad
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Moein Farshchian
- Molecular Medicine Research Department, ACECR-Khorasan Razavi Branch, Mashhad, Iran
| | - Negin Taghehchian
- Division of Human Genetics, Immunology Research Center, Avicenna Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehran Gholamin
- Division of Human Genetics, Immunology Research Center, Avicenna Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Abbaszadegan
- Division of Human Genetics, Immunology Research Center, Avicenna Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Medical Genetics Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|