1
|
Khan FA, Fang N, Zhang W, Ji S. The multifaceted role of Fragile X-Related Protein 1 (FXR1) in cellular processes: an updated review on cancer and clinical applications. Cell Death Dis 2024; 15:72. [PMID: 38238286 PMCID: PMC10796922 DOI: 10.1038/s41419-023-06413-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/22/2024]
Abstract
RNA-binding proteins (RBPs) modulate the expression level of several target RNAs (such as mRNAs) post-transcriptionally through interactions with unique binding sites in the 3'-untranslated region. There is mounting information that suggests RBP dysregulation plays a significant role in carcinogenesis. However, the function of FMR1 autosomal homolog 1(FXR1) in malignancies is just beginning to be unveiled. Due to the diversity of their RNA-binding domains and functional adaptability, FXR1 can regulate diverse transcript processing. Changes in FXR1 interaction with RNA networks have been linked to the emergence of cancer, although the theoretical framework defining these alterations in interaction is insufficient. Alteration in FXR1 expression or localization has been linked to the mRNAs of cancer suppressor genes, cancer-causing genes, and genes involved in genomic expression stability. In particular, FXR1-mediated gene regulation involves in several cellular phenomena related to cancer growth, metastasis, epithelial-mesenchymal transition, senescence, apoptosis, and angiogenesis. FXR1 dysregulation has been implicated in diverse cancer types, suggesting its diagnostic and therapeutic potential. However, the molecular mechanisms and biological effects of FXR1 regulation in cancer have yet to be understood. This review highlights the current knowledge of FXR1 expression and function in various cancer situations, emphasizing its functional variety and complexity. We further address the challenges and opportunities of targeting FXR1 for cancer diagnosis and treatment and propose future directions for FXR1 research in oncology. This work intends to provide an in-depth review of FXR1 as an emerging oncotarget with multiple roles and implications in cancer biology and therapy.
Collapse
Affiliation(s)
- Faiz Ali Khan
- Huaihe Hospital,Medical School, Henan University, Kaifeng, China
- Laboratory of Cell Signal Transduction, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Department of Basic Sciences Research, Shaukat Khanum Memorial Cancer Hospital and Research Centre (SKMCH&RC), Lahore, Pakistan
| | - Na Fang
- Huaihe Hospital,Medical School, Henan University, Kaifeng, China.
- Laboratory of Cell Signal Transduction, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China.
| | - Weijuan Zhang
- Huaihe Hospital,Medical School, Henan University, Kaifeng, China.
- Laboratory of Cell Signal Transduction, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China.
| | - Shaoping Ji
- Huaihe Hospital,Medical School, Henan University, Kaifeng, China.
- Laboratory of Cell Signal Transduction, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China.
- Zhengzhou Shuqing Medical College, Zhengzhou, China.
| |
Collapse
|
2
|
Shi DL, Grifone R. RNA-Binding Proteins in the Post-transcriptional Control of Skeletal Muscle Development, Regeneration and Disease. Front Cell Dev Biol 2021; 9:738978. [PMID: 34616743 PMCID: PMC8488162 DOI: 10.3389/fcell.2021.738978] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/31/2021] [Indexed: 12/21/2022] Open
Abstract
Embryonic myogenesis is a temporally and spatially regulated process that generates skeletal muscle of the trunk and limbs. During this process, mononucleated myoblasts derived from myogenic progenitor cells within the somites undergo proliferation, migration and differentiation to elongate and fuse into multinucleated functional myofibers. Skeletal muscle is the most abundant tissue of the body and has the remarkable ability to self-repair by re-activating the myogenic program in muscle stem cells, known as satellite cells. Post-transcriptional regulation of gene expression mediated by RNA-binding proteins is critically required for muscle development during embryogenesis and for muscle homeostasis in the adult. Differential subcellular localization and activity of RNA-binding proteins orchestrates target gene expression at multiple levels to regulate different steps of myogenesis. Dysfunctions of these post-transcriptional regulators impair muscle development and homeostasis, but also cause defects in motor neurons or the neuromuscular junction, resulting in muscle degeneration and neuromuscular disease. Many RNA-binding proteins, such as members of the muscle blind-like (MBNL) and CUG-BP and ETR-3-like factors (CELF) families, display both overlapping and distinct targets in muscle cells. Thus they function either cooperatively or antagonistically to coordinate myoblast proliferation and differentiation. Evidence is accumulating that the dynamic interplay of their regulatory activity may control the progression of myogenic program as well as stem cell quiescence and activation. Moreover, the role of RNA-binding proteins that regulate post-transcriptional modification in the myogenic program is far less understood as compared with transcription factors involved in myogenic specification and differentiation. Here we review past achievements and recent advances in understanding the functions of RNA-binding proteins during skeletal muscle development, regeneration and disease, with the aim to identify the fundamental questions that are still open for further investigations.
Collapse
Affiliation(s)
- De-Li Shi
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Developmental Biology Laboratory, CNRS-UMR 7622, Institut de Biologie de Paris-Seine, Sorbonne University, Paris, France
| | - Raphaëlle Grifone
- Developmental Biology Laboratory, CNRS-UMR 7622, Institut de Biologie de Paris-Seine, Sorbonne University, Paris, France
| |
Collapse
|
3
|
Smith JA, Curry EG, Blue RE, Roden C, Dundon SER, Rodríguez-Vargas A, Jordan DC, Chen X, Lyons SM, Crutchley J, Anderson P, Horb ME, Gladfelter AS, Giudice J. FXR1 splicing is important for muscle development and biomolecular condensates in muscle cells. J Cell Biol 2020; 219:133869. [PMID: 32328638 PMCID: PMC7147106 DOI: 10.1083/jcb.201911129] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/30/2020] [Accepted: 01/31/2020] [Indexed: 02/06/2023] Open
Abstract
Fragile-X mental retardation autosomal homologue-1 (FXR1) is a muscle-enriched RNA-binding protein. FXR1 depletion is perinatally lethal in mice, Xenopus, and zebrafish; however, the mechanisms driving these phenotypes remain unclear. The FXR1 gene undergoes alternative splicing, producing multiple protein isoforms and mis-splicing has been implicated in disease. Furthermore, mutations that cause frameshifts in muscle-specific isoforms result in congenital multi-minicore myopathy. We observed that FXR1 alternative splicing is pronounced in the serine- and arginine-rich intrinsically disordered domain; these domains are known to promote biomolecular condensation. Here, we show that tissue-specific splicing of fxr1 is required for Xenopus development and alters the disordered domain of FXR1. FXR1 isoforms vary in the formation of RNA-dependent biomolecular condensates in cells and in vitro. This work shows that regulation of tissue-specific splicing can influence FXR1 condensates in muscle development and how mis-splicing promotes disease.
Collapse
Affiliation(s)
- Jean A Smith
- Department of Biology, Stetson University, DeLand, FL.,Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Ennessa G Curry
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC.,Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - R Eric Blue
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Christine Roden
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Samantha E R Dundon
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT
| | - Anthony Rodríguez-Vargas
- Eugene Bell Center for Regenerative Biology and Tissue Engineering and National Xenopus Resource, Marine Biology Laboratory, Woods Hole, MA
| | - Danielle C Jordan
- Eugene Bell Center for Regenerative Biology and Tissue Engineering and National Xenopus Resource, Marine Biology Laboratory, Woods Hole, MA
| | - Xiaomin Chen
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Shawn M Lyons
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA.,Department of Medicine, Harvard Medical School, Boston, MA
| | - John Crutchley
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Paul Anderson
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA.,Department of Medicine, Harvard Medical School, Boston, MA
| | - Marko E Horb
- Eugene Bell Center for Regenerative Biology and Tissue Engineering and National Xenopus Resource, Marine Biology Laboratory, Woods Hole, MA
| | - Amy S Gladfelter
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC.,Eugene Bell Center for Regenerative Biology and Tissue Engineering and National Xenopus Resource, Marine Biology Laboratory, Woods Hole, MA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Jimena Giudice
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC.,Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC.,McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
4
|
Kim SS, Lee JA, Yeo MK. Reduction in Toxicity of Nano-Ag-Polyvinyl-pyrrolidone Using Hydra Proteins and Peptides during Zebrafish Embryogenesis. NANOMATERIALS 2019; 9:nano9091210. [PMID: 31462001 PMCID: PMC6780337 DOI: 10.3390/nano9091210] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 08/12/2019] [Accepted: 08/23/2019] [Indexed: 12/21/2022]
Abstract
Hydra magnipapillata cells reduce the toxicity of silver nanomaterials to zebrafish (Danio rerio) embryos. In this study, we investigated whether Hydra protein (HP) and Hydra basal disc peptide (Hym176) materials reduce nano-Ag-polyvinylpyrrolidone (N-Ag-PVP) toxicity during embryogenesis of the nanosensitive organism zebrafish. Protein (HP) was extracted from Hydra, and peptide (Hym176) was extracted from the hydra basal disc, which is attractive to nanomaterials and related to the immune system. The experimental conditions were exposure to N-Ag-PVP, HP, N-Ag-PVP+HP, Hym176, or N-Ag-PVP+Hym176 during embryo development. N-Ag-PVP+HP group showed lower toxicity than N-Ag-PVP group. In addition, in the N-Ag-PVP+HP group formed aggregated nanomaterials (≥200 nm size) through electrostatic bonding. In the gene expression profile, HP group differed in gene expression profile compared the other experimental groups and it was no genetic toxicity. HP showed a tendency to reduce side effects and abnormal gene expression produced by N-Ag-PVP with no evidence of inherent toxicity. Considering the potential nanotoxicity effects of released nanomaterials on the ecosystem, the reduction of nanotoxicity observed with HP natural materials should be regarded with great interest in terms of the overall health of the ecosystem.
Collapse
Affiliation(s)
- Soon Seok Kim
- Department of Environmental Science and Engineering, College of Engineering, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do Seoul 17104, Korea
| | - Jin Ah Lee
- Department of Environmental Science and Engineering, College of Engineering, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do Seoul 17104, Korea
| | - Min-Kyeong Yeo
- Department of Environmental Science and Engineering, College of Engineering, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do Seoul 17104, Korea.
| |
Collapse
|
5
|
Nikonova E, Kao SY, Ravichandran K, Wittner A, Spletter ML. Conserved functions of RNA-binding proteins in muscle. Int J Biochem Cell Biol 2019; 110:29-49. [PMID: 30818081 DOI: 10.1016/j.biocel.2019.02.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 02/21/2019] [Accepted: 02/23/2019] [Indexed: 12/13/2022]
Abstract
Animals require different types of muscle for survival, for example for circulation, motility, reproduction and digestion. Much emphasis in the muscle field has been placed on understanding how transcriptional regulation generates diverse types of muscle during development. Recent work indicates that alternative splicing and RNA regulation are as critical to muscle development, and altered function of RNA-binding proteins causes muscle disease. Although hundreds of genes predicted to bind RNA are expressed in muscles, many fewer have been functionally characterized. We present a cross-species view summarizing what is known about RNA-binding protein function in muscle, from worms and flies to zebrafish, mice and humans. In particular, we focus on alternative splicing regulated by the CELF, MBNL and RBFOX families of proteins. We discuss the systemic nature of diseases associated with loss of RNA-binding proteins in muscle, focusing on mis-regulation of CELF and MBNL in myotonic dystrophy. These examples illustrate the conservation of RNA-binding protein function and the marked utility of genetic model systems in understanding mechanisms of RNA regulation.
Collapse
Affiliation(s)
- Elena Nikonova
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-University München, Großhaderner Str. 9, 82152, Martinsried-Planegg, Germany
| | - Shao-Yen Kao
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-University München, Großhaderner Str. 9, 82152, Martinsried-Planegg, Germany
| | - Keshika Ravichandran
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-University München, Großhaderner Str. 9, 82152, Martinsried-Planegg, Germany
| | - Anja Wittner
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-University München, Großhaderner Str. 9, 82152, Martinsried-Planegg, Germany
| | - Maria L Spletter
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-University München, Großhaderner Str. 9, 82152, Martinsried-Planegg, Germany; Center for Integrated Protein Science Munich (CIPSM) at the Department of Chemistry, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
6
|
Recessive mutations in muscle-specific isoforms of FXR1 cause congenital multi-minicore myopathy. Nat Commun 2019; 10:797. [PMID: 30770808 PMCID: PMC6377633 DOI: 10.1038/s41467-019-08548-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 01/18/2019] [Indexed: 02/06/2023] Open
Abstract
FXR1 is an alternatively spliced gene that encodes RNA binding proteins (FXR1P) involved in muscle development. In contrast to other tissues, cardiac and skeletal muscle express two FXR1P isoforms that incorporate an additional exon-15. We report that recessive mutations in this particular exon of FXR1 cause congenital multi-minicore myopathy in humans and mice. Additionally, we show that while Myf5-dependent depletion of all FXR1P isoforms is neonatal lethal, mice carrying mutations in exon-15 display non-lethal myopathies which vary in severity depending on the specific effect of each mutation on the protein.
Collapse
|
7
|
Bao J, Ye C, Zheng Z, Zhou Z. Fmr1 protects cardiomyocytes against lipopolysaccharide-induced myocardial injury. Exp Ther Med 2018; 16:1825-1833. [PMID: 30186407 PMCID: PMC6122302 DOI: 10.3892/etm.2018.6386] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 02/16/2018] [Indexed: 12/22/2022] Open
Abstract
The present study explored the mechanisms by which fragile X mental retardation 1 (fmr1) overexpression inhibits lipopolysaccharide (LPS)-induced cardiomyocyte injury. Factors including oxidative stress reaction, mitochondrial membrane potential variation and cell apoptosis were evaluated. The viability of H9c2 cells was evaluated with a Cell Counting Kit-8 assay after cells were treated with LPS at different concentrations (0, 1, 3, 6 and 9 µg/ml) for various durations (4, 12 and 24 h). Flow cytometry was used to determine variations in reactive oxygen species (ROS), mitochondrial membrane potential and cell apoptosis. Reverse transcription-quantitative polymerase chain reaction and western blot analysis were performed to detect the levels of apoptosis-associated factors, and western blot analysis was used to determine the phosphorylation levels of phosphoinositide-3 kinase (PI3K), Akt and forkhead box (Fox)O3a. The results indicated that LPS decreased the viability of H9c2 cells in a dose- and time-dependent manner. Overexpression of fmr1 inhibited the LPS-induced decrease in the mitochondrial membrane potential and the production of ROS as well as apoptosis in H9c2 cells. Fmr1 also inhibited LPS-induced reductions in antioxidant enzyme activities, including those of superoxide dismutase and reduced/oxidized glutathione ratio, and decreased LPS-associated increases in the lipid peroxidation product malondialdehyde. Apoptosis-associated factors were identified to be involved in the effects of Fmr1. Overexpression of Fmr1 attenuated LPS-associated increases in the apoptosis-activating factors B-cell lymphoma 2 (Bcl-2)-associated X protein and caspase-3 and decreases in apoptosis inhibitors, including Bcl-2 and X-linked inhibitor of apoptosis protein. Fmr1 overexpression also reduced LPS-induced increases in the phosphorylation levels of PI3K, Akt and FoxO3a. In conclusion, fmr1 overexpression alleviated oxidative stress and apoptosis in H9c2 cardiomyocytes injured by LPS via regulating oxidative stress and apoptosis-associated factors, as well as the PI3K/Akt pathway. This information may provide a novel and effective therapeutic strategy for heart diseases.
Collapse
Affiliation(s)
- Jiasheng Bao
- Department of Electrocardiogram Diagnosis, Zhejiang Hospital, Hangzhou, Zhejiang 310013, P.R. China
| | - Chen Ye
- Department of Cardiovascular Medicine, Zhejiang Hospital, Hangzhou, Zhejiang 310013, P.R. China
| | - Zhelan Zheng
- Department of Cardiovascular Ultrasonic Center, The First Affiliated Hospital of Zhejiang University, The First Hospital of Zhejiang Province, Hangzhou, Zhejiang 310003, P.R. China
| | - Zhengwen Zhou
- Department of Electrocardiogram Diagnosis, Zhejiang Hospital, Hangzhou, Zhejiang 310013, P.R. China
| |
Collapse
|
8
|
Drozd M, Bardoni B, Capovilla M. Modeling Fragile X Syndrome in Drosophila. Front Mol Neurosci 2018; 11:124. [PMID: 29713264 PMCID: PMC5911982 DOI: 10.3389/fnmol.2018.00124] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 03/29/2018] [Indexed: 01/18/2023] Open
Abstract
Intellectual disability (ID) and autism are hallmarks of Fragile X Syndrome (FXS), a hereditary neurodevelopmental disorder. The gene responsible for FXS is Fragile X Mental Retardation gene 1 (FMR1) encoding the Fragile X Mental Retardation Protein (FMRP), an RNA-binding protein involved in RNA metabolism and modulating the expression level of many targets. Most cases of FXS are caused by silencing of FMR1 due to CGG expansions in the 5'-UTR of the gene. Humans also carry the FXR1 and FXR2 paralogs of FMR1 while flies have only one FMR1 gene, here called dFMR1, sharing the same level of sequence homology with all three human genes, but functionally most similar to FMR1. This enables a much easier approach for FMR1 genetic studies. Drosophila has been widely used to investigate FMR1 functions at genetic, cellular, and molecular levels since dFMR1 mutants have many phenotypes in common with the wide spectrum of FMR1 functions that underlay the disease. In this review, we present very recent Drosophila studies investigating FMRP functions at genetic, cellular, molecular, and electrophysiological levels in addition to research on pharmacological treatments in the fly model. These studies have the potential to aid the discovery of pharmacological therapies for FXS.
Collapse
Affiliation(s)
- Małgorzata Drozd
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France.,CNRS LIA (Neogenex), Valbonne, France
| | - Barbara Bardoni
- CNRS LIA (Neogenex), Valbonne, France.,Université Côte d'Azur, INSERM, CNRS, IPMC, Valbonne, France
| | - Maria Capovilla
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France.,CNRS LIA (Neogenex), Valbonne, France
| |
Collapse
|
9
|
Chu M, Novak SM, Cover C, Wang AA, Chinyere IR, Juneman EB, Zarnescu DC, Wong PK, Gregorio CC. Increased Cardiac Arrhythmogenesis Associated With Gap Junction Remodeling With Upregulation of RNA-Binding Protein FXR1. Circulation 2017; 137:605-618. [PMID: 29101288 DOI: 10.1161/circulationaha.117.028976] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 10/23/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Gap junction remodeling is well established as a consistent feature of human heart disease involving spontaneous ventricular arrhythmia. The mechanisms responsible for gap junction remodeling that include alterations in the distribution of, and protein expression within, gap junctions are still debated. Studies reveal that multiple transcriptional and posttranscriptional regulatory pathways are triggered in response to cardiac disease, such as those involving RNA-binding proteins. The expression levels of FXR1 (fragile X mental retardation autosomal homolog 1), an RNA-binding protein, are critical to maintain proper cardiac muscle function; however, the connection between FXR1 and disease is not clear. METHODS To identify the mechanisms regulating gap junction remodeling in cardiac disease, we sought to identify the functional properties of FXR1 expression, direct targets of FXR1 in human left ventricle dilated cardiomyopathy (DCM) biopsy samples and mouse models of DCM through BioID proximity assay and RNA immunoprecipitation, how FXR1 regulates its targets through RNA stability and luciferase assays, and functional consequences of altering the levels of this important RNA-binding protein through the analysis of cardiac-specific FXR1 knockout mice and mice injected with 3xMyc-FXR1 adeno-associated virus. RESULTS FXR1 expression is significantly increased in tissue samples from human and mouse models of DCM via Western blot analysis. FXR1 associates with intercalated discs, and integral gap junction proteins Cx43 (connexin 43), Cx45 (connexin 45), and ZO-1 (zonula occludens-1) were identified as novel mRNA targets of FXR1 by using a BioID proximity assay and RNA immunoprecipitation. Our findings show that FXR1 is a multifunctional protein involved in translational regulation and stabilization of its mRNA targets in heart muscle. In addition, introduction of 3xMyc-FXR1 via adeno-associated virus into mice leads to the redistribution of gap junctions and promotes ventricular tachycardia, showing the functional significance of FXR1 upregulation observed in DCM. CONCLUSIONS In DCM, increased FXR1 expression appears to play an important role in disease progression by regulating gap junction remodeling. Together this study provides a novel function of FXR1, namely, that it directly regulates major gap junction components, contributing to proper cell-cell communication in the heart.
Collapse
Affiliation(s)
- Miensheng Chu
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program (M.C., S.M.N., C.C., A.A.W., C.C.G.)
| | - Stefanie Mares Novak
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program (M.C., S.M.N., C.C., A.A.W., C.C.G.)
| | - Cathleen Cover
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program (M.C., S.M.N., C.C., A.A.W., C.C.G.)
| | - Anne A Wang
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program (M.C., S.M.N., C.C., A.A.W., C.C.G.)
| | | | | | | | - Pak Kin Wong
- University of Arizona, Tucson. Department of Biomedical Engineering at Pennsylvania State University, University Park (P.K.W.)
| | - Carol C Gregorio
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program (M.C., S.M.N., C.C., A.A.W., C.C.G.)
| |
Collapse
|
10
|
Fan Y, Yue J, Xiao M, Han-Zhang H, Wang YV, Ma C, Deng Z, Li Y, Yu Y, Wang X, Niu S, Hua Y, Weng Z, Atadja P, Li E, Xiang B. FXR1 regulates transcription and is required for growth of human cancer cells with TP53/FXR2 homozygous deletion. eLife 2017; 6:26129. [PMID: 28767039 PMCID: PMC5595435 DOI: 10.7554/elife.26129] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 08/01/2017] [Indexed: 11/25/2022] Open
Abstract
Tumor suppressor p53 prevents cell transformation by inducing apoptosis and other responses. Homozygous TP53 deletion occurs in various types of human cancers for which no therapeutic strategies have yet been reported. TCGA database analysis shows that the TP53 homozygous deletion locus mostly exhibits co-deletion of the neighboring gene FXR2, which belongs to the Fragile X gene family. Here, we demonstrate that inhibition of the remaining family member FXR1 selectively blocks cell proliferation in human cancer cells containing homozygous deletion of both TP53 and FXR2 in a collateral lethality manner. Mechanistically, in addition to its RNA-binding function, FXR1 recruits transcription factor STAT1 or STAT3 to gene promoters at the chromatin interface and regulates transcription thus, at least partially, mediating cell proliferation. Our study anticipates that inhibition of FXR1 is a potential therapeutic approach to targeting human cancers harboring TP53 homozygous deletion. Healthy human cells employ many tricks to avoid becoming cancerous. For example, they produce proteins known as tumor suppressors, which sense if a cell shows early signs of cancer and instruct the cell to die. A gene known as TP53 produces one of the most important tumor suppressor proteins, and this gene is inactive or missing in many types of human cancer. Treating cancers that have completely lost the TP53 gene is particularly difficult. One way to develop new treatments for these conditions would be to target other proteins that these cancers need to survive; but these proteins first need to be identified. Fan et al. have now identified one such protein in human cancer cells lacking TP53. Searching databases of DNA sequences from human cancer cells revealed that those without the TP53 gene often also lose a neighboring gene called FXR2. Cancer cells survive without FXR2 because a similar gene, called FXR1, can compensate. Fan et al. therefore decided to experimentally lower the activity of the FXR1 gene and, as expected, cancer cells without TP53 and FXR2 stopped growing. Normal cells, on the other hand, were unaffected by the deletion of the FXR1 gene since FXR2 is still there. This phenomenon, in which cancer cells become vulnerable after the loss of certain genes but only because they have already lost important tumor suppressors, is called “collateral lethality”. Further experiments showed that the protein encoded by FXR1 coordinates with other proteins to activate genes that contribute to cell growth. These findings suggest new ways to treat human cancers that have lost TP53. For example, if scientists can find small molecules that inhibit the protein encoded by FXR1 and show that these molecules can block the growth of tumors lacking TP53 and FXR2, this could eventually lead to a new anticancer drug. However, like any new drug, these small molecule inhibitors would also need to be extensively tested before they could be taken into human clinical trials.
Collapse
Affiliation(s)
- Yichao Fan
- Epigenetic Discovery, China Novartis Institutes for BioMedical Research, Shanghai, China
| | - Jiao Yue
- Epigenetic Discovery, China Novartis Institutes for BioMedical Research, Shanghai, China
| | - Mengtao Xiao
- Epigenetic Discovery, China Novartis Institutes for BioMedical Research, Shanghai, China
| | - Han Han-Zhang
- Epigenetic Discovery, China Novartis Institutes for BioMedical Research, Shanghai, China
| | - Yao Vickie Wang
- Epigenetic Discovery, China Novartis Institutes for BioMedical Research, Shanghai, China
| | - Chun Ma
- Epigenetic Discovery, China Novartis Institutes for BioMedical Research, Shanghai, China
| | - Zhilin Deng
- Epigenetic Discovery, China Novartis Institutes for BioMedical Research, Shanghai, China
| | - Yingxiang Li
- Department of Bioinformatics, Tongji University, Shanghai, China
| | - Yanyan Yu
- Epigenetic Discovery, China Novartis Institutes for BioMedical Research, Shanghai, China
| | - Xinghao Wang
- Epigenetic Discovery, China Novartis Institutes for BioMedical Research, Shanghai, China
| | - Shen Niu
- Epigenetic Discovery, China Novartis Institutes for BioMedical Research, Shanghai, China
| | - Youjia Hua
- Epigenetic Discovery, China Novartis Institutes for BioMedical Research, Shanghai, China
| | - Zhiping Weng
- Department of Bioinformatics, Tongji University, Shanghai, China
| | - Peter Atadja
- Epigenetic Discovery, China Novartis Institutes for BioMedical Research, Shanghai, China
| | - En Li
- Epigenetic Discovery, China Novartis Institutes for BioMedical Research, Shanghai, China
| | - Bin Xiang
- Epigenetic Discovery, China Novartis Institutes for BioMedical Research, Shanghai, China
| |
Collapse
|
11
|
Oldenburg A, Briand N, Sørensen AL, Cahyani I, Shah A, Moskaug JØ, Collas P. A lipodystrophy-causing lamin A mutant alters conformation and epigenetic regulation of the anti-adipogenic MIR335 locus. J Cell Biol 2017; 216:2731-2743. [PMID: 28751304 PMCID: PMC5584164 DOI: 10.1083/jcb.201701043] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 05/04/2017] [Accepted: 06/20/2017] [Indexed: 12/30/2022] Open
Abstract
Mutations in the Lamin A/C (LMNA) gene-encoding nuclear LMNA cause laminopathies, which include partial lipodystrophies associated with metabolic syndromes. The lipodystrophy-associated LMNA p.R482W mutation is known to impair adipogenic differentiation, but the mechanisms involved are unclear. We show in this study that the lamin A p.R482W hot spot mutation prevents adipogenic gene expression by epigenetically deregulating long-range enhancers of the anti-adipogenic MIR335 microRNA gene in human adipocyte progenitor cells. The R482W mutation results in a loss of function of differentiation-dependent lamin A binding to the MIR335 locus. This impairs H3K27 methylation and instead favors H3K27 acetylation on MIR335 enhancers. The lamin A mutation further promotes spatial clustering of MIR335 enhancer and promoter elements along with overexpression of the MIR355 gene after adipogenic induction. Our results link a laminopathy-causing lamin A mutation to an unsuspected deregulation of chromatin states and spatial conformation of an miRNA locus critical for adipose progenitor cell fate.
Collapse
Affiliation(s)
- Anja Oldenburg
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Nolwenn Briand
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Anita L Sørensen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Inswasti Cahyani
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Akshay Shah
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jan Øivind Moskaug
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway.,Norwegian Center for Stem Cell Research, Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Philippe Collas
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway .,Norwegian Center for Stem Cell Research, Department of Immunology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
12
|
Nutter CA, Jaworski E, Verma SK, Perez-Carrasco Y, Kuyumcu-Martinez MN. Developmentally regulated alternative splicing is perturbed in type 1 diabetic skeletal muscle. Muscle Nerve 2017; 56:744-749. [PMID: 28164326 DOI: 10.1002/mus.25599] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 01/19/2017] [Accepted: 01/31/2017] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Type 1 diabetic patients can develop skeletal muscle weakness and atrophy by molecular mechanisms that are not well understood. Alternative splicing (AS) is critical for gene expression in the skeletal muscle, and its dysregulation is implicated in muscle weakness and atrophy. Therefore, we investigated whether AS patterns are affected in type 1 diabetic skeletal muscle contributing to skeletal muscle defects. METHODS AS patterns were determined by reverse transcription-polymerase chain reaction and levels of RNA binding proteins were assessed by Western blot in type 1 diabetic mouse skeletal muscle and during normal mouse skeletal muscle development. RESULTS Five genes with critical functions in the skeletal muscle are misspliced in type 1 diabetic skeletal muscle, resembling their AS patterns at embryonic stages. AS of these genes undergoes dramatic transitions during skeletal muscle development, correlating with changes in specific RNA binding proteins. CONCLUSION Embryonic spliced variants are inappropriately expressed in type 1 diabetic skeletal muscle. Muscle Nerve 56: 744-749, 2017.
Collapse
Affiliation(s)
- Curtis A Nutter
- Biochemistry and Molecular Biology, University of Texas Medical Branch, 301 University Boulevard, Galveston, Texas, 77555, USA
| | - Elizabeth Jaworski
- Biochemistry and Molecular Biology, University of Texas Medical Branch, 301 University Boulevard, Galveston, Texas, 77555, USA
| | - Sunil K Verma
- Biochemistry and Molecular Biology, University of Texas Medical Branch, 301 University Boulevard, Galveston, Texas, 77555, USA
| | | | - Muge N Kuyumcu-Martinez
- Biochemistry and Molecular Biology, University of Texas Medical Branch, 301 University Boulevard, Galveston, Texas, 77555, USA.,Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, USA.,Institute for Translational Sciences University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
13
|
Gonzalez-Pena D, Gao G, Baranski M, Moen T, Cleveland BM, Kenney PB, Vallejo RL, Palti Y, Leeds TD. Genome-Wide Association Study for Identifying Loci that Affect Fillet Yield, Carcass, and Body Weight Traits in Rainbow Trout ( Oncorhynchus mykiss). Front Genet 2016; 7:203. [PMID: 27920797 PMCID: PMC5118429 DOI: 10.3389/fgene.2016.00203] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 11/02/2016] [Indexed: 11/22/2022] Open
Abstract
Fillet yield (FY, %) is an economically-important trait in rainbow trout aquaculture that affects production efficiency. Despite that, FY has received little attention in breeding programs because it is difficult to measure on a large number of fish and cannot be directly measured on breeding candidates. The recent development of a high-density SNP array for rainbow trout has provided the needed tool for studying the underlying genetic architecture of this trait. A genome-wide association study (GWAS) was conducted for FY, body weight at 10 (BW10) and 13 (BW13) months post-hatching, head-off carcass weight (CAR), and fillet weight (FW) in a pedigreed rainbow trout population selectively bred for improved growth performance. The GWAS analysis was performed using the weighted single-step GBLUP method (wssGWAS). Phenotypic records of 1447 fish (1.5 kg at harvest) from 299 full-sib families in three successive generations, of which 875 fish from 196 full-sib families were genotyped, were used in the GWAS analysis. A total of 38,107 polymorphic SNPs were analyzed in a univariate model with hatch year and harvest group as fixed effects, harvest weight as a continuous covariate, and animal and common environment as random effects. A new linkage map was developed to create windows of 20 adjacent SNPs for use in the GWAS. The two windows with largest effect for FY and FW were located on chromosome Omy9 and explained only 1.0-1.5% of genetic variance, thus suggesting a polygenic architecture affected by multiple loci with small effects in this population. One window on Omy5 explained 1.4 and 1.0% of the genetic variance for BW10 and BW13, respectively. Three windows located on Omy27, Omy17, and Omy9 (same window detected for FY) explained 1.7, 1.7, and 1.0%, respectively, of genetic variance for CAR. Among the detected 100 SNPs, 55% were located directly in genes (intron and exons). Nucleotide sequences of intragenic SNPs were blasted to the Mus musculus genome to create a putative gene network. The network suggests that differences in the ability to maintain a proliferative and renewable population of myogenic precursor cells may affect variation in growth and fillet yield in rainbow trout.
Collapse
Affiliation(s)
- Dianelys Gonzalez-Pena
- United States Department of Agriculture, National Center for Cool and Cold Water Aquaculture, Agricultural Research ServiceKearneysville, WV, USA
| | - Guangtu Gao
- United States Department of Agriculture, National Center for Cool and Cold Water Aquaculture, Agricultural Research ServiceKearneysville, WV, USA
| | | | | | - Beth M. Cleveland
- United States Department of Agriculture, National Center for Cool and Cold Water Aquaculture, Agricultural Research ServiceKearneysville, WV, USA
| | - P. Brett Kenney
- Division of Animal and Nutritional Sciences, West Virginia UniversityMorgantown, WV, USA
| | - Roger L. Vallejo
- United States Department of Agriculture, National Center for Cool and Cold Water Aquaculture, Agricultural Research ServiceKearneysville, WV, USA
| | - Yniv Palti
- United States Department of Agriculture, National Center for Cool and Cold Water Aquaculture, Agricultural Research ServiceKearneysville, WV, USA
| | - Timothy D. Leeds
- United States Department of Agriculture, National Center for Cool and Cold Water Aquaculture, Agricultural Research ServiceKearneysville, WV, USA
| |
Collapse
|
14
|
Ma Y, Tian S, Wang Z, Wang C, Chen X, Li W, Yang Y, He S. CMP‑N‑acetylneuraminic acid synthetase interacts with fragile X related protein 1. Mol Med Rep 2016; 14:1501-8. [PMID: 27357083 PMCID: PMC4940058 DOI: 10.3892/mmr.2016.5438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 05/25/2016] [Indexed: 11/30/2022] Open
Abstract
Fragile X mental retardation protein (FMRP), fragile X related 1 protein (FXR1P) and FXR2P are the members of the FMR protein family. These proteins contain two KH domains and a RGG box, which are characteristic of RNA binding proteins. The absence of FMRP, causes fragile X syndrome (FXS), the leading cause of hereditary mental retardation. FXR1P is expressed throughout the body and important for normal muscle development, and its absence causes cardiac abnormality. To investigate the functions of FXR1P, a screen was performed to identify FXR1P-interacting proteins and determine the biological effect of the interaction. The current study identified CMP-N-acetylneuraminic acid synthetase (CMAS) as an interacting protein using the yeast two-hybrid system, and the interaction between FXR1P and CMAS was validated in yeast using a β-galactosidase assay and growth studies with selective media. Furthermore, co-immunoprecipitation was used to analyze the FXR1P/CMAS association and immunofluorescence microscopy was performed to detect expression and intracellular localization of the proteins. The results of the current study indicated that FXR1P and CMAS interact, and colocalize in the cytoplasm and the nucleus of HEK293T and HeLa cells. Accordingly, a fragile X related 1 (FXR1) gene overexpression vector was constructed to investigate the effect of FXR1 overexpression on the level of monosialotetrahexosylganglioside 1 (GM1). The results of the current study suggested that FXR1P is a tissue-specific regulator of GM1 levels in SH-SY5Y cells, but not in HEK293T cells. Taken together, the results initially indicate that FXR1P interacts with CMAS, and that FXR1P may enhance the activation of sialic acid via interaction with CMAS, and increase GM1 levels to affect the development of the nervous system, thus providing evidence for further research into the pathogenesis of FXS.
Collapse
Affiliation(s)
- Yun Ma
- Department of Biochemistry & Biology, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Shuai Tian
- Department of Biochemistry & Biology, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Zongbao Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmaceutical and Biological Sciences, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Changbo Wang
- Department of Biochemistry & Biology, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xiaowei Chen
- Department of Biochemistry & Biology, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Wei Li
- Department of Biochemistry & Biology, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yang Yang
- Department of Biochemistry & Biology, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Shuya He
- Department of Biochemistry & Biology, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
15
|
Ladd AN. New Insights Into the Role of RNA-Binding Proteins in the Regulation of Heart Development. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 324:125-85. [PMID: 27017008 DOI: 10.1016/bs.ircmb.2015.12.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The regulation of gene expression during development takes place both at the transcriptional and posttranscriptional levels. RNA-binding proteins (RBPs) regulate pre-mRNA processing, mRNA localization, stability, and translation. Many RBPs are expressed in the heart and have been implicated in heart development, function, or disease. This chapter will review the current knowledge about RBPs in the developing heart, focusing on those that regulate posttranscriptional gene expression. The involvement of RBPs at each stage of heart development will be considered in turn, including the establishment of specific cardiac cell types and formation of the primitive heart tube, cardiac morphogenesis, and postnatal maturation and aging. The contributions of RBPs to cardiac birth defects and heart disease will also be considered in these contexts. Finally, the interplay between RBPs and other regulatory factors in the developing heart, such as transcription factors and miRNAs, will be discussed.
Collapse
Affiliation(s)
- A N Ladd
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States of America.
| |
Collapse
|
16
|
Novak SM, Joardar A, Gregorio CC, Zarnescu DC. Regulation of Heart Rate in Drosophila via Fragile X Mental Retardation Protein. PLoS One 2015; 10:e0142836. [PMID: 26571124 PMCID: PMC4646288 DOI: 10.1371/journal.pone.0142836] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 10/27/2015] [Indexed: 01/07/2023] Open
Abstract
RNA binding proteins play a pivotal role in post-transcriptional gene expression regulation, however little is understood about their role in cardiac function. The Fragile X (FraX) family of RNA binding proteins is most commonly studied in the context of neurological disorders, as mutations in Fragile X Mental Retardation 1 (FMR1) are the leading cause of inherited mental retardation. More recently, alterations in the levels of Fragile X Related 1 protein, FXR1, the predominant FraX member expressed in vertebrate striated muscle, have been linked to structural and functional defects in mice and zebrafish models. FraX proteins are established regulators of translation and are known to regulate specific targets in different tissues. To decipher the direct role of FraX proteins in the heart in vivo, we turned to Drosophila, which harbors a sole, functionally conserved and ubiquitously expressed FraX protein, dFmr1. Using classical loss of function alleles as well as muscle specific RNAi knockdown, we show that Drosophila FMRP, dFmr1, is required for proper heart rate during development. Functional analyses in the context of cardiac-specific dFmr1 knockdown by RNAi demonstrate that dFmr1 is required cell autonomously in cardiac cells for regulating heart rate. Interestingly, these functional defects are not accompanied by any obvious structural abnormalities, suggesting that dFmr1 may regulate a different repertoire of targets in Drosophila than in vertebrates. Taken together, our findings support the hypothesis that dFmr1 protein is essential for proper cardiac function and establish the fly as a new model for studying the role(s) of FraX proteins in the heart.
Collapse
Affiliation(s)
- Stefanie Mares Novak
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, 85724, United States of America
| | - Archi Joardar
- Department of Molecular and Cellular Biology The University of Arizona, Tucson, Arizona, 85721, United States of America
| | - Carol C. Gregorio
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, 85724, United States of America
| | - Daniela C. Zarnescu
- Department of Molecular and Cellular Biology The University of Arizona, Tucson, Arizona, 85721, United States of America
- * E-mail:
| |
Collapse
|
17
|
Wei N, Cheng Y, Wang Z, Liu Y, Luo C, Liu L, Chen L, Xie Z, Lu Y, Feng Y. SRSF10 Plays a Role in Myoblast Differentiation and Glucose Production via Regulation of Alternative Splicing. Cell Rep 2015; 13:1647-57. [DOI: 10.1016/j.celrep.2015.10.038] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 07/02/2015] [Accepted: 10/12/2015] [Indexed: 12/26/2022] Open
|
18
|
Jin X, Zhai B, Fang T, Guo X, Xu L. FXR1 is elevated in colorectal cancer and acts as an oncogene. Tumour Biol 2015; 37:2683-90. [DOI: 10.1007/s13277-015-4068-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 09/13/2015] [Indexed: 01/28/2023] Open
|
19
|
Borji R, Zghal F, Zarrouk N, Sahli S, Rebai H. Individuals with intellectual disability have lower voluntary muscle activation level. RESEARCH IN DEVELOPMENTAL DISABILITIES 2014; 35:3574-3581. [PMID: 25241117 DOI: 10.1016/j.ridd.2014.08.038] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 08/26/2014] [Accepted: 08/28/2014] [Indexed: 06/03/2023]
Abstract
The aim of this study was to explore the voluntary activation level during maximal voluntary contraction (MVC) in individuals with intellectual disability (ID) versus individuals without ID using the twitch interpolation technique. Ten individuals with mild ID (ID group) and 10 sedentary men without ID (control group) participated in this study. The evaluation of neuromuscular function consisted in three brief MVCs (3s) of the knee extension superimposed with electrical nerve stimulation (NES) to measure voluntary activation. Muscle activity levels were also measured with surface EMG. The root mean square (RMS) was extracted from the EMG signal. The RMS/Mmax ratio and the neuromuscular efficiency (NME) were calculated. Our results reported that individuals with ID present lower muscle strength (p < 0.001), lower voluntary activation level (p < 0.001), lower RMS values of vastus lateralis (p < 0.05), vastus medialis (p < 0.05), and rectus femoris (p < 0.001) muscles. In addition, our results showed lower RMS/Mmax values in the ID group than in the control group for the VM (0.05 ± 0.01 mV vs. 0.04 ± 0.01 mV; p < 0.05) and the RF (0.06 ± 0.02 mV vs. 0.05 ± 0.02 mV; p < 0.05) muscles. However, no significant difference was reported for the VL muscle (0.05 ± 0.02 mV vs. 0.05 ± 0.02 mV; p=0.463). Moreover, Individuals with ID present smaller potentiated twitch (p < 0.001). However, no significant difference was reported in the NME ratio. These results suggest that the lower muscle strength known in individuals with ID is related to a central nervous system failure to activate motor units and to some abnormal intrinsic muscle properties. It seems that the inactive lifestyle adopted by individuals with ID is one of the most important factors of their lower voluntary activation levels. Therefore, physical activities should be introduced in life style of individuals with ID to improve their neuromuscular function.
Collapse
Affiliation(s)
- Rihab Borji
- Unité de Recherche Education, Motricité, Sports et santé, Institut Supérieur du Sport et de l'Education Physique de Sfax, Université de Sfax, Tunisia.
| | - Firas Zghal
- Laboratoire Adaptations Métaboliques à l'Exercice en Conditions Physiologiques et Pathologiques (AME2P, EA 3533), Université Blaise Pascal, Clermont-Ferrand, France.
| | - Nidhal Zarrouk
- Laboratoire des techniques d'imagerie médicale (LR 12ES06, LTIM), Faculté de Médicine de Monastir, Université de Monastir, Tunisia.
| | - Sonia Sahli
- Unité de Recherche Education, Motricité, Sports et santé, Institut Supérieur du Sport et de l'Education Physique de Sfax, Université de Sfax, Tunisia.
| | - Haithem Rebai
- Unité de Recherche Education, Motricité, Sports et santé, Institut Supérieur du Sport et de l'Education Physique de Sfax, Université de Sfax, Tunisia.
| |
Collapse
|
20
|
Ma Y, Wang C, Li B, Qin L, Su J, Yang M, He S. Bcl-2-associated transcription factor 1 interacts with fragile X-related protein 1. Acta Biochim Biophys Sin (Shanghai) 2014; 46:119-27. [PMID: 24389646 PMCID: PMC7109863 DOI: 10.1093/abbs/gmt134] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The absence of fragile X mental retardation protein (FMRP) causes fragile X syndrome (FXS), which is the leading cause of hereditary mental retardation. Fragile X-related protein 1 (FXR1P), which plays an important role in normal muscle development, is one of the two autosomal paralogs of FMRP. To understand the functions of FXR1P, we screened FXR1P-interacting proteins by using a yeast two-hybrid system. The fragile X-related gene 1 (FXR1) was fused to pGBKT7 and then used as the bait to screen the human fetal brain cDNA library. The screening results revealed 10 FXR1P-interacting proteins including Bcl-2-associated transcription factor 1 (BTF). The interaction between FXR1P and BTF was confirmed by using both β-galactosidase assay and growth test in selective media. Co-immunoprecipitation assay in mammalian cells was also carried out to confirm the FXR1P/BTF interaction. Moreover, we confirmed that BTF co-localized with FXR1P in the cytoplasm around the nucleus in rat vascular smooth muscle cells by using confocal fluorescence microscopy. These results provide clues to elucidate the relationship between FXR1P and FXS.
Collapse
Affiliation(s)
- Yun Ma
- Department of Biochemistry and Biology, South China University, Hengyang 421001, China
| | - Changbo Wang
- Department of Biochemistry and Biology, South China University, Hengyang 421001, China
| | - Binyuan Li
- Department of Biochemistry and Biology, South China University, Hengyang 421001, China
| | - Lingxue Qin
- Department of Biochemistry and Biology, South China University, Hengyang 421001, China
| | - Jiao Su
- Department of Biochemistry and Biology, South China University, Hengyang 421001, China
| | - Manjun Yang
- Department of Biochemistry and Biology, South China University, Hengyang 421001, China
| | - Shuya He
- Department of Biochemistry and Biology, South China University, Hengyang 421001, China
- Correspondence address. Tel: +86-13807348502; E-mail:
| |
Collapse
|
21
|
Wilkinson RN, Jopling C, van Eeden FJM. Zebrafish as a model of cardiac disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 124:65-91. [PMID: 24751427 DOI: 10.1016/b978-0-12-386930-2.00004-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The zebrafish has been rapidly adopted as a model for cardiac development and disease. The transparency of the embryo, its limited requirement for active oxygen delivery, and ease of use in genetic manipulations and chemical exposure have made it a powerful alternative to rodents. Novel technologies like TALEN/CRISPR-mediated genome engineering and advanced imaging methods will only accelerate its use. Here, we give an overview of heart development and function in the fish and highlight a number of areas where it is most actively contributing to the understanding of cardiac development and disease. We also review the current state of research on a feature that we only could wish to be conserved between fish and human; cardiac regeneration.
Collapse
Affiliation(s)
- Robert N Wilkinson
- Department of Cardiovascular Science, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Chris Jopling
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, Département de Physiologie, Labex Ion Channel Science and Therapeutics, Montpellier, France; INSERM, U661, Montpellier, France; Universités de Montpellier 1&2, UMR-5203, Montpellier, France
| | - Fredericus J M van Eeden
- MRC Centre for Biomedical Genetics, Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
22
|
Abstract
Takotsubo cardiomyopathy (TTC) is an enigmatic disease with a multifactorial and still unresolved pathogenesis. Recent experimental and clinical observation has suggested a role for genetics in the pathogenesis of TTC. Ethnic as well as seasonal variation in the prevalence of TTC is well described, but it is only recently that familial cases of TTC have been reported. In recent years technological advances in exome capture and DNA sequencing have offered clinicians a new opportunity to discover genetics-related disease. This article explores the role of genetic mechanisms that might explain or modulate the pathogenesis of TTC.
Collapse
|
23
|
Verma SK, Deshmukh V, Liu P, Nutter CA, Espejo R, Hung ML, Wang GS, Yeo GW, Kuyumcu-Martinez MN. Reactivation of fetal splicing programs in diabetic hearts is mediated by protein kinase C signaling. J Biol Chem 2013; 288:35372-86. [PMID: 24151077 DOI: 10.1074/jbc.m113.507426] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Diabetic cardiomyopathy is one of the complications of diabetes that eventually leads to heart failure and death. Aberrant activation of PKC signaling contributes to diabetic cardiomyopathy by mechanisms that are poorly understood. Previous reports indicate that PKC is implicated in alternative splicing regulation. Therefore, we wanted to test whether PKC activation in diabetic hearts induces alternative splicing abnormalities. Here, using RNA sequencing we identified a set of 22 alternative splicing events that undergo a developmental switch in splicing, and we confirmed that splicing reverts to an embryonic pattern in adult diabetic hearts. This network of genes has important functions in RNA metabolism and in developmental processes such as differentiation. Importantly, PKC isozymes α/β control alternative splicing of these genes via phosphorylation and up-regulation of the RNA-binding proteins CELF1 and Rbfox2. Using a mutant of CELF1, we show that phosphorylation of CELF1 by PKC is necessary for regulation of splicing events altered in diabetes. In summary, our studies indicate that activation of PKCα/β in diabetic hearts contributes to the genome-wide splicing changes through phosphorylation and up-regulation of CELF1/Rbfox2 proteins. These findings provide a basis for PKC-mediated cardiac pathogenesis under diabetic conditions.
Collapse
Affiliation(s)
- Sunil K Verma
- From the Departments of Biochemistry and Molecular Biology and
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Oldenburg AR, Delbarre E, Thiede B, Vigouroux C, Collas P. Deregulation of Fragile X-related protein 1 by the lipodystrophic lamin A p.R482W mutation elicits a myogenic gene expression program in preadipocytes. Hum Mol Genet 2013; 23:1151-62. [PMID: 24108105 DOI: 10.1093/hmg/ddt509] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The nuclear lamina is implicated in the regulation of various nuclear functions. Several laminopathy-causing mutations in the LMNA gene, notably the p.R482W substitution linked to familial partial lipodystrophy type 2 (FPLD2), are clustered in the immunoglobulin fold of lamin A. We report a functional association between lamin A and fragile X-related protein 1 (FXR1P), a protein of the fragile X-related family involved in fragile X syndrome. Searching for proteins differentially interacting with the immunoglobulin fold of wild-type and R482W mutant lamin A, we identify FXR1P as a novel component of the lamin A protein network. The p.R482W mutation abrogates interaction of FXR1P with lamin A. Fibroblasts from FPLD2 patients display elevated levels of FXR1P and delocalized FXR1P. In human adipocyte progenitors, deregulation of lamin A expression leads to FXR1P up-regulation, impairment of adipogenic differentiation and induction of myogenin expression. FXR1P overexpression also stimulates a myogenic gene expression program in these cells. Our results demonstrate a cross-talk between proteins hitherto implicated in two distinct mesodermal pathologies. We propose a model where the FPLD2 lamin A p.R482W mutation elicits, through up-regulation of FXR1P, a remodeling of an adipogenic differentiation program into a myogenic program.
Collapse
Affiliation(s)
- Anja R Oldenburg
- Stem Cell Epigenetics Laboratory, Institute of Basic Medical Sciences and Norwegian Center for Stem Cell Research, Faculty of Medicine, University of Oslo, PO Box 1112, Blindern, Oslo 0317, Norway
| | | | | | | | | |
Collapse
|
25
|
Blech-Hermoni Y, Ladd AN. RNA binding proteins in the regulation of heart development. Int J Biochem Cell Biol 2013; 45:2467-78. [PMID: 23973289 DOI: 10.1016/j.biocel.2013.08.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 08/09/2013] [Accepted: 08/13/2013] [Indexed: 11/28/2022]
Abstract
In vivo, RNA molecules are constantly accompanied by RNA binding proteins (RBPs), which are intimately involved in every step of RNA biology, including transcription, editing, splicing, transport and localization, stability, and translation. RBPs therefore have opportunities to shape gene expression at multiple levels. This capacity is particularly important during development, when dynamic chemical and physical changes give rise to complex organs and tissues. This review discusses RBPs in the context of heart development. Since the targets and functions of most RBPs--in the heart and at large--are not fully understood, this review focuses on the expression and roles of RBPs that have been implicated in specific stages of heart development or developmental pathology. RBPs are involved in nearly every stage of cardiogenesis, including the formation, morphogenesis, and maturation of the heart. A fuller understanding of the roles and substrates of these proteins could ultimately provide attractive targets for the design of therapies for congenital heart defects, cardiovascular disease, or cardiac tissue repair.
Collapse
Affiliation(s)
- Yotam Blech-Hermoni
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Program in Cell Biology, Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | | |
Collapse
|
26
|
Zarnescu DC, Gregorio CC. Fragile hearts: new insights into translational control in cardiac muscle. Trends Cardiovasc Med 2013; 23:275-81. [PMID: 23582851 DOI: 10.1016/j.tcm.2013.03.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 03/01/2013] [Accepted: 03/05/2013] [Indexed: 01/20/2023]
Abstract
Current investigations focused on RNA-binding proteins in striated muscle, which provide a scenario whereby muscle function and development are governed by the interplay of post-transcriptional RNA regulation, including transcript localization, splicing, stability, and translational control. New data have recently emerged, linking the RNA-binding protein FXR1 to the translation of key cytoskeletal components such as talin and desmoplakin in heart muscle. These findings, together with a plethora of recent reports implicating RNA-binding proteins and their RNA targets in both basic aspects of muscle development and differentiation as well as heart disease and muscular dystrophies, point to a critical role of RNA-based regulatory mechanisms in muscle biology. Here we focus on FXR1, the striated muscle-specific member of the Fragile X family of RNA-binding proteins and discuss its newly reported cytoskeletal targets as well as potential implications for heart disease.
Collapse
Affiliation(s)
- Daniela C Zarnescu
- Department of Molecular and Cellular Biology, the Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ 85724.
| | | |
Collapse
|
27
|
Davidovic L, Durand N, Khalfallah O, Tabet R, Barbry P, Mari B, Sacconi S, Moine H, Bardoni B. A novel role for the RNA-binding protein FXR1P in myoblasts cell-cycle progression by modulating p21/Cdkn1a/Cip1/Waf1 mRNA stability. PLoS Genet 2013; 9:e1003367. [PMID: 23555284 PMCID: PMC3605292 DOI: 10.1371/journal.pgen.1003367] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 01/21/2013] [Indexed: 12/14/2022] Open
Abstract
The Fragile X-Related 1 gene (FXR1) is a paralog of the Fragile X Mental Retardation 1 gene (FMR1), whose absence causes the Fragile X syndrome, the most common form of inherited intellectual disability. FXR1P plays an important role in normal muscle development, and its absence causes muscular abnormalities in mice, frog, and zebrafish. Seven alternatively spliced FXR1 transcripts have been identified and two of them are skeletal muscle-specific. A reduction of these isoforms is found in myoblasts from Facio-Scapulo Humeral Dystrophy (FSHD) patients. FXR1P is an RNA–binding protein involved in translational control; however, so far, no mRNA target of FXR1P has been linked to the drastic muscular phenotypes caused by its absence. In this study, gene expression profiling of C2C12 myoblasts reveals that transcripts involved in cell cycle and muscular development pathways are modulated by Fxr1-depletion. We observed an increase of p21—a regulator of cell-cycle progression—in Fxr1-knocked-down mouse C2C12 and FSHD human myoblasts. Rescue of this molecular phenotype is possible by re-expressing human FXR1P in Fxr1-depleted C2C12 cells. FXR1P muscle-specific isoforms bind p21 mRNA via direct interaction with a conserved G-quadruplex located in its 3′ untranslated region. The FXR1P/G-quadruplex complex reduces the half-life of p21 mRNA. In the absence of FXR1P, the upregulation of p21 mRNA determines the elevated level of its protein product that affects cell-cycle progression inducing a premature cell-cycle exit and generating a pool of cells blocked at G0. Our study describes a novel role of FXR1P that has crucial implications for the understanding of its role during myogenesis and muscle development, since we show here that in its absence a reduced number of myoblasts will be available for muscle formation/regeneration, shedding new light into the pathophysiology of FSHD. Muscle development is a complex process controlled by the timely expression of genes encoding crucial regulators of the muscle cell precursors called myoblasts. We know from previous studies that inactivation of the Fragile X related 1 (FXR1) gene in various animal models (mouse, frog, and zebrafish) causes muscular and cardiac abnormalities. Also, FXR1P is reduced in a human myopathy called Fascio-Scapulo Humeral Dystrophy (FSHD), suggesting its critical role in muscle that findings presented in this study contribute to elucidating. Cell-cycle arrest is a prerequisite to differentiation of myoblasts into mature myotubes, which will form the muscle. One key regulator is the p21/Cdkn1a/Cip1/Waf1 protein, which commands myoblasts to stop proliferating, and this action is particularly important during muscle regeneration. In this study, we have identified FXR1P as a novel regulator of p21 expression. We show that FXR1P absence in mouse myoblasts and FSHD-derived myopathic myoblasts increases abnormally the levels of p21, causing a premature cell cycle exit of myoblasts. Our study predicts that FXR1P absence leads to a reduced number of myoblasts available for muscle formation and regeneration. This explains the drastic effects of FXR1 inactivation on muscle and brings a better understanding of the molecular/cellular bases of FSHD.
Collapse
Affiliation(s)
- Laetitia Davidovic
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
- Université de Nice-Sophia Antipolis, Nice, France
| | - Nelly Durand
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
- Université de Nice-Sophia Antipolis, Nice, France
| | - Olfa Khalfallah
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
- Université de Nice-Sophia Antipolis, Nice, France
| | - Ricardo Tabet
- IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire), CNRS, UMR7104, Inserm U596, Collège de France, Strasbourg University, Illkirch-Graffenstaden, France
| | - Pascal Barbry
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
- Université de Nice-Sophia Antipolis, Nice, France
| | - Bernard Mari
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
- Université de Nice-Sophia Antipolis, Nice, France
| | - Sabrina Sacconi
- INSERM U638, Faculté de Médecine, Université de Nice Sophia-Antipolis, Centre de Référence pour les Maladies Neuromusculaires, CHU de Nice, Nice, France
| | - Hervé Moine
- IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire), CNRS, UMR7104, Inserm U596, Collège de France, Strasbourg University, Illkirch-Graffenstaden, France
| | - Barbara Bardoni
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS UMR 7275, Valbonne, France
- Université de Nice-Sophia Antipolis, Nice, France
- * E-mail:
| |
Collapse
|
28
|
Bardoni B, Abekhoukh S, Zongaro S, Melko M. Intellectual disabilities, neuronal posttranscriptional RNA metabolism, and RNA-binding proteins: three actors for a complex scenario. PROGRESS IN BRAIN RESEARCH 2012; 197:29-51. [PMID: 22541287 DOI: 10.1016/b978-0-444-54299-1.00003-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Intellectual disability (ID) is the most frequent cause of serious handicap in children and young adults and interests 2-3% of worldwide population, representing a serious problem from the medical, social, and economic points of view. The causes are very heterogeneous. Genes involved in ID have various functions altering different pathways important in neuronal function. Regulation of mRNA metabolism is particularly important in neurons for synaptic structure and function. Here, we review ID due to alteration of mRNA metabolism. Functional absence of some RNA-binding proteins--namely, FMRP, FMR2P, PQBP1, UFP3B, VCX-A--causes different forms of ID. These proteins are involved in different steps of RNA metabolism and, even if a detailed analysis of their RNA targets has been performed so far only for FMRP, it appears clear that they modulate some aspects (translation, stability, transport, and sublocalization) of a subset of RNAs coding for proteins, whose function must be relevant for neurons. Two other proteins, DYRK1A and CDKL5, involved in Down syndrome and Rett syndrome, respectively, have been shown to have an impact on splicing efficiency of specific mRNAs. Both proteins are kinases and their effect is indirect. Interestingly, both are localized in nuclear speckles, the nuclear domains where splicing factors are assembled, stocked, and recycled and influence their biogenesis and/or their organization.
Collapse
Affiliation(s)
- Barbara Bardoni
- Institute of Molecular and Cellular Pharmacology, CNRS-UMR6097, Université de Nice Sophia-Antipolis,Valbonne, France.
| | | | | | | |
Collapse
|
29
|
Winograd C, Ceman S. Fragile X family members have important and non-overlapping functions. Biomol Concepts 2011; 2:343-52. [DOI: 10.1515/bmc.2011.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Accepted: 06/29/2011] [Indexed: 01/15/2023] Open
Abstract
AbstractThe fragile X family of genes encodes a small family of RNA binding proteins including FMRP, FXR1P and FXR2P that were identified in the 1990s. All three members are encoded by 17 exons and show alternative splicing at the 3′ ends of their respective transcripts. They share significant homology in the protein functional domains, including the Tudor domains, the nuclear localization sequence, a protein-protein interaction domain, the KH1 and KH2 domains and the nuclear export sequence. Fragile X family members are found throughout the animal kingdom, although all three members are not consistently present in species outside of mammals: only two family members are present in the avian species examined, Gallus gallus and Taeniopygia guttata, and in the frog Xenopus tropicalis. Although present in many tissues, the functions of the fragile X family members differ, which are particularly evident in knockout studies performed in animals. The fragile X family members play roles in normal neuronal function and in the case of FXR1, in muscle function.
Collapse
Affiliation(s)
- Claudia Winograd
- 2Neuroscience Program and College of Medicine, University of Illinois, 601 S. Goodwin Avenue, Urbana–Champaign, IL 61801, USA
| | | |
Collapse
|
30
|
Xu XL, Zong R, Li Z, Biswas MHU, Fang Z, Nelson DL, Gao FB. FXR1P but not FMRP regulates the levels of mammalian brain-specific microRNA-9 and microRNA-124. J Neurosci 2011; 31:13705-9. [PMID: 21957233 PMCID: PMC3446782 DOI: 10.1523/jneurosci.2827-11.2011] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 07/08/2011] [Accepted: 07/14/2011] [Indexed: 11/21/2022] Open
Abstract
Mammalian brain-specific miR-9 and miR-124 have been implicated in several aspects of neuronal development and function. However, it is not known how their expression levels are regulated in vivo. We found that the levels of miR-9 and miR-124 are regulated by FXR1P but not by the loss of FXR2P or FMRP in vivo, a mouse model of fragile X syndrome. Surprisingly, the levels of miR-9 and miR-124 are elevated in fmr1/fxr2 double-knock-out mice, in part reflecting posttranscriptional upregulation of FXR1P. Indeed, FXR1P is required for efficient processing of pre-miR-9 and pre-miR-124 in vitro and forms a complex with Dicer and pre-miRNAs. These findings reveal differential roles of FMRP family proteins in controlling the expression levels of brain-specific miRNAs.
Collapse
Affiliation(s)
- Xia-Lian Xu
- Gladstone Institute of Neurological Disease, San Francisco, California 94158
| | - Ruiting Zong
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, and
| | - Zhaodong Li
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Md Helal Uddin Biswas
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Zhe Fang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, and
| | - David L. Nelson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, and
| | - Fen-Biao Gao
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| |
Collapse
|
31
|
Whitman SA, Cover C, Yu L, Nelson DL, Zarnescu DC, Gregorio CC. Desmoplakin and talin2 are novel mRNA targets of fragile X-related protein-1 in cardiac muscle. Circ Res 2011; 109:262-71. [PMID: 21659647 DOI: 10.1161/circresaha.111.244244] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
RATIONALE The proper function of cardiac muscle requires the precise assembly and interactions of numerous cytoskeletal and regulatory proteins into specialized structures that orchestrate contraction and force transmission. Evidence suggests that posttranscriptional regulation is critical for muscle function, but the mechanisms involved remain understudied. OBJECTIVE To investigate the molecular mechanisms and targets of the muscle-specific fragile X mental retardation, autosomal homolog 1 (FXR1), an RNA binding protein whose loss leads to perinatal lethality in mice and cardiomyopathy in zebrafish. METHODS AND RESULTS Using RNA immunoprecipitation approaches we found that desmoplakin and talin2 mRNAs associate with FXR1 in a complex. In vitro assays indicate that FXR1 binds these mRNA targets directly and represses their translation. Fxr1 KO hearts exhibit an up-regulation of desmoplakin and talin2 proteins, which is accompanied by severe disruption of desmosome as well as costamere architecture and composition in the heart, as determined by electron microscopy and deconvolution immunofluorescence analysis. CONCLUSIONS Our findings reveal the first direct mRNA targets of FXR1 in striated muscle and support translational repression as a novel mechanism for regulating heart muscle development and function, in particular the assembly of specialized cytoskeletal structures.
Collapse
Affiliation(s)
- Samantha A Whitman
- Department of Cellular and Molecular Medicine, University of Arizona, 1656 East Mabel, Tucson, AZ 85724, USA
| | | | | | | | | | | |
Collapse
|
32
|
Pandey UB, Nichols CD. Human disease models in Drosophila melanogaster and the role of the fly in therapeutic drug discovery. Pharmacol Rev 2011; 63:411-36. [PMID: 21415126 PMCID: PMC3082451 DOI: 10.1124/pr.110.003293] [Citation(s) in RCA: 706] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The common fruit fly, Drosophila melanogaster, is a well studied and highly tractable genetic model organism for understanding molecular mechanisms of human diseases. Many basic biological, physiological, and neurological properties are conserved between mammals and D. melanogaster, and nearly 75% of human disease-causing genes are believed to have a functional homolog in the fly. In the discovery process for therapeutics, traditional approaches employ high-throughput screening for small molecules that is based primarily on in vitro cell culture, enzymatic assays, or receptor binding assays. The majority of positive hits identified through these types of in vitro screens, unfortunately, are found to be ineffective and/or toxic in subsequent validation experiments in whole-animal models. New tools and platforms are needed in the discovery arena to overcome these limitations. The incorporation of D. melanogaster into the therapeutic discovery process holds tremendous promise for an enhanced rate of discovery of higher quality leads. D. melanogaster models of human diseases provide several unique features such as powerful genetics, highly conserved disease pathways, and very low comparative costs. The fly can effectively be used for low- to high-throughput drug screens as well as in target discovery. Here, we review the basic biology of the fly and discuss models of human diseases and opportunities for therapeutic discovery for central nervous system disorders, inflammatory disorders, cardiovascular disease, cancer, and diabetes. We also provide information and resources for those interested in pursuing fly models of human disease, as well as those interested in using D. melanogaster in the drug discovery process.
Collapse
Affiliation(s)
- Udai Bhan Pandey
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, 1901 Perdido St., New Orleans, LA 70112, USA
| | | |
Collapse
|
33
|
Cheever A, Blackwell E, Ceman S. Fragile X protein family member FXR1P is regulated by microRNAs. RNA (NEW YORK, N.Y.) 2010; 16:1530-1539. [PMID: 20519410 PMCID: PMC2905753 DOI: 10.1261/rna.2022210] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Accepted: 04/26/2010] [Indexed: 05/29/2023]
Abstract
FXR1P is one of two autosomal paralogs of the fragile X mental retardation protein FMRP. The absence of FMRP causes fragile X syndrome, the leading cause of hereditary mental retardation. FXR1P plays an important role in normal muscle development and has been implicated in facioscapulohumeral muscular dystrophy (FSHD). Its absence also causes cardiac abnormalities in both mice and zebrafish. To examine miRNA-mediated regulation of FMRP and FXR1P, we studied their expression in a conditional Dicer knockdown cell line, DT40. We found that FXR1P, but not FMRP, is significantly increased upon Dicer knockdown and the consequent reduction of miRNAs, suggesting that FXR1P is regulated by miRNAs while FMRP is not in DT40 cells. Expression of a luciferase reporter bearing the 3' untranslated region (3'UTR) of FXR1 was significantly increased in the absence of miRNAs, confirming miRNA-mediated regulation of FXR1P, while a luciferase reporter bearing the FMR1 3'UTR was not. We identified one of the regulatory regions in the 3'UTR of FXR1 by removing a conserved, 8-nucleotide miRNA seed sequence common to miRNAs 25, 32, 92, 363, and 367 and demonstrated loss of miRNA-mediated suppression. Treatment with specific miRNA hairpin inhibitors to each of the miRNAs in the seed sequence showed that miRs 92b, 363, and 367 regulated FXR1P expression. Accordingly, overexpression of the miRNA 367 mimic significantly decreased endogenous FXR1P expression in human cell lines HEK-293T and HeLa. We report for the first time that FXR1P is regulated through miRNA binding, with one site being the miR-25/32/92/363/367 seed sequence.
Collapse
Affiliation(s)
- Anne Cheever
- Department of Cell and Developmental Biology, University of Illinois, Urbana, IL 61801, USA
| | | | | |
Collapse
|
34
|
Abstract
Alternative splicing of pre-mRNAs is a major contributor to proteomic diversity and to the control of gene expression in higher eukaryotic cells. For this reasons, alternative splicing is tightly regulated in different tissues and developmental stages and its disruption can lead to a wide range of human disorders. The aim of this review is to focus on the relevance of alternative splicing for muscle function and muscle disease. We begin by giving a brief overview of alternative splicing, muscle-specific gene expression and muscular dystrophy. Next, to illustrate these concepts we focus on two muscular dystrophy, myotonic muscular dystrophy and facioscapulohumeral muscular dystrophy, both associated to disruption of splicing regulation in muscle.
Collapse
Affiliation(s)
- Mariaelena Pistoni
- Division of Regenerative Medicine; San Raffaele Scientific Institute; Milano, Italy
| | - Claudia Ghigna
- Istituto di Genetica Molecolare—Consiglio Nazionale delle Ricerche (iGM-CNR); Pavia, Italy
| | - Davide Gabellini
- Division of Regenerative Medicine; San Raffaele Scientific Institute; Milano, Italy
- Dulbecco Telethon Insitute; Milano, Italy
| |
Collapse
|
35
|
Banerjee P, Schoenfeld BP, Bell AJ, Choi CH, Bradley MP, Hinchey P, Kollaros M, Park JH, McBride SMJ, Dockendorff TC. Short- and long-term memory are modulated by multiple isoforms of the fragile X mental retardation protein. J Neurosci 2010; 30:6782-92. [PMID: 20463240 PMCID: PMC2880182 DOI: 10.1523/jneurosci.6369-09.2010] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Revised: 03/09/2010] [Accepted: 04/02/2010] [Indexed: 12/28/2022] Open
Abstract
The diversity of protein isoforms arising from alternative splicing is thought to modulate fine-tuning of synaptic plasticity. Fragile X mental retardation protein (FMRP), a neuronal RNA binding protein, exists in isoforms as a result of alternative splicing, but the contribution of these isoforms to neural plasticity are not well understood. We show that two isoforms of Drosophila melanogaster FMRP (dFMR1) have differential roles in mediating neural development and behavior functions conferred by the dfmr1 gene. These isoforms differ in the presence of a protein interaction module that is related to prion domains and is functionally conserved between FMRPs. Expression of both isoforms is necessary for optimal performance in tests of short- and long-term memory of courtship training. The presence or absence of the protein interaction domain may govern the types of ribonucleoprotein (RNP) complexes dFMR1 assembles into, with different RNPs regulating gene expression in a manner necessary for establishing distinct phases of memory formation.
Collapse
Affiliation(s)
- Paromita Banerjee
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Brian P. Schoenfeld
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Aaron J. Bell
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Catherine H. Choi
- Department of Medicine, Lehigh Valley Health Network, Allentown, Pennsylvania 18105
| | - Michael P. Bradley
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan 48201, and
| | - Paul Hinchey
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Maria Kollaros
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Jae H. Park
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee 37996
| | - Sean M. J. McBride
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Thomas C. Dockendorff
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee 37996
| |
Collapse
|
36
|
Coffee RL, Tessier CR, Woodruff EA, Broadie K. Fragile X mental retardation protein has a unique, evolutionarily conserved neuronal function not shared with FXR1P or FXR2P. Dis Model Mech 2010; 3:471-85. [PMID: 20442204 DOI: 10.1242/dmm.004598] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Fragile X syndrome (FXS), resulting solely from the loss of function of the human fragile X mental retardation 1 (hFMR1) gene, is the most common heritable cause of mental retardation and autism disorders, with syndromic defects also in non-neuronal tissues. In addition, the human genome encodes two closely related hFMR1 paralogs: hFXR1 and hFXR2. The Drosophila genome, by contrast, encodes a single dFMR1 gene with close sequence homology to all three human genes. Drosophila that lack the dFMR1 gene (dfmr1 null mutants) recapitulate FXS-associated molecular, cellular and behavioral phenotypes, suggesting that FMR1 function has been conserved, albeit with specific functions possibly sub-served by the expanded human gene family. To test evolutionary conservation, we used tissue-targeted transgenic expression of all three human genes in the Drosophila disease model to investigate function at (1) molecular, (2) neuronal and (3) non-neuronal levels. In neurons, dfmr1 null mutants exhibit elevated protein levels that alter the central brain and neuromuscular junction (NMJ) synaptic architecture, including an increase in synapse area, branching and bouton numbers. Importantly, hFMR1 can, comparably to dFMR1, fully rescue both the molecular and cellular defects in neurons, whereas hFXR1 and hFXR2 provide absolutely no rescue. For non-neuronal requirements, we assayed male fecundity and testes function. dfmr1 null mutants are effectively sterile owing to disruption of the 9+2 microtubule organization in the sperm tail. Importantly, all three human genes fully and equally rescue mutant fecundity and spermatogenesis defects. These results indicate that FMR1 gene function is evolutionarily conserved in neural mechanisms and cannot be compensated by either FXR1 or FXR2, but that all three proteins can substitute for each other in non-neuronal requirements. We conclude that FMR1 has a neural-specific function that is distinct from its paralogs, and that the unique FMR1 function is responsible for regulating neuronal protein expression and synaptic connectivity.
Collapse
Affiliation(s)
- R Lane Coffee
- Department of Biological Sciences, Vanderbilt Brain Institute, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37235-1634, USA
| | | | | | | |
Collapse
|
37
|
A Functional Requirement for PAK1 Binding to the KH(2) Domain of the Fragile X Protein-Related FXR1. Mol Cell 2010; 38:236-49. [DOI: 10.1016/j.molcel.2010.04.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2009] [Revised: 12/21/2009] [Accepted: 02/21/2010] [Indexed: 01/14/2023]
|