1
|
Valina AA, Belashova TA, Yuzman AK, Zadorsky SP, Sysoev EI, Mitkevich VA, Makarov AA, Galkin AP. Functional amyloid protein FXR1 is recruited into neuronal stress granules. Prion 2025; 19:1-16. [PMID: 40411539 PMCID: PMC12118398 DOI: 10.1080/19336896.2025.2505422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 05/06/2025] [Accepted: 05/08/2025] [Indexed: 05/26/2025] Open
Abstract
The FXR1 protein regulates the stability and translation of a number of RNA molecules and plays an important role in the regulation of cellular processes under normal conditions and stress. In particular, this protein is known to be a negative regulator of the key proinflammatory cytokine TNF alpha. We had previously shown that FXR1 functioned in the amyloid form in neurons of the brain of jawed vertebrates. Under stress conditions, FXR1 is incorporated into stress granules in some cell lines, but such studies have not been conducted for neuronal cells. Here, we showed the ability of the FXR1 protein to form cytoplasmic granules in a neuroblastoma cell line under various types of stress. This protein colocalizes with core proteins of neuronal stress granules upon heat shock and sodium arsenite treatment. We also showed that FXR1 colocalizes with anti-amyloid antibodies OC under both normal and stress conditions. Given that stress granules are dynamic structures, we propose that amyloid FXR1-containing RNP particles interact with other stress granule proteins through weak intermolecular hydrogen bonds. Using a yeast model system, we found that FXR1 colocalizes and physically interacts with stress granule proteins such as TIA-1, FMRP, FXR2, and SFPQ. Overall, our results provide new insights into the role of the RNA-binding protein FXR1 in neuronal stress response. We believe that FXR1 inactivation in neuronal stress granules can contribute to an increase in the level of the proinflammatory cytokine TNF alpha in neurodegenerative diseases.
Collapse
Affiliation(s)
- Anna A. Valina
- St. Petersburg Branch, Vavilov Institute of General Genetics, Russian Academy of Sciences, St. Petersburg, Russian Federation
- Department of Genetics and Biotechnology, Faculty of Biology, St. Petersburg State University, St. Petersburg, Russian Federation
| | - Tatyana A. Belashova
- St. Petersburg Branch, Vavilov Institute of General Genetics, Russian Academy of Sciences, St. Petersburg, Russian Federation
- Laboratory of Amyloid Biology, St. Petersburg State University, St. Petersburg, Russian Federation
| | - Anastasia K. Yuzman
- Department of Genetics and Biotechnology, Faculty of Biology, St. Petersburg State University, St. Petersburg, Russian Federation
| | - Sergey P. Zadorsky
- St. Petersburg Branch, Vavilov Institute of General Genetics, Russian Academy of Sciences, St. Petersburg, Russian Federation
- Department of Genetics and Biotechnology, Faculty of Biology, St. Petersburg State University, St. Petersburg, Russian Federation
| | - Evgeniy I. Sysoev
- St. Petersburg Branch, Vavilov Institute of General Genetics, Russian Academy of Sciences, St. Petersburg, Russian Federation
- Department of Genetics and Biotechnology, Faculty of Biology, St. Petersburg State University, St. Petersburg, Russian Federation
| | - Vladimir A. Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russian Federation
| | - Alexander A. Makarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russian Federation
| | - Alexey P. Galkin
- St. Petersburg Branch, Vavilov Institute of General Genetics, Russian Academy of Sciences, St. Petersburg, Russian Federation
- Department of Genetics and Biotechnology, Faculty of Biology, St. Petersburg State University, St. Petersburg, Russian Federation
| |
Collapse
|
2
|
Scheuer KS, Jansson AM, Shen M, Zhao X, Jackson MB. Fxr1 Deletion from Cortical Parvalbumin Interneurons Modifies Their Excitatory Synaptic Responses. eNeuro 2025; 12:ENEURO.0363-24.2024. [PMID: 39753370 PMCID: PMC11735682 DOI: 10.1523/eneuro.0363-24.2024] [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: 08/22/2024] [Revised: 12/10/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025] Open
Abstract
Fragile X autosomal homolog 1 (FXR1), a member of the fragile X messenger riboprotein 1 family, has been linked to psychiatric disorders including autism and schizophrenia. Parvalbumin (PV) interneurons play critical roles in cortical processing and have been implicated in FXR1-linked mental illnesses. Targeted deletion of FXR1 from PV interneurons in mice has been shown to alter cortical excitability and elicit schizophrenia-like behavior. This indicates that FXR1 regulates behaviorally relevant electrophysiological functions in PV interneurons. We therefore expressed a genetically encoded hybrid voltage sensor in PV interneurons and used voltage imaging in slices of mouse somatosensory cortex to assess the impact of targeted FXR1 deletion. These experiments showed that PV interneurons lacking FXR1 had excitatory synaptic potentials with larger amplitudes and shorter latencies compared with wild type. Synaptic potential rise-times, decay-times, and half-widths were also impacted to degrees that varied between cortical layer and synaptic input. Thus, FXR1 modulates the responsiveness of PV interneurons to excitatory synaptic inputs. This will enable FXR1 to control cortical processing in subtle ways, with the potential to influence behavior and contribute to psychiatric dysfunction.
Collapse
Affiliation(s)
- Katherine S Scheuer
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Anna M Jansson
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Minjie Shen
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin 53705
- The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Xinyu Zhao
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin 53705
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Meyer B Jackson
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin 53705
| |
Collapse
|
3
|
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
|
4
|
Galkin AP, Sysoev EI, Valina AA. Amyloids and prions in the light of evolution. Curr Genet 2023; 69:189-202. [PMID: 37165144 DOI: 10.1007/s00294-023-01270-6] [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: 04/14/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/12/2023]
Abstract
Functional amyloids have been identified in a wide variety of organisms including bacteria, fungi, plants, and vertebrates. Intracellular and extracellular amyloid fibrils of different proteins perform storage, protective, structural, and regulatory functions. The structural organization of amyloid fibrils determines their unique physical and biochemical properties. The formation of these fibrillar structures can provide adaptive advantages that are picked up by natural selection. Despite the great interest in functional and pathological amyloids, questions about the conservatism of the amyloid properties of proteins and the regularities in the appearance of these fibrillar structures in evolution remain almost unexplored. Using bioinformatics approaches and summarizing the data published previously, we have shown that amyloid fibrils performing similar functions in different organisms have been arising repeatedly and independently in the course of evolution. On the other hand, we show that the amyloid properties of a number of bacterial and eukaryotic proteins are evolutionarily conserved. We also discuss the role of protein-based inheritance in the evolution of microorganisms. Considering that missense mutations and the emergence of prions cause the same consequences, we propose the concept that the formation of prions, similarly to mutations, generally causes a negative effect, although it can also lead to adaptations in rare cases. In general, our analysis revealed certain patterns in the emergence and spread of amyloid fibrillar structures in the course of evolution.
Collapse
Affiliation(s)
- Alexey P Galkin
- Vavilov Institute of General Genetics, St. Petersburg Branch, Russian Academy of Sciences, St. Petersburg, Russian Federation, 199034.
- Department of Genetics and Biotechnology, St. Petersburg State University, St. Petersburg, Russian Federation, 199034.
| | - Evgeniy I Sysoev
- Vavilov Institute of General Genetics, St. Petersburg Branch, Russian Academy of Sciences, St. Petersburg, Russian Federation, 199034
- Department of Genetics and Biotechnology, St. Petersburg State University, St. Petersburg, Russian Federation, 199034
| | - Anna A Valina
- Department of Genetics and Biotechnology, St. Petersburg State University, St. Petersburg, Russian Federation, 199034
| |
Collapse
|
5
|
Amyloid Properties of the FXR1 Protein Are Conserved in Evolution of Vertebrates. Int J Mol Sci 2022; 23:ijms23147997. [PMID: 35887344 PMCID: PMC9319111 DOI: 10.3390/ijms23147997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/16/2022] [Accepted: 07/18/2022] [Indexed: 12/10/2022] Open
Abstract
Functional amyloids are fibrillary proteins with a cross-β structure that play a structural or regulatory role in pro- and eukaryotes. Previously, we have demonstrated that the RNA-binding FXR1 protein functions in an amyloid form in the rat brain. This RNA-binding protein plays an important role in the regulation of long-term memory, emotions, and cancer. Here, we evaluate the amyloid properties of FXR1 in organisms representing various classes of vertebrates. We show the colocalization of FXR1 with amyloid-specific dyes in the neurons of amphibians, reptiles, and birds. Moreover, FXR1, as with other amyloids, forms detergent-resistant insoluble aggregates in all studied animals. The FXR1 protein isolated by immunoprecipitation from the brains of different vertebrate species forms fibrils, which show yellow-green birefringence after staining with Congo red. Our data indicate that in the evolution of vertebrates, FXR1 acquired amyloid properties at least 365 million years ago. Based on the obtained data, we discuss the possible role of FXR1 amyloid fibrils in the regulation of vital processes in the brain of vertebrates.
Collapse
|
6
|
The Fragile X Proteins Differentially Regulate Translation of Reporter mRNAs with G-quadruplex Structures. J Mol Biol 2022; 434:167396. [PMID: 34896112 PMCID: PMC8892671 DOI: 10.1016/j.jmb.2021.167396] [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: 07/19/2021] [Revised: 10/29/2021] [Accepted: 12/01/2021] [Indexed: 02/01/2023]
Abstract
Fragile X Syndrome, as well as some manifestations of autism spectrum disorder, results from improper RNA regulation due to a deficiency of fragile X mental retardation protein (FMRP). FMRP and its autosomal paralogs, fragile X related proteins 1 & 2 (FXR1P/2P), have been implicated in many aspects of RNA regulation, from protein synthesis to mRNA stability and decay. The literature on the fragile X related proteins' (FXPs) role in mRNA regulation and their potential mRNA targets is vast. Therefore, we developed an approach to investigate the function of FXPs in translational control using three potential mRNA targets. Briefly, we first selected top mRNA candidates found to be associated with the FXPs and whose translation are influenced by one or more of the FXPs. We then narrowed down the FXPs' binding site(s) within the mRNA, analyzed the strength of this binding in vitro, and determined how each FXP affects the translation of a minimal reporter mRNA with the binding site. Overall, all FXPs bound with high affinity to RNAs containing G-quadruplexes, such as Cyclin Dependent Kinase Inhibitor p21 and FMRP's own coding region. Interestingly, FMRP inhibited the translation of each mRNA distinctly and in a manner that appears to correlate with its binding to each mRNA. In contrast, FXR1P/2P inhibited all mRNAs tested. Finally, although binding of our RNAs was due to the RGG (arginine-glycine-glycine) motif-containing C-terminal region of the FXPs, this region was not sufficient to cause inhibition of translation.
Collapse
|
7
|
Grad M, Nir A, Levy G, Trangle SS, Shapira G, Shomron N, Assaf Y, Barak B. Altered White Matter and microRNA Expression in a Murine Model Related to Williams Syndrome Suggests That miR-34b/c Affects Brain Development via Ptpru and Dcx Modulation. Cells 2022; 11:cells11010158. [PMID: 35011720 PMCID: PMC8750756 DOI: 10.3390/cells11010158] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/15/2021] [Accepted: 12/28/2021] [Indexed: 11/16/2022] Open
Abstract
Williams syndrome (WS) is a multisystem neurodevelopmental disorder caused by a de novo hemizygous deletion of ~26 genes from chromosome 7q11.23, among them the general transcription factor II-I (GTF2I). By studying a novel murine model for the hypersociability phenotype associated with WS, we previously revealed surprising aberrations in myelination and cell differentiation properties in the cortices of mutant mice compared to controls. These mutant mice had selective deletion of Gtf2i in the excitatory neurons of the forebrain. Here, we applied diffusion magnetic resonance imaging and fiber tracking, which showed a reduction in the number of streamlines in limbic outputs such as the fimbria/fornix fibers and the stria terminalis, as well as the corpus callosum of these mutant mice compared to controls. Furthermore, we utilized next-generation sequencing (NGS) analysis of cortical small RNAs' expression (RNA-Seq) levels to identify altered expression of microRNAs (miRNAs), including two from the miR-34 cluster, known to be involved in prominent processes in the developing nervous system. Luciferase reporter assay confirmed the direct binding of miR-34c-5p to the 3'UTR of PTPRU-a gene involved in neural development that was elevated in the cortices of mutant mice relative to controls. Moreover, we found an age-dependent variation in the expression levels of doublecortin (Dcx)-a verified miR-34 target. Thus, we demonstrate the substantial effect a single gene deletion can exert on miRNA regulation and brain structure, and advance our understanding and, hopefully, treatment of WS.
Collapse
Affiliation(s)
- Meitar Grad
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel; (M.G.); (A.N.); (G.L.); (N.S.); (Y.A.)
| | - Ariel Nir
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel; (M.G.); (A.N.); (G.L.); (N.S.); (Y.A.)
| | - Gilad Levy
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel; (M.G.); (A.N.); (G.L.); (N.S.); (Y.A.)
| | - Sari Schokoroy Trangle
- Faculty of Social Sciences, School of Psychological Sciences, Tel Aviv University, Tel Aviv 6997801, Israel;
| | - Guy Shapira
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel;
- Edmond J. Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Noam Shomron
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel; (M.G.); (A.N.); (G.L.); (N.S.); (Y.A.)
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel;
- Edmond J. Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Yaniv Assaf
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel; (M.G.); (A.N.); (G.L.); (N.S.); (Y.A.)
- Faculty of Life Sciences, School of Neurobiology, Biochemistry & Biophysics, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Boaz Barak
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel; (M.G.); (A.N.); (G.L.); (N.S.); (Y.A.)
- Faculty of Social Sciences, School of Psychological Sciences, Tel Aviv University, Tel Aviv 6997801, Israel;
- Correspondence:
| |
Collapse
|
8
|
Li L, Jin Y, Wang X, Wang L, Sun Y, Luo Y, Ni X, Lu Q, Wei W, Zhi X, Yu J, Zhu W, Zhou P. Long noncoding RNA ENST00000508435 promotes migration of breast cancer via FXR 1. Cell Adh Migr 2021; 15:140-151. [PMID: 34057025 PMCID: PMC8168597 DOI: 10.1080/19336918.2021.1921402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/24/2021] [Accepted: 04/20/2021] [Indexed: 11/19/2022] Open
Abstract
LncRNA plays a critical role in tumor progression. However, the role it executes in breast cancer is still unclear. Here, we report a newly discovered lncRNA, ENST00000508435, which could be remarkably up-regulated in breast cancer cells and tissues. We found that the expression of ENST00000508435 was positively correlated with tumor size, lymph node metastasis and HER2. More interesting, overexpression of ENST00000508435 significantly increased cell migration, while specific knockdown led to the opposite. RNA pull-down and RNA immunoprecipitation assays demonstrated that ENST00000508435 could directly bind to FXR1 to promote tumor metastasis. ENST00000508435 and FXR1 were positively correlated. FXR1 was also significantly up-regulated in breast tumors. Taken together, we propose that ENST00000508435 regulates FXR1 to promote breast cancer metastasis.
Collapse
Affiliation(s)
- Luying Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Youping Jin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xue Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Li Wang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Pudong Hospital, Fudan University, Shanghai, China
| | - Yangbai Sun
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yihong Luo
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaojian Ni
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qi Lu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wenbo Wei
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiuling Zhi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jerry Yu
- Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Wei Zhu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ping Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
9
|
FXR1 regulation of parvalbumin interneurons in the prefrontal cortex is critical for schizophrenia-like behaviors. Mol Psychiatry 2021; 26:6845-6867. [PMID: 33863995 PMCID: PMC8521570 DOI: 10.1038/s41380-021-01096-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 03/18/2021] [Accepted: 03/31/2021] [Indexed: 12/13/2022]
Abstract
Parvalbumin interneurons (PVIs) are affected in many psychiatric disorders including schizophrenia (SCZ), however the mechanism remains unclear. FXR1, a high confident risk gene for SCZ, is indispensable but its role in the brain is largely unknown. We show that deleting FXR1 from PVIs of medial prefrontal cortex (mPFC) leads to reduced PVI excitability, impaired mPFC gamma oscillation, and SCZ-like behaviors. PVI-specific translational profiling reveals that FXR1 regulates the expression of Cacna1h/Cav3.2 a T-type calcium channel implicated in autism and epilepsy. Inhibition of Cav3.2 in PVIs of mPFC phenocopies whereas elevation of Cav3.2 in PVIs of mPFC rescues behavioral deficits resulted from FXR1 deficiency. Stimulation of PVIs using a gamma oscillation-enhancing light flicker rescues behavioral abnormalities caused by FXR1 deficiency in PVIs. This work unveils the function of a newly identified SCZ risk gene in SCZ-relevant neurons and identifies a therapeutic target and a potential noninvasive treatment for psychiatric disorders.
Collapse
|
10
|
Chen C, Zhang M, Zhang Y. Circ_0000079 Decoys the RNA-Binding Protein FXR1 to Interrupt Formation of the FXR1/PRCKI Complex and Decline Their Mediated Cell Invasion and Drug Resistance in NSCLC. Cell Transplant 2021; 29:963689720961070. [PMID: 32951448 PMCID: PMC7784611 DOI: 10.1177/0963689720961070] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Nonsmall cell lung cancer (NSCLC) has gradually become one of the deadliest threats to human health and life worldwide. Although reports have shown that circular RNAs (circRNAs) are associated with progression and metastasis of NSCLC, the biological functions of circRNAs during these processes remain largely unknown. Our study showed that circ_0000079 (CiR79) levels were significantly downregulated in NSCLC patients, especially in cisplatin (DDP)-resistant NSCLC patients, and low circ_0000079 levels were significantly associated with poor overall survival of NSCLC patients. Then, results from Cell Counting Kit-8 (CCK-8) cell viability assay and transwell cell invasion assay in A549/DDP and H460/DDP cells transfected with pCDH-CiR79 expression vector showed that circ_0000079 overexpression significantly inhibited cell proliferation and invasion of these DDP-resistant NSCLC cells. The online bioinformatic program StarBase and RNA-binding protein immunoprecipitation predicted and demonstrated that circ_0000079 could bind with the Fragile X-Related 1 (FXR1) protein rather than with protein kinase C, iota (PRKCI), which was shown to form a complex with FXR1 to promote invasion and growth of NSCLC cells. Co-immunoprecipitation combined with Western blot assays indicated that FXR1 levels were remarkably decreased, but PRKCI levels remained unchanged in pCDH-ciR79 transfected NSCLC cells. Moreover, circ_0000079 negatively regulated FXR1/PRKCI-mediated phosphorylation of glycogen synthesis kinase 3β and activator protein 1, thus suppressing the protein level of the Snail gene, an important promoter gene regulating cancer cell growth and epithelial-mesenchymal transition. Furthermore, DDP resistance of A549/DDP and H460/DDP cells was inhibited by circ_0000079 overexpression but was restored by FXR1. Hence, our findings demonstrated that circ_0000079 might inhibit cell invasion and drug resistance in NSCLC by interrupting the formation of the FXR1/PRCKI complex by interacting with FXR1, and circ_0000079 could act as a potential biomarker and therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Chen Chen
- Department of Pathology, 569063the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| | - Min Zhang
- Department of Pathology, 569063the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| | - Yan Zhang
- Department of Pathology, 569063the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| |
Collapse
|
11
|
Wang B, Yin H, Zhang H, Wang T. circNRIP1 facilitates keloid progression via FXR1‑mediated upregulation of miR‑503‑3p and miR‑503‑5p. Int J Mol Med 2021; 47:70. [PMID: 33649815 PMCID: PMC7952250 DOI: 10.3892/ijmm.2021.4903] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 01/25/2021] [Indexed: 01/11/2023] Open
Abstract
Circular nuclear receptor interacting protein 1 (circNRIP1) is implicated in tumor initiation and progression; however, the underlying mechanism of keloid progression is unclear. To the best of our knowledge, the present study is the first to characterize the contribution of circNRIP1 to keloid progression and evaluate the potential underlying molecular mechanisms using keloid-derived fibroblasts. The expression profile of circNRIP1 was confirmed in keloid tissue. The contribution of circNRIP1 to keloid progression was investigated via loss-of-function assays. Furthermore, the molecular mechanism by which circNRIP1 contributes to pre-microRNA (miR)-503 maturation through blocking Fbxo4-mediated Fragile-X mental retardation 1 (FXR1) ubiquitination was verified. Finally, the biological functions of FXR1, miR-503-3p, and miR-503-5p in keloid-derived fibroblast proliferation, apoptosis and extracellular matrix accumulation were confirmed. circNRIP1 was highly expressed in keloid tissue and keloid-derived fibroblasts. Functional analysis showed that circNRIP1 knockdown successfully blocked the proliferation and expression of extracellular matrix-associated proteins while increasing the rate of apoptosis in keloid-derived fibroblasts. Mechanistically, circNRIP1 maintained FXR1 stability by impeding Fbxo4-mediated FXR1 ubiquitination and degradation. Additionally, FXR1 increased the abundance of miR-503-3p and miR-503-5p by contributing to pre-miR-503 maturation. Knockdown of FXR1, miR-503-3p and miR-503-5p also inhibited proliferation and extracellular matrix accumulation in keloid-derived fibroblasts and increased levels of cell apoptosis. Collectively, the present study confirmed that circNRIP1 contributed to pre-miR-503 maturation via blocking Fbxo4-mediated FXR1 ubiquitination and degradation, which facilitates keloid progression. These results indicate that circNRIP1 has potential as a novel therapeutic target for the control and/or treatment of keloids.
Collapse
Affiliation(s)
- Baolin Wang
- Department of Dermatology, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, P.R. China
| | - Hang Yin
- Department of Neurosurgery, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, P.R. China
| | - Hongmei Zhang
- Department of Pharmacy, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, P.R. China
| | - Tiantian Wang
- Department of Dermatology, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, P.R. China
| |
Collapse
|
12
|
A simple procedure for bacterial expression and purification of the fragile X protein family. Sci Rep 2020; 10:15858. [PMID: 32985615 PMCID: PMC7522082 DOI: 10.1038/s41598-020-72984-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 09/07/2020] [Indexed: 11/23/2022] Open
Abstract
The fragile X protein family consists of three RNA-binding proteins involved in translational regulation. Fragile X mental retardation protein (FMRP) is well-studied, as its loss leads to fragile X syndrome, a neurodevelopmental disorder which is the most prevalent form of inherited mental retardation and the primary monogenetic cause of autism. Fragile X related proteins 1 and 2 (FXR1P and FXR2P) are autosomal paralogs of FMRP that are involved in promoting muscle development and neural development, respectively. There is great interest in studying this family of proteins, yet researchers have faced much difficulty in expressing and purifying the full-length versions of these proteins in sufficient quantities. We have developed a simple, rapid, and inexpensive procedure that allows for the recombinant expression and purification of full-length human FMRP, FXR1P, and FXR2P from Escherichia coli in high yields, free of protein and nucleic acid contamination. In order to assess the proteins’ function after purification, we confirmed their binding to pseudoknot and G-quadruplex forming RNAs as well as their ability to regulate translation in vitro.
Collapse
|
13
|
Dubes S, Favereaux A, Thoumine O, Letellier M. miRNA-Dependent Control of Homeostatic Plasticity in Neurons. Front Cell Neurosci 2019; 13:536. [PMID: 31866828 PMCID: PMC6906196 DOI: 10.3389/fncel.2019.00536] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/19/2019] [Indexed: 11/13/2022] Open
Abstract
Homeostatic plasticity is a form of plasticity in which neurons compensate for changes in neuronal activity through the control of key physiological parameters such as the number and the strength of their synaptic inputs and intrinsic excitability. Recent studies revealed that miRNAs, which are small non-coding RNAs repressing mRNA translation, participate in this process by controlling the translation of multiple effectors such as glutamate transporters, receptors, signaling molecules and voltage-gated ion channels. In this review, we present and discuss the role of miRNAs in both cell-wide and compartmentalized forms of homeostatic plasticity as well as their implication in pathological processes associated with homeostatic failure.
Collapse
Affiliation(s)
- Sandra Dubes
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
| | - Alexandre Favereaux
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
| | - Olivier Thoumine
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
| | - Mathieu Letellier
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
| |
Collapse
|
14
|
Santos-Cortez RLP, Khan V, Khan FS, Mughal ZUN, Chakchouk I, Lee K, Rasheed M, Hamza R, Acharya A, Ullah E, Saqib MAN, Abbe I, Ali G, Hassan MJ, Khan S, Azeem Z, Ullah I, Bamshad MJ, Nickerson DA, Schrauwen I, Ahmad W, Ansar M, Leal SM. Novel candidate genes and variants underlying autosomal recessive neurodevelopmental disorders with intellectual disability. Hum Genet 2018; 137:735-752. [PMID: 30167849 PMCID: PMC6201268 DOI: 10.1007/s00439-018-1928-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/10/2018] [Indexed: 01/30/2023]
Abstract
Identification of Mendelian genes for neurodevelopmental disorders using exome sequencing to study autosomal recessive (AR) consanguineous pedigrees has been highly successful. To identify causal variants for syndromic and non-syndromic intellectual disability (ID), exome sequencing was performed using DNA samples from 22 consanguineous Pakistani families with ARID, of which 21 have additional phenotypes including microcephaly. To aid in variant identification, homozygosity mapping and linkage analysis were performed. DNA samples from affected family member(s) from every pedigree underwent exome sequencing. Identified rare damaging exome variants were tested for co-segregation with ID using Sanger sequencing. For seven ARID families, variants were identified in genes not previously associated with ID, including: EI24, FXR1 and TET3 for which knockout mouse models have brain defects; and CACNG7 and TRAPPC10 where cell studies suggest roles in important neural pathways. For two families, the novel ARID genes CARNMT1 and GARNL3 lie within previously reported ID microdeletion regions. We also observed homozygous variants in two ID candidate genes, GRAMD1B and TBRG1, for which each has been previously reported in a single family. An additional 14 families have homozygous variants in established ID genes, of which 11 variants are novel. All ARID genes have increased expression in specific structures of the developing and adult human brain and 91% of the genes are differentially expressed in utero or during early childhood. The identification of novel ARID candidate genes and variants adds to the knowledge base that is required to further understand human brain function and development.
Collapse
Affiliation(s)
- Regie Lyn P Santos-Cortez
- Department of Molecular and Human Genetics, Center for Statistical Genetics, Baylor College of Medicine, 1 Baylor Plaza 700D, Houston, TX, 77030, USA
- Department of Otolaryngology, University of Colorado School of Medicine, 12700 E. 19th Ave., Aurora, CO, 80045, USA
| | - Valeed Khan
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Falak Sher Khan
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Zaib-Un-Nisa Mughal
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Imen Chakchouk
- Department of Molecular and Human Genetics, Center for Statistical Genetics, Baylor College of Medicine, 1 Baylor Plaza 700D, Houston, TX, 77030, USA
| | - Kwanghyuk Lee
- Department of Molecular and Human Genetics, Center for Statistical Genetics, Baylor College of Medicine, 1 Baylor Plaza 700D, Houston, TX, 77030, USA
| | - Memoona Rasheed
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Rifat Hamza
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Anushree Acharya
- Department of Molecular and Human Genetics, Center for Statistical Genetics, Baylor College of Medicine, 1 Baylor Plaza 700D, Houston, TX, 77030, USA
| | - Ehsan Ullah
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Muhammad Arif Nadeem Saqib
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
- Pakistan Health Research Council, Shahrah-e-Jamhuriat, G-5/2, Islamabad, Pakistan
| | - Izoduwa Abbe
- Department of Molecular and Human Genetics, Center for Statistical Genetics, Baylor College of Medicine, 1 Baylor Plaza 700D, Houston, TX, 77030, USA
| | - Ghazanfar Ali
- Department of Biotechnology, University of Azad Jammu and Kashmir, Muzaffarabad, Pakistan
| | - Muhammad Jawad Hassan
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Saadullah Khan
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology, Kohat, KPK, Pakistan
| | - Zahid Azeem
- Department of Biochemistry, Azad Jammu and Kashmir Medical College, Muzaffarabad, Pakistan
| | - Irfan Ullah
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Michael J Bamshad
- Department of Genome Sciences, University of Washington, Foege Building S-250, 3720 15th Ave. NE, Seattle, WA, 98195, USA
- Department of Pediatrics, University of Washington, 1959 NE Pacific St., Seattle, WA, 98195, USA
| | - Deborah A Nickerson
- Department of Genome Sciences, University of Washington, Foege Building S-250, 3720 15th Ave. NE, Seattle, WA, 98195, USA
| | - Isabelle Schrauwen
- Department of Molecular and Human Genetics, Center for Statistical Genetics, Baylor College of Medicine, 1 Baylor Plaza 700D, Houston, TX, 77030, USA
| | - Wasim Ahmad
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Muhammad Ansar
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Suzanne M Leal
- Department of Molecular and Human Genetics, Center for Statistical Genetics, Baylor College of Medicine, 1 Baylor Plaza 700D, Houston, TX, 77030, USA.
| |
Collapse
|
15
|
Patzlaff NE, Shen M, Zhao X. Regulation of Adult Neurogenesis by the Fragile X Family of RNA Binding Proteins. Brain Plast 2018; 3:205-223. [PMID: 30151344 PMCID: PMC6091053 DOI: 10.3233/bpl-170061] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The fragile X mental retardation protein (FMRP) has an important role in neural development. Functional loss of FMRP in humans leads to fragile X syndrome, and it is the most common monogenetic contributor to intellectual disability and autism. FMRP is part of a larger family of RNA-binding proteins known as FXRs, which also includes fragile X related protein 1 (FXR1P) and fragile X related protein 2 (FXR2P). Despite the similarities of the family members, the functions of FXR1P and FXR2P in human diseases remain unclear. Although most studies focus on FMRP's role in mature neurons, all three FXRs regulate adult neurogenesis. Extensive studies have demonstrated important roles of adult neurogenesis in neuroplasticity, learning, and cognition. Impaired adult neurogenesis is implicated in neuropsychiatric disorders, neurodegenerative diseases, and neurodevelopmental disorders. Interventions aimed at regulating adult neurogenesis are thus being evaluated as potential therapeutic strategies. Here, we review and discuss the functions of FXRs in adult neurogenesis and their known similarities and differences. Understanding the overlapping regulatory functions of FXRs in adult neurogenesis can give us insights into the adult brain and fragile X syndrome.
Collapse
Affiliation(s)
- Natalie E. Patzlaff
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Minjie Shen
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Xinyu Zhao
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
16
|
Khlghatyan J, Beaulieu JM. Are FXR Family Proteins Integrators of Dopamine Signaling and Glutamatergic Neurotransmission in Mental Illnesses? Front Synaptic Neurosci 2018; 10:22. [PMID: 30087606 PMCID: PMC6066532 DOI: 10.3389/fnsyn.2018.00022] [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: 04/01/2018] [Accepted: 06/29/2018] [Indexed: 01/11/2023] Open
Abstract
Dopamine receptors and related signaling pathways have long been implicated in pathophysiology and treatment of mental illnesses, including schizophrenia and bipolar disorder. Dopamine signaling may impact neuronal activity by modulation of glutamate neurotransmission. Recent evidence indicates a direct and/or indirect involvement of fragile X-related family proteins (FXR) in the regulation and mediation of dopamine receptor functions. FXRs consists of fragile X mental retardation protein 1 (Fmr1/FMRP) and its autosomal homologs Fxr1 and Fxr2. These RNA-binding proteins are enriched in the brain. Loss of function mutation in human FMR1 is the major genetic contributor to Fragile X mental retardation syndrome. Therefore, the role of FXR proteins has mostly been studied in the context of autism spectrum disorders. However, recent genome-wide association studies have linked this family to schizophrenia, bipolar disorders, and mood regulation pointing toward a broader involvement in mental illnesses. FXR family proteins play an important role in the regulation of glutamate-mediated neuronal activity and plasticity. Here, we discuss the brain-specific functions of FXR family proteins by focusing on the regulation of dopamine receptor functions, ionotropic glutamate receptors-mediated synaptic plasticity and contribution to mental illnesses. Based on recent evidence, we propose that FXR proteins are potential integrators of dopamine signaling and ionotropic glutamate transmission.
Collapse
Affiliation(s)
- Jivan Khlghatyan
- Department of Pharmacology and Toxicology, Medical Sciences Building, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec, QC, Canada
| | - Jean-Martin Beaulieu
- Department of Pharmacology and Toxicology, Medical Sciences Building, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec, QC, Canada
| |
Collapse
|
17
|
Eissa N, Al-Houqani M, Sadeq A, Ojha SK, Sasse A, Sadek B. Current Enlightenment About Etiology and Pharmacological Treatment of Autism Spectrum Disorder. Front Neurosci 2018; 12:304. [PMID: 29867317 PMCID: PMC5964170 DOI: 10.3389/fnins.2018.00304] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 04/19/2018] [Indexed: 12/22/2022] Open
Abstract
Autistic Spectrum Disorder (ASD) is a complex neurodevelopmental brain disorder characterized by two core behavioral symptoms, namely impairments in social communication and restricted/repetitive behavior. The molecular mechanisms underlying ASD are not well understood. Recent genetic as well as non-genetic animal models contributed significantly in understanding the pathophysiology of ASD, as they establish autism-like behavior in mice and rats. Among the genetic causes, several chromosomal mutations including duplications or deletions could be possible causative factors of ASD. In addition, the biochemical basis suggests that several brain neurotransmitters, e.g., dopamine (DA), serotonin (5-HT), gamma-amino butyric acid (GABA), acetylcholine (ACh), glutamate (Glu) and histamine (HA) participate in the onset and progression of ASD. Despite of convincible understanding, risperidone and aripiprazole are the only two drugs available clinically for improving behavioral symptoms of ASD following approval by Food and Drug Administration (FDA). Till date, up to our knowledge there is no other drug approved for clinical usage specifically for ASD symptoms. However, many novel drug candidates and classes of compounds are underway for ASD at different phases of preclinical and clinical drug development. In this review, the diversity of numerous aetiological factors and the alterations in variety of neurotransmitter generation, release and function linked to ASD are discussed with focus on drugs currently used to manage neuropsychiatric symptoms related to ASD. The review also highlights the clinical development of drugs with emphasis on their pharmacological targets aiming at improving core symptoms in ASD.
Collapse
Affiliation(s)
- Nermin Eissa
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohammed Al-Houqani
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Adel Sadeq
- Department of Clinical Pharmacy, College of Pharmacy, Al Ain University of Science and Technology, Al Ain, United Arab Emirates
| | - Shreesh K. Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Astrid Sasse
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, University of Dublin, Dublin, Ireland
| | - Bassem Sadek
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
18
|
Khlghatyan J, Evstratova A, Chamberland S, Marakhovskaia A, Bahremand A, Toth K, Beaulieu JM. Mental Illnesses-Associated Fxr1 and Its Negative Regulator Gsk3β Are Modulators of Anxiety and Glutamatergic Neurotransmission. Front Mol Neurosci 2018; 11:119. [PMID: 29706865 PMCID: PMC5906571 DOI: 10.3389/fnmol.2018.00119] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 03/28/2018] [Indexed: 12/13/2022] Open
Abstract
Genetic variants of the fragile X mental retardation syndrome-related protein 1 (FXR1) have been associated to mood regulation, schizophrenia, and bipolar disorders. Nonetheless, genetic association does not indicate a functional link of a given gene to neuronal activity and associated behaviors. In addition, interaction between multiple genes is often needed to sculpt complex traits such as behavior. Thus, modulation of neuronal functions by a given gene product, such as Fxr1, has to be thoroughly studied in the context of its interactions with other gene products. Glycogen synthase kinase-3 beta (GSK3β) is a shared target of several psychoactive drugs. In addition, interaction between functional polymorphisms of GSK3b and FXR1 has been implicated in mood regulation in healthy subjects and bipolar patients. However, the mechanistic underpinnings of this interaction remain unknown. We used somatic CRISPR/Cas9 mediated knockout and overexpression to investigate the impact of Fxr1 and its regulator Gsk3β on neuronal functions directly in the adult mouse brain. Suppression of Gsk3β or increase of Fxr1 expression in medial prefrontal cortex neurons leads to anxiolytic-like responses associated with a decrease in AMPA mediated excitatory postsynaptic currents. Furthermore, Fxr1 and Gsk3β modulate glutamatergic neurotransmission via regulation of AMPA receptor subunits GluA1 and GluA2 as well as vesicular glutamate transporter VGlut1. These results underscore a potential mechanism underlying the action of Fxr1 on neuronal activity and behaviors. Association between the Gsk3β-Fxr1 pathway and glutamatergic signaling also suggests how it may contribute to emotional regulation in response to mood stabilizers, or in illnesses like mood disorders and schizophrenia.
Collapse
Affiliation(s)
- Jivan Khlghatyan
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Alesya Evstratova
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Simon Chamberland
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | | | - Arash Bahremand
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Katalin Toth
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Jean-Martin Beaulieu
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| |
Collapse
|
19
|
Alwin Prem Anand A, Huber C, Asnet Mary J, Gallus N, Leucht C, Klafke R, Hirt B, Wizenmann A. Expression and function of microRNA-9 in the mid-hindbrain area of embryonic chick. BMC DEVELOPMENTAL BIOLOGY 2018; 18:3. [PMID: 29471810 PMCID: PMC5824543 DOI: 10.1186/s12861-017-0159-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 11/27/2017] [Indexed: 12/21/2022]
Abstract
Background MiR-9 is a small non-coding RNA that is highly conserved between species and primarily expressed in the central nervous system (CNS). It is known to influence proliferation and neuronal differentiation in the brain and spinal cord of different vertebrates. Different studies have pointed to regional and species-specific differences in the response of neural progenitors to miR-9. Methods In ovo and ex ovo electroporation was used to overexpress or reduce miR-9 followed by mRNA in situ hybridisation and immunofluorescent stainings to evaluate miR- expression and the effect of changed miR-9 expression. Results We have investigated the expression and function of miR-9 during early development of the mid-hindbrain region (MH) in chick. Our analysis reveals a closer relationship of chick miR-9 to mammalian miR-9 than to fish and a dynamic expression pattern in the chick neural tube. Early in development, miR-9 is diffusely expressed in the entire brain, bar the forebrain, and it becomes more restricted to specific areas of the CNS at later stages. MiR-9 overexpression at HH9–10 results in a reduction of FGF8 expression and premature neuronal differentiation in the mid-hindbrain boundary (MHB). Within the midbrain miR-9 does not cause premature neuronal differentiation it rather reduces proliferation in the midbrain. Conclusion Our findings indicate that miR-9 has regional specific effects in the developing mid-hindbrain region with a divergence of response of regional progenitors. Electronic supplementary material The online version of this article (10.1186/s12861-017-0159-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- A Alwin Prem Anand
- Institute of Clinical Anatomy and Cell Analysis, University of Tuebingen, Oesterbergstrasse 3, D-72074, Tuebingen, Germany
| | - Carola Huber
- Institute of Clinical Anatomy and Cell Analysis, University of Tuebingen, Oesterbergstrasse 3, D-72074, Tuebingen, Germany.,Robert-Bosch-Krankenhaus, Auerbachstraße 110, 70376, Stuttgart, Germany
| | - John Asnet Mary
- Department of Zoology, Fatima College, Madurai, Tamilnadu, 625018, India
| | - Nancy Gallus
- Institute of Clinical Anatomy and Cell Analysis, University of Tuebingen, Oesterbergstrasse 3, D-72074, Tuebingen, Germany.,Department of Neurobiology, McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Christoph Leucht
- Institute of Developmental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Ruth Klafke
- Institute of Developmental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Bernhard Hirt
- Institute of Clinical Anatomy and Cell Analysis, University of Tuebingen, Oesterbergstrasse 3, D-72074, Tuebingen, Germany
| | - Andrea Wizenmann
- Institute of Clinical Anatomy and Cell Analysis, University of Tuebingen, Oesterbergstrasse 3, D-72074, Tuebingen, Germany.
| |
Collapse
|
20
|
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
|
21
|
Rajman M, Schratt G. MicroRNAs in neural development: from master regulators to fine-tuners. Development 2017; 144:2310-2322. [DOI: 10.1242/dev.144337] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The proper formation and function of neuronal networks is required for cognition and behavior. Indeed, pathophysiological states that disrupt neuronal networks can lead to neurodevelopmental disorders such as autism, schizophrenia or intellectual disability. It is well-established that transcriptional programs play major roles in neural circuit development. However, in recent years, post-transcriptional control of gene expression has emerged as an additional, and probably equally important, regulatory layer. In particular, it has been shown that microRNAs (miRNAs), an abundant class of small regulatory RNAs, can regulate neuronal circuit development, maturation and function by controlling, for example, local mRNA translation. It is also becoming clear that miRNAs are frequently dysregulated in neurodevelopmental disorders, suggesting a role for miRNAs in the etiology and/or maintenance of neurological disease states. Here, we provide an overview of the most prominent regulatory miRNAs that control neural development, highlighting how they act as ‘master regulators’ or ‘fine-tuners’ of gene expression, depending on context, to influence processes such as cell fate determination, cell migration, neuronal polarization and synapse formation.
Collapse
Affiliation(s)
- Marek Rajman
- Biochemisch-Pharmakologisches Centrum, Institut für Physiologische Chemie, Philipps-Universität Marburg, Marburg 35043, Germany
| | - Gerhard Schratt
- Biochemisch-Pharmakologisches Centrum, Institut für Physiologische Chemie, Philipps-Universität Marburg, Marburg 35043, Germany
| |
Collapse
|
22
|
Hara Y, Ago Y, Takano E, Hasebe S, Nakazawa T, Hashimoto H, Matsuda T, Takuma K. Prenatal exposure to valproic acid increases miR-132 levels in the mouse embryonic brain. Mol Autism 2017; 8:33. [PMID: 28670439 PMCID: PMC5490164 DOI: 10.1186/s13229-017-0149-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 06/09/2017] [Indexed: 11/10/2022] Open
Abstract
Background MicroRNAs, small non-coding RNAs, are highly expressed in the mammalian brain, and the dysregulation of microRNA levels may be involved in neurodevelopmental disorders such as autism spectrum disorder (ASD). In the present study, we examined whether prenatal valproic acid (VPA) exposure affects levels of microRNAs, especially the brain specific and enriched microRNAs, in the mouse embryonic brain. Results Prenatal exposure to VPA at E12.5 immediately increased miR-132 levels, but not miR-9 or miR-124 levels, in the male embryonic brain. Prenatal exposure to VPA at E12.5 also increased miR-132 levels in the female embryonic brain. We further found that the prenatal exposure to VPA at E12.5 increased mRNA levels of Arc, c-Fos and brain-derived neurotrophic factor in both male and female embryonic brains, prior to miR-132 expression. In contrast, prenatal exposure to VPA at E14.5 did not affect miR-132 levels in either male or female embryonic brain. The prenatal VPA exposure at E12.5 also decreased mRNA levels of methyl-CpG-binding protein 2 and Rho GTPase-activating protein p250GAP, both of which are molecular targets of miR-132. Furthermore, RNA sequence analysis revealed that prenatal VPA exposure caused changes in several microRNA levels other than miR-132 in the embryonic whole brain. Conclusions These findings suggest that the alterations in neuronal activity-dependent microRNAs levels, including an increased level of miR-132, in the embryonic period, at least in part, underlie the ASD-like behaviors and cortical pathology produced by prenatal VPA exposure.
Collapse
Affiliation(s)
- Yuta Hara
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871 Japan.,Laboratory of Medicinal Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Yukio Ago
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871 Japan.,Laboratory of Medicinal Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Erika Takano
- Laboratory of Medicinal Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Shigeru Hasebe
- Department of Pharmacology, Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Takanobu Nakazawa
- Department of Pharmacology, Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871 Japan.,United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, 2-2 Yamadaoka, Suita, Osaka 565-0871 Japan.,Division of Bioscience, Institute for Datability Science, Osaka University, 1-1 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Toshio Matsuda
- Laboratory of Medicinal Pharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Kazuhiro Takuma
- Department of Pharmacology, Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka 565-0871 Japan.,United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, 2-2 Yamadaoka, Suita, Osaka 565-0871 Japan
| |
Collapse
|
23
|
Patzlaff NE, Nemec KM, Malone SG, Li Y, Zhao X. Fragile X related protein 1 (FXR1P) regulates proliferation of adult neural stem cells. Hum Mol Genet 2017; 26:1340-1352. [PMID: 28204491 PMCID: PMC6075589 DOI: 10.1093/hmg/ddx034] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/16/2017] [Accepted: 01/19/2017] [Indexed: 11/14/2022] Open
Abstract
Fragile X related protein 1 (FXR1P) is a member of the fragile X family of RNA-binding proteins, which includes FMRP and FXR2P. Both FMRP and FXR2P regulate neurogenesis, a process affected in a number of neurological and neuropsychiatric disorders, including fragile X syndrome. Although FXR1P has been implicated in various developmental processes and neuropsychiatric diseases, its role in neurodevelopment is not well understood. The goal of the present study was to elucidate the function of FXR1P in adult neurogenesis. We used an inducible mouse model that allows us to investigate how FXR1P deficiency in adult neural stem cells (aNSCs) affects proliferation and neuronal differentiation. Deletion of FXR1 in aNSCs resulted in fewer adult-born cells in the dentate gyrus (DG) overall, reducing populations across different stages of neurogenesis, including radial glia-like cells, intermediate progenitors, neuroblasts, immature neurons and neurons. We hypothesized that this reduction in new cell numbers resulted from impaired proliferation, which we confirmed both in vivo and in vitro. We discovered that FXR1P-deficient aNSCs have altered expression of a select number of cell-cycle genes, and we identified the mRNA of cyclin-dependent kinase inhibitor 1A (Cdkn1a, p21) as a direct target of FXR1P. Restoration of p21 mRNA to wild-type levels rescued the proliferation deficit in cells lacking FXR1P, demonstrating that p21 is a mediator of FXR1P in aNSCs. These results indicate that FXR1P plays an important role in regulating aNSC self-renewal and maintenance in the adult brain, which may have implications for a number of neurodevelopmental and psychiatric disorders.
Collapse
Affiliation(s)
- Natalie E. Patzlaff
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Kelsey M. Nemec
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Sydney G. Malone
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Yue Li
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Xinyu Zhao
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
24
|
Ma Y, Tian S, He S, Chen Q, Wang Z, Xiao X, Fu L, Lei X. The mechanism of action of FXR1P-related miR-19b-3p in SH-SY5Y. Gene 2016; 588:62-8. [DOI: 10.1016/j.gene.2016.04.037] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 04/19/2016] [Indexed: 11/28/2022]
|
25
|
RNA Secondary Structure Modulates FMRP's Bi-Functional Role in the MicroRNA Pathway. Int J Mol Sci 2016; 17:ijms17060985. [PMID: 27338369 PMCID: PMC4926514 DOI: 10.3390/ijms17060985] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/23/2016] [Accepted: 06/14/2016] [Indexed: 11/28/2022] Open
Abstract
MicroRNAs act by post-transcriptionally regulating the gene expression of 30%–60% of mammalian genomes. MicroRNAs are key regulators in all cellular processes, though the mechanism by which the cell activates or represses microRNA-mediated translational regulation is poorly understood. In this review, we discuss the RNA binding protein Fragile X Mental Retardation Protein (FMRP) and its role in microRNA-mediated translational regulation. Historically, FMRP is known to function as a translational suppressor. However, emerging data suggests that FMRP has both an agonistic and antagonistic role in regulating microRNA-mediated translational suppression. This bi-functional role is dependent on FMRP’s interaction with the RNA helicase Moloney leukemia virus 10 (MOV10), which modifies the structural landscape of bound mRNA, therefore facilitating or inhibiting its association with the RNA-Induced Silencing Complex.
Collapse
|
26
|
Hauberg ME, Roussos P, Grove J, Børglum AD, Mattheisen M. Analyzing the Role of MicroRNAs in Schizophrenia in the Context of Common Genetic Risk Variants. JAMA Psychiatry 2016; 73:369-77. [PMID: 26963595 PMCID: PMC7005318 DOI: 10.1001/jamapsychiatry.2015.3018] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
IMPORTANCE The recent implication of 108 genomic loci in schizophrenia marked a great advancement in our understanding of the disease. Against the background of its polygenic nature there is a necessity to identify how schizophrenia risk genes interplay. As regulators of gene expression, microRNAs (miRNAs) have repeatedly been implicated in schizophrenia etiology. It is therefore of interest to establish their role in the regulation of schizophrenia risk genes in disease-relevant biological processes. OBJECTIVE To examine the role of miRNAs in schizophrenia in the context of disease-associated genetic variation. DESIGN, SETTING, AND PARTICIPANTS The basis of this study was summary statistics from the largest schizophrenia genome-wide association study meta-analysis to date (83 550 individuals in a meta-analysis of 52 genome-wide association studies) completed in 2014 along with publicly available data for predicted miRNA targets. We examined whether schizophrenia risk genes were more likely to be regulated by miRNA. Further, we used gene set analyses to identify miRNAs that are regulators of schizophrenia risk genes. MAIN OUTCOMES AND MEASURES Results from association tests for miRNA targetomes and related analyses. RESULTS In line with previous studies, we found that similar to other complex traits, schizophrenia risk genes were more likely to be regulated by miRNAs (P < 2 × 10-16). Further, the gene set analyses revealed several miRNAs regulating schizophrenia risk genes, with the strongest enrichment for targets of miR-9-5p (P = .0056 for enrichment among the top 1% most-associated single-nucleotide polymorphisms, corrected for multiple testing). It is further of note that MIR9-2 is located in a genomic region showing strong evidence for association with schizophrenia (P = 7.1 × 10-8). The second and third strongest gene set signals were seen for the targets of miR-485-5p and miR-137, respectively. CONCLUSIONS AND RELEVANCE This study provides evidence for a role of miR-9-5p in the etiology of schizophrenia. Its implication is of particular interest as the functions of this neurodevelopmental miRNA tie in with established disease biology: it has a regulatory loop with the fragile X mental retardation homologue FXR1 and regulates dopamine D2 receptor density.
Collapse
Affiliation(s)
- Mads Engel Hauberg
- Department of Biomedicine, Aarhus University, Aarhus, Denmark2Lundbeck Foundation Initiative of Integrative Psychiatric Research, Lundbeck, Denmark3Centre for Integrative Sequencing, Aarhus University, Aarhus, Denmark
| | - Panos Roussos
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York5Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York6Institute for Genomics and Multiscale Biology, Icahn School of M
| | - Jakob Grove
- Department of Biomedicine, Aarhus University, Aarhus, Denmark2Lundbeck Foundation Initiative of Integrative Psychiatric Research, Lundbeck, Denmark3Centre for Integrative Sequencing, Aarhus University, Aarhus, Denmark9Bioinformatics Research Centre, Aarhu
| | - Anders Dupont Børglum
- Department of Biomedicine, Aarhus University, Aarhus, Denmark2Lundbeck Foundation Initiative of Integrative Psychiatric Research, Lundbeck, Denmark3Centre for Integrative Sequencing, Aarhus University, Aarhus, Denmark10Research Department P, Aarhus Univer
| | - Manuel Mattheisen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark2Lundbeck Foundation Initiative of Integrative Psychiatric Research, Lundbeck, Denmark3Centre for Integrative Sequencing, Aarhus University, Aarhus, Denmark
| |
Collapse
|
27
|
Role of MicroRNA in Governing Synaptic Plasticity. Neural Plast 2016; 2016:4959523. [PMID: 27034846 PMCID: PMC4808557 DOI: 10.1155/2016/4959523] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 01/06/2016] [Accepted: 02/14/2016] [Indexed: 12/21/2022] Open
Abstract
Although synaptic plasticity in neural circuits is orchestrated by an ocean of genes, molecules, and proteins, the underlying mechanisms remain poorly understood. Recently, it is well acknowledged that miRNA exerts widespread regulation over the translation and degradation of target gene in nervous system. Increasing evidence suggests that quite a few specific miRNAs play important roles in various respects of synaptic plasticity including synaptogenesis, synaptic morphology alteration, and synaptic function modification. More importantly, the miRNA-mediated regulation of synaptic plasticity is not only responsible for synapse development and function but also involved in the pathophysiology of plasticity-related diseases. A review is made here on the function of miRNAs in governing synaptic plasticity, emphasizing the emerging regulatory role of individual miRNAs in synaptic morphological and functional plasticity, as well as their implications in neurological disorders. Understanding of the way in which miRNAs contribute to synaptic plasticity provides rational clues in establishing the novel therapeutic strategy for plasticity-related diseases.
Collapse
|
28
|
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
|
29
|
Abstract
Drug addiction is characterized by uncontrolled drug consumption and high rates of relapse to drug taking during periods of attempted abstinence. Addiction is now largely considered a disorder of experience-dependent neuroplasticity, driven by remodeling of synapses in reward and motivation relevant brain circuits in response to a history of prolonged drug intake. Alterations in gene expression play a central role in addiction-relevant neuroplasticity, but the mechanisms by which additive drugs remodel brain motivation circuits remains unclear. MicroRNAs (miRNAs) are a class of noncoding RNA that can regulate the expression of large numbers of protein-coding mRNA transcripts by binding to the 3' untranslated region (3' UTR) of target transcripts and blocking their translation into the encoded protein or triggering their destabilization and degradation. Emerging evidence has implicated miRNAs in regulating addiction-relevant neuroplasticity in the brain, and in controlling the motivational properties of cocaine and other drugs of abuse. Here, the role for miRNAs in regulating basic aspects of neuronal function is reviewed. The involvement of miRNAs in controlling the motivational properties of addictive drugs is also summarized. Finally, mechanisms by which miRNAs exert their actions on drug intake, when known, are considered.
Collapse
Affiliation(s)
- Paul J Kenny
- Laboratory of Behavioral & Molecular Neuroscience, Department of Pharmacology & Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
30
|
Huang F, Long Z, Chen Z, Li J, Hu Z, Qiu R, Zhuang W, Tang B, Xia K, Jiang H. Investigation of Gene Regulatory Networks Associated with Autism Spectrum Disorder Based on MiRNA Expression in China. PLoS One 2015; 10:e0129052. [PMID: 26061495 PMCID: PMC4462583 DOI: 10.1371/journal.pone.0129052] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 05/03/2015] [Indexed: 11/25/2022] Open
Abstract
Autism spectrum disorder (ASD) comprise a group of neurodevelopmental disorders characterized by deficits in social and communication capacities and repetitive behaviors. Increasing neuroscientific evidence indicates that the neuropathology of ASD is widespread and involves epigenetic regulation in the brain. Differentially expressed miRNAs in the peripheral blood from autism patients were identified by high-throughput miRNA microarray analyses. Five of these miRNAs were confirmed through quantitative reverse transcription-polymerase chain reaction (qRT-PCR) analysis. A search for candidate target genes of the five confirmed miRNAs was performed through a Kyoto encyclopedia of genes and genomes (KEGG) biological pathways and Gene Ontology enrichment analysis of gene function to identify gene regulatory networks. To the best of our knowledge, this study provides the first global miRNA expression profile of ASD in China. The differentially expressed miR-34b may potentially explain the higher percentage of male ASD patients, and the aberrantly expressed miR-103a-3p may contribute to the abnormal ubiquitin-mediated proteolysis observed in ASD.
Collapse
Affiliation(s)
- Fengzhen Huang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
- Department of Neurology at University of South China, The First People’s Hospital of Chenzhou, Chenzhou, Hunan, 423000, P. R. China
- Institute of Translational Medicine at University of South China, The First People’s Hospital of Chenzhou, Chenzhou, Hunan, 423000, P. R. China
| | - Zhe Long
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Zhao Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Jiada Li
- State Key Laboratory of Medical Genetics of China, Central South University, Changsha, Hunan,410078, P. R. China
| | - Zhengmao Hu
- State Key Laboratory of Medical Genetics of China, Central South University, Changsha, Hunan,410078, P. R. China
| | - Rong Qiu
- School of Information Science and Engineering, Central South University, Hunan, 410083, P. R. China
- Hunan Engineering Laboratory for Advanced Control and Intelligent Automation, Hunan, 410083, P. R. China
| | - Wei Zhuang
- Department of Thoracic surgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
- State Key Laboratory of Medical Genetics of China, Central South University, Changsha, Hunan,410078, P. R. China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Kun Xia
- State Key Laboratory of Medical Genetics of China, Central South University, Changsha, Hunan,410078, P. R. China
| | - Hong Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
- State Key Laboratory of Medical Genetics of China, Central South University, Changsha, Hunan,410078, P. R. China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, Hunan, 410008, P. R. China
- * E-mail:
| |
Collapse
|
31
|
Taniguchi K, Ito Y, Sugito N, Kumazaki M, Shinohara H, Yamada N, Nakagawa Y, Sugiyama T, Futamura M, Otsuki Y, Yoshida K, Uchiyama K, Akao Y. Organ-specific PTB1-associated microRNAs determine expression of pyruvate kinase isoforms. Sci Rep 2015; 5:8647. [PMID: 25721733 PMCID: PMC4342556 DOI: 10.1038/srep08647] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 01/26/2015] [Indexed: 01/21/2023] Open
Abstract
The Warburg effect is a well-known feature of cancer cells. However, its' functional significance hasn't been elucidated yet. Pyruvate kinase muscle (PKM), which is a rate-limiting glycolytic enzyme, has 2 isoforms, PKM1 and PKM2. It has been reported that PKM2 is a tumor-specific isoform and promotes the Warburg effect. Also, it has been thought that tumor cells switch their PKM isoform from PKM1 to PKM2 during tumor development. Here, we showed that this switching machinery was induced only in limited cases, based on PKM expression in normal tissues, and that brain-specific microRNA (miR)-124 and muscle-specific miR-133b regulated this machinery by controlling PKM expression through targeting polypyrimidine tract-binding protein 1 (PTB1), which is a splicer of the PKM gene. Also, we confirmed that the PKM2/PKM1 ratio was further elevated in other PKM2-dominant organs such as colon through the down-regulation of these PTB1-associated microRNAs during tumor development.
Collapse
Affiliation(s)
- Kohei Taniguchi
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
- Department of General and Gastroenterological Surgery, Osaka Medical College, Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Yuko Ito
- Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Nobuhiko Sugito
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Minami Kumazaki
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Haruka Shinohara
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Nami Yamada
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Yoshihito Nakagawa
- Department of Gastroenterology, Fujita Health University, School of Medicine, Kutsukake-cho, Toyoake, Aichi 470-1192, Japan
| | - Tarou Sugiyama
- Department of Oncological surgery, Gifu University School of medicine, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Manabu Futamura
- Department of Oncological surgery, Gifu University School of medicine, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Yoshinori Otsuki
- Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Kazuhiro Yoshida
- Department of Oncological surgery, Gifu University School of medicine, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Kazuhisa Uchiyama
- Department of General and Gastroenterological Surgery, Osaka Medical College, Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Yukihiro Akao
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| |
Collapse
|
32
|
Ryan B, Joilin G, Williams JM. Plasticity-related microRNA and their potential contribution to the maintenance of long-term potentiation. Front Mol Neurosci 2015; 8:4. [PMID: 25755632 PMCID: PMC4337328 DOI: 10.3389/fnmol.2015.00004] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/04/2015] [Indexed: 12/24/2022] Open
Abstract
Long-term potentiation (LTP) is a form of synaptic plasticity that is an excellent model for the molecular mechanisms that underlie memory. LTP, like memory, is persistent, and both are widely believed to be maintained by a coordinated genomic response. Recently, a novel class of non-coding RNA, microRNA, has been implicated in the regulation of LTP. MicroRNA negatively regulate protein synthesis by binding to specific messenger RNA response elements. The aim of this review is to summarize experimental evidence for the proposal that microRNA play a major role in the regulation of LTP. We discuss a growing body of research which indicates that specific microRNA regulate synaptic proteins relevant to LTP maintenance, as well as studies that have reported differential expression of microRNA in response to LTP induction. We conclude that microRNA are ideally suited to contribute to the regulation of LTP-related gene expression; microRNA are pleiotropic, synaptically located, tightly regulated, and function in response to synaptic activity. The potential impact of microRNA on LTP maintenance as regulators of gene expression is enormous.
Collapse
Affiliation(s)
- Brigid Ryan
- Brain Health Research Centre, University of Otago, Dunedin New Zealand ; Department of Anatomy, Otago School of Medical Sciences, University of Otago, Dunedin New Zealand
| | - Greig Joilin
- Brain Health Research Centre, University of Otago, Dunedin New Zealand ; Department of Anatomy, Otago School of Medical Sciences, University of Otago, Dunedin New Zealand
| | - Joanna M Williams
- Brain Health Research Centre, University of Otago, Dunedin New Zealand ; Department of Anatomy, Otago School of Medical Sciences, University of Otago, Dunedin New Zealand
| |
Collapse
|
33
|
Chandran PA, Keller A, Weinmann L, Seida AA, Braun M, Andreev K, Fischer B, Horn E, Schwinn S, Junker M, Houben R, Dombrowski Y, Dietl J, Finotto S, Wölfl M, Meister G, Wischhusen J. The TGF-β-inducible miR-23a cluster attenuates IFN-γ levels and antigen-specific cytotoxicity in human CD8⁺ T cells. J Leukoc Biol 2014; 96:633-45. [PMID: 25030422 DOI: 10.1189/jlb.3a0114-025r] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cytokine secretion and degranulation represent key components of CD8(+) T-cell cytotoxicity. While transcriptional blockade of IFN-γ and inhibition of degranulation by TGF-β are well established, we wondered whether TGF-β could also induce immune-regulatory miRNAs in human CD8(+) T cells. We used miRNA microarrays and high-throughput sequencing in combination with qRT-PCR and found that TGF-β promotes expression of the miR-23a cluster in human CD8(+) T cells. Likewise, TGF-β up-regulated expression of the cluster in CD8(+) T cells from wild-type mice, but not in cells from mice with tissue-specific expression of a dominant-negative TGF-β type II receptor. Reporter gene assays including site mutations confirmed that miR-23a specifically targets the 3'UTR of CD107a/LAMP1 mRNA, whereas the further miRNAs expressed in this cluster-namely, miR-27a and -24-target the 3'UTR of IFN-γ mRNA. Upon modulation of the miR-23a cluster by the respective miRNA antagomirs and mimics, we observed significant changes in IFN-γ expression, but only slight effects on CD107a/LAMP1 expression. Still, overexpression of the cluster attenuated the cytotoxic activity of antigen-specific CD8(+) T cells. These functional data thus reveal that the miR-23a cluster not only is induced by TGF-β, but also exerts a suppressive effect on CD8(+) T-cell effector functions, even in the absence of TGF-β signaling.
Collapse
Affiliation(s)
- P Anoop Chandran
- Graduate School of Life Sciences (GSLS), University of Würzburg, Germany; Department of Obstetrics and Gynecology
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Lasse Weinmann
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Ahmed Adel Seida
- Department of Obstetrics and Gynecology, Interdisciplinary Center for Clinical Research
| | - Matthias Braun
- Pediatric Hematology, Oncology, and Stem Cell Transplantation, Children's Hospital
| | - Katerina Andreev
- Laboratory of Cellular and Molecular Lung Immunology, Institute of Molecular Pneumology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; and
| | | | - Evi Horn
- Department of Obstetrics and Gynecology
| | - Stefanie Schwinn
- Pediatric Hematology, Oncology, and Stem Cell Transplantation, Children's Hospital
| | - Markus Junker
- Department of Obstetrics and Gynecology, Interdisciplinary Center for Clinical Research
| | - Roland Houben
- Department of Dermatology, University of Würzburg Medical School, Würzburg, Germany
| | - Yvonne Dombrowski
- Department of Obstetrics and Gynecology, Interdisciplinary Center for Clinical Research
| | | | - Susetta Finotto
- Laboratory of Cellular and Molecular Lung Immunology, Institute of Molecular Pneumology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; and
| | - Matthias Wölfl
- Pediatric Hematology, Oncology, and Stem Cell Transplantation, Children's Hospital
| | - Gunter Meister
- Max Planck Institute of Biochemistry, Martinsried, Germany; Department of Biochemistry, University of Regensburg, Germany
| | - Jörg Wischhusen
- Department of Obstetrics and Gynecology, Interdisciplinary Center for Clinical Research,
| |
Collapse
|
34
|
Herai RR, Stefanacci L, Hrvoj-Mihic B, Chailangkarn T, Hanson K, Semendeferi K, Muotri AR. Micro RNA detection in long-term fixed tissue of cortical glutamatergic pyramidal neurons after targeted laser-capture neuroanatomical microdissection. J Neurosci Methods 2014; 235:76-82. [PMID: 24992573 DOI: 10.1016/j.jneumeth.2014.06.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Revised: 06/18/2014] [Accepted: 06/20/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND Formalin fixation (FF) is the standard and most common method for preserving postmortem brain tissue. FF stabilizes cellular morphology and tissue architecture, and can be used to study the distinct morphologic and genetic signatures of different cell types. Although the procedure involved in FF degrades messenger RNA over time, an alternative approach is to use small RNAs (sRNAs) for genetic analysis associated with cell morphology. Although genetic analysis is carried out on fresh or frozen tissue, there is limited availability or impossibility on targeting specific cell populations, respectively. NEW METHOD The goal of this study is to detect miRNA and other classes of sRNA stored in formalin or in paraffin embedded for over decades. Two brain samples, one formed by a mixed population of cortical and subcortical cells, and one formed by pyramidal shaped cells collected by laser-capture microdissection, were subjected to sRNA sequencing. RESULTS Performing bioinformatics analysis over the sequenced sRNA from brain tissue, we detected several classes of sRNA, such as miRNAs that play key roles in brain neurodevelopmental and maintenance pathways, and hsa-mir-155 expression in neurons. Comparison with existing method: Our method is the first to combine the approaches for: laser-capture of pyramidal neurons from long-term formalin-fixed brain; extract sRNA from laser-captured pyramidal neurons; apply a suite of bioinformatics tools to detect miRNA and other classes of sRNAs on sequenced samples having high levels of RNA degradation. CONCLUSION This is the first study to show that sRNA can be rescued from laser-captured FF pyramidal neurons.
Collapse
Affiliation(s)
- Roberto R Herai
- University of California San Diego, School of Medicine, Department of Pediatrics/Rady Children's Hospital San Diego, Department of Cellular & Molecular Medicine, Stem Cell Program, La Jolla, CA 92093, MC 0695, USA
| | - Lisa Stefanacci
- University of California San Diego, Department of Anthropology, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Branka Hrvoj-Mihic
- University of California San Diego, Department of Anthropology, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Thanathom Chailangkarn
- University of California San Diego, School of Medicine, Department of Pediatrics/Rady Children's Hospital San Diego, Department of Cellular & Molecular Medicine, Stem Cell Program, La Jolla, CA 92093, MC 0695, USA
| | - Kari Hanson
- University of California San Diego, Department of Anthropology, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Katerina Semendeferi
- University of California San Diego, Department of Anthropology, 9500 Gilman Drive, La Jolla, CA, 92093, USA.,Center for Academic Research and Training in Anthropogeny (CARTA), University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093. USA.,Neuroscience Graduate Program, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Alysson R Muotri
- University of California San Diego, School of Medicine, Department of Pediatrics/Rady Children's Hospital San Diego, Department of Cellular & Molecular Medicine, Stem Cell Program, La Jolla, CA 92093, MC 0695, USA.,Center for Academic Research and Training in Anthropogeny (CARTA), University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093. USA.,Neuroscience Graduate Program, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| |
Collapse
|
35
|
Fang J, Zhang T, Liu Y, Li Y, Zhou S, Song D, Zhao Y, Feng R, Zhang X, Li L, Wen J. PAX6 downregulates miR-124 expression to promote cell migration during embryonic stem cell differentiation. Stem Cells Dev 2014; 23:2297-310. [PMID: 24773074 DOI: 10.1089/scd.2013.0410] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
PAX6-null mice exhibit defects in multiple organs leading to neonatal lethality, but the mechanism by which this occurs has not yet fully elucidated. In this study, we generated induced pluripotent stem cells (iPSCs) from Pax6-mutant mice and investigated the effect of PAX6 on cell fate during embryoid body (EB) formation. We found that PAX6 promotes cell migration by directly downregulating miR-124, which is important for the fate transition of migratory cells during gastrulation of embryonic stem (ES) cells. Although several downstream targets of miR-124 have been reported, little is known regarding the upstream regulation of miR-124. When we observed EB formation of iPSCs from Pax6-mutant mice, we found that higher levels of miR-124 in Pax6 homozygous EBs (Homo-EBs) inhibited cell migration, whereas inhibition of miR-124 in Homo-EBs rescued the migratory phenotypes associated with PAX6 deficiency. Further, we found that PAX6 binds to the promoter regions of the miR-124-3 gene and directly represses its expression. Therefore, we propose a novel PAX6-miR-124 pathway that controls ES cell migration. Our findings may provide important information for studies on ES cell differentiation and embryonic development.
Collapse
Affiliation(s)
- Jing Fang
- 1 Department of Cell Biology, Peking University Stem Cell Research Center, Peking University Health Science Center , School of Basic Medical Sciences, Beijing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
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
|
37
|
Di Liegro CM, Schiera G, Di Liegro I. Regulation of mRNA transport, localization and translation in the nervous system of mammals (Review). Int J Mol Med 2014; 33:747-62. [PMID: 24452120 PMCID: PMC3976132 DOI: 10.3892/ijmm.2014.1629] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 12/09/2013] [Indexed: 12/13/2022] Open
Abstract
Post-transcriptional control of mRNA trafficking and metabolism plays a critical role in the actualization and fine tuning of the genetic program of cells, both in development and in differentiated tissues. Cis-acting signals, responsible for post-transcriptional regulation, reside in the RNA message itself, usually in untranslated regions, 5′ or 3′ to the coding sequence, and are recognized by trans-acting factors: RNA-binding proteins (RBPs) and/or non-coding RNAs (ncRNAs). ncRNAs bind short mRNA sequences usually present in the 3′-untranslated (3′-UTR) region of their target messages. RBPs recognize specific nucleotide sequences and/or secondary/tertiary structures. Most RBPs assemble on mRNA at the moment of transcription and shepherd it to its destination, somehow determining its final fate. Regulation of mRNA localization and metabolism has a particularly important role in the nervous system where local translation of pre-localized mRNAs has been implicated in developing axon and dendrite pathfinding, and in synapse formation. Moreover, activity-dependent mRNA trafficking and local translation may underlie long-lasting changes in synaptic efficacy, responsible for learning and memory. This review focuses on the role of RBPs in neuronal development and plasticity, as well as possible connections between ncRNAs and RBPs.
Collapse
Affiliation(s)
- Carlo Maria Di Liegro
- Department of Biological Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), I-90128 Palermo, Italy
| | - Gabriella Schiera
- Department of Biological Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), I-90128 Palermo, Italy
| | - Italia Di Liegro
- Department of Experimental Biomedicine and Clinical Neurosciences (BIONEC), University of Palermo, I-90127 Palermo, Italy
| |
Collapse
|
38
|
Coolen M, Katz S, Bally-Cuif L. miR-9: a versatile regulator of neurogenesis. Front Cell Neurosci 2013; 7:220. [PMID: 24312010 PMCID: PMC3834235 DOI: 10.3389/fncel.2013.00220] [Citation(s) in RCA: 217] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 10/31/2013] [Indexed: 12/21/2022] Open
Abstract
Soon after its discovery, microRNA-9 (miR-9) attracted the attention of neurobiologists, since it is one of the most highly expressed microRNAs in the developing and adult vertebrate brain. Functional analyses in different vertebrate species have revealed a prominent role of this microRNA in balancing proliferation in embryonic neural progenitor populations. Key transcriptional regulators such as FoxG1, Hes1 or Tlx, were identified as direct targets of miR-9, placing it at the core of the gene network controlling the progenitor state. Recent data also suggest that this function could extend to adult neural stem cells. Other studies point to a role of miR-9 in differentiated neurons. Moreover miR-9 has been implicated in human brain pathologies, either displaying a protective role, such as in Progeria, or participating in disease progression in brain cancers. Altogether functional studies highlight a prominent feature of this highly conserved microRNA, its functional versatility, both along its evolutionary history and across cellular contexts.
Collapse
Affiliation(s)
- Marion Coolen
- Zebrafish Neurogenetics Team, Laboratory of Neurobiology and Development, Institute of Neurobiology Alfred Fessard, CNRS Gif-sur-Yvette, France
| | | | | |
Collapse
|
39
|
Zhang Z, Almeida S, Lu Y, Nishimura AL, Peng L, Sun D, Wu B, Karydas AM, Tartaglia MC, Fong JC, Miller BL, Farese RV, Moore MJ, Shaw CE, Gao FB. Downregulation of microRNA-9 in iPSC-derived neurons of FTD/ALS patients with TDP-43 mutations. PLoS One 2013; 8:e76055. [PMID: 24143176 PMCID: PMC3797144 DOI: 10.1371/journal.pone.0076055] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 08/17/2013] [Indexed: 12/12/2022] Open
Abstract
Transactive response DNA-binding protein 43 (TDP-43) is a major pathological protein in frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). There are many disease-associated mutations in TDP-43, and several cellular and animal models with ectopic overexpression of mutant TDP-43 have been established. Here we sought to study altered molecular events in FTD and ALS by using induced pluripotent stem cell (iPSC) derived patient neurons. We generated multiple iPSC lines from an FTD/ALS patient with the TARDBP A90V mutation and from an unaffected family member who lacked the mutation. After extensive characterization, two to three iPSC lines from each subject were selected, differentiated into postmitotic neurons, and screened for relevant cell-autonomous phenotypes. Patient-derived neurons were more sensitive than control neurons to 100 nM straurosporine but not to other inducers of cellular stress. Three disease-relevant cellular phenotypes were revealed under staurosporine-induced stress. First, TDP-43 was localized in the cytoplasm of a higher percentage of patient neurons than control neurons. Second, the total TDP-43 level was lower in patient neurons with the A90V mutation. Third, the levels of microRNA-9 (miR-9) and its precursor pri-miR-9-2 decreased in patient neurons but not in control neurons. The latter is likely because of reduced TDP-43, as shRNA-mediated TDP-43 knockdown in rodent primary neurons also decreased the pri-miR-9-2 level. The reduction in miR-9 expression was confirmed in human neurons derived from iPSC lines containing the more pathogenic TARDBP M337V mutation, suggesting miR-9 downregulation might be a common pathogenic event in FTD/ALS. These results show that iPSC models of FTD/ALS are useful for revealing stress-dependent cellular defects of human patient neurons containing rare TDP-43 mutations in their native genetic contexts.
Collapse
Affiliation(s)
- Zhijun Zhang
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Sandra Almeida
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Yubing Lu
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Agnes L. Nishimura
- Department of Clinical Neuroscience, Institute of Psychiatry, London, United Kingdom
| | - Lingtao Peng
- Department of Biological Chemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Howard Hughes Medical Institute, Worcester, Massachusetts, United States of America
| | - Danqiong Sun
- Gladstone Institute of Cardiovascular Disease, San Francisco, California, United States of America
| | - Bei Wu
- Center for Neurologic Diseases, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Anna M. Karydas
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
| | - Maria C. Tartaglia
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
| | - Jamie C. Fong
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
| | - Bruce L. Miller
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
| | - Robert V. Farese
- Gladstone Institute of Cardiovascular Disease, San Francisco, California, United States of America
- Departments of Medicine and Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, United States of America
| | - Melissa J. Moore
- Department of Biological Chemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Howard Hughes Medical Institute, Worcester, Massachusetts, United States of America
| | - Christopher E. Shaw
- Department of Clinical Neuroscience, Institute of Psychiatry, London, United Kingdom
| | - Fen-Biao Gao
- Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| |
Collapse
|
40
|
Qiao Y, Badduke C, Mercier E, Lewis SME, Pavlidis P, Rajcan-Separovic E. miRNA and miRNA target genes in copy number variations occurring in individuals with intellectual disability. BMC Genomics 2013; 14:544. [PMID: 23937676 PMCID: PMC3750877 DOI: 10.1186/1471-2164-14-544] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Accepted: 08/06/2013] [Indexed: 12/20/2022] Open
Abstract
Background MicroRNAs (miRNAs) are a family of short, non-coding RNAs modulating expression of human protein coding genes (miRNA target genes). Their dysfunction is associated with many human diseases, including neurodevelopmental disorders. It has been recently shown that genomic copy number variations (CNVs) can cause aberrant expression of integral miRNAs and their target genes, and contribute to intellectual disability (ID). Results To better understand the CNV-miRNA relationship in ID, we investigated the prevalence and function of miRNAs and miRNA target genes in five groups of CNVs. Three groups of CNVs were from 213 probands with ID (24 de novo CNVs, 46 familial and 216 common CNVs), one group of CNVs was from a cohort of 32 cognitively normal subjects (67 CNVs) and one group of CNVs represented 40 ID related syndromic regions listed in DECIPHER (30 CNVs) which served as positive controls for CNVs causing or predisposing to ID. Our results show that 1). The number of miRNAs is significantly higher in de novo or DECIPHER CNVs than in familial or common CNV subgroups (P < 0.01). 2). miRNAs with brain related functions are more prevalent in de novo CNV groups compared to common CNV groups. 3). More miRNA target genes are found in de novo, familial and DECIPHER CNVs than in the common CNV subgroup (P < 0.05). 4). The MAPK signaling cascade is found to be enriched among the miRNA target genes from de novo and DECIPHER CNV subgroups. Conclusions Our findings reveal an increase in miRNA and miRNA target gene content in de novo versus common CNVs in subjects with ID. Their expression profile and participation in pathways support a possible role of miRNA copy number change in cognition and/or CNV-mediated developmental delay. Systematic analysis of expression/function of miRNAs in addition to coding genes integral to CNVs could uncover new causes of ID.
Collapse
Affiliation(s)
- Ying Qiao
- Department of Pathology and Lab Medicine, BC Child and Family Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | | | | | | | | | | |
Collapse
|
41
|
Plakophilin-associated RNA-binding proteins in prostate cancer and their implications in tumor progression and metastasis. Virchows Arch 2013; 463:379-90. [PMID: 23881279 DOI: 10.1007/s00428-013-1452-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 07/01/2013] [Accepted: 07/08/2013] [Indexed: 01/04/2023]
Abstract
Both plakophilins (PKP) 1 and 3 play a role in the progression of prostate cancer. The RNA-binding proteins (RBPs) GAP-SH3-binding protein (G3BP), fragile-X-related protein 1 (FXR1), poly(A)-binding protein, cytoplasmic 1 (PABPC1), and up-frameshift factor 1 (UPF1) are associated with PKP3. All these RBPs have an impact on RNA metabolism. Until recently, the PKP-associated RBPs have not been analyzed in prostate cancer. In the current study, we showed by affinity purification that the PKP3-associated RBPs were also binding partners of PKP1. We examined the expression of PKP1/3-associated RBPs and PKP1/3 in prostate cell lines, tumor-free prostate, and 136 prostatic adenocarcinomas by immunofluorescence and immunoblot. All four RBPs G3BP, FXR1, UPF1, and PABPC1 were expressed in the glandular epithelium of the normal prostate. PKP1 and FXR1 were strongly reduced in tumor tissues with Gleason score >7 and diminished expression of PKP1 and FXR1 also appeared to be associated with a metastatic phenotype. Additionally, the predominant nuclear localization of UPF1 in normal glandular cells and low grade tumors was switched to a more cytoplasmic pattern in carcinomas with Gleason score >7. Our findings suggest that PKP1 and FXR1 may have a tumor-suppressive function and are downregulated in more aggressive tumors. Collectively, PKP1/3-associated RBPs FXR1 and UPF1 may have a functional role in prostate cancer progression and metastasis and highlight the potential importance of posttranscriptional regulation of gene expression and nonsense-mediated decay in cancer.
Collapse
|
42
|
Hörnberg H, Holt C. RNA-binding proteins and translational regulation in axons and growth cones. Front Neurosci 2013; 7:81. [PMID: 23734093 PMCID: PMC3661996 DOI: 10.3389/fnins.2013.00081] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 05/06/2013] [Indexed: 01/11/2023] Open
Abstract
RNA localization and regulation play an important role in the developing and adult nervous system. In navigating axons, extrinsic cues can elicit rapid local protein synthesis that mediates directional or morphological responses. The mRNA repertoire in axons is large and dynamically changing, yet studies suggest that only a subset of these mRNAs are translated after cue stimulation, suggesting the need for a high level of translational regulation. Here, we review the role of RNA-binding proteins (RBPs) as local regulators of translation in developing axons. We focus on their role in growth, guidance, and synapse formation, and discuss the mechanisms by which they regulate translation in axons.
Collapse
Affiliation(s)
- Hanna Hörnberg
- Department of Physiology Development and Neuroscience, University of Cambridge Cambridge, UK
| | | |
Collapse
|
43
|
Siew WH, Tan KL, Babaei MA, Cheah PS, Ling KH. MicroRNAs and intellectual disability (ID) in Down syndrome, X-linked ID, and Fragile X syndrome. Front Cell Neurosci 2013; 7:41. [PMID: 23596395 PMCID: PMC3625835 DOI: 10.3389/fncel.2013.00041] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 03/27/2013] [Indexed: 01/01/2023] Open
Abstract
Intellectual disability (ID) is one of the many features manifested in various genetic syndromes leading to deficits in cognitive function among affected individuals. ID is a feature affected by polygenes and multiple environmental factors. It leads to a broad spectrum of affected clinical and behavioral characteristics among patients. Until now, the causative mechanism of ID is unknown and the progression of the condition is poorly understood. Advancement in technology and research had identified various genetic abnormalities and defects as the potential cause of ID. However, the link between these abnormalities with ID is remained inconclusive and the roles of many newly discovered genetic components such as non-coding RNAs have not been thoroughly investigated. In this review, we aim to consolidate and assimilate the latest development and findings on a class of small non-coding RNAs known as microRNAs (miRNAs) involvement in ID development and progression with special focus on Down syndrome (DS) and X-linked ID (XLID) [including Fragile X syndrome (FXS)].
Collapse
Affiliation(s)
- Wei-Hong Siew
- NeuroBiology and Genetics Group, Genetic Medicine Research Centre, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia UPM Serdang, Malaysia ; Clinical Genetics Unit, Department of Obstetrics and Gynaecology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia UPM Serdang, Malaysia
| | | | | | | | | |
Collapse
|
44
|
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
|
45
|
Stappert L, Borghese L, Roese-Koerner B, Weinhold S, Koch P, Terstegge S, Uhrberg M, Wernet P, Brüstle O. MicroRNA-based promotion of human neuronal differentiation and subtype specification. PLoS One 2013; 8:e59011. [PMID: 23527072 PMCID: PMC3601127 DOI: 10.1371/journal.pone.0059011] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 01/17/2013] [Indexed: 11/19/2022] Open
Abstract
MicroRNAs are key regulators of neural cell proliferation, differentiation and fate choice. Due to the limited access to human primary neural tissue, the role of microRNAs in human neuronal differentiation remains largely unknown. Here, we use a population of long-term self-renewing neuroepithelial-like stem cells (lt-NES cells) derived from human embryonic stem cells to study the expression and function of microRNAs at early stages of human neural stem cell differentiation and neuronal lineage decision. Based on microRNA expression profiling followed by gain- and loss-of-function analyses in lt-NES cells and their neuronal progeny, we demonstrate that miR-153, miR-324-5p/3p and miR-181a/a contribute to the shift of lt-NES cells from self-renewal to neuronal differentiation. We further show that miR-125b and miR-181a specifically promote the generation of neurons of dopaminergic fate, whereas miR-181a inhibits the development of this neurotransmitter subtype. Our data demonstrate that time-controlled modulation of specific microRNA activities not only regulates human neural stem cell self-renewal and differentiation but also contributes to the development of defined neuronal subtypes.
Collapse
Affiliation(s)
- Laura Stappert
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn and Hertie Foundation, Bonn, Germany
| | - Lodovica Borghese
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn and Hertie Foundation, Bonn, Germany
- * E-mail: (LB); (OB)
| | - Beate Roese-Koerner
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn and Hertie Foundation, Bonn, Germany
| | - Sandra Weinhold
- Institute for Transplantation Diagnostics and Cell Therapeutics, University Düsseldorf, Düsseldorf, Germany
| | - Philipp Koch
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn and Hertie Foundation, Bonn, Germany
| | - Stefanie Terstegge
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn and Hertie Foundation, Bonn, Germany
| | - Markus Uhrberg
- Institute for Transplantation Diagnostics and Cell Therapeutics, University Düsseldorf, Düsseldorf, Germany
| | - Peter Wernet
- Institute for Transplantation Diagnostics and Cell Therapeutics, University Düsseldorf, Düsseldorf, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn and Hertie Foundation, Bonn, Germany
- * E-mail: (LB); (OB)
| |
Collapse
|
46
|
Abstract
Regulation of miRNA (microRNA) biogenesis shapes the profiles of miRNAs in the living cell, contributing to cell identity and function. Importantly, aberrant miRNA levels have been linked to a variety of human pathological states. In recent years, a number of proteins have been shown to regulate the miRNA biogenesis at the level of Drosha and Dicer cleavage. A large proportion of these factors regulate miRNA production through binding to the TL (terminal loop) regions of miRNA progenitors. In the present paper, we review the current knowledge about pri-miRNA (primary miRNA) and pre-miRNA (precursor miRNA) TL involvement in the regulation of miRNA biogenesis.
Collapse
|
47
|
Li Z, Lu Y, Xu XL, Gao FB. The FTD/ALS-associated RNA-binding protein TDP-43 regulates the robustness of neuronal specification through microRNA-9a in Drosophila. Hum Mol Genet 2012; 22:218-25. [PMID: 23042786 DOI: 10.1093/hmg/dds420] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
TDP-43 is an evolutionarily conserved RNA-binding protein currently under intense investigation for its involvement in the molecular pathogenesis of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). TDP-43 is normally localized in the nucleus, but translocated to the cytoplasm in diseased neurons. The endogenous functions of TDP-43 in the nervous system remain poorly understood. Here, we show that the loss of Drosophila TDP-43 (dTDP-43) results in an increased production of sensory bristles and sensory organ precursor (SOP) cells on the notum of some but not all flies. The location of ectopic SOPs varies among mutant flies. The penetrance of this novel phenotype is dependent on the gender and sensitive to environmental influences. A similar SOP phenotype was also observed on the wing and in the embryos. Overexpression of dTDP-43 causes both loss and ectopic production of SOPs. Ectopic expression of ALS-associated mutant human TDP-43 (hTDP-43(M337V) and hTDP-43(Q331K)) produces a less severe SOP phenotype than hTDP-43(WT), indicating a partial loss of function of mutant hTDP-43. In dTDP-43 mutants, miR-9a expression is significantly reduced. Genetic interaction studies further support the notion that dTDP-43 acts through miR-9a to control the precision of SOP specification. These findings reveal a novel role for endogenous TDP-43 in neuronal specification and suggest that the FTD/ALS-associated RNA-binding protein TDP-43 functions to ensure the robustness of genetic control programs.
Collapse
Affiliation(s)
- Zhaodong Li
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | |
Collapse
|
48
|
Emerging roles of non-coding RNAs in brain evolution, development, plasticity and disease. Nat Rev Neurosci 2012; 13:528-41. [PMID: 22814587 DOI: 10.1038/nrn3234] [Citation(s) in RCA: 430] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Novel classes of small and long non-coding RNAs (ncRNAs) are being characterized at a rapid pace, driven by recent paradigm shifts in our understanding of genomic architecture, regulation and transcriptional output, as well as by innovations in sequencing technologies and computational and systems biology. These ncRNAs can interact with DNA, RNA and protein molecules; engage in diverse structural, functional and regulatory activities; and have roles in nuclear organization and transcriptional, post-transcriptional and epigenetic processes. This expanding inventory of ncRNAs is implicated in mediating a broad spectrum of processes including brain evolution, development, synaptic plasticity and disease pathogenesis.
Collapse
|
49
|
Abstract
MicroRNAs (miRNAs) are short non-coding RNAs that regulate gene expression at the post-transcriptional level by mediating mRNA degradation or translational inhibition. MiRNAs are implicated in many biological functions, including neurogenesis. It has been shown that miRNAs regulate multiple steps of neurogenesis, from neural stem cell proliferation to neuronal differentiation and maturation. MiRNAs execute their functions in a dynamic and context-dependent manner by targeting diverse downstream target genes, from transcriptional factors to epigenetic regulators. Identifying context-specific target genes is instrumental for understanding the roles that miRNAs play in neurogenesis. This review summarizes our current state of knowledge on the dynamic roles that miRNAs play in neural stem cells and neurogenesis.
Collapse
Affiliation(s)
- Ming-Fei Lang
- Department of Neurosciences, Center for Gene Expression and Drug Discovery, Cancer Center, Beckman Research Institute of City of Hope Duarte, CA, USA
| | | |
Collapse
|
50
|
Im HI, Kenny PJ. MicroRNAs in neuronal function and dysfunction. Trends Neurosci 2012; 35:325-34. [PMID: 22436491 PMCID: PMC3565236 DOI: 10.1016/j.tins.2012.01.004] [Citation(s) in RCA: 333] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 01/24/2012] [Accepted: 01/24/2012] [Indexed: 01/07/2023]
Abstract
MicroRNAs (miRNAs) are small noncoding RNA transcripts expressed throughout the brain that can regulate neuronal gene expression at the post-transcriptional level. Here, we provide an overview of the role for miRNAs in brain development and function, and review evidence suggesting that dysfunction in miRNA signaling contributes to neurodevelopment disorders such as Rett and fragile X syndromes, as well as complex behavioral disorders including schizophrenia, depression and drug addiction. A better understanding of how miRNAs influence the development of neuropsychiatric disorders may reveal fundamental insights into the causes of these devastating illnesses and offer novel targets for therapeutic development.
Collapse
Affiliation(s)
- Heh-In Im
- Laboratory of Behavioral and Molecular Neuroscience, Department of Molecular Therapeutics, The Scripps Research Institute - Scripps Florida, Jupiter, FL 33458, USA
| | | |
Collapse
|