1
|
Simmonds E, Leonenko G, Yaman U, Bellou E, Myers A, Morgan K, Brookes K, Hardy J, Salih D, Escott-Price V. Chromosome X-wide association study in case control studies of pathologically confirmed Alzheimer's disease in a European population. Transl Psychiatry 2024; 14:358. [PMID: 39231932 PMCID: PMC11375158 DOI: 10.1038/s41398-024-03058-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/06/2024] Open
Abstract
Although there are several genome-wide association studies available which highlight genetic variants associated with Alzheimer's disease (AD), often the X chromosome is excluded from the analysis. We conducted an X-chromosome-wide association study (XWAS) in three independent studies with a pathologically confirmed phenotype (total 1970 cases and 1113 controls). The XWAS was performed in males and females separately, and these results were then meta-analysed. Four suggestively associated genes were identified which may be of potential interest for further study in AD, these are DDX53 (rs12006935, OR = 0.52, p = 6.9e-05), IL1RAPL1 (rs6628450, OR = 0.36, p = 4.2e-05; rs137983810, OR = 0.52, p = 0.0003), TBX22 (rs5913102, OR = 0.74, p = 0.0003) and SH3BGRL (rs186553004, OR = 0.35, p = 0.0005; rs113157993, OR = 0.52, p = 0.0003), which replicate across at least two studies. The SNP rs5913102 in TBX22 achieves chromosome-wide significance in meta-analysed data. DDX53 shows highest expression in astrocytes, IL1RAPL1 is most highly expressed in oligodendrocytes and neurons and SH3BGRL is most highly expressed in microglia. We have also identified SNPs in the NXF5 gene at chromosome-wide significance in females (rs5944989, OR = 0.62, p = 1.1e-05) but not in males (p = 0.83). The discovery of relevant AD associated genes on the X chromosome may identify AD risk differences and similarities based on sex and lead to the development of sex-stratified therapeutics.
Collapse
Affiliation(s)
- Emily Simmonds
- Dementia Research Institute, Cardiff University, Cardiff, UK
| | - Ganna Leonenko
- Dementia Research Institute, Cardiff University, Cardiff, UK
| | - Umran Yaman
- Institute of Neurology, University College London, London, UK
| | - Eftychia Bellou
- Dementia Research Institute, Cardiff University, Cardiff, UK
| | - Amanda Myers
- Department of Cell Biology, University of Miami, Miller School of Medicine, Miami, FL, USA
| | | | - Keeley Brookes
- Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - John Hardy
- Institute of Neurology, University College London, London, UK
| | - Dervis Salih
- Institute of Neurology, University College London, London, UK
| | - Valentina Escott-Price
- Dementia Research Institute, Cardiff University, Cardiff, UK.
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK.
| |
Collapse
|
2
|
Qin S, Zeng J, Wang J, Ye M, Deng Q, Wang X, Zhang Z, Yi D, Wu Y, Li-Ling J. Delineation of an inverted tandem Xq23-26.3 duplication in a female featuring extremely short stature and mild mental deficiency. Mol Cytogenet 2023; 16:33. [PMID: 38031145 PMCID: PMC10685508 DOI: 10.1186/s13039-023-00663-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/06/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Partial duplications involving the long arm of the X chromosome are associated with mental retardation, short stature, microcephaly, and a wide range of physical findings. Female carriers usually have no clinical phenotype. Occasionally, they may also have heterogeneous features due to non-random inactivation of the X chromosome. METHODS The peripheral blood sample was collected from the patient and subjected to a few genetic testing, including chromosomal karyotyping, Chromosomal microarray analysis (CMA), Optical genome mapping, short tandem repeat (STR) analysis for Determination of parental origin, and X chromosome inactivation (XCI) analysis. RESULTS We have identified a de novo Xq23-Xq26.3 duplication in an adult female featuring extremely short stature and mild mental deficiency. Chromosome analysis detected a duplication on Xq23-q26.3 with a size of approximately 20 Mb. The duplication region has encompassed a number of genes, among which ARHGEF6, PHF6, HPRT1 and SLC9A6 are associated with X-linked mental retardation. Further analysis suggested that the duplication has derived from her father, was of the inversion duplication type and involved various degrees of skewed X chromosome inactivation. CONCLUSION Correlation with her phenotypes might indicate new mechanisms by which the X chromosome may lead to short stature and mental retardation. Our findings thereby may shed more light on the phenotypic implication of functional disomy of X-chromosome genes.
Collapse
Affiliation(s)
- Shengfang Qin
- Department of Medical Genetics and Prenatal Diagnosis, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, Sichuan, China.
| | - Jiuzhi Zeng
- West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, Sichuan, China
| | - Jin Wang
- Department of Medical Genetics and Prenatal Diagnosis, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, Sichuan, China
| | - Mengling Ye
- Department of Medical Genetics and Prenatal Diagnosis, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, Sichuan, China
| | - Qin Deng
- Department of Medical Genetics and Prenatal Diagnosis, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, Sichuan, China
| | - Xueyan Wang
- Department of Medical Genetics and Prenatal Diagnosis, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, Sichuan, China
| | - Zhuo Zhang
- Department of Medical Genetics and Prenatal Diagnosis, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, Sichuan, China
| | - Dangying Yi
- Department of Medical Genetics and Prenatal Diagnosis, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, Sichuan, China
| | - Yang Wu
- Department of Medical Genetics and Prenatal Diagnosis, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, Sichuan, China
| | - Jesse Li-Ling
- West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Department of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, 610045, Sichuan, China.
| |
Collapse
|
3
|
Sending the message: specialized RNA export mechanisms in trypanosomes. Trends Parasitol 2022; 38:854-867. [PMID: 36028415 PMCID: PMC9894534 DOI: 10.1016/j.pt.2022.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 02/04/2023]
Abstract
Export of RNA from the nucleus is essential for all eukaryotic cells and has emerged as a major step in the control of gene expression. mRNA molecules are required to complete a complex series of processing events and pass a quality control system to protect the cytoplasm from the translation of aberrant proteins. Many of these events are highly conserved across eukaryotes, reflecting their ancient origin, but significant deviation from a canonical pathway as described from animals and fungi has emerged in the trypanosomatids. With significant implications for the mechanisms that control gene expression and hence differentiation, responses to altered environments and fitness as a parasite, these deviations may also reveal additional, previously unsuspected, mRNA export pathways.
Collapse
|
4
|
Miwa T, Ohtani K, Inoue K, Sakamoto H. The germ cell-specific TAP-like protein NXF-2 forms a novel granular structure and is required for tra-2 3'UTR-dependent mRNA export in Caenorhabditis elegans. Genes Cells 2022; 27:621-628. [PMID: 35950937 DOI: 10.1111/gtc.12978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/04/2022] [Accepted: 08/07/2022] [Indexed: 11/29/2022]
Abstract
TAP is a general mRNA export receptor and is highly conserved among eukaryotes. The nematode Caenorhabditis elegans has another TAP-like protein, NXF-2, but little is known about its function. In this study, we show that NXF-2 is specifically expressed in germ cells and forms a novel granular structure that is different from that of P granules and that NXF-2 granules are anchored to the nuclear periphery in the mitotic region of the hermaphrodite gonad. In contrast, NXF-2 granules are released within the whole cytoplasm in the meiotic region, where the feminization gene tra-2 starts to function. Both inhibition of XPO-1 (an ortholog of the export receptor CRM1) and mutation of the nuclear export signal of NXF-2 caused the release of NXF-2 granules from the nuclear periphery, indicating that anchoring of NXF-2 granules depends on XPO-1 function. Moreover, inhibition of NXF-2 resulted in a substantial nuclear accumulation of the reporter mRNA carrying the tra-2 3'UTR. These results suggest that, together with XPO-1, NXF-2 exports and anchors tra-2 mRNA to the nuclear periphery to avoid precocious translation until the germ cells reach the meiotic region, thereby contributing to the regulation of tra-2 mRNA expression. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Takashi Miwa
- Biology, Kobe University Graduate School of Science Faculty of Science, Grad. Sch. Sci. Tech.1-1 Rokkodai, Nada-ku, Kobe Hyogo, Japan
| | - Keigo Ohtani
- Biology, Kobe University Graduate School of Science Faculty of Science, Grad. Sch. Sci. Tech.1-1 Rokkodai, Nada-ku, Kobe Hyogo, Japan
| | - Kunio Inoue
- Biology, Kobe University Graduate School of Science Faculty of Science, Grad. Sch. Sci. Tech.1-1 Rokkodai, Nada-ku, Kobe Hyogo, Japan
| | - Hiroshi Sakamoto
- Biology, Kobe University Graduate School of Science Faculty of Science, Grad. Sch. Sci. Tech.1-1 Rokkodai, Nada-ku, Kobe Hyogo, Japan
| |
Collapse
|
5
|
Sajid A, Saeed MS, Malik RM, Fazal S, Malik S, Kamal MA. Prediction of Secondary and Tertiary Structure and Docking of Rb1WT
And Rb1R661W Proteins. CURRENT BIOTECHNOLOGY 2022. [DOI: 10.2174/2211550111666220127100203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Background:
Retinoblastoma, a malignancy occurring in the juvenile cells of the retina,
is responsible for light detection. It is one of the most emerging ra re childhood and infant cancer.
It is initiated by the mutation in Rb1, a first tumor suppressor gene located on chromosome 13q14.
Rb1 protein is responsible for cell cycle regulation.
Methods:
In our study, secondary and 3D-Structural predictions of Rb1WT and Rb1R661W were made
by comparative or homology modeling to find any structural change leading to the disruption in its
further interactions. Quality assurance of the structures was done by Ramachandran Plot for a stable
structure. Both the proteins were then applied by docking process with proteins of interest.
Results:
Secondary structure showed a number of mutations in helixes, β-Hairpins of Rb1R661W. The
major change was the loss of β-Hairpin loop, extension and shortening of helixes. 3D comparison
structure showed a change in the groove of Rb1R661W. Docking results, unlike RB1 WT, had different
and no interactions with some of the proteins of interest. This mutation in Rb1 protein had a deleterious
effect on the protein functionality.
Conclusion:
This study will help to design the appropriate therapy and also understand the mechanism
of disease of retinoblastoma, for researchers and pharmaceuticals.
Collapse
Affiliation(s)
- Aimen Sajid
- Capital University of Science and Technology, Islamabad, Pakistan
| | | | - Rabbiah Manzoor Malik
- Capital University of Science and Technology, Islamabad, Pakistan
- Wah Medical College, Wah Cantt, Pakistan
| | - Sahar Fazal
- Capital University of Science and Technology, Islamabad, Pakistan
| | - Shaukat Malik
- Capital University of Science and Technology, Islamabad, Pakistan
| | - Mohammad Amjad Kamal
- West China School of Nursing / Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- King Fahd Medical Research
Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Saudi Arabia
- Enzymoics, 7 Peterlee
Place, Hebersham, NSW 2770; Novel Global Community Educational Foundation, Australia
| |
Collapse
|
6
|
An F-Box Protein, Mdm30, Interacts with TREX Subunit Sub2 To Regulate Cellular Abundance Cotranscriptionally in Orchestrating mRNA Export Independently of Splicing and Mitochondrial Function. Mol Cell Biol 2020; 40:MCB.00570-19. [PMID: 31932480 DOI: 10.1128/mcb.00570-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 01/03/2020] [Indexed: 02/02/2023] Open
Abstract
Although an F-box protein, Mdm30, is found to regulate ubiquitylation of the Sub2 component of TREX (transcription-export) complex for proteasomal degradation in stimulation of mRNA export, it remains unknown whether such ubiquitin-proteasome system (UPS) regulation of Sub2 occurs cotranscriptionally via its interaction with Mdm30. Further, it is unclear whether impaired UPS regulation of Sub2 in the absence of Mdm30 alters mRNA export via splicing defects of export factors and/or mitochondrial dynamics/function, since Sub2 controls mRNA splicing and Mdm30 regulates mitochondrial aggregation. Here, we show that Mdm30 interacts with Sub2, and temporary shutdown of Mdm30 enhances Sub2's abundance and impairs mRNA export. Likewise, Sub2's abundance is increased following transcriptional inhibition. These results support Mdm30's direct role in regulation of Sub2's cellular abundance in a transcription-dependent manner. Consistently, the chromatin-bound Sub2 level is increased in the absence of Mdm30. Further, we find that Mdm30 does not facilitate splicing of export factors. Moreover, Mdm30 does not have a dramatic effect on mitochondrial respiration/function, and mRNA export occurs in the absence of Fzo1, which is required for mitochondrial dynamics/respiration. Collective results reveal that Mdm30 interacts with Sub2 for proteasomal degradation in a transcription-dependent manner to promote mRNA export independently of splicing or mitochondrial function, thus advancing our understanding of mRNA export.
Collapse
|
7
|
Zeng T, Wang D, Chen J, Chen K, Yu G, Chen Q, Liu Y, Yan S, Zhu L, Zhou H, Huang A, Tang H. AF119895 regulates NXF3 expression to promote migration and invasion of hepatocellular carcinoma through an interaction with miR-6508-3p. Exp Cell Res 2017; 363:129-139. [PMID: 29274323 DOI: 10.1016/j.yexcr.2017.12.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 12/15/2017] [Accepted: 12/16/2017] [Indexed: 12/25/2022]
Abstract
Various studies revealed that numerous long noncoding RNAs (lncRNAs) have been found dysregulated in HCC and played important role in hepatocarcinogenesis, although the underlying mechanism still remains unclear. Herein, we reported AF119895, a new lncRNA which was identified from microarray and amplified in HCC. Functionally, AF119895 promoted migration and invasion of HCC cells both in vitro and in vivo. Furthermore, we identified that NXF3 was a downstream target of AF119895. NXF3 depletion could decrease HCC cells migration and invasion. In addition, AF119895 could act as an endogenous sponge by binding to miR-6508-3p and reduce miR-6508-3p expression. And miR-6508-3p could regulate NXF3 by interacting with its 3'UTR. These observations collectively demonstrate that AF119895 modulates the repression of NXF3 by binding to miR-6508-3p. Our results outline a novel signaling pathway mediated by AF119895 and suggest its candidacy as a new prognostic biomarker and therapeutic target of HCC.
Collapse
Affiliation(s)
- Tao Zeng
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China
| | - Dan Wang
- Department of Clinical Laboratory, The People's Hospital of Rongchang, Chongqing, China
| | - Juan Chen
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ke Chen
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Gangfeng Yu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Qiuxu Chen
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yuyang Liu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Shaoyin Yan
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Liying Zhu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Huihao Zhou
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ailong Huang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, China.
| | - Hua Tang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
8
|
Li MW, Sletten AC, Lee J, Pyles KD, Matkovich SJ, Ory DS, Schaffer JE. Nuclear export factor 3 regulates localization of small nucleolar RNAs. J Biol Chem 2017; 292:20228-20239. [PMID: 29021253 PMCID: PMC5724009 DOI: 10.1074/jbc.m117.818146] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 10/05/2017] [Indexed: 01/04/2023] Open
Abstract
Small nucleolar RNAs (snoRNAs) guide chemical modifications of ribosomal and small nuclear RNAs, functions that are carried out in the nucleus. Although most snoRNAs reside in the nucleolus, a growing body of evidence indicates that snoRNAs are also present in the cytoplasm and that snoRNAs move between the nucleus and cytoplasm by a mechanism that is regulated by lipotoxic and oxidative stress. Here, in a genome-wide shRNA-based screen, we identified nuclear export factor 3 (NXF3) as a transporter that alters the nucleocytoplasmic distribution of box C/D snoRNAs from the ribosomal protein L13a (Rpl13a) locus. Using RNA-sequencing analysis, we show that NXF3 associates not only with Rpl13a snoRNAs, but also with a broad range of box C/D and box H/ACA snoRNAs. Under homeostatic conditions, gain- or loss-of-function of NXF3, but not related family member NXF1, decreases or increases cytosolic Rpl13a snoRNAs, respectively. Furthermore, treatment with the adenylyl cyclase activator forskolin diminishes cytosolic localization of the Rpl13a snoRNAs through a mechanism that is dependent on NXF3 but not NXF1. Our results provide evidence of a new role for NXF3 in regulating the distribution of snoRNAs between the nuclear and cytoplasmic compartments.
Collapse
Affiliation(s)
- Melissa W Li
- Diabetes Research Center, Department of Medicine, St. Louis, Missouri 63110
| | - Arthur C Sletten
- Diabetes Research Center, Department of Medicine, St. Louis, Missouri 63110
| | - Jiyeon Lee
- Diabetes Research Center, Department of Medicine, St. Louis, Missouri 63110
| | - Kelly D Pyles
- Diabetes Research Center, Department of Medicine, St. Louis, Missouri 63110
| | - Scot J Matkovich
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Daniel S Ory
- Diabetes Research Center, Department of Medicine, St. Louis, Missouri 63110
| | - Jean E Schaffer
- Diabetes Research Center, Department of Medicine, St. Louis, Missouri 63110.
| |
Collapse
|
9
|
Mamon LA, Ginanova VR, Kliver SF, Yakimova AO, Atsapkina AA, Golubkova EV. RNA-binding proteins of the NXF (nuclear export factor) family and their connection with the cytoskeleton. Cytoskeleton (Hoboken) 2017; 74:161-169. [PMID: 28296067 DOI: 10.1002/cm.21362] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 03/04/2017] [Accepted: 03/08/2017] [Indexed: 02/05/2023]
Abstract
The mutual relationship between mRNA and the cytoskeleton can be seen from two points of view. On the one hand, the cytoskeleton is necessary for mRNA trafficking and anchoring to subcellular domains. On the other hand, cytoskeletal growth and rearrangement require the translation of mRNAs that are connected to the cytoskeleton. β-actin mRNA localization may influence dynamic changes in the actin cytoskeleton. In the cytoplasm, long-lived mRNAs exist in the form of RNP (ribonucleoprotein) complexes, where they interact with RNA-binding proteins, including NXF (Nuclear eXport Factor). Dm NXF1 is an evolutionarily conserved protein in Drosophila melanogaster that has orthologs in different animals. The universal function of nxf1 genes is the nuclear export of different mRNAs in various organisms. In this mini-review, we briefly discuss the evidence demonstrating that Dm NXF1 fulfils not only universal but also specialized cytoplasmic functions. This protein is detected not only in the nucleus but also in the cytoplasm. It is a component of neuronal granules. Dm NXF1 marks nuclear division spindles during early embryogenesis and the dense body on one side of the elongated spermatid nuclei. The characteristic features of sbr mutants (sbr10 and sbr5 ) are impairment of chromosome segregation and spindle formation anomalies during female meiosis. sbr12 mutant sterile males with immobile spermatozoa exhibit disturbances in the axoneme, mitochondrial derivatives and cytokinesis. These data allow us to propose that the Dm NXF1 proteins transport certain mRNAs in neurites and interact with localized mRNAs that are necessary for dynamic changes of the cytoskeleton.
Collapse
Affiliation(s)
- L A Mamon
- Department of Genetics and Biotechnology, Faculty of Biology, St Petersburg State University Universitetskaya nab. 7-9, Saint-Petersburg, 199034, Russia
| | - V R Ginanova
- Department of Genetics and Biotechnology, Faculty of Biology, St Petersburg State University Universitetskaya nab. 7-9, Saint-Petersburg, 199034, Russia
| | - S F Kliver
- Department of Genetics and Biotechnology, Faculty of Biology, St Petersburg State University Universitetskaya nab. 7-9, Saint-Petersburg, 199034, Russia
| | - A O Yakimova
- Department of Genetics and Biotechnology, Faculty of Biology, St Petersburg State University Universitetskaya nab. 7-9, Saint-Petersburg, 199034, Russia
| | - A A Atsapkina
- Department of Genetics and Biotechnology, Faculty of Biology, St Petersburg State University Universitetskaya nab. 7-9, Saint-Petersburg, 199034, Russia
| | - E V Golubkova
- Department of Genetics and Biotechnology, Faculty of Biology, St Petersburg State University Universitetskaya nab. 7-9, Saint-Petersburg, 199034, Russia
| |
Collapse
|
10
|
Li Y, Bor YC, Fitzgerald MP, Lee KS, Rekosh D, Hammarskjold ML. An NXF1 mRNA with a retained intron is expressed in hippocampal and neocortical neurons and is translated into a protein that functions as an Nxf1 cofactor. Mol Biol Cell 2016; 27:3903-3912. [PMID: 27708137 PMCID: PMC5170612 DOI: 10.1091/mbc.e16-07-0515] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/14/2016] [Accepted: 09/26/2016] [Indexed: 12/22/2022] Open
Abstract
A small Nxf1 protein, expressed from an NXF1 mRNA with a retained intron is highly expressed in rodent hippocampal and neocortical neurons, colocalizes with Staufen2 proteins in neuronal RNA granules, is present in polysomes, and replaces Nxt1 as an Nxf1 cofactor in export and expression of mRNA with retained introns. The Nxf1 protein is a major nuclear export receptor for the transport of mRNA, and it also is essential for export of retroviral mRNAs with retained introns. In the latter case, it binds to RNA elements known as constitutive transport elements (CTEs) and functions in conjunction with a cofactor known as Nxt1. The NXF1 gene also regulates expression of its own intron-containing RNA through the use of a functional CTE within intron 10. mRNA containing this intron is exported to the cytoplasm, where it can be translated into the 356–amino acid short Nxf1(sNxf1) protein, despite the fact that it is a prime candidate for nonsense-mediated decay (NMD). Here we demonstrate that sNxf1 is highly expressed in nuclei and dendrites of hippocampal and neocortical neurons in rodent brain. Additionally, we show that sNxf1 localizes in RNA granules in neurites of differentiated N2a mouse neuroblastoma cells, where it shows partial colocalization with Staufen2 isoform SS, a protein known to play a role in dendritic mRNA trafficking. We also show that sNxf1 forms heterodimers in conjunction with the full-length Nxf1 and that sNxf1 can replace Nxt1 to enhance the expression of CTE-containing mRNA and promote its association with polyribosomes.
Collapse
Affiliation(s)
- Ying Li
- Myles H. Thaler Center for AIDS and Human Retrovirus Research and Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Yeou-Cherng Bor
- Myles H. Thaler Center for AIDS and Human Retrovirus Research and Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Mark P Fitzgerald
- Departments of Neuroscience and Neurosurgery, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Kevin S Lee
- Departments of Neuroscience and Neurosurgery, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - David Rekosh
- Myles H. Thaler Center for AIDS and Human Retrovirus Research and Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Marie-Louise Hammarskjold
- Myles H. Thaler Center for AIDS and Human Retrovirus Research and Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908
| |
Collapse
|
11
|
Mutation of genes controlling mRNA metabolism and protein synthesis predisposes to neurodevelopmental disorders. Biochem Soc Trans 2016; 43:1259-65. [PMID: 26614670 DOI: 10.1042/bst20150168] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Brain development is a tightly controlled process that depends upon differentiation and function of neurons to allow for the formation of functional neural networks. Mutation of genes encoding structural proteins is well recognized as causal for neurodevelopmental disorders (NDDs). Recent studies have shown that aberrant gene expression can also lead to disorders of neural development. Here we summarize recent evidence implicating in the aetiology of NDDs mutation of factors acting at the level of mRNA splicing, mRNA nuclear export, translation and mRNA degradation. This highlights the importance of these fundamental processes for human health and affords new strategies and targets for therapeutic intervention.
Collapse
|
12
|
Yakimova AO, Pugacheva OM, Golubkova EV, Mamon LA. Cytoplasmic localization of SBR (Dm NXF1) protein and its zonal distribution in the ganglia of Drosophila melanogaster larvae. INVERTEBRATE NEUROSCIENCE 2016; 16:9. [DOI: 10.1007/s10158-016-0192-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 06/24/2016] [Indexed: 10/21/2022]
|
13
|
Callaerts-Vegh Z, Ahmed T, Vermaercke B, Marynen P, Balschun D, Froyen G, D'Hooge R. Nxf7 deficiency impairs social exploration and spatio-cognitive abilities as well as hippocampal synaptic plasticity in mice. Front Behav Neurosci 2015. [PMID: 26217206 PMCID: PMC4498129 DOI: 10.3389/fnbeh.2015.00179] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Nuclear RNA export factors (NXF) are conserved in all metazoans and are deemed essential for shuttling RNA across the nuclear envelope and other post-transcriptional processes (such as mRNA metabolism, storage and stability). Disruption of human NXF5 has been implicated in intellectual and psychosocial disabilities. In the present report, we use recently described Nxf7 knockout (KO) mice as an experimental model to analyze in detail the behavioral consequences of clinical NXF5 deficiency. We examined male Nxf7 KO mice using an extended cognitive and behavioral test battery, and recorded extracellular field potentials in the hippocampal CA1 region. We observed various cognitive and behavioral changes including alterations in social exploration, impaired spatial learning and spatio-cognitive abilities. We also defined a new experimental paradigm to discriminate search strategies in Morris water maze and showed significant differences between Nxf7 KO and control animals. Furthermore, while we observed no difference in a nose poke suppression in an conditioned emotional response (CER) protocol, Nxf7 KO mice were impaired in discriminating between differentially reinforced cues in an auditory fear conditioning protocol. This distinct neurocognitive phenotype was accompanied by impaired hippocampal Long-term potentiation (LTP), while long-term depression (LTD) was not affected by Nxf7 deficiency. Our data demonstrate that disruption of murine Nxf7 leads to behavioral phenotypes that may relate to the intellectual and social deficits in patients with NXF5 deficiency.
Collapse
Affiliation(s)
| | - Tariq Ahmed
- Laboratory of Biological Psychology, University of Leuven, KU Leuven Leuven, Belgium
| | - Ben Vermaercke
- Laboratory of Biological Psychology, University of Leuven, KU Leuven Leuven, Belgium
| | - Peter Marynen
- Human Genome Laboratory, University of Leuven and VIB Center for the Biology of Disease Leuven, Belgium
| | - Detlef Balschun
- Laboratory of Biological Psychology, University of Leuven, KU Leuven Leuven, Belgium
| | - Guy Froyen
- Human Genome Laboratory, University of Leuven and VIB Center for the Biology of Disease Leuven, Belgium
| | - Rudi D'Hooge
- Laboratory of Biological Psychology, University of Leuven, KU Leuven Leuven, Belgium
| |
Collapse
|
14
|
Nuclear export of messenger RNA. Genes (Basel) 2015; 6:163-84. [PMID: 25836925 PMCID: PMC4488659 DOI: 10.3390/genes6020163] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/23/2015] [Accepted: 03/24/2015] [Indexed: 11/29/2022] Open
Abstract
Transport of messenger RNA (mRNA) from the nucleus to the cytoplasm is an essential step of eukaryotic gene expression. In the cell nucleus, a precursor mRNA undergoes a series of processing steps, including capping at the 5' ends, splicing and cleavage/polyadenylation at the 3' ends. During this process, the mRNA associates with a wide variety of proteins, forming a messenger ribonucleoprotein (mRNP) particle. Association with factors involved in nuclear export also occurs during transcription and processing, and thus nuclear export is fully integrated into mRNA maturation. The coupling between mRNA maturation and nuclear export is an important mechanism for providing only fully functional and competent mRNA to the cytoplasmic translational machinery, thereby ensuring accuracy and swiftness of gene expression. This review describes the molecular mechanism of nuclear mRNA export mediated by the principal transport factors, including Tap-p15 and the TREX complex.
Collapse
|
15
|
Wünsch D, Hahlbrock A, Heiselmayer C, Bäcker S, Heun P, Goesswein D, Stöcker W, Schirmeister T, Schneider G, Krämer OH, Knauer SK, Stauber RH. Fly versus man: evolutionary impairment of nucleolar targeting affects the degradome of Drosophila's Taspase1. FASEB J 2015; 29:1973-85. [PMID: 25634959 DOI: 10.1096/fj.14-262451] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 12/29/2014] [Indexed: 12/12/2022]
Abstract
Human Taspase1 is essential for development and cancer by processing critical regulators, such as the mixed-lineage leukemia protein. Likewise, its ortholog, trithorax, is cleaved by Drosophila Taspase1 (dTaspase1), implementing a functional coevolution. To uncover novel mechanism regulating protease function, we performed a functional analysis of dTaspase1 and its comparison to the human ortholog. dTaspase1 contains an essential nucleophile threonine(195), catalyzing cis cleavage into its α- and β-subunits. A cell-based assay combined with alanine scanning mutagenesis demonstrated that the target cleavage motif for dTaspase1 (Q(3)[F/I/L/M](2)D(1)↓G(1')X(2')X(3')) differs significantly from the human ortholog (Q(3)[F,I,L,V](2)D(1)↓G(1')x(2')D(3')D(4')), predicting an enlarged degradome containing 70 substrates for Drosophila. In contrast to human Taspase1, dTaspase1 shows no discrete localization to the nucleus/nucleolus due to the lack of the importin-α/nucleophosmin1 interaction domain (NoLS) conserved in all vertebrates. Consequently, dTaspase1 interacts with neither the Drosophila nucleoplasmin-like protein nor human nucleophosmin1. The impact of localization on the protease's degradome was confirmed by demonstrating that dTaspase1 did not efficiently process nuclear substrates, such as upstream stimulatory factor 2. However, genetic introduction of the NoLS into dTaspase1 restored its nucleolar localization, nucleophosmin1 interaction, and efficient cleavage of nuclear substrates. We report that evolutionary functional divergence separating vertebrates from invertebrates can be achieved for proteases by a transport/localization-regulated mechanism.
Collapse
Affiliation(s)
- Désirée Wünsch
- *Molecular and Cellular Oncology, Ear, Nose and Throat Department, University Medical Center of Mainz, Mainz, Germany; Institute for Molecular Biology, Centre for Medical Biotechnology, Mainz Scientific Screening Center UG & Co. KG, University of Duisburg-Essen, Essen, Germany; Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany; Cell and Matrix Biology, Institute for Zoology, and Institute for Pharmacy and Biochemistry, University of Mainz, Mainz, Germany; II. Medizinische Klinik, Technische Universität München, Munich, Germany; and Department of Toxicology, University Medical Center, Mainz, Germany
| | - Angelina Hahlbrock
- *Molecular and Cellular Oncology, Ear, Nose and Throat Department, University Medical Center of Mainz, Mainz, Germany; Institute for Molecular Biology, Centre for Medical Biotechnology, Mainz Scientific Screening Center UG & Co. KG, University of Duisburg-Essen, Essen, Germany; Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany; Cell and Matrix Biology, Institute for Zoology, and Institute for Pharmacy and Biochemistry, University of Mainz, Mainz, Germany; II. Medizinische Klinik, Technische Universität München, Munich, Germany; and Department of Toxicology, University Medical Center, Mainz, Germany
| | - Christina Heiselmayer
- *Molecular and Cellular Oncology, Ear, Nose and Throat Department, University Medical Center of Mainz, Mainz, Germany; Institute for Molecular Biology, Centre for Medical Biotechnology, Mainz Scientific Screening Center UG & Co. KG, University of Duisburg-Essen, Essen, Germany; Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany; Cell and Matrix Biology, Institute for Zoology, and Institute for Pharmacy and Biochemistry, University of Mainz, Mainz, Germany; II. Medizinische Klinik, Technische Universität München, Munich, Germany; and Department of Toxicology, University Medical Center, Mainz, Germany
| | - Sandra Bäcker
- *Molecular and Cellular Oncology, Ear, Nose and Throat Department, University Medical Center of Mainz, Mainz, Germany; Institute for Molecular Biology, Centre for Medical Biotechnology, Mainz Scientific Screening Center UG & Co. KG, University of Duisburg-Essen, Essen, Germany; Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany; Cell and Matrix Biology, Institute for Zoology, and Institute for Pharmacy and Biochemistry, University of Mainz, Mainz, Germany; II. Medizinische Klinik, Technische Universität München, Munich, Germany; and Department of Toxicology, University Medical Center, Mainz, Germany
| | - Patrick Heun
- *Molecular and Cellular Oncology, Ear, Nose and Throat Department, University Medical Center of Mainz, Mainz, Germany; Institute for Molecular Biology, Centre for Medical Biotechnology, Mainz Scientific Screening Center UG & Co. KG, University of Duisburg-Essen, Essen, Germany; Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany; Cell and Matrix Biology, Institute for Zoology, and Institute for Pharmacy and Biochemistry, University of Mainz, Mainz, Germany; II. Medizinische Klinik, Technische Universität München, Munich, Germany; and Department of Toxicology, University Medical Center, Mainz, Germany
| | - Dorothee Goesswein
- *Molecular and Cellular Oncology, Ear, Nose and Throat Department, University Medical Center of Mainz, Mainz, Germany; Institute for Molecular Biology, Centre for Medical Biotechnology, Mainz Scientific Screening Center UG & Co. KG, University of Duisburg-Essen, Essen, Germany; Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany; Cell and Matrix Biology, Institute for Zoology, and Institute for Pharmacy and Biochemistry, University of Mainz, Mainz, Germany; II. Medizinische Klinik, Technische Universität München, Munich, Germany; and Department of Toxicology, University Medical Center, Mainz, Germany
| | - Walter Stöcker
- *Molecular and Cellular Oncology, Ear, Nose and Throat Department, University Medical Center of Mainz, Mainz, Germany; Institute for Molecular Biology, Centre for Medical Biotechnology, Mainz Scientific Screening Center UG & Co. KG, University of Duisburg-Essen, Essen, Germany; Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany; Cell and Matrix Biology, Institute for Zoology, and Institute for Pharmacy and Biochemistry, University of Mainz, Mainz, Germany; II. Medizinische Klinik, Technische Universität München, Munich, Germany; and Department of Toxicology, University Medical Center, Mainz, Germany
| | - Tanja Schirmeister
- *Molecular and Cellular Oncology, Ear, Nose and Throat Department, University Medical Center of Mainz, Mainz, Germany; Institute for Molecular Biology, Centre for Medical Biotechnology, Mainz Scientific Screening Center UG & Co. KG, University of Duisburg-Essen, Essen, Germany; Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany; Cell and Matrix Biology, Institute for Zoology, and Institute for Pharmacy and Biochemistry, University of Mainz, Mainz, Germany; II. Medizinische Klinik, Technische Universität München, Munich, Germany; and Department of Toxicology, University Medical Center, Mainz, Germany
| | - Günter Schneider
- *Molecular and Cellular Oncology, Ear, Nose and Throat Department, University Medical Center of Mainz, Mainz, Germany; Institute for Molecular Biology, Centre for Medical Biotechnology, Mainz Scientific Screening Center UG & Co. KG, University of Duisburg-Essen, Essen, Germany; Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany; Cell and Matrix Biology, Institute for Zoology, and Institute for Pharmacy and Biochemistry, University of Mainz, Mainz, Germany; II. Medizinische Klinik, Technische Universität München, Munich, Germany; and Department of Toxicology, University Medical Center, Mainz, Germany
| | - Oliver H Krämer
- *Molecular and Cellular Oncology, Ear, Nose and Throat Department, University Medical Center of Mainz, Mainz, Germany; Institute for Molecular Biology, Centre for Medical Biotechnology, Mainz Scientific Screening Center UG & Co. KG, University of Duisburg-Essen, Essen, Germany; Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany; Cell and Matrix Biology, Institute for Zoology, and Institute for Pharmacy and Biochemistry, University of Mainz, Mainz, Germany; II. Medizinische Klinik, Technische Universität München, Munich, Germany; and Department of Toxicology, University Medical Center, Mainz, Germany
| | - Shirley K Knauer
- *Molecular and Cellular Oncology, Ear, Nose and Throat Department, University Medical Center of Mainz, Mainz, Germany; Institute for Molecular Biology, Centre for Medical Biotechnology, Mainz Scientific Screening Center UG & Co. KG, University of Duisburg-Essen, Essen, Germany; Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany; Cell and Matrix Biology, Institute for Zoology, and Institute for Pharmacy and Biochemistry, University of Mainz, Mainz, Germany; II. Medizinische Klinik, Technische Universität München, Munich, Germany; and Department of Toxicology, University Medical Center, Mainz, Germany
| | - Roland H Stauber
- *Molecular and Cellular Oncology, Ear, Nose and Throat Department, University Medical Center of Mainz, Mainz, Germany; Institute for Molecular Biology, Centre for Medical Biotechnology, Mainz Scientific Screening Center UG & Co. KG, University of Duisburg-Essen, Essen, Germany; Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany; Cell and Matrix Biology, Institute for Zoology, and Institute for Pharmacy and Biochemistry, University of Mainz, Mainz, Germany; II. Medizinische Klinik, Technische Universität München, Munich, Germany; and Department of Toxicology, University Medical Center, Mainz, Germany
| |
Collapse
|
16
|
Truant R, Raymond LA, Xia J, Pinchev D, Burtnik A, Atwal RS. Canadian Association of Neurosciences Review: Polyglutamine Expansion Neurodegenerative Diseases. Can J Neurol Sci 2014; 33:278-91. [PMID: 17001815 DOI: 10.1017/s031716710000514x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
ABSTRACT:Since the early 1990s, DNA triplet repeat expansions have been found to be the cause in an ever increasing number of genetic neurologic diseases. A subset of this large family of genetic diseases has the expansion of a CAG DNA triplet in the open reading frame of a coding exon. The result of this DNA expansion is the expression of expanded glutamine amino acid repeat tracts in the affected proteins, leading to the term, Polyglutamine Diseases, which is applied to this sub-family of diseases. To date, nine distinct genes are known to be linked to polyglutamine diseases, including Huntington's disease, Machado-Joseph Disease and spinobulbar muscular atrophy or Kennedy's disease. Most of the polyglutamine diseases are characterized clinically as spinocerebellar ataxias. Here we discuss recent successes and advancements in polyglutamine disease research, comparing these different diseases with a common genetic flaw at the level of molecular biology and early drug design for a family of diseases where many new research tools for these genetic disorders have been developed. Polyglutamine disease research has successfully used interdisciplinary collaborative efforts, informative multiple mouse genetic models and advanced tools of pharmaceutical industry research to potentially serve as the prototype model of therapeutic research and development for rare neurodegenerative diseases.
Collapse
Affiliation(s)
- Ray Truant
- Department of Biochemistry and Biomedical Sciences McMaster University, Hamilton, ON, Canada
| | | | | | | | | | | |
Collapse
|
17
|
An emerging phenotype of Xq22 microdeletions in females with severe intellectual disability, hypotonia and behavioral abnormalities. J Hum Genet 2014; 59:300-6. [DOI: 10.1038/jhg.2014.21] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 02/21/2014] [Accepted: 02/25/2014] [Indexed: 12/28/2022]
|
18
|
Zhou J, Goldberg EM, Leu NA, Zhou L, Coulter DA, Wang PJ. Respiratory failure, cleft palate and epilepsy in the mouse model of human Xq22.1 deletion syndrome. Hum Mol Genet 2014; 23:3823-9. [PMID: 24569167 DOI: 10.1093/hmg/ddu095] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Chromosomal segmental deletion is a frequent cause of human diseases. A familial 1.1 Mb deletion of human chromosome Xq22.1 associates with epilepsy, cleft palate and developmental defects in heterozygous female patients. Here, we describe a mouse mutant with a targeted deletion of the syntenic segment of the mouse X chromosome that phenocopies the human syndrome. Male mice with a deletion of a 1.1 Mb Nxf2-Nxf3 X-chromosomal segment exhibit respiratory failure, neonatal lethality and cleft palate. In female mice, heterozygosity for the deletion manifests cleft palate, early postnatal lethality, postnatal growth delay and spontaneous seizures in surviving animals, apparently due to X-chromosome inactivation. Furthermore, loss of a 0.35 Mb subregion containing Armcx5, Gprasp1, Gprasp2 and Bhlhb9 is sufficient to cause the Xq22.1 syndrome phenotype. Our results support that the 1.1 Mb deletion of human Xq22.1 is the genetic cause of the associated syndrome.
Collapse
Affiliation(s)
- Jian Zhou
- Department of Animal Biology, University of Pennsylvania School of Veterinary Medicine, 3800 Spruce Street, Philadelphia, PA 19104, USA and
| | - Ethan M Goldberg
- The Children's Hospital of Philadelphia, Division of Neurology, Colket Translational Research Building, 3501 Civic Center Boulevard, Philadelphia, PA 19104-4399, USA
| | - N Adrian Leu
- Department of Animal Biology, University of Pennsylvania School of Veterinary Medicine, 3800 Spruce Street, Philadelphia, PA 19104, USA and
| | - Lei Zhou
- The Children's Hospital of Philadelphia, Division of Neurology, Colket Translational Research Building, 3501 Civic Center Boulevard, Philadelphia, PA 19104-4399, USA
| | - Douglas A Coulter
- The Children's Hospital of Philadelphia, Division of Neurology, Colket Translational Research Building, 3501 Civic Center Boulevard, Philadelphia, PA 19104-4399, USA
| | - P Jeremy Wang
- Department of Animal Biology, University of Pennsylvania School of Veterinary Medicine, 3800 Spruce Street, Philadelphia, PA 19104, USA and
| |
Collapse
|
19
|
Piton A, Redin C, Mandel JL. XLID-causing mutations and associated genes challenged in light of data from large-scale human exome sequencing. Am J Hum Genet 2013; 93:368-83. [PMID: 23871722 DOI: 10.1016/j.ajhg.2013.06.013] [Citation(s) in RCA: 199] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 05/29/2013] [Accepted: 06/08/2013] [Indexed: 12/30/2022] Open
Abstract
Because of the unbalanced sex ratio (1.3-1.4 to 1) observed in intellectual disability (ID) and the identification of large ID-affected families showing X-linked segregation, much attention has been focused on the genetics of X-linked ID (XLID). Mutations causing monogenic XLID have now been reported in over 100 genes, most of which are commonly included in XLID diagnostic gene panels. Nonetheless, the boundary between true mutations and rare non-disease-causing variants often remains elusive. The sequencing of a large number of control X chromosomes, required for avoiding false-positive results, was not systematically possible in the past. Such information is now available thanks to large-scale sequencing projects such as the National Heart, Lung, and Blood (NHLBI) Exome Sequencing Project, which provides variation information on 10,563 X chromosomes from the general population. We used this NHLBI cohort to systematically reassess the implication of 106 genes proposed to be involved in monogenic forms of XLID. We particularly question the implication in XLID of ten of them (AGTR2, MAGT1, ZNF674, SRPX2, ATP6AP2, ARHGEF6, NXF5, ZCCHC12, ZNF41, and ZNF81), in which truncating variants or previously published mutations are observed at a relatively high frequency within this cohort. We also highlight 15 other genes (CCDC22, CLIC2, CNKSR2, FRMPD4, HCFC1, IGBP1, KIAA2022, KLF8, MAOA, NAA10, NLGN3, RPL10, SHROOM4, ZDHHC15, and ZNF261) for which replication studies are warranted. We propose that similar reassessment of reported mutations (and genes) with the use of data from large-scale human exome sequencing would be relevant for a wide range of other genetic diseases.
Collapse
Affiliation(s)
- Amélie Piton
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique Unité Mixte de Recherche 7104, Institut National de la Santé et de la Recherche Médicale Unité 964, University of Strasbourg, 67404 Illkirch Cedex, France; Chaire de Génétique Humaine, Collège de France, 75231 Paris Cedex 05, France.
| | | | | |
Collapse
|
20
|
Esposito T, Lea RA, Maher BH, Moses D, Cox HC, Magliocca S, Angius A, Nyholt DR, Titus T, Kay T, Gray NA, Rastaldi MP, Parnham A, Gianfrancesco F, Griffiths LR. Unique X-linked familial FSGS with co-segregating heart block disorder is associated with a mutation in the NXF5 gene. Hum Mol Genet 2013; 22:3654-66. [PMID: 23686279 DOI: 10.1093/hmg/ddt215] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) is the consequence of a disease process that attacks the kidney's filtering system, causing serious scarring. More than half of FSGS patients develop chronic kidney failure within 10 years, ultimately requiring dialysis or renal transplantation. There are currently several genes known to cause the hereditary forms of FSGS (ACTN4, TRPC6, CD2AP, INF2, MYO1E and NPHS2). This study involves a large, unique, multigenerational Australian pedigree in which FSGS co-segregates with progressive heart block with apparent X-linked recessive inheritance. Through a classical combined approach of linkage and haplotype analysis, we identified a 21.19 cM interval implicated on the X chromosome. We then used a whole exome sequencing approach to identify two mutated genes, NXF5 and ALG13, which are located within this linkage interval. The two mutations NXF5-R113W and ALG13-T141L segregated perfectly with the disease phenotype in the pedigree and were not found in a large healthy control cohort. Analysis using bioinformatics tools predicted the R113W mutation in the NXF5 gene to be deleterious and cellular studies support a role in the stability and localization of the protein suggesting a causative role of this mutation in these co-morbid disorders. Further studies are now required to determine the functional consequence of these novel mutations to development of FSGS and heart block in this pedigree and to determine whether these mutations have implications for more common forms of these diseases in the general population.
Collapse
Affiliation(s)
- Teresa Esposito
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso, National Research Council of Italy, Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Vanmarsenille L, Verbeeck J, Belet S, Roebroek AJ, Van de Putte T, Nevelsteen J, Callaerts-Vegh Z, D’Hooge R, Marynen P, Froyen G. Generation and characterization of an Nxf7 knockout mouse to study NXF5 deficiency in a patient with intellectual disability. PLoS One 2013; 8:e64144. [PMID: 23675524 PMCID: PMC3652825 DOI: 10.1371/journal.pone.0064144] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 04/09/2013] [Indexed: 12/11/2022] Open
Abstract
Members of the Nuclear eXport Factor (NXF) family are involved in the export of mRNA from the nucleus to the cytoplasm, or hypothesized to play a role in transport of cytoplasmic mRNA. We previously reported on the loss of NXF5 in a male patient with a syndromic form of intellectual disability. To study the functional role of NXF5 we identified the mouse counterpart. Based on synteny, mouse Nxf2 is the ortholog of human NXF5. However, we provide several lines of evidence that mouse Nxf7 is the actual functional equivalent of NXF5. Both Nxf7 and NXF5 are predominantly expressed in the brain, show cytoplasmic localization, and present as granules in neuronal dendrites suggesting a role in cytoplasmic mRNA metabolism in neurons. Nxf7 was primarily detected in the pyramidal cells of the hippocampus and in layer V of the cortex. Similar to human NXF2, mouse Nxf2 is highly expressed in testis and shows a nuclear localization. Interestingly, these findings point to a different evolutionary path for both NXF genes in human and mouse. We thus generated and validated Nxf7 knockout mice, which were fertile and did not present any gross anatomical or morphological abnormalities. Expression profiling in the hippocampus and the cortex did not reveal significant changes between wild-type and Nxf7 knockout mice. However, impaired spatial memory was observed in these KO mice when evaluated in the Morris water maze test. In conclusion, our findings provide strong evidence that mouse Nxf7 is the functional counterpart of human NXF5, which might play a critical role in mRNA metabolism in the brain.
Collapse
Affiliation(s)
- Lieselot Vanmarsenille
- Human Genome Laboratory, VIB Center for the Biology of Disease, Leuven, Belgium
- Human Genome Laboratory, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Jelle Verbeeck
- Human Genome Laboratory, VIB Center for the Biology of Disease, Leuven, Belgium
- Human Genome Laboratory, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Stefanie Belet
- Human Genome Laboratory, VIB Center for the Biology of Disease, Leuven, Belgium
- Human Genome Laboratory, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Anton J. Roebroek
- Experimental Mouse Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Tom Van de Putte
- Laboratory of Molecular Biology (Celgen), Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Joke Nevelsteen
- Human Genome Laboratory, VIB Center for the Biology of Disease, Leuven, Belgium
- Human Genome Laboratory, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | | | - Rudi D’Hooge
- Laboratory of Biological Psychology, KU Leuven, Leuven, Belgium
- Leuven Institute for Neuroscience and Disease (LIND), KU Leuven, Leuven, Belgium
| | - Peter Marynen
- Human Genome Laboratory, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Guy Froyen
- Human Genome Laboratory, VIB Center for the Biology of Disease, Leuven, Belgium
- Human Genome Laboratory, Department of Human Genetics, KU Leuven, Leuven, Belgium
- * E-mail:
| |
Collapse
|
22
|
Mamon LA, Kliver SF, Golubkova EV. Evolutionarily conserved features of the retained intron in alternative transcripts of the <i>nxf1</i> (nuclear export factor) genes in different organisms. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/ojgen.2013.33018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
23
|
Piton A, Jouan L, Rochefort D, Dobrzeniecka S, Lachapelle K, Dion PA, Gauthier J, Rouleau GA. Analysis of the effects of rare variants on splicing identifies alterations in GABAA receptor genes in autism spectrum disorder individuals. Eur J Hum Genet 2012; 21:749-56. [PMID: 23169495 DOI: 10.1038/ejhg.2012.243] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
A large-scale sequencing screen of X-linked synaptic genes in individuals with autism spectrum disorder (ASD) or schizophrenia (SCZ), two common neurodevelopmental disorders, identified many variants most of which have no easily predictable effect on gene function. In this report, we evaluated the impact of these rare missense and silent variants on gene splicing. For this purpose, we used complementary in silico analyses, in vitro minigene-based assays and RNA prepared from lymphoblastoid cells derived from patients with these mutations. Our goal was to identify the variants which might either create or disrupt an acceptor splice site, a donor splice site or an exonic splicing enhancer, thus leading to aberrant splicing that could be involved in the pathogenesis of ASD or SCZ. We identified truncating mutations in distinct X-linked gamma-aminobutyric acid A (GABAA) receptor subunit-encoding genes, GABRQ and GABRA3, in two different families. Furthermore, missense and silent variants in nuclear RNA export factor 5 and histone deacetylase 6 were shown to partially disrupt the protein. While genes from the GABAergic pathway have previously been thought to be involved in the pathophysiology of ASD, this is the first report of ASD patients with truncating mutations in GABA receptors genes.
Collapse
Affiliation(s)
- Amélie Piton
- Department of Medicine, Centre of Excellence in Neuroscience of Université de Montréal, CRCHUM Notre-Dame Hospital, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Chen CP, Su YN, Lin HH, Chern SR, Tsai FJ, Wu PC, Lee CC, Chen YT, Wang W. De novo duplication of Xq22.1→q24 with a disruption of the NXF gene cluster in a mentally retarded woman with short stature and premature ovarian failure. Taiwan J Obstet Gynecol 2012; 50:339-44. [PMID: 22030050 DOI: 10.1016/j.tjog.2011.01.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2010] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE To present molecular cytogenetic characterization of a de novo duplication of Xq22.1→q24 in a mentally retarded woman with short stature and premature ovarian failure. MATERIALS AND METHODS A 19-year-old woman presented with psychomotor retardation, developmental delay, mental retardation, short stature, low body weight, general muscle hypotonia, distal muscle hypotrophy of the lower extremities, elongated digits, scanty pubic and axillary hair, hypoplastic external female genitalia, and secondary amenorrhea but no clinical features of Pelizaeus-Merzbacher disease. Conventional cytogenetic analysis revealed a karyotype of 46,X,dup(X)(q22.1q24). Fluorescence in situ hybridization determined a direct duplication with a linear tandem orientation. Array comparative genomic hybridization demonstrated partial trisomy Xq [arr cgh Xq22.1q24 (101,490,234-119,070,188 bp)×3] with a 17.6-Mb duplication. RESULTS The duplicated region contained NXF2B, NXF4, NXF3, PLP1, and PGRMC1 genes. There was a disruption of the NXF gene cluster of Xcen-NXF5-NXF2-NXF2B-NXF4-NXF3-Xqter. CONCLUSION A duplication of Xq22.1→q24 with a disruption of the NXF gene cluster in female patients can be associated with clinical manifestations of mental retardation in addition to short stature and premature ovarian failure.
Collapse
Affiliation(s)
- Chih-Ping Chen
- Department of Obstetrics and Gynecology, Mackay Memorial Hospital, Taipei, Taiwan.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Bier C, Knauer SK, Klapthor A, Schweitzer A, Rekik A, Krämer OH, Marschalek R, Stauber RH. Cell-based analysis of structure-function activity of threonine aspartase 1. J Biol Chem 2011; 286:3007-17. [PMID: 21084304 PMCID: PMC3024795 DOI: 10.1074/jbc.m110.161646] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 10/22/2010] [Indexed: 12/24/2022] Open
Abstract
Taspase1 is a threonine protease responsible for cleaving intracellular substrates. As such, (de)regulated Taspase1 function is expected not only to be vital for ordered development but may also be relevant for disease. However, the full repertoires of Taspase1 targets as well as the exact biochemical requirements for its efficient and substrate-specific cleavage are not yet resolved. Also, no cellular assays for this protease are currently available, hampering the exploitation of the (patho)biological relevance of Taspase1. Here, we developed highly efficient cell-based translocation biosensor assays to probe Taspase1 trans-cleavage in vivo. These modular sensors harbor variations of Taspase1 cleavage sites and localize to the cytoplasm. Expression of Taspase1 but not of inactive Taspase1 mutants or of unrelated proteases triggers proteolytic cleavage and nuclear accumulation of the biosensors. Employing our assay combined with scanning mutagenesis, we identified the sequence and spatial requirements for efficient Taspase1 processing in liquid and solid tumor cell lines. Collectively, our results defined an improved Taspase1 consensus recognition sequence, Q(3)(F/I/L/V)(2)D(1)↓G(1)'X(2)'D(3)'D(4)', allowing the first genome-wide bioinformatic identification of the human Taspase1 degradome. Among the 27 most likely Taspase1 targets are cytoplasmic but also nuclear proteins, such as the upstream stimulatory factor 2 (USF2) or the nuclear RNA export factors 2/5 (NXF2/5). Cleavage site recognition and proteolytic processing of selected targets were verified in the context of the biosensor and for the full-length proteins. We provide novel mechanistic insights into the function and bona fide targets of Taspase1 allowing for a focused investigation of the (patho)biological relevance of this type 2 asparaginase.
Collapse
Affiliation(s)
- Carolin Bier
- From the Molecular and Cellular Oncology/Mainzer Screening Center, University Hospital of Mainz, Langenbeckstrasse 1, 55101 Mainz
| | - Shirley K. Knauer
- the Institute for Molecular Biology, Centre for Medical Biotechnology, University Duisburg-Essen, Universitätsstrasse, 45117 Essen
| | - Alexander Klapthor
- From the Molecular and Cellular Oncology/Mainzer Screening Center, University Hospital of Mainz, Langenbeckstrasse 1, 55101 Mainz
| | - Andrea Schweitzer
- From the Molecular and Cellular Oncology/Mainzer Screening Center, University Hospital of Mainz, Langenbeckstrasse 1, 55101 Mainz
| | - Alexander Rekik
- From the Molecular and Cellular Oncology/Mainzer Screening Center, University Hospital of Mainz, Langenbeckstrasse 1, 55101 Mainz
| | - Oliver H. Krämer
- the Institute for Biochemistry and Biophysics/Centre for Molecular Biomedicine, Friedrich-Schiller-University Jena, Hans-Knöll-Strasse 2, 07745 Jena, and
| | - Rolf Marschalek
- the Institute for Pharmaceutical Biology/ZAFES, Goethe-University, Max-von Laue-Strasse 9, 60438 Frankfurt/Main, Germany
| | - Roland H. Stauber
- From the Molecular and Cellular Oncology/Mainzer Screening Center, University Hospital of Mainz, Langenbeckstrasse 1, 55101 Mainz
| |
Collapse
|
26
|
Atsapkina AA, Golubkova EV, Kasatkina VV, Avanesyan EO, Ivankova NA, Mamon LA. Peculiarities of spermatogenesis in Drosophila melanogaster: Role of main transport receptor of mRNA (Dm NXF1). ACTA ACUST UNITED AC 2010. [DOI: 10.1134/s1990519x10050044] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
27
|
Anthony K, Gallo JM. Aberrant RNA processing events in neurological disorders. Brain Res 2010; 1338:67-77. [DOI: 10.1016/j.brainres.2010.03.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Revised: 02/24/2010] [Accepted: 03/03/2010] [Indexed: 12/12/2022]
|
28
|
Ivankova N, Tretyakova I, Lyozin GT, Avanesyan E, Zolotukhin A, Zatsepina OG, Evgen'ev MB, Mamon LA. Alternative transcripts expressed by small bristles, the Drosophila melanogaster nxf1 gene. Gene 2010; 458:11-9. [DOI: 10.1016/j.gene.2010.02.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2007] [Revised: 02/16/2010] [Accepted: 02/25/2010] [Indexed: 11/30/2022]
|
29
|
Molecular cytogenetic definition of a translocation t(X;15) associated with premature ovarian failure. Fertil Steril 2010; 94:1097.e5-8. [PMID: 20338563 DOI: 10.1016/j.fertnstert.2010.02.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Revised: 02/02/2010] [Accepted: 02/04/2010] [Indexed: 10/19/2022]
Abstract
OBJECTIVE To characterize the breakpoints of a t(X;15) found in a woman with premature ovarian failure (POF). DESIGN Case report. SETTING Molecular and cytogenetics unit in a university-affiliated hospital. PATIENT(S) A 19-year-old infertile woman presenting with a normal female phenotype but primary amenorrhea. INTERVENTION(S) Molecular cytogenetic analyses and genetic counseling. MAIN OUTCOME MEASURE(S) Translocation t(X;15) defined by fluorescence in situ hybridization (FISH) and array comparative genomic hybridization (array CGH). RESULT(S) Chromosome and FISH analysis revealed 46,XX, t(X;15)(Xq22.1;p11); the active X was translocated and had been inherited from her mother. Detailed molecular characterization by FISH showed that the NXF5 (nuclear RNA export factor 5) gene was contained in the clone spanning the breakpoint on the X chromosome. CONCLUSION(S) The NXF5 gene is an appealing candidate for POF because it shows functional homology with the FMR1 (fragile X mental retardation 1) gene. Further analyses of its expression as well as mutation screening in other POF patients will help to elucidate its role.
Collapse
|
30
|
Familial 1.1 Mb deletion in chromosome Xq22.1 associated with mental retardation and behavioural disorders in female patients. Eur J Med Genet 2010; 53:113-6. [PMID: 20096387 DOI: 10.1016/j.ejmg.2010.01.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2009] [Accepted: 01/09/2010] [Indexed: 11/21/2022]
Abstract
We report on a 7-year-old girl with severe mental retardation (MR), autism, micro-brachycephaly, generalized muscle hypotonia with distal hypotrophy of lower limbs, scoliosis and facial dysmorphisms. Array-CGH analysis identified a 1.1 Mb deletion of chromosome Xq22.1. Further analysis demonstrated that the deletion was inherited from her mother who showed mild MR, short stature, brachycephaly, epilepsy and a Borderline Personality Disorder. Microsatellite segregation analysis revealed that the rearrangement arose de novo in the mother on the paternal X chromosome. The deleted Xq22.1 region contains part of the NXF gene cluster which is involved in mRNA nuclear export and metabolism. Among them, the NXF5 gene has already been linked to mental retardation whereas NXF2 protein has been recently found to be partner of FMRP in regulating Nxf1 mRNA stability in neuronal cells. The dosage imbalance of NXF5 and NXF2 genes may explain the severe phenotype in our patient.
Collapse
|
31
|
Ding D, Dave KR, Bhattacharya SK. On Message Ribonucleic Acids Targeting to Mitochondria. BIOCHEMISTRY INSIGHTS 2009. [DOI: 10.4137/bci.s3745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Mitochondria are subcellular organelles that provide energy for a variety of basic cellular processes in eukaryotic cells. Mitochondria maintain their own genomes and many of their endosymbiont genes are encoded by nuclear genomes. The crosstalk between the mitochondrial and nuclear genomes ensures mitochondrial biogenesis, dynamics and maintenance. Mitochondrial proteins are partly encoded by nucleus and synthesized in the cytosol and partly in the mitochondria coded by mitochondrial genome. The efficiency of transport systems that transport nuclear encoded gene products such as proteins and mRNAs to the mitochondrial vicinity to allow for their translation and/or import are recently receiving wide attention. There is currently no concrete evidence that nuclear encoded mRNA is transported into the mitochondria, however, they can be transported onto the mitochondrial surface and translated at the surface of mitochondria utilizing cytosolic machinery. In this review we present an overview of the recent advances in the mRNA transport, with emphasis on the transport of nuclear-encoded mitochondrial protein mRNA into the mitochondria.
Collapse
Affiliation(s)
- Di Ding
- Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL, USA
- Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA
| | - Kunjan R. Dave
- Department of Neurology, University of Miami, Miami, FL, USA
| | - Sanjoy K. Bhattacharya
- Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL, USA
- Department of Neurology, University of Miami, Miami, FL, USA
| |
Collapse
|
32
|
Dm nxf1/sbr gene affects the formation of meiotic spindle in female Drosophila melanogaster. Chromosome Res 2009; 17:833-45. [PMID: 19779841 DOI: 10.1007/s10577-009-9046-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Revised: 06/08/2009] [Accepted: 06/08/2009] [Indexed: 10/20/2022]
Abstract
The small bristles (sbr) gene of Drosophila melanogaster belongs to the family of nuclear export factor (NXF) genes that participate in mRNA nuclear export. During meiosis, females of Drosophila melanogaster that carry various combinations of mutant alleles of the Dm nxf1/sbr gene exhibit disruption of the division spindle and misalignment of chromosomes at the metaphase plate. Meiosis of sbr ( 5 ) /+ females is characterized by the formation of tripolar spindles during the first cell division. According to the sequencing results, the sbr ( 5 ) (l(1)K4) lethal allele is a deletion of 492 nucleotides. In SBR(5) protein, 57 of the 146 amino acids that have been lost by deletion belong to the NTF2-like domain.
Collapse
|
33
|
Uranishi H, Zolotukhin AS, Lindtner S, Warming S, Zhang GM, Bear J, Copeland NG, Jenkins NA, Pavlakis GN, Felber BK. The RNA-binding motif protein 15B (RBM15B/OTT3) acts as cofactor of the nuclear export receptor NXF1. J Biol Chem 2009; 284:26106-16. [PMID: 19586903 PMCID: PMC2758010 DOI: 10.1074/jbc.m109.040113] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The human SPEN family proteins SHARP, RBM15/OTT1, and RBM15B/OTT3 share the structural domain architecture but show distinct functional properties. Here, we examined the function of OTT3 and compared it with its paralogues RBM15 and SHARP. We found that OTT3, like RBM15, has post-transcriptional regulatory activity, whereas SHARP does not, supporting a divergent role of RBM15 and OTT3. OTT3 shares with RBM15 the association with the splicing factor compartment and the nuclear envelope as well as the binding to mRNA export factors NXF1 and Aly/REF. Mutational analysis revealed direct interaction of OTT3 and RBM15 with NXF1 via their C-terminal regions. Biochemical and subcellular localization studies showed that OTT3 and RBM15 also interact with each other in vivo, further supporting a shared function. Genetic knockdown of RBM15 in mouse is embryonically lethal, indicating that OTT3 cannot compensate for the RBM15 loss, which supports the notion that these proteins, in addition to sharing similar activities, likely have distinct biological roles.
Collapse
Affiliation(s)
- Hiroaki Uranishi
- Human Retrovirus Section, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland 21702-1201, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Stouffs K, Tournaye H, Van der Elst J, Liebaers I, Lissens W. Is there a role for the nuclear export factor 2 gene in male infertility? Fertil Steril 2008; 90:1787-91. [PMID: 18258234 DOI: 10.1016/j.fertnstert.2007.08.071] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2007] [Revised: 08/30/2007] [Accepted: 08/30/2007] [Indexed: 12/01/2022]
Abstract
OBJECTIVE To determine the presence of mutations in the NXF2 gene of patients with nonobstructive azoospermia. DESIGN Molecular analysis of male infertility. SETTING University genetic laboratory and reproductive clinic. PATIENT(S) Sixty-five patients with Sertoli cell-only syndrome (SCOS) and 20 control men. INTERVENTION(S) Polymerase chain reaction, sequencing analysis, RNA extraction, and reverse transcription polymerase chain reaction. MAIN OUTCOME MEASURE(S) Expression of NXF2 messenger RNA and analysis of the NXF2 gene for the presence of mutations and polymorphisms. RESULT(S) Messenger RNA derived from the NXF2 gene could be amplified from normal human testicular tissue. Sequencing analysis showed the presence of two polymorphisms in the NXF2 gene. A first alteration, c.1779C>T, was observed in one man who had complete SCOS. Although it is located near an intron-exon boundary, this change probably does not affect splicing. The second alteration, c.1857A>G, was detected in 22 patients with complete SCOS and in 13 patients with incomplete SCOS. Also, 15 of 20 men with normal spermatogenesis had this alteration. Neither of these alterations causes a change at the amino acid level. CONCLUSION(S) No mutations were detected in the NXF2 gene, from which we concluded that there is no need to screen for mutations in the NXF2 gene in a routine IVF program.
Collapse
Affiliation(s)
- Katrien Stouffs
- Research Centre for Reproduction and Genetics, Vrije Universiteit Brussel, Brussels, Belgium.
| | | | | | | | | |
Collapse
|
35
|
Katahira J, Miki T, Takano K, Maruhashi M, Uchikawa M, Tachibana T, Yoneda Y. Nuclear RNA export factor 7 is localized in processing bodies and neuronal RNA granules through interactions with shuttling hnRNPs. Nucleic Acids Res 2007; 36:616-28. [PMID: 18063567 PMCID: PMC2241847 DOI: 10.1093/nar/gkm556] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The nuclear RNA export factor (NXF) family proteins have been implicated in various aspects of post-transcriptional gene expression. This study shows that mouse NXF7 exhibits heterologous localization, i.e. NXF7 associates with translating ribosomes, stress granules (SGs) and processing bodies (P-bodies), the latter two of which are believed to be cytoplasmic sites of storage, degradation and/or sorting of mRNAs. By yeast two-hybrid screening, a series of heterogeneous nuclear ribonucleoproteins (hnRNPs) were identified as possible binding partners for NXF7. Among them, hnRNP A3, which is believed to be involved in translational control and/or cytoplasmic localization of certain mRNAs, formed a stable complex with NXF7 in vitro. Although hnRNP A3 was not associated with translating ribosomes, it was co-localized with NXF7 in P-bodies. After exposing to oxidative stress, NXF7 trans-localized to SGs, whereas hnRNP A3 did not. In differentiated neuroblastoma Neuro2a cells, NXF7 was co-localized with hnRNP A3 in cell body and neurites. The amino terminal half of NXF7, which was required for stable complex formation with hnRNP A3, coincided with the region required for localization in both P-bodies and neuronal RNA granules. These findings suggest that NXF7 plays a role in sorting, transport and/or storage of mRNAs through interactions with hnRNP A3.
Collapse
Affiliation(s)
- Jun Katahira
- Biomolecular Networks Laboratories, Biomolecular Dynamics Laboratory, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Japan.
| | | | | | | | | | | | | |
Collapse
|
36
|
Truant R, Atwal RS, Burtnik A. Nucleocytoplasmic trafficking and transcription effects of huntingtin in Huntington's disease. Prog Neurobiol 2007; 83:211-27. [PMID: 17240517 DOI: 10.1016/j.pneurobio.2006.11.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2006] [Revised: 11/29/2006] [Accepted: 11/30/2006] [Indexed: 01/08/2023]
Abstract
There are nine genetic neurodegenerative diseases caused by a similar genetic defect, a CAG DNA triplet-repeat expansion in the disease gene's open reading frame resulting in a polyglutamine expansion in the disease proteins. Despite the commonality of polyglutamine expansion, each of the polyglutamine diseases manifest as unique diseases, with some similarities, but important differences. These differences suggest that the context of the polyglutamine expansion is important to the mechanism of pathology of the disease proteins. Therefore, it is becoming increasingly paramount to understand the normal functions of these polyglutamine disease proteins, which include huntingtin, the polyglutamine-expanded protein in Huntington's disease (HD). Transcriptional dysregulation is seen in HD. Here we discuss the role of normal huntingtin in transcriptional regulation and misregulation in Huntington's disease in relation to potentially analogous model systems, and to other polyglutamine disease proteins. Huntingtin has functional roles in both the cytoplasm and the nucleus. One commonality of activity of polyglutamine disease proteins is at the level of protein dynamics and ability to import and export to and from the nucleus. Knowing the temporal location of huntingtin protein in response to signaling and neuronal communication could lead to valuable insights into an important trigger of HD pathology.
Collapse
Affiliation(s)
- Ray Truant
- McMaster University, Department of Biochemistry and Biomedical Sciences, HSC4H24A, 1200 Main Street West, Hamilton, Ontario, Canada L8N3Z5.
| | | | | |
Collapse
|
37
|
Matzat LH, Berberoglu S, Lévesque L. Formation of a Tap/NXF1 homotypic complex is mediated through the amino-terminal domain of Tap and enhances interaction with nucleoporins. Mol Biol Cell 2007; 19:327-38. [PMID: 17978099 DOI: 10.1091/mbc.e07-03-0255] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Nuclear export of mRNAs is mediated by the Tap/Nxt1 pathway. Tap moves its RNA cargo through the nuclear pore complex by direct interaction with nucleoporin phenylalanine-glycine repeats. This interaction is strengthened by the formation of a Tap/Nxt1 heterodimer. We now present evidence that Tap can form a multimeric complex with itself and with other members of the NXF family. We also show that the homotypic Tap complex can interact with both Nxt1 and nucleoporins in vitro. The region mediating this oligomerization is localized to the first 187 amino acids of Tap, which overlaps with its RNA-binding domain. Removal of this domain greatly reduces the ability of Tap to bind nucleoporins in vitro and in vivo. This is the first report showing that the Tap amino terminus modulates the interaction of Tap with nucleoporins. We speculate that this mechanism has a regulatory role for RNA export independent of RNA binding.
Collapse
Affiliation(s)
- Leah H Matzat
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | | | | |
Collapse
|
38
|
Froyen G, Van Esch H, Bauters M, Hollanders K, Frints SGM, Vermeesch JR, Devriendt K, Fryns JP, Marynen P. Detection of genomic copy number changes in patients with idiopathic mental retardation by high-resolution X-array-CGH: important role for increased gene dosage of XLMR genes. Hum Mutat 2007; 28:1034-42. [PMID: 17546640 DOI: 10.1002/humu.20564] [Citation(s) in RCA: 146] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
A tiling X-chromosome-specific genomic array with a theoretical resolution of 80 kb was developed to screen patients with idiopathic mental retardation (MR) for submicroscopic copy number differences. Four patients with aberrations previously detected at lower resolution were first analyzed. This facilitated delineation of the location and extent of the aberration at high resolution and subsequently, more precise genotype-phenotype analyses. A cohort of 108 patients was screened, 57 of which were suspected of X-linked mental retardation (XLMR), 26 were probands of brother pairs, and 25 were sporadic cases. A total of 15 copy number changes in 14 patients (13%) were detected, which included two deletions and 13 duplications ranging from 0.1 to 2.7 Mb. The aberrations are associated with the phenotype in five patients (4.6%), based on the following criteria: de novo aberration; involvement of a known or candidate X-linked nonsyndromic(syndromic) MR (MRX(S)) gene; segregation with the disease in the family; absence in control individuals; and skewed X-inactivation in carrier females. These include deletions that contain the MRX(S) genes CDKL5, OPHN1, and CASK, and duplications harboring CDKL5, NXF5, MECP2, and GDI1. In addition, seven imbalances were apparent novel polymorphic regions because they do not fulfill the proposed criteria. Taken together, our data strongly suggest that not only deletions but also duplications on the X chromosome contribute to the phenotype more often than expected, supporting the increased gene dosage mechanism for deregulation of normal cognitive development.
Collapse
Affiliation(s)
- Guy Froyen
- Human Genome Laboratory, Department for Molecular and Developmental Genetics, VIB, Leuven, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Takano K, Miki T, Katahira J, Yoneda Y. NXF2 is involved in cytoplasmic mRNA dynamics through interactions with motor proteins. Nucleic Acids Res 2007; 35:2513-21. [PMID: 17403691 PMCID: PMC1885657 DOI: 10.1093/nar/gkm125] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Tap/NXF1, the founding member of the evolutionarily conserved NXF (Nuclear RNA export Factor) family of proteins, is required for the nuclear export of bulk poly(A)+ RNAs. In mice, three additional NXF family genes (NXF2, NXF3, NXF7) have been identified and characterized to date. Cumulative data suggest that NXF family members play roles, not only in nuclear mRNA export, but also in various aspects of post-transcriptional mRNA metabolism. In order to better understand the functional role of NXF2, we searched for its binding partners by yeast two-hybrid screening and identified several cytoplasmic motor proteins, including KIF17. The interaction of NXF2 with KIF17, which was confirmed by GST pull-down and co-immunoprecipitation assays, is mediated by the N-terminal domain of NXF2, which is required for the punctate localization patterns in dendrites of primary neurons. We also found that the NXF2-containing dendritic granules, which were co-localized with KIF17, mRNA and Staufen1, a known component of neuronal mRNA granules, moved bidirectionally along dendrites in a microtubule-dependent manner. These results suggest that NXF2, a nucleo-cytoplasmic mRNA transporter, plays additional roles in the cytoplasmic localization of mRNAs through interactions with cytoplasmic motor proteins.
Collapse
Affiliation(s)
- Keizo Takano
- Department of Cell Biology and Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan and Laboratories for Biomolecular Networks, Department of Frontier Biosciences, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takashi Miki
- Department of Cell Biology and Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan and Laboratories for Biomolecular Networks, Department of Frontier Biosciences, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Jun Katahira
- Department of Cell Biology and Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan and Laboratories for Biomolecular Networks, Department of Frontier Biosciences, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
- *To whom correspondence should be addressed +81-6-6879-4606+81-6-6879-4609
| | - Yoshihiro Yoneda
- Department of Cell Biology and Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan and Laboratories for Biomolecular Networks, Department of Frontier Biosciences, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
40
|
Sheffield LG, Miskiewicz HB, Tannenbaum LB, Mirra SS. Nuclear pore complex proteins in Alzheimer disease. J Neuropathol Exp Neurol 2006; 65:45-54. [PMID: 16410748 DOI: 10.1097/01.jnen.0000195939.40410.08] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Ultrastructural studies of neurofibrillary tangles in Alzheimer disease (AD) have demonstrated a close relationship between nuclear pores and the cytoplasmic paired helical filaments comprising the tangles, as well as nuclear irregularity in many tangle-bearing neurons; nuclear pore aggregation has been observed in nearby neurons. These observations prompted examination of the nuclear pore complex (NPC) and proteins critical to nucleocytoplasmic transport in neurons with and without tangles in AD and control cases. Light microscopic study of hippocampus and neocortex in AD and controls revealed that all nuclei were labeled by antibodies to NPC proteins, including the central transporter nucleoporin Nup62. Nucleoporin and tau label revealed significantly more nuclear irregularity in AD, often associated with neurofibrillary tangles. Double label of Nup62 with apoptotic markers (TUNEL and active caspase-3) and a cell-cycle protein (cyclin B1) revealed no clear relationship of nuclear irregularity to apoptosis or cell-cycle protein expression. However, cytoplasmic accumulation of nuclear transport factor 2 (NTF2), a protein that transports cargo from the cytoplasm into the nucleus, was observed in a subset of hippocampal neurons with and without tangles in AD but not control cases. Further investigation of the NPC and nucleocytoplasmic transport in AD is warranted.
Collapse
Affiliation(s)
- Lynette G Sheffield
- Department of Pathology, SUNY Downstate Medical Center, Brooklyn, New York, NY 11203, USA
| | | | | | | |
Collapse
|
41
|
De Raedt T, Maertens O, Chmara M, Brems H, Heyns I, Sciot R, Majounie E, Upadhyaya M, De Schepper S, Speleman F, Messiaen L, Vermeesch JR, Legius E. Somatic loss of wild typeNF1 allele in neurofibromas: Comparison ofNF1 microdeletion and non-microdeletion patients. Genes Chromosomes Cancer 2006; 45:893-904. [PMID: 16830335 DOI: 10.1002/gcc.20353] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Neurofibromatosis type I (NF1) is an autosomal dominant familial tumor syndrome characterized by the presence of multiple benign neurofibromas. In 95% of NF1 individuals, a mutation is found in the NF1 gene, and in 5% of the patients, the germline mutation consists of a microdeletion that includes the NF1 gene and several flanking genes. We studied the frequency of loss of heterozygosity (LOH) in the NF1 region as a mechanism of somatic NF1 inactivation in neurofibromas from NF1 patients with and without a microdeletion. There was a statistically significant difference between these two patient groups in the proportion of neurofibromas with LOH. None of the 40 neurofibromas from six different NF1 microdeletion patients showed LOH, whereas LOH was observed in 6/28 neurofibromas from five patients with an intragenic NF1 mutation (P = 0.0034, Fisher's exact). LOH of the NF1 microdeletion region in NF1 microdeletion patients would de facto lead to a nullizygous state of the genes located in the deletion region and might be lethal. The mechanisms leading to LOH were further analyzed in six neurofibromas. In two out of six neurofibromas, a chromosomal microdeletion was found; in three, a mitotic recombination was responsible for the observed LOH; and in one, a chromosome loss with reduplication was present. These data show an important difference in the mechanisms of second hit formation in the 2 NF1 patient groups. We conclude that NF1 is a familial tumor syndrome in which the type of germline mutation influences the type of second hit in the tumors.
Collapse
Affiliation(s)
- Thomas De Raedt
- Center for Human Genetics, University Hospital Leuven, Catholic University of Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Tretyakova I, Zolotukhin AS, Tan W, Bear J, Propst F, Ruthel G, Felber BK. Nuclear Export Factor Family Protein Participates in Cytoplasmic mRNA Trafficking. J Biol Chem 2005; 280:31981-90. [PMID: 16014633 DOI: 10.1074/jbc.m502736200] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In eukaryotes, the nuclear export of mRNA is mediated by nuclear export factor 1 (NXF1) receptors. Metazoans encode additional NXF1-related proteins of unknown function, which share homology and domain organization with NXF1. Some mammalian NXF1-related genes are expressed preferentially in the brain and are thought to participate in neuronal mRNA metabolism. To address the roles of NXF1-related factors, we studied the two mouse NXF1 homologues, mNXF2 and mNXF7. In neuronal cells, mNXF2, but not mNXF7, exhibited mRNA export activity similar to that of Tip-associated protein/NXF1. Surprisingly, mNXF7 incorporated into mobile particles in the neurites that contained poly(A) and ribosomal RNA and colocalized with Staufen1-containing transport granules, indicating a role in neuronal mRNA trafficking. Yeast two-hybrid interaction, coimmunoprecipitation, and in vitro binding studies showed that NXF proteins bound to brain-specific microtubule-associated proteins (MAP) such as MAP1B and the WD repeat protein Unrip. Both in vitro and in vivo, MAP1B also bound to NXF export cofactor U2AF as well as to Staufen1 and Unrip. These findings revealed a network of interactions likely coupling the export and cytoplasmic trafficking of mRNA. We propose a model in which MAP1B tethers the NXF-associated mRNA to microtubules and facilitates their translocation along dendrites while Unrip provides a scaffold for the assembly of these transport intermediates.
Collapse
Affiliation(s)
- Irina Tretyakova
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, NCI-Frederick, National Institutes of Health, Frederick, Maryland 21702, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Van Esch H, Bauters M, Ignatius J, Jansen M, Raynaud M, Hollanders K, Lugtenberg D, Bienvenu T, Jensen LR, Gecz J, Moraine C, Marynen P, Fryns JP, Froyen G. Duplication of the MECP2 region is a frequent cause of severe mental retardation and progressive neurological symptoms in males. Am J Hum Genet 2005; 77:442-53. [PMID: 16080119 PMCID: PMC1226209 DOI: 10.1086/444549] [Citation(s) in RCA: 468] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2005] [Accepted: 07/05/2005] [Indexed: 12/16/2022] Open
Abstract
Loss-of-function mutations of the MECP2 gene at Xq28 are associated with Rett syndrome in females and with syndromic and nonsyndromic forms of mental retardation (MR) in males. By array comparative genomic hybridization (array-CGH), we identified a small duplication at Xq28 in a large family with a severe form of MR associated with progressive spasticity. Screening by real-time quantitation of 17 additional patients with MR who have similar phenotypes revealed three more duplications. The duplications in the four patients vary in size from 0.4 to 0.8 Mb and harbor several genes, which, for each duplication, include the MR-related L1CAM and MECP2 genes. The proximal breakpoints are located within a 250-kb region centromeric of L1CAM, whereas the distal breakpoints are located in a 300-kb interval telomeric of MECP2. The precise size and location of each duplication is different in the four patients. The duplications segregate with the disease in the families, and asymptomatic carrier females show complete skewing of X inactivation. Comparison of the clinical features in these patients and in a previously reported patient enables refinement of the genotype-phenotype correlation and strongly suggests that increased dosage of MECP2 results in the MR phenotype. Our findings demonstrate that, in humans, not only impaired or abolished gene function but also increased MeCP2 dosage causes a distinct phenotype. Moreover, duplication of the MECP2 region occurs frequently in male patients with a severe form of MR, which justifies quantitative screening of MECP2 in this group of patients.
Collapse
Affiliation(s)
- Hilde Van Esch
- Centre for Human Genetics, University Hospital Gasthuisberg, Leuven, Belgium.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Sasaki M, Takeda E, Takano K, Yomogida K, Katahira J, Yoneda Y. Molecular cloning and functional characterization of mouse Nxf family gene products. Genomics 2005; 85:641-53. [PMID: 15820316 DOI: 10.1016/j.ygeno.2005.01.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2004] [Accepted: 01/14/2005] [Indexed: 11/29/2022]
Abstract
Tap, a member of the evolutionarily conserved nuclear RNA export factor (NXF) family of proteins, has been implicated in the nuclear export of bulk poly(A)+ RNAs. cDNAs encoding the mouse NXF proteins (Tap, NXF7, NXF2, and NXF3) were prepared and the gene products were characterized in terms of their genomic organization, expression patterns, and biochemical properties. Mouse Tap was found to be ubiquitously expressed, whereas tissue- and developmental stage specific expression of mouse Nxf2, Nxf3, and Nxf7 was observed. Although mouse Tap and NXF2 bound to the phenylalanine-glycine repeat sequences of nucleoporins, NXF7 and NXF3 did not. GFP-tagged mouse Tap and NXF2 were localized predominantly in the nucleus. In contrast, GFP-tagged NXF7 and NXF3 were localized exclusively in the cytoplasm. As shown for the human counterpart, disruption of the leucine-rich nuclear export signal or leptomycin B treatment abolishes the cytoplasmic localization of mouse NXF3. p15/NXT1, an essential cofactor for human Tap in the export of mRNAs, was able to bind to mouse Tap, NXF2, and NXF3, but NXF7 did not form a stable heterodimeric complex. Transient transfection experiments indicated that only mouse Tap and NXF2 enhance the nuclear export of an otherwise inefficiently exported mRNA substrate. The orthologous relationship between human and mouse Nxf genes is discussed on the basis of these data.
Collapse
Affiliation(s)
- Mitsuho Sasaki
- Department of Cell Biology and Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | |
Collapse
|
45
|
Tan W, Zolotukhin AS, Tretyakova I, Bear J, Lindtner S, Smulevitch SV, Felber BK. Identification and characterization of the mouse nuclear export factor (Nxf) family members. Nucleic Acids Res 2005; 33:3855-65. [PMID: 16027110 PMCID: PMC1175460 DOI: 10.1093/nar/gki706] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
TAP/hNXF1 is a key factor that mediates general cellular mRNA export from the nucleus, and its orthologs are structurally and functionally conserved from yeast to humans. Metazoans encode additional proteins that share homology and domain organization with TAP/hNXF1, suggesting their participation in mRNA metabolism; however, the precise role(s) of these proteins is not well understood. Here, we found that the human mRNA export factor hNXF2 is specifically expressed in the brain, suggesting a brain-specific role in mRNA metabolism. To address the roles of additional NXF factors, we have identified and characterized the two Nxf genes, Nxf2 and Nxf7, which together with the TAP/hNXF1's ortholog Nxf1 comprise the murine Nxf family. Both mNXF2 and mNXF7 have a domain structure typical of the NXF family. We found that mNXF2 protein is expressed during mouse brain development. Similar to TAP/hNXF1, the mNXF2 protein is found in the nucleus, the nuclear envelope and cytoplasm, and is an active mRNA export receptor. In contrast, mNXF7 localizes exclusively to cytoplasmic granules and, despite its overall conserved sequence, lacks mRNA export activity. We concluded that mNXF2 is an active mRNA export receptor similar to the prototype TAP/hNXF1, whereas mNXF7 may have a more specialized role in the cytoplasm.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Barbara K. Felber
- To whom correspondence should be addressed. Tel: +1 301 846 5159; Fax: +1 301 846 7146;
| |
Collapse
|
46
|
Irwin S, Vandelft M, Pinchev D, Howell JL, Graczyk J, Orr HT, Truant R. RNA association and nucleocytoplasmic shuttling by ataxin-1. J Cell Sci 2005; 118:233-42. [PMID: 15615787 DOI: 10.1242/jcs.01611] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Spinocerebellar ataxia type 1 (SCA1) is a dominant neurodegenerative disease caused by the expression of mutant ataxin-1 containing an expanded polyglutamine tract. Ataxin-1 is a nuclear protein that localizes to punctate inclusions similar to neuronal nuclear inclusions seen in many polyglutamine expansion disease proteins. We demonstrate that ataxin-1 localization to inclusions and inclusion dynamics within the nucleus are RNA and transcription dependent, but not dependent on the polyglutamine tract. Ataxin-1 nuclear inclusions are distinct from other described nuclear bodies but recruit the mRNA export factor, TAP/NXF1, in a manner that is enhanced by cell heat shock. By FRAP protein dynamic studies in live cells, we found that wild-type, but not mutant, ataxin-1 was capable of nuclear export. These results suggest that the normal role of ataxin-1 may be in RNA processing, perhaps nuclear RNA export. Thus, nuclear retention of mutant ataxin-1 may be an important toxic gain of function in SCA1 disease.
Collapse
Affiliation(s)
- Stuart Irwin
- McMaster University, HSC 4H45, Department of Biochemistry, Hamilton, Ontario, L8N 3Z5, Canada
| | | | | | | | | | | | | |
Collapse
|
47
|
Williams B, Boyne J, Goodwin D, Roaden L, Hautbergue G, Wilson S, Whitehouse A. The prototype gamma-2 herpesvirus nucleocytoplasmic shuttling protein, ORF 57, transports viral RNA through the cellular mRNA export pathway. Biochem J 2005; 387:295-308. [PMID: 15537388 PMCID: PMC1134957 DOI: 10.1042/bj20041223] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2004] [Revised: 10/27/2004] [Accepted: 11/10/2004] [Indexed: 12/27/2022]
Abstract
HVS (herpesvirus saimiri) is the prototype gamma-2 herpesvirus. This is a subfamily of herpesviruses gaining importance since the identification of the first human gamma-2 herpesvirus, Kaposi's sarcoma-associated herpesvirus. The HVS ORF 57 (open reading frame 57) protein is a multifunctional transregulatory protein homologous with genes identified in all classes of herpesviruses. Recent work has demonstrated that ORF 57 has the ability to bind viral RNA, shuttles between the nucleus and cytoplasm and promotes the nuclear export of viral transcripts. In the present study, we show that ORF 57 shuttles between the nucleus and cytoplasm in a CRM-1 (chromosomal region maintenance 1)-independent manner. ORF 57 interacts with the mRNA export factor REF (RNA export factor) and two other components of the exon junction complex, Y14 and Magoh. The association of ORF 57 with REF stimulates recruitment of the cellular mRNA export factor TAP (Tip-associated protein), and HVS infection triggers the relocalization of REF and TAP from the nuclear speckles to several large clumps within the cell. Using a dominant-negative form of TAP and RNA interference to deplete TAP, we show that it is essential for bulk mRNA export in mammalian cells and is required for ORF 57-mediated viral RNA export. Furthermore, we show that the disruption of TAP reduces viral replication. These results indicate that HVS utilizes ORF 57 to recruit components of the exon junction complex and subsequently TAP to promote viral RNA export through the cellular mRNA export pathway.
Collapse
Key Words
- chromosomal region maintenance 1 (crm-1)
- exon junction complex
- herpesvirus
- mrna export
- open reading frame 57 (orf 57)
- tip-associated protein (tap)
- cmv, cytomegalovirus
- crm-1, chromosomal region maintenance 1
- cte, constitutive transport element
- ejc, exon junction complex
- fish, fluorescence in situ hybridization
- gb, glycoprotein b
- gfp, green fluorescent protein
- gst, glutathione s-transferase
- hek-293t cells, human embryonic kidney 293t cells
- hsv, herpes simplex virus
- hvs, herpesvirus saimiri
- kshv, kaposi's sarcoma associated herpesvirus
- lmb, leptomycin b
- moi, multiplicity of infection
- ref, rna export factor
- mref2-1, murine ref2-1
- nes, nuclear export signal
- nmd, nonsense-mediated decay
- omk cells, owl monkey kidney cells
- orf 57, open reading frame 57
- rnai, rna interference
- rnps1, rna-binding protein prevalent during s phase
- rrm, rna recognition motif
- tap, tip-associated protein
- trex complex, transcription and export complex
- yfp, yellow fluorescent protein
Collapse
Affiliation(s)
- Ben J. L. Williams
- *Department of Biomolecular Sciences, University of Manchester Institute of Science and Technology, Manchester M60 1QD, U.K
| | - James R. Boyne
- †School of Biochemistry and Microbiology, University of Leeds, Leeds LS2 9JT, U.K
| | - Delyth J. Goodwin
- †School of Biochemistry and Microbiology, University of Leeds, Leeds LS2 9JT, U.K
| | - Louise Roaden
- *Department of Biomolecular Sciences, University of Manchester Institute of Science and Technology, Manchester M60 1QD, U.K
| | - Guillaume M. Hautbergue
- *Department of Biomolecular Sciences, University of Manchester Institute of Science and Technology, Manchester M60 1QD, U.K
| | - Stuart A. Wilson
- *Department of Biomolecular Sciences, University of Manchester Institute of Science and Technology, Manchester M60 1QD, U.K
| | - Adrian Whitehouse
- †School of Biochemistry and Microbiology, University of Leeds, Leeds LS2 9JT, U.K
- ‡Molecular and Cellular Biology Research Group, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K
| |
Collapse
|
48
|
Huang H, Zhang B, Hartenstein PA, Chen JN, Lin S. NXT2 is required for embryonic heart development in zebrafish. BMC DEVELOPMENTAL BIOLOGY 2005; 5:7. [PMID: 15790397 PMCID: PMC1079804 DOI: 10.1186/1471-213x-5-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2004] [Accepted: 03/24/2005] [Indexed: 11/10/2022]
Abstract
Background NXT2 is a member of NXT family proteins that are generally involved in exporting nuclear RNA in eukaryotic cells. It is not known if NXT2 has any function in specific biological processes. Results A zebrafish mutant exhibiting specific heart defects during embryogenesis was generated by animal cloning-mediated retroviral insertions. Molecular analysis indicated that the mutant phenotype was caused by a disruption of NXT2. Whole-mount RNA in situ hybridization showed that NXT2 transcripts were clearly detectable in embryonic heart as well as other tissues. Further analysis revealed that expression level of one form of alternative splicing NXT2 mRNA transcripts was significantly reduced, resulting in deficient myocardial cell differentiation and the malformation of cardiac valve at the atrioventricular boundary. The defects could be reproduced by morpholino anti-sense oligo knockdown of NXT2. Conclusion NXT2 has a critical role in maintaining morphogenetic integrity of embryonic heart in vertebrate species.
Collapse
MESH Headings
- Active Transport, Cell Nucleus
- Alternative Splicing
- Animals
- Cell Differentiation/genetics
- Cloning, Organism
- DNA Transposable Elements
- Edema, Cardiac/genetics
- Edema, Cardiac/pathology
- Heart/embryology
- Heart/physiology
- Heart Defects, Congenital/genetics
- Heart Defects, Congenital/pathology
- Heart Valves/pathology
- Myocardium/chemistry
- Myocardium/cytology
- Myocardium/pathology
- Nuclear Export Signals/genetics
- Nuclear Export Signals/physiology
- Phenotype
- RNA, Antisense
- RNA, Messenger/analysis
- RNA, Messenger/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Transcription, Genetic
- Zebrafish/embryology
- Zebrafish/genetics
- Zebrafish Proteins/genetics
- Zebrafish Proteins/physiology
Collapse
Affiliation(s)
- Haigen Huang
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095, USA
| | - Bo Zhang
- Center of Developmental Biology and Genetics, College of Life Sciences Peking University, Beijing 100871, P. R. CHINA
| | - Parvana A Hartenstein
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095, USA
| | - Jau-nian Chen
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095, USA
| | - Shuo Lin
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095, USA
- Center of Developmental Biology and Genetics, College of Life Sciences Peking University, Beijing 100871, P. R. CHINA
| |
Collapse
|
49
|
Jin L, Guzik BW, Bor YC, Rekosh D, Hammarskjöld ML. Tap and NXT promote translation of unspliced mRNA. Genes Dev 2004; 17:3075-86. [PMID: 14701875 PMCID: PMC305259 DOI: 10.1101/gad.1155703] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Tap has been proposed to play a role in general mRNA export and also functions in expression of RNA with retained introns that contain the MPMV CTE (constitutive transport element). Tap forms a functional heterodimer with NXT/p15. We have previously demonstrated that unspliced intron-containing CTE RNA is efficiently exported to the cytoplasm in mammalian cells. Here we show that Tap and NXT proteins function together to enhance translation of proteins from the exported CTE RNA. Pulse chase experiments show that Tap/NXT significantly increases the rate of protein synthesis. Sucrose gradient analysis demonstrates that Tap and NXT efficiently shift the unspliced RNA into polyribosomal fractions. Furthermore, Tap, but not NXT is detected in polyribosomes. Taken together, our results indicate that Tap and NXT serve a role in translational regulation of RNA after export to the cytoplasm. They further suggest that Tap/NXT may play a role in remodeling of cytoplasmic RNP complexes, providing a link between export pathways and cytoplasmic fate.
Collapse
Affiliation(s)
- Li Jin
- Myles H. Thaler Center for AIDS and Human Retrovirus Research, and Department of Microbiology, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | | | |
Collapse
|
50
|
|