301
|
Tuoc TC, Pavlakis E, Tylkowski MA, Stoykova A. Control of cerebral size and thickness. Cell Mol Life Sci 2014; 71:3199-218. [PMID: 24614969 PMCID: PMC11113230 DOI: 10.1007/s00018-014-1590-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 02/10/2014] [Accepted: 02/13/2014] [Indexed: 11/24/2022]
Abstract
The mammalian neocortex is a sheet of cells covering the cerebrum that provides the structural basis for the perception of sensory inputs, motor output responses, cognitive function, and mental capacity of primates. Recent discoveries promote the concept that increased cortical surface size and thickness in phylogenetically advanced species is a result of an increased generation of neurons, a process that underlies higher cognitive and intellectual performance in higher primates and humans. Here, we review some of the advances in the field, focusing on the diversity of neocortical progenitors in different species and the cellular mechanisms of neurogenesis. We discuss recent views on intrinsic and extrinsic molecular determinants, including the role of epigenetic chromatin modifiers and microRNA, in the control of neuronal output in developing cortex and in the establishment of normal cortical architecture.
Collapse
Affiliation(s)
- Tran Cong Tuoc
- Institute of Neuroanatomy, Universitätsmedizin Göttingen, Kreuzbergring 40, 37075, Göttingen, Germany,
| | | | | | | |
Collapse
|
302
|
Aoki H, Hara A, Oomori Y, Shimizu Y, Yamada Y, Kunisada T. Neonatal lethality of neural crest cell-specificRestknockout mice is associated with gastrointestinal distension caused by aberrations of myenteric plexus. Genes Cells 2014; 19:723-42. [DOI: 10.1111/gtc.12172] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 07/13/2014] [Indexed: 11/29/2022]
Affiliation(s)
- Hitomi Aoki
- Department of Tissue and Organ Development, Regeneration, and Advanced Medical Science; Gifu University Graduate School of Medicine; 1-1 Yanagido Gifu 501-1194 Japan
| | - Akira Hara
- Department of Tumor Pathology; Gifu University Graduate School of Medicine; 1-1 Yanagido Gifu 501-1194 Japan
| | - Yoshiyuki Oomori
- TAIYO NIPPON SANSO Corporation; 3054-3 Shimokurosawa Takane-cho Hokuto-shi Yamanashi 408-0015 Japan
| | - Yasutake Shimizu
- Department of Basic Veterinary Science; Laboratory of Physiology; The United Graduate School of Veterinary Sciences Gifu University; 1-1 Yanagido Gifu 501-1193 Japan
| | - Yasuhiro Yamada
- Center for iPS Cell Research and Application (CiRA); Institute for Integrated Cell-Material Sciences (iCeMS); Kyoto University; Kyoto 606-8507 Japan
| | - Takahiro Kunisada
- Department of Tissue and Organ Development, Regeneration, and Advanced Medical Science; Gifu University Graduate School of Medicine; 1-1 Yanagido Gifu 501-1194 Japan
| |
Collapse
|
303
|
Smith J, Hing A, Clarke C, Johnson N, Perez F, Park S, Horst J, Mecham B, Maves L, Nickerson D, Cunningham M, Cunningham ML. Exome sequencing identifies a recurrent de novo ZSWIM6 mutation associated with acromelic frontonasal dysostosis. Am J Hum Genet 2014; 95:235-40. [PMID: 25105228 DOI: 10.1016/j.ajhg.2014.07.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Accepted: 07/15/2014] [Indexed: 10/24/2022] Open
Abstract
Acromelic frontonasal dysostosis (AFND) is a rare disorder characterized by distinct craniofacial, brain, and limb malformations, including frontonasal dysplasia, interhemispheric lipoma, agenesis of the corpus callosum, tibial hemimelia, preaxial polydactyly of the feet, and intellectual disability. Exome sequencing of one trio and two unrelated probands revealed the same heterozygous variant (c.3487C>T [p. Arg1163Trp]) in a highly conserved protein domain of ZSWIM6; this variant has not been seen in the 1000 Genomes data, dbSNP, or the Exome Sequencing Project. Sanger validation of the three trios confirmed that the variant was de novo and was also present in a fourth isolated proband. In situ hybridization of early zebrafish embryos at 24 hr postfertilization (hpf) demonstrated telencephalic expression of zswim6 and onset of midbrain, hindbrain, and retinal expression at 48 hpf. Immunohistochemistry of later-stage mouse embryos demonstrated tissue-specific expression in the derivatives of all three germ layers. qRT-PCR expression analysis of osteoblast and fibroblast cell lines available from two probands was suggestive of Hedgehog pathway activation, indicating that the ZSWIM6 mutation associated with AFND may lead to the craniofacial, brain and limb malformations through the disruption of Hedgehog signaling.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Michael L Cunningham
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Craniofacial Center, Seattle Children's Hospital, Seattle, WA 98105, USA.
| |
Collapse
|
304
|
RE-1 silencing transcription factor (REST): a regulator of neuronal development and neuronal/endocrine function. Cell Tissue Res 2014; 359:99-109. [PMID: 25092546 DOI: 10.1007/s00441-014-1963-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 07/08/2014] [Indexed: 01/04/2023]
Abstract
RE-1 silencing transcription factor (REST) is a transcriptional repressor that has been proposed to function as a master negative regulator of neurogenesis, as REST target genes encode neuronal receptors, ion channels, neuropeptides and synaptic proteins. During neuronal differentiation, REST expression levels are reduced, allowing expression of selected REST target genes. The analysis of neural stem/progenitor cells that are either devoid of REST or overexpress REST revealed that REST is not the master regulator that is solely responsible for the acquisition of the neuronal fate. Rather, REST provides a regulatory hub that coordinately regulates multiple tiers of neuronal development in vitro. In addition, REST may play an important role for maintaining the integrity of adult neurons. REST confers oxidative stress resistance and is essential for maintaining neuronal viability. Furthermore, the concentration of REST has been reported to influence the pathogenic outcome by neuronal diseases, including stroke, epilepsy and Alzheimer's disease. Experiments performed with PC12 pheochromocytoma cells indicate that REST may function as a key regulator of the neurosecretory phenotype. Moreover, transgenic mice overexpressing REST in pancreatic β-cells showed impaired insulin secretion leading to significantly reduced plasma insulin levels. Based on the fact that REST plays a prominent role in controlling stimulus-induced secretion in endocrine cells, we propose that REST may also be important for neurotransmitter release via regulation of genes that encode important proteins of the exocytotic machinery.
Collapse
|
305
|
Sedaghat Y, Bui HH, Mazur C, Monia BP. Identification of REST-regulated genes and pathways using a REST-targeted antisense approach. Nucleic Acid Ther 2014; 23:389-400. [PMID: 24329414 DOI: 10.1089/nat.2013.0445] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The repressor element-1 silencing transcription factor/neuron-restrictive silencer factor (REST/NRSF) is one of the first negative-acting transcriptional regulators implicated in vertebrate development thought to regulate hundreds of neuron-specific genes. However, its function in the adult system remains elusive. Here we employ second-generation antisense oligonucleotides (ASOs) to study the impact of rest-mediated suppression on gene expression. We demonstrate specific reductions in REST levels in vitro, and in vivo in mouse liver following treatment with ASOs, and we show that ASO mediated-REST suppression results in the elevation in expression of many neuronal genes including brain-derived neurotrophic factor, Synapsin1 (syn1) and β3-tubulin in BALB/c liver. Furthermore, we show the elevation of the affected proteins in plasma following ASO treatment. Finally, microarray analysis was applied to identify a broad range of genes modulated by REST suppression in mouse liver. Our findings suggest that REST may be an important target for neurodegenerative diseases like Huntington's disease, is also involved in the regulation of a broad range of additional cellular pathways, and that the antisense approach is a viable strategy for selectively modulating REST activity in vivo.
Collapse
Affiliation(s)
- Yalda Sedaghat
- 1 Department of Molecular Neurobiology, Evotec AG , Hamburg, Germany
| | | | | | | |
Collapse
|
306
|
Sugawara H, Tominaga A, Inoue K, Takeda Y, Yamada K, Miyata A. Functional characterization of neural-restrictive silencer element in mouse pituitary adenylate cyclase-activating polypeptide (PACAP) gene expression. J Mol Neurosci 2014; 54:526-34. [PMID: 24939248 DOI: 10.1007/s12031-014-0348-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 06/02/2014] [Indexed: 11/28/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is predominantly localized in the nervous system, but the underlying mechanism in its neuron-specific expression remains unclear. In addition to two neural-restrictive silencer-like element (NRSLE1 and 2), as reported previously, we have identified the third element in -1,601 to -1,581 bp from the translational initiation site of mouse PACAP gene and termed it as NRSLE3, of which, the sequence and location were highly conserved among mouse, rat, and human PACAP genes. In luciferase reporter assay, the deletion or site-directed mutagenesis of NRSLE3 in the reporter gene construct, driven by heterologous SV40 promoter, cancelled the repression of luciferase activity in non-neuronal Swiss-3T3 cells. Furthermore, its promoter activity was significantly repressed in Swiss-3T3 cells, but not in neuronal-differentiated PC12 cells. The electrophoretic mobility shift assay (EMSA) with nuclear extracts of Swiss-3T3 cells demonstrated a specific complex with NRSLE3 probe that exhibited the same migration with the neural-restrictive silencer element (NRSE) probe of rat type II sodium channel gene. During neuronal differentiation of PC12 cells, the increment of PACAP mRNA exhibited the correlation with that of REST4 mRNA, which is a neuron-specific variant form of neural-restrictive silencer factor (NRSF). In undifferentiated PC12 cells, trichostatin A (TSA), a histone deacetylase (HDAC) inhibitor, which indirectly inhibits NRSF-mediated gene silencing, increased PACAP mRNA level and attenuated the repression of promoter activity of 5' flanking region of mouse PACAP gene containing NRSLEs. These suggest that the NRSE-NRSF system implicates in the regulatory mechanism of neuron-specific expression of PACAP gene.
Collapse
Affiliation(s)
- Hideki Sugawara
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | | | | | | | | | | |
Collapse
|
307
|
Gasper WC, Marinov GK, Pauli-Behn F, Scott MT, Newberry K, DeSalvo G, Ou S, Myers RM, Vielmetter J, Wold BJ. Fully automated high-throughput chromatin immunoprecipitation for ChIP-seq: identifying ChIP-quality p300 monoclonal antibodies. Sci Rep 2014; 4:5152. [PMID: 24919486 PMCID: PMC4053718 DOI: 10.1038/srep05152] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 05/13/2014] [Indexed: 01/11/2023] Open
Abstract
Chromatin immunoprecipitation coupled with DNA sequencing (ChIP-seq) is the major contemporary method for mapping in vivo protein-DNA interactions in the genome. It identifies sites of transcription factor, cofactor and RNA polymerase occupancy, as well as the distribution of histone marks. Consortia such as the ENCyclopedia Of DNA Elements (ENCODE) have produced large datasets using manual protocols. However, future measurements of hundreds of additional factors in many cell types and physiological states call for higher throughput and consistency afforded by automation. Such automation advances, when provided by multiuser facilities, could also improve the quality and efficiency of individual small-scale projects. The immunoprecipitation process has become rate-limiting, and is a source of substantial variability when performed manually. Here we report a fully automated robotic ChIP (R-ChIP) pipeline that allows up to 96 reactions. A second bottleneck is the dearth of renewable ChIP-validated immune reagents, which do not yet exist for most mammalian transcription factors. We used R-ChIP to screen new mouse monoclonal antibodies raised against p300, a histone acetylase, well-known as a marker of active enhancers, for which ChIP-competent monoclonal reagents have been lacking. We identified, validated for ChIP-seq, and made publicly available a monoclonal reagent called ENCITp300-1.
Collapse
Affiliation(s)
- William C Gasper
- 1] Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA 91125, USA [2]
| | - Georgi K Marinov
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA 91125, USA
| | | | - Max T Scott
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA 91125, USA
| | | | - Gilberto DeSalvo
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Susan Ou
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Richard M Myers
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | - Jost Vielmetter
- 1] Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA 91125, USA [2]
| | - Barbara J Wold
- 1] Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA 91125, USA [2]
| |
Collapse
|
308
|
Rockowitz S, Lien WH, Pedrosa E, Wei G, Lin M, Zhao K, Lachman HM, Fuchs E, Zheng D. Comparison of REST cistromes across human cell types reveals common and context-specific functions. PLoS Comput Biol 2014; 10:e1003671. [PMID: 24922058 PMCID: PMC4055426 DOI: 10.1371/journal.pcbi.1003671] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Accepted: 05/01/2014] [Indexed: 11/18/2022] Open
Abstract
Recent studies have shown that the transcriptional functions of REST are much broader than repressing neuronal genes in non-neuronal systems. Whether REST occupies similar chromatin regions in different cell types and how it interacts with other transcriptional regulators to execute its functions in a context-dependent manner has not been adequately investigated. We have applied ChIP-seq analysis to identify the REST cistrome in human CD4+ T cells and compared it with published data from 15 other cell types. We found that REST cistromes were distinct among cell types, with REST binding to several tumor suppressors specifically in cancer cells, whereas 7% of the REST peaks in non-neuronal cells were ubiquitously called and <25% were identified for ≥ 5 cell types. Nevertheless, using a quantitative metric directly comparing raw ChIP-seq signals, we found the majority (∼80%) was shared by ≥ 2 cell types. Integration with RNA-seq data showed that REST binding was generally correlated with low gene expression. Close examination revealed that multiple contexts were correlated with reduced expression of REST targets, e.g., the presence of a cognate RE1 motif and cellular specificity of REST binding. These contexts were shown to play a role in differential corepressor recruitment. Furthermore, transcriptional outcome was highly influenced by REST cofactors, e.g., SIN3 and EZH2 co-occupancy marked higher and lower expression of REST targets, respectively. Unexpectedly, the REST cistrome in differentiated neurons exhibited unique features not observed in non-neuronal cells, e.g., the lack of RE1 motifs and an association with active gene expression. Finally, our analysis demonstrated how REST could differentially regulate a transcription network constituted of miRNAs, REST complex and neuronal factors. Overall, our findings of contexts playing critical roles in REST occupancy and regulatory outcome provide insights into the molecular interactions underlying REST's diverse functions, and point to novel roles of REST in differentiated neurons.
Collapse
Affiliation(s)
- Shira Rockowitz
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Wen-Hui Lien
- Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology & Development, The Rockefeller University, New York, New York, United States of America
| | - Erika Pedrosa
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Gang Wei
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda, Maryland, United States of America
| | - Mingyan Lin
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Keji Zhao
- Systems Biology Center, National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda, Maryland, United States of America
| | - Herbert M. Lachman
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Elaine Fuchs
- Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology & Development, The Rockefeller University, New York, New York, United States of America
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
- The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
309
|
Bai Y, Lathia JD, Zhang P, Flavahan W, Rich JN, Mattson MP. Molecular targeting of TRF2 suppresses the growth and tumorigenesis of glioblastoma stem cells. Glia 2014; 62:1687-98. [PMID: 24909307 DOI: 10.1002/glia.22708] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 05/23/2014] [Accepted: 05/23/2014] [Indexed: 12/23/2022]
Abstract
Glioblastoma is the most prevalent primary brain tumor and is essentially universally fatal within 2 years of diagnosis. Glioblastomas contain cellular hierarchies with self-renewing glioblastoma stem cells (GSCs) that are often resistant to chemotherapy and radiation therapy. GSCs express high amounts of repressor element 1 silencing transcription factor (REST), which may contribute to their resistance to standard therapies. Telomere repeat-binding factor 2 (TRF2) stablizes telomeres and REST to maintain self-renewal of neural stem cells and tumor cells. Here we show viral vector-mediated delivery of shRNAs targeting TRF2 mRNA depletes TRF2 and REST from GSCs isolated from patient specimens. As a result, GSC proliferation is reduced and the level of proteins normally expressed by postmitotic neurons (L1CAM and β3-tubulin) is increased, suggesting that loss of TRF2 engages a cell differentiation program in the GSCs. Depletion of TRF2 also sensitizes GSCs to temozolomide, a DNA-alkylating agent currently used to treat glioblastoma. Targeting TRF2 significantly increased the survival of mice bearing GSC xenografts. These findings reveal a role for TRF2 in the maintenance of REST-associated proliferation and chemotherapy resistance of GSCs, suggesting that TRF2 is a potential therapeutic target for glioblastoma.
Collapse
Affiliation(s)
- Yun Bai
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China; Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, Maryland
| | | | | | | | | | | |
Collapse
|
310
|
Liu CT, Young KL, Brody J, Olden M, Wojczynski MK, Heard-Costa N, Li G, Morrison AC, Muzny D, Gibbs RA, Reid JG, Shao Y, Zhou Y, Boerwinkle E, Heiss G, Wagenknecht L, McKnight B, Borecki IB, Fox CS, North KE, Cupples LA. Sequence variation in TMEM18 in association with body mass index: Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) Consortium Targeted Sequencing Study. CIRCULATION. CARDIOVASCULAR GENETICS 2014; 7:344-9. [PMID: 24951660 PMCID: PMC4135723 DOI: 10.1161/circgenetics.13.000067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Genome-wide association studies for body mass index (BMI) previously identified a locus near TMEM18. We conducted targeted sequencing of this region to investigate the role of common, low-frequency, and rare variants influencing BMI. METHODS AND RESULTS We sequenced TMEM18 and regions downstream of TMEM18 on chromosome 2 in 3976 individuals of European ancestry from 3 community-based cohorts (Atherosclerosis Risk in Communities, Cardiovascular Health Study, and Framingham Heart Study), including 200 adults selected for high BMI. We examined the association between BMI and variants identified in the region from nucleotide position 586 432 to 677 539 (hg18). Rare variants (minor allele frequency, <1%) were analyzed using a burden test and the sequence kernel association test. Results from the 3 cohort studies were meta-analyzed. We estimate that mean BMI is 0.43 kg/m(2) higher for each copy of the G allele of single-nucleotide polymorphism rs7596758 (minor allele frequency, 29%; P=3.46×10(-4)) using a Bonferroni threshold of P<4.6×10(-4). Analyses conditional on previous genome-wide association study single-nucleotide polymorphisms associated with BMI in the region led to attenuation of this signal and uncovered another independent (r(2)<0.2), statistically significant association, rs186019316 (P=2.11×10(-4)). Both rs186019316 and rs7596758 or proxies are located in transcription factor binding regions. No significant association with rare variants was found in either the exons of TMEM18 or the 3' genome-wide association study region. CONCLUSIONS Targeted sequencing around TMEM18 identified 2 novel BMI variants with possible regulatory function.
Collapse
Affiliation(s)
- Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Kristin L. Young
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC
- Carolina Population Center, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | - Jennifer Brody
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
| | | | - Mary K. Wojczynski
- Division of Statistical Genomics, Department of Genetics, Washington University, St. Louis, MO
| | - Nancy Heard-Costa
- NHLBI Framingham Heart Study, Framingham, MA
- Department of Neurology, Boston University School of Medicine, Boston, MA
| | - Guo Li
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
| | - Alanna C. Morrison
- Division of Epidemiology, School of Public Health, University of Texas Health Science Center, Houston, TX
| | - Donna Muzny
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX
| | - Richard A. Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX
| | - Jeffrey G. Reid
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX
| | - Yaming Shao
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC
| | - Yanhua Zhou
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Eric Boerwinkle
- Division of Epidemiology, School of Public Health, University of Texas Health Science Center, Houston, TX
| | - Geraldo Heiss
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC
| | - Lynne Wagenknecht
- Department of Epidemiology and Prevention, Wake Forest Baptist Medical Center, Winston-Salem, NC
| | - Barbara McKnight
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA
| | - Ingrid B. Borecki
- Division of Statistical Genomics, Department of Genetics, Washington University, St. Louis, MO
| | | | - Kari E. North
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC
| | - L. Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
- NHLBI Framingham Heart Study, Framingham, MA
| |
Collapse
|
311
|
50 years of research on the phenomena and epigenetic mechanism of neurogenesis. Neurosci Res 2014. [DOI: 10.1016/j.neures.2014.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
312
|
Tint GS, Pan L, Shang Q, Sharpe LJ, Brown AJ, Li M, Yu H. Desmosterol in brain is elevated because DHCR24 needs REST for Robust Expression but REST is poorly expressed. Dev Neurosci 2014; 36:132-42. [PMID: 24861183 DOI: 10.1159/000362363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 03/19/2014] [Indexed: 11/19/2022] Open
Abstract
Cholesterol synthesis in the fetal brain is inhibited because activity of DHCR24 (24-dehydrocholesterol reductase) is insufficient, causing concentrations of the precursor desmosterol to increase temporarily to 15-25% of total sterols at birth. We demonstrate that failure of DHCR24 to be adequately upregulated during periods of elevated cholesterol synthesis in the brain results from the presence in its promoter of the repressor element 1 (RE1) nucleotide sequence that binds the RE1-silencing transcription factor (REST) and that REST, generally reduced in neural tissues, uncharacteristically but not without precedent, enhances DHCR24 transcription. DHCR24 and REST mRNA levels are reduced 3- to 4-fold in fetal mouse brain compared to liver (p < 0.001). Chromatin immunoprecipitation assays suggested that REST binds to the human DHCR24 promoter in the vicinity of the predicted human RE1 sequence. Luminescent emission from a human DHCR24 promoter construct with a mutated RE1 sequence was reduced 2-fold compared to output from a reporter with wild-type RE1 (p < 0.005). Silencing REST in HeLa cells resulted in significant reductions of DHCR24 mRNA (2-fold) and DHCR24 protein (4-fold). As expected, relative concentrations of Δ(24)-cholesterol precursor sterols increased 3- to 4-fold, reflecting the inhibition of DHCR24 enzyme activity. In contrast, mRNA levels of DHCR7 (sterol 7-dehydrocholesterol reductase), a gene essential for cholesterol synthesis lacking an RE1 sequence, and concentrations of HMGR (3-hydroxy-3-methyl-glutaryl-CoA reductase) enzyme protein were both unaffected. Surprisingly, a dominant negative fragment of REST consisting of just the DNA binding domain (about 20% of the protein) and full-length REST enhanced DHCR24 expression equally well. Furthermore, RE1 and the sterol response element (SRE), the respective binding sites for REST and the SRE binding protein (SREBP), are contiguous. These observations led us to hypothesize that REST acts because it is bound in close proximity to SREBP, thus amplifying its ability to upregulate DHCR24. It is likely that modulation of DHCR24 expression by REST persisted in the mammalian genome either because it does no harm or because suppressing metabolically active DHCR24 while providing abundant quantities of the multifunctional sterol desmosterol during neural development proved useful.
Collapse
Affiliation(s)
- G S Tint
- Research Service, Department of Veterans Affairs Medical Center, East Orange, N.J., USA
| | | | | | | | | | | | | |
Collapse
|
313
|
Charbord J, Poydenot P, Bonnefond C, Feyeux M, Casagrande F, Brinon B, Francelle L, Aurégan G, Guillermier M, Cailleret M, Viegas P, Nicoleau C, Martinat C, Brouillet E, Cattaneo E, Peschanski M, Lechuga M, Perrier AL. High throughput screening for inhibitors of REST in neural derivatives of human embryonic stem cells reveals a chemical compound that promotes expression of neuronal genes. Stem Cells 2014; 31:1816-28. [PMID: 23712629 DOI: 10.1002/stem.1430] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 04/05/2013] [Accepted: 04/09/2013] [Indexed: 01/12/2023]
Abstract
Decreased expression of neuronal genes such as brain-derived neurotrophic factor (BDNF) is associated with several neurological disorders. One molecular mechanism associated with Huntington disease (HD) is a discrete increase in the nuclear activity of the transcriptional repressor REST/NRSF binding to repressor element-1 (RE1) sequences. High-throughput screening of a library of 6,984 compounds with luciferase-assay measuring REST activity in neural derivatives of human embryonic stem cells led to identify two benzoimidazole-5-carboxamide derivatives that inhibited REST silencing in a RE1-dependent manner. The most potent compound, X5050, targeted REST degradation, but neither REST expression, RNA splicing nor binding to RE1 sequence. Differential transcriptomic analysis revealed the upregulation of neuronal genes targeted by REST in wild-type neural cells treated with X5050. This activity was confirmed in neural cells produced from human induced pluripotent stem cells derived from a HD patient. Acute intraventricular delivery of X5050 increased the expressions of BDNF and several other REST-regulated genes in the prefrontal cortex of mice with quinolinate-induced striatal lesions. This study demonstrates that the use of pluripotent stem cell derivatives can represent a crucial step toward the identification of pharmacological compounds with therapeutic potential in neurological affections involving decreased expression of neuronal genes associated to increased REST activity, such as Huntington disease.
Collapse
Affiliation(s)
- Jérémie Charbord
- Inserm U861, AFM Evry Cedex, France; UEVE U861, AFM Evry Cedex, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
314
|
Lux CA, Mark P, Klopsch C, Laupheimer M, Tu-Rapp H, Li W, Ma N, Steinhoff G, David R. Impact of short-term liquid storage on human CD133(+) stem cells. Cell Transplant 2014; 24:2409-22. [PMID: 24800805 DOI: 10.3727/096368914x681577] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Stem cell transplantation is a viable strategy for regenerative medicine. However, it is inevitable to have cells undergo storage for several hours or days due to processing and transportation. Therefore, it is crucial to have rigidly controlled conditions ensuring the therapeutic benefit of isolated stem cells. In the present study, we investigated the impact of short-term storage on human CD133(+) cells. CD133(+) cells were isolated from human bone marrow and kept at standardized nonfreezing storage conditions for up to 72 h. Cell viability (apoptosis/necrosis) and expression of CD133 and CXCR4 were analyzed by flow cytometry. Metabolic activity was determined using an MTT assay; colony-forming ability, as well as endothelial-like differentiation, was further evaluated. A qRT-PCR array was employed to investigate the expression of stemness genes. CD133 and CXCR4 expressions were preserved at all time points. After 30 h, cell number and metabolic activity decreased, although no significant changes were detected in cell viability and proliferation as well as endothelial-like differentiation. Cell viability and proliferation decreased significantly only after 72 h of storage. Our results indicate that storage of isolated human CD133(+) bone marrow stem cells in liquid allows for high viability and functionality. However, storage time should be limited in order to avoid cell loss.
Collapse
Affiliation(s)
- Cornelia A Lux
- Reference and Translation Center for Cardiac Stem Cell Therapy, University of Rostock, Rostock, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
315
|
Adachi M, Monteggia LM. Decoding transcriptional repressor complexes in the adult central nervous system. Neuropharmacology 2014; 80:45-52. [PMID: 24418103 PMCID: PMC3984594 DOI: 10.1016/j.neuropharm.2013.12.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 12/18/2013] [Accepted: 12/30/2013] [Indexed: 12/27/2022]
Abstract
Cells maintain precise gene expression by balancing transcriptional activation and repression. While much work has focused on elucidating transcriptional activation in the central nervous system (CNS), little is known about transcriptional repression. One means to repress gene expression is to initiate binding of transcription factors to DNA, which then recruit co-repressors as well as other accessory proteins, forming a multi-protein repressor complex. These multi-protein repressor complexes include histone modifying enzymes that trigger processes such as histone acetylation, methylation, and ubiquitylation, altering chromatin structures to impact gene expression. Within these complexes transcriptional repressor proteins per se do not exhibit enzymatic reactions to remodel chromatin structure, whereas histone modifying enzymes lack intrinsic DNA binding activity but have an ability to process post-translational modifications on histones. Thus, the mutual association between transcriptional repressors and histone modifying enzymes is essential to sculpt chromatin to favor transcriptional repression and down regulate gene expression. Additionally, co-repressors are integral components in the context of gene repression as they bridge the association of transcriptional repressors and histone modifying enzymes. In this review, we will discuss the roles of some of the major components of these repressor complex in the CNS as well as their cellular functions that may underlie fundamental behavior in animals.
Collapse
Affiliation(s)
- Megumi Adachi
- Department of Neuroscience, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390-9111, USA
| | - Lisa M Monteggia
- Department of Neuroscience, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390-9111, USA.
| |
Collapse
|
316
|
Seki M, Masaki H, Arauchi T, Nakauchi H, Sugano S, Suzuki Y. A comparison of the rest complex binding patterns in embryonic stem cells and epiblast stem cells. PLoS One 2014; 9:e95374. [PMID: 24752154 PMCID: PMC3994037 DOI: 10.1371/journal.pone.0095374] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 03/26/2014] [Indexed: 10/29/2022] Open
Abstract
We detected and characterized the binding sites of the representative Rest complex components Rest, Sin3A, and Lsd1. We compared their binding patterns in mouse embryonic stem (ES) cells and epiblast stem (EpiS) cells. We found few Rest sites unique to the EpiS cells. The ES-unique site features were distinct from those of the common sites, namely, the signal intensities were weaker, and the characteristic gene function categories differed. Our analyses showed that the Rest binding sites do not always overlap with the Sin3A and Lsd1 binding sites. The Sin3A binding pattern differed remarkably between the ES and EpiS cells and was accompanied by significant changes in acetylated-histone patterns in the surrounding regions. A series of transcriptome analyses in the same cell types unexpectedly showed that the putative target gene transcript levels were not dramatically different despite dynamic changes in the Rest complex binding patterns and chromatin statuses, which suggests that Rest is not the sole determinant of repression at its targets. Nevertheless, we identified putative Rest targets with explicitly enhanced transcription upon Rest knock-down in 143 and 60 common and ES-unique Rest target genes, respectively. Among such sites, several genes are involved in ES cell proliferation. In addition, we also found that long, intergenic non-coding RNAs were apparent Rest targets and shared similar features with the protein-coding target genes. Interestingly, such non-coding target genes showed less conservation through evolution than protein-coding targets. As a result of differences in the components and targets of the Rest complex, its functional roles may differ in ES and EpiS cells.
Collapse
Affiliation(s)
- Masahide Seki
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, the University of Tokyo, Kashiwa, Chiba, Japan
| | - Hideki Masaki
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, the University of Tokyo, Minato-ku, Tokyo, Japan
| | - Takako Arauchi
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, the University of Tokyo, Kashiwa, Chiba, Japan
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, the University of Tokyo, Minato-ku, Tokyo, Japan
| | - Sumio Sugano
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, the University of Tokyo, Kashiwa, Chiba, Japan
| | - Yutaka Suzuki
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, the University of Tokyo, Kashiwa, Chiba, Japan
- * E-mail:
| |
Collapse
|
317
|
Saritas-Yildirim B, Silva EM. The role of targeted protein degradation in early neural development. Genesis 2014; 52:287-99. [PMID: 24623518 DOI: 10.1002/dvg.22771] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 03/05/2014] [Accepted: 03/07/2014] [Indexed: 11/08/2022]
Abstract
As neural stem cells differentiate into neurons during neurogenesis, the proteome of the cells is restructured by de novo expression and selective removal of regulatory proteins. The control of neurogenesis at the level of gene regulation is well documented and the regulation of protein abundance through protein degradation via the Ubiquitin/26S proteasome pathway is a rapidly developing field. This review describes our current understanding of the role of the proteasome pathway in neurogenesis. Collectively, the studies show that targeted protein degradation is an important regulatory mechanism in the generation of new neurons.
Collapse
|
318
|
Lu T, Aron L, Zullo J, Pan Y, Kim H, Chen Y, Yang TH, Kim HM, Drake D, Liu XS, Bennett DA, Colaiácovo MP, Yankner BA. REST and stress resistance in ageing and Alzheimer's disease. Nature 2014; 507:448-54. [PMID: 24670762 PMCID: PMC4110979 DOI: 10.1038/nature13163] [Citation(s) in RCA: 562] [Impact Index Per Article: 51.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 02/21/2014] [Indexed: 12/13/2022]
Abstract
Human neurons are functional over an entire lifetime, yet the mechanisms that preserve function and protect against neurodegeneration during aging are unknown. Here we show that induction of the repressor element 1-silencing transcription/neuron-restrictive silencer factor (REST/NRSF) is a universal feature of normal aging in human cortical and hippocampal neurons. REST is lost, however, in mild cognitive impairment (MCI) and Alzheimer’s disease (AD). Chromatin immunoprecipitation with deep sequencing (ChIP-seq) and expression analysis show that REST represses genes that promote cell death and AD pathology, and induces the expression of stress response genes. Moreover, REST potently protects neurons from oxidative stress and amyloid β-protein (Aβ) toxicity, and conditional deletion of REST in the mouse brain leads to age-related neurodegeneration. A functional ortholog of REST, C. elegans SPR-4, also protects against oxidative stress and Aβ toxicity. During normal aging, REST is induced in part by cell non-autonomous Wnt signaling. However, in AD, frontotemporal dementia and dementia with Lewy bodies, REST is lost from the nucleus and appears in autophagosomes together with pathologic misfolded proteins. Finally, REST levels during aging are closely correlated with cognitive preservation and longevity. Thus, the activation state of REST may distinguish neuroprotection from neurodegeneration in the aging brain.
Collapse
Affiliation(s)
- Tao Lu
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Liviu Aron
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Joseph Zullo
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Ying Pan
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Haeyoung Kim
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Yiwen Chen
- Department of Biostatistics and Computational Biology, Dana-Faber Cancer Institute and Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | - Tun-Hsiang Yang
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Hyun-Min Kim
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Derek Drake
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - X Shirley Liu
- Department of Biostatistics and Computational Biology, Dana-Faber Cancer Institute and Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois 60612, USA
| | - Monica P Colaiácovo
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Bruce A Yankner
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
319
|
Chang PC, Wang TY, Chang YT, Chu CY, Lee CL, Hsu HW, Zhou TA, Wu Z, Kim RH, Desai SJ, Liu S, Kung HJ. Autophagy pathway is required for IL-6 induced neuroendocrine differentiation and chemoresistance of prostate cancer LNCaP cells. PLoS One 2014; 9:e88556. [PMID: 24551118 PMCID: PMC3925144 DOI: 10.1371/journal.pone.0088556] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 01/07/2014] [Indexed: 12/29/2022] Open
Abstract
Prostate cancer (PCa) cells undergoing neuroendocrine differentiation (NED) are clinically relevant to the development of relapsed castration-resistant PCa. Increasing evidences show that autophagy involves in the development of neuroendocrine (NE) tumors, including PCa. To clarify the effect of autophagy on NED, androgen-sensitive PCa LNCaP cells were examined. Treatment of LNCaP cells with IL-6 resulted in an induction of autophagy. In the absence of androgen, IL-6 caused an even stronger activation of autophagy. Similar result was identified in NED induction. Inhibition of autophagy with chloroquine (CQ) markedly decreased NED. This observation was confirmed by beclin1 and Atg5 silencing experiments. Further supporting the role of autophagy in NED, we found that LC3 was up-regulated in PCa tissue that had relapsed after androgen-deprivation therapy when compared with their primary tumor counterpart. LC3 staining in relapsed PCa tissue showed punctate pattern similar to the staining of chromogranin A (CgA), a marker for NED cells. Moreover, autophagy inhibition induced the apoptosis of IL-6 induced NE differentiated PCa cells. Consistently, inhibition of autophagy by knockdown of beclin1 or Atg5 sensitized NE differentiated LNCaP cells to etoposide, a chemotherapy drug. To identify the mechanisms, phosphorylation of IL-6 downstream targets was analyzed. An increase in phospho-AMPK and a decrease in phospho-mTOR were found, which implies that IL-6 regulates autophagy through the AMPK/mTOR pathway. Most important to this study is the discovery of REST, a neuronal gene-specific transcriptional repressor that is involved in autophagy activation. REST was down-regulated in IL-6 treatment. Knockdown experiments suggest that REST is critical to NED and autophagy activation by IL-6. Together, our studies imply that autophagy is involved in PCa progression and plays a cytoprotective role when NED is induced in PCa cells by IL-6 treatment. These results reveal the potential of targeting autophagy as part of a combined therapeutic regime for NE tumors.
Collapse
Affiliation(s)
- Pei-Ching Chang
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan, R.O.C
- * E-mail: (PCC); (HJK)
| | - Tao-Yeuan Wang
- Department of Pathology, Mackay Medical College and Mackay Memorial Hospital, New Taipei City, Taiwan, R.O.C
- Mackay Junior College of Medicine, Nursing, and Management, New Taipei City, Taiwan, R.O.C
| | - Yi-Ting Chang
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan, R.O.C
| | - Cheng-Ying Chu
- Institute for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, R.O.C
| | - Chin-Ling Lee
- Department of Pathology, Mackay Medical College and Mackay Memorial Hospital, New Taipei City, Taiwan, R.O.C
- Mackay Junior College of Medicine, Nursing, and Management, New Taipei City, Taiwan, R.O.C
| | - Hung-Wei Hsu
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan, R.O.C
| | - Tyng-An Zhou
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan, R.O.C
| | - Zhaoju Wu
- Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, California, United States of America
- UC Davis Cancer Center, University of California Davis, Davis, California, United States of America
| | - Randie H. Kim
- Department of Dermatology, New York University School of Medicine, New York, New York, United States of America
| | - Sonal J. Desai
- UC Davis Cancer Center, University of California Davis, Davis, California, United States of America
| | - Shangqin Liu
- Department of Hematlogy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hsing-Jien Kung
- Institute for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, R.O.C
- Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, California, United States of America
- Division of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli County, Taiwan, R.O.C
- * E-mail: (PCC); (HJK)
| |
Collapse
|
320
|
Barca-Mayo O, De Pietri Tonelli D. Convergent microRNA actions coordinate neocortical development. Cell Mol Life Sci 2014; 71:2975-95. [PMID: 24519472 PMCID: PMC4111863 DOI: 10.1007/s00018-014-1576-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 01/11/2014] [Accepted: 01/27/2014] [Indexed: 12/19/2022]
Abstract
Neocortical development is a complex process that, at the cellular level, involves tight control of self-renewal, cell fate commitment, survival, differentiation and delamination/migration. These processes require, at the molecular level, the precise regulation of intrinsic signaling pathways and extrinsic factors with coordinated action in a spatially and temporally specific manner. Transcriptional regulation plays an important role during corticogenesis; however, microRNAs (miRNAs) are emerging as important post-transcriptional regulators of various aspects of central nervous system development. miRNAs are a class of small, single-stranded noncoding RNA molecules that control the expression of the majority of protein coding genes (i.e., targets). How do different miRNAs achieve precise control of gene networks during neocortical development? Here, we critically review all the miRNA–target interactions validated in vivo, with relevance to the generation and migration of pyramidal-projection glutamatergic neurons, and for the initial formation of cortical layers in the embryonic development of rodent neocortex. In particular, we focus on convergent miRNA actions, which are still a poorly understood layer of complexity in miRNA signaling, but potentially one of the keys to disclosing how miRNAs achieve the precise coordination of complex biological processes such as neocortical development.
Collapse
Affiliation(s)
- Olga Barca-Mayo
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Via Morego 30, 16163, Genoa, Italy
| | | |
Collapse
|
321
|
Survival of motor neuron protein downregulates miR-9 expression in patients with spinal muscular atrophy. Kaohsiung J Med Sci 2014; 30:229-34. [PMID: 24751385 DOI: 10.1016/j.kjms.2013.12.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 09/26/2013] [Indexed: 11/23/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a lethal hereditary disease caused by homozygous absence of the survival of the motor neuron (SMN) 1 gene (SMN1), and it is the leading genetic cause of infant mortality. The severity of SMA is directly correlated with SMN protein levels in affected patients; however, the cellular regulatory mechanisms for SMN protein expression are not completely understood. In this study, we investigated the regulatory effects between SMN expression and miR-9a, a downstream noncoding small RNA. Using an inducible SMN short hairpin RNA interference (shRNAi) system in NSC 34 and human skin fibroblast cells, cellular miR-9 levels and SMN protein repression were time-dependently upregulated. Conversely, cellular miR-9 levels decreased when HeLa cells were transfected with SMN protein fused with green fluorescent protein. In SMA-like mice spinal cords and human primary skin fibroblasts isolated from patients with different degrees of SMA, human SMN exhibited a disease severity-dependent decrease, whereas cellular miR-9 levels increased. These results clearly suggested that cellular SMN proteins regulated miR-9 expression and that miR-9 expression was related to SMA severity. Thus, miR-9 may be a marker for SMA prognosis.
Collapse
|
322
|
Dong ZF, Tang LJ, Deng GF, Zeng T, Liu SJ, Wan RP, Liu T, Zhao QH, Yi YH, Liao WP, Long YS. Transcription of the human sodium channel SCN1A gene is repressed by a scaffolding protein RACK1. Mol Neurobiol 2014; 50:438-48. [PMID: 24436055 DOI: 10.1007/s12035-014-8633-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 01/02/2014] [Indexed: 11/29/2022]
Abstract
Voltage-gated sodium channel α subunit type I (Nav1.1, encoded by SCN1A gene) plays a critical role in the initiation of action potential in the central nervous system. Downregulated expression of SCN1A is believed to be associated with epilepsy. Here, we found that the SCN1A promoter (P1c), located at the 5' untranslated exon 1c, drove the reporter gene expression in human NT2 cells, and a region between nt +53 and +62 downstream of the P1c promoter repressed the promoter activity. Further analyses showed that a scaffolding protein RACK1 (receptor for activated C kinase 1) was involved in binding to this silencer. Knockdown of RACK1 expression in NT2 cells deprived the repressive role of the silencer on the P1c promoter and increased SCN1A transcription, suggesting the potential involvement of RACK1 in negatively regulating SCN1A transcription via interaction with the silencer. Furthermore, we demonstrated that the binding of the protein complex including RACK1 to the SCN1A promoter motif was decreased in neuron-like differentiation of the NT2 cells induced by retinoic acid and resulted in the upregulation of SCN1A transcription. Taken together, this study reports a novel role of RACK1 in regulating SCN1A expression that participates in retinoic acid-induced neuronal differentiation of NT2 cells.
Collapse
Affiliation(s)
- Zhao-Fei Dong
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang East Road, Guangzhou, 510260, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
323
|
Abstract
The pseudounipolar sensory neurons of the dorsal root ganglia (DRG) give rise to peripheral branches that convert thermal, mechanical, and chemical stimuli into electrical signals that are transmitted via central branches to the spinal cord. These neurons express unique combinations of tetrodotoxin-sensitive (TTX-S) and tetrodotoxin-resistant (TTX-R) Na(+) channels that contribute to the resting membrane potential, action potential threshold, and regulate neuronal firing frequency. The small-diameter neurons (<25 μm) isolated from the DRG represent the cell bodies of C-fiber nociceptors that express both TTX-S and TTX-R Na(+) currents. The large-diameter neurons (>35 μm) are typically low-threshold A-fibers that predominately express TTX-S Na(+) currents. Peripheral nerve damage, inflammation, and metabolic diseases alter the expression and function of these Na(+) channels leading to increases in neuronal excitability and pain. The Na(+) channels expressed in these neurons are the target of intracellular signaling cascades that regulate the trafficking, cell surface expression, and gating properties of these channels. Post-translational regulation of Na(+) channels by protein kinases (PKA, PKC, MAPK) alter the expression and function of the channels. Injury-induced changes in these signaling pathways have been linked to sensory neuron hyperexcitability and pain. This review examines the signaling pathways and regulatory mechanisms that modulate the voltage-gated Na(+) channels of sensory neurons.
Collapse
Affiliation(s)
- Mohamed Chahine
- Centre de recherche, Institut en santé mentale de Québec, Local F-6539, 2601, chemin de la Canardière, QC City, QC, Canada, G1J 2G3,
| | | |
Collapse
|
324
|
Lopez-Ramirez MA, Nicoli S. Role of miRNAs and epigenetics in neural stem cell fate determination. Epigenetics 2013; 9:90-100. [PMID: 24342893 DOI: 10.4161/epi.27536] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The regulation of gene expression that determines stem cell fate determination is tightly controlled by both epigenetic and posttranscriptional mechanisms. Indeed, small non-coding RNAs such as microRNAs (miRNAs) are able to regulate neural stem cell fate by targeting chromatin-remodeling pathways. Here, we aim to summarize the latest findings regarding the feedback network of epigenetics and miRNAs during embryonic and adult neurogenesis.
Collapse
Affiliation(s)
- Miguel Alejandro Lopez-Ramirez
- Yale Cardiovascular Research Center; Section of Cardiovascular Medicine; Yale University School of Medicine; New Haven, CT USA
| | - Stefania Nicoli
- Yale Cardiovascular Research Center; Section of Cardiovascular Medicine; Yale University School of Medicine; New Haven, CT USA
| |
Collapse
|
325
|
Bersten DC, Wright JA, McCarthy PJ, Whitelaw ML. Regulation of the neuronal transcription factor NPAS4 by REST and microRNAs. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2013; 1839:13-24. [PMID: 24291638 DOI: 10.1016/j.bbagrm.2013.11.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 11/18/2013] [Accepted: 11/19/2013] [Indexed: 12/31/2022]
Abstract
NPAS4 is a brain restricted, activity-induced transcription factor which regulates the expression of inhibitory synapse genes to control homeostatic excitatory/inhibitory balance in neurons. NPAS4 is required for normal social interaction and contextual memory formation in mice. Protein and mRNA expression of NPAS4 is tightly coupled to neuronal depolarization and most prevalent in the cortical and hippocampal regions in the brain, however the precise mechanisms by which the NPAS4 gene is controlled remain unexplored. Here we show that expression of NPAS4 mRNA is actively repressed by RE-1 silencing transcription factor/neuron-restrictive silencer factor (REST/NRSF) in embryonic stem cells and non-neuronal cells by binding multiple sites within the promoter and Intron I of NPAS4. Repression by REST also appears to correlate with the binding of the zinc finger DNA binding protein CTCF within Intron I of NPAS4. In addition, we show that the 3' untranslated region (3'UTR) of NPAS4 can be targeted by two microRNAs, miR-203 and miR-224 to further regulate its expression. miR-224 is a midbrain/hypothalamus enriched microRNA which is expressed from an intron within the GABAA receptor epsilon (GABRE) gene and may further regionalize NPAS4 expression. Our results reveal REST and microRNA dependent mechanisms that restrict NPAS4 expression to the brain.
Collapse
Affiliation(s)
- David C Bersten
- School of Molecular and Biomedical Science (Biochemistry) and Australian Research, Council Special Research Centre for the Molecular Genetics of Development, The University of Adelaide, Adelaide, South Australia, Australia.
| | - Josephine A Wright
- School of Molecular and Biomedical Science (Biochemistry) and Australian Research, Council Special Research Centre for the Molecular Genetics of Development, The University of Adelaide, Adelaide, South Australia, Australia
| | - Peter J McCarthy
- School of Molecular and Biomedical Science (Biochemistry) and Australian Research, Council Special Research Centre for the Molecular Genetics of Development, The University of Adelaide, Adelaide, South Australia, Australia
| | - Murray L Whitelaw
- School of Molecular and Biomedical Science (Biochemistry) and Australian Research, Council Special Research Centre for the Molecular Genetics of Development, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
326
|
REST/NRSF-mediated intrinsic homeostasis protects neuronal networks from hyperexcitability. EMBO J 2013; 32:2994-3007. [PMID: 24149584 DOI: 10.1038/emboj.2013.231] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 09/24/2013] [Indexed: 11/08/2022] Open
Abstract
Intrinsic homeostasis enables neuronal circuits to maintain activity levels within an appropriate range by modulating neuronal voltage-gated conductances, but the signalling pathways involved in this process are largely unknown. We characterized the process of intrinsic homeostasis induced by sustained electrical activity in cultured hippocampal neurons based on the activation of the Repressor Element-1 Silencing Transcription Factor/Neuron-Restrictive Silencer Factor (REST/NRSF). We showed that 4-aminopyridine-induced hyperactivity enhances the expression of REST/NRSF, which in turn, reduces the expression of voltage-gated Na(+) channels, thereby decreasing the neuronal Na(+) current density. This mechanism plays an important role in the downregulation of the firing activity at the single-cell level, re-establishing a physiological spiking activity in the entire neuronal network. Conversely, interfering with REST/NRSF expression impaired this homeostatic response. Our results identify REST/NRSF as a critical factor linking neuronal activity to the activation of intrinsic homeostasis and restoring a physiological level of activity in the entire neuronal network.
Collapse
|
327
|
Foley JW, Sidow A. Transcription-factor occupancy at HOT regions quantitatively predicts RNA polymerase recruitment in five human cell lines. BMC Genomics 2013; 14:720. [PMID: 24138567 PMCID: PMC3826616 DOI: 10.1186/1471-2164-14-720] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 10/04/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND High-occupancy target (HOT) regions are compact genome loci occupied by many different transcription factors (TFs). HOT regions were initially defined in invertebrate model organisms, and we here show that they are a ubiquitous feature of the human gene-regulation landscape. RESULTS We identified HOT regions by a comprehensive analysis of ChIP-seq data from 96 DNA-associated proteins in 5 human cell lines. Most HOT regions co-localize with RNA polymerase II binding sites, but many are not near the promoters of annotated genes. At HOT promoters, TF occupancy is strongly predictive of transcription preinitiation complex recruitment and moderately predictive of initiating Pol II recruitment, but only weakly predictive of elongating Pol II and RNA transcript abundance. TF occupancy varies quantitatively within human HOT regions; we used this variation to discover novel associations between TFs. The sequence motif associated with any given TF's direct DNA binding is somewhat predictive of its empirical occupancy, but a great deal of occupancy occurs at sites without the TF's motif, implying indirect recruitment by another TF whose motif is present. CONCLUSIONS Mammalian HOT regions are regulatory hubs that integrate the signals from diverse regulatory pathways to quantitatively tune the promoter for RNA polymerase II recruitment.
Collapse
Affiliation(s)
- Joseph W Foley
- Department of Genetics, Stanford University, 300 Pasteur Drive, Stanford, California 94305, USA
- Current address: Douglas Mental Health University Institute, McGill University, 6875 Boulevard LaSalle, Verdun, Québec H4H 1R3, Canada
| | - Arend Sidow
- Department of Genetics, Stanford University, 300 Pasteur Drive, Stanford, California 94305, USA
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, California 94305, USA
| |
Collapse
|
328
|
Doghman M, Figueiredo BC, Volante M, Papotti M, Lalli E. Integrative analysis of SF-1 transcription factor dosage impact on genome-wide binding and gene expression regulation. Nucleic Acids Res 2013; 41:8896-8907. [PMID: 23907384 PMCID: PMC3799431 DOI: 10.1093/nar/gkt658] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 07/04/2013] [Accepted: 07/04/2013] [Indexed: 12/05/2022] Open
Abstract
Steroidogenic Factor-1 (SF-1) is a nuclear receptor that has a pivotal role in the development of adrenal glands and gonads and in the control of steroid hormone production, being also implicated in the pathogenesis of adrenocortical tumors. We have analyzed the mechanisms how SF-1 controls gene expression in adrenocortical cells and showed that it regulates different categories of genes according to its dosage. Significant correlations exist between the localization of SF-1-binding sites in chromatin under different dosage conditions and dosage-dependent regulation of gene expression. Our study revealed unexpected functional interactions between SF-1 and Neuron-Restrictive Silencer Factor/RE1-Silencing Transcription Factor (NRSF/REST), which was first characterized as a repressor of neuronal gene expression in non-neuronal tissues, in the regulation of gene expression in steroidogenic cells. When overexpressed, SF-1 reshapes the repertoire of NRSF/REST-regulated genes, relieving repression of key steroidogenic genes. These data show that NRSF/REST has a novel function in regulating gene expression in steroidogenic cells and suggest that it may have a broad role in regulating tissue-specific gene expression programs.
Collapse
Affiliation(s)
- Mabrouka Doghman
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS, Valbonne 06560, France, Associated International Laboratory (LIA) NEOGENEX CNRS, Valbonne 06560, France, University of Nice-Sophia-Antipolis, Valbonne 06560, France, Federal University of Paraná, Curitiba, Paraná 80060-000, Brazil, Instituto de Pesquisa Pelé Pequeno Principe, Curitiba, Paraná 80250-060, Brazil and Department of Oncology, University of Turin, Orbassano 10043, Italy
| | - Bonald C. Figueiredo
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS, Valbonne 06560, France, Associated International Laboratory (LIA) NEOGENEX CNRS, Valbonne 06560, France, University of Nice-Sophia-Antipolis, Valbonne 06560, France, Federal University of Paraná, Curitiba, Paraná 80060-000, Brazil, Instituto de Pesquisa Pelé Pequeno Principe, Curitiba, Paraná 80250-060, Brazil and Department of Oncology, University of Turin, Orbassano 10043, Italy
| | - Marco Volante
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS, Valbonne 06560, France, Associated International Laboratory (LIA) NEOGENEX CNRS, Valbonne 06560, France, University of Nice-Sophia-Antipolis, Valbonne 06560, France, Federal University of Paraná, Curitiba, Paraná 80060-000, Brazil, Instituto de Pesquisa Pelé Pequeno Principe, Curitiba, Paraná 80250-060, Brazil and Department of Oncology, University of Turin, Orbassano 10043, Italy
| | - Mauro Papotti
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS, Valbonne 06560, France, Associated International Laboratory (LIA) NEOGENEX CNRS, Valbonne 06560, France, University of Nice-Sophia-Antipolis, Valbonne 06560, France, Federal University of Paraná, Curitiba, Paraná 80060-000, Brazil, Instituto de Pesquisa Pelé Pequeno Principe, Curitiba, Paraná 80250-060, Brazil and Department of Oncology, University of Turin, Orbassano 10043, Italy
| | - Enzo Lalli
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS, Valbonne 06560, France, Associated International Laboratory (LIA) NEOGENEX CNRS, Valbonne 06560, France, University of Nice-Sophia-Antipolis, Valbonne 06560, France, Federal University of Paraná, Curitiba, Paraná 80060-000, Brazil, Instituto de Pesquisa Pelé Pequeno Principe, Curitiba, Paraná 80250-060, Brazil and Department of Oncology, University of Turin, Orbassano 10043, Italy
| |
Collapse
|
329
|
Vega AV, Avila G, Matthews G. Interaction between the transcriptional corepressor Sin3B and voltage-gated sodium channels modulates functional channel expression. Sci Rep 2013; 3:2809. [PMID: 24077057 PMCID: PMC3786298 DOI: 10.1038/srep02809] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 09/05/2013] [Indexed: 11/09/2022] Open
Abstract
Proteins that interact with voltage-gated sodium (Na(v)) channels are important in channel sorting and modulation. In this study, we identified the transcriptional regulator, Sin3B, as a novel binding partner of Na(v) channels in a yeast two-hybrid screen and confirmed the interaction using pull-down assays, co-immunoprecipitation, and immunofluorescence-colocalization. Because both long (~1100-residue) and short (N-terminal 293 residues) Sin3B variants interacted with Na(v) channels, binding occurred within the N-terminal region containing two paired-amphipathic helix domains. In Na(v) channels, Sin3B bound to a 132-residue portion of the cytoplasmic C-terminus. Expression of the short Sin3B variant strongly reduced native sodium current and Na(v)-channel gating charge in the neuronal cell line N1E-115, without affecting the voltage-dependence of activation. Because the total amount of channel protein was unchanged by Sin3B, binding of Sin3B likely decreases the number of channels in the plasma membrane, suggesting that interaction with Sin3B influences Na(v)-channel trafficking or stability in the membrane.
Collapse
Affiliation(s)
- Ana V Vega
- Carrera de Médico Cirujano. UBIMED. FES Iztacala, UNAM. Los Reyes Iztacala. Edo, Mex. 54090 México
| | | | | |
Collapse
|
330
|
Abstract
Spontaneous folding into a specific native structure is the most important property of protein to perform their biological functions within organisms. Spontaneous folding is understood on the basis of an energy landscape picture based on the minimum frustration principle. Therefore, frustration seemingly only leads to protein functional disorder. However, frustration has recently been suggested to have a function in allosteric regulation. Functional frustration has the possibility to be a key to our deeper understanding of protein function. To explore another functional frustration, we theoretically examined structural frustration, which is designed to induce intrinsic disorder of a protein and its function through the coupled folding and binding. We extended the Wako-Saitô-Muñoz-Eaton model to take into account a frustration effect. With the model, we analyzed the binding part of neuron-restrictive silencer factor and showed that designed structural frustration in it induces intrinsic disorder. Furthermore, we showed that the folding and the binding are cooperative in interacting with a target protein. The cooperativity enables an intrinsically disordered protein to exhibit a sharp switch-like folding response to binding chemical potential change. Through this switch-like response, the structural frustration may contribute to the regulation function of interprotein interaction of the intrinsically disordered protein.
Collapse
|
331
|
Kinetic analysis of npBAF to nBAF switching reveals exchange of SS18 with CREST and integration with neural developmental pathways. J Neurosci 2013; 33:10348-61. [PMID: 23785148 DOI: 10.1523/jneurosci.1258-13.2013] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
During the development of the vertebrate nervous system, neural progenitors divide, generate progeny that exit mitosis, and then migrate to sites where they elaborate specific morphologies and synaptic connections. Mitotic exit in neurons is accompanied by an essential switch in ATP-dependent chromatin regulatory complexes from the neural progenitor Brg/Brm-associated factor (npBAF) to neuron-specific nBAF complexes that is in part driven by miR-9/9* and miR-124. Recapitulating this microRNA/chromatin switch in fibroblasts leads to their direct conversion to neurons. We have defined the kinetics of neuron-specific BAF complex assembly in the formation of induced neurons from mouse embryonic stem cells, human fibroblasts, and normal mouse neural differentiation and, using proteomic analysis, found that this switch also includes the removal of SS18 and its replacement by CREST at mitotic exit. We found that switching of chromatin remodeling mechanisms is highly correlated with a broad switch in the use of neurogenic transcription factors. Knock-down of SS18 in neural stem cells causes cell-cycle exit and failure to self-renew, whereas continued expression of SS18 in neurons blocks dendritic outgrowth, underlining the importance of subunit switching. Because dominant mutations in BAF subunits underlie widely different human neurologic diseases arising in different neuronal types, our studies suggest that the characteristics of these diseases must be interpreted in the context of the different BAF assemblies in neurons rather than a singular mammalian SWItch/sucrose nonfermentable (mSWI/SNF) complex.
Collapse
|
332
|
Shimojo M, Shudo Y, Ikeda M, Kobashi T, Ito S. The small cell lung cancer-specific isoform of RE1-silencing transcription factor (REST) is regulated by neural-specific Ser/Arg repeat-related protein of 100 kDa (nSR100). Mol Cancer Res 2013; 11:1258-68. [PMID: 23928058 DOI: 10.1158/1541-7786.mcr-13-0269] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED Small cell lung cancer (SCLC) is a highly malignant form of cancer, which originates from primitive neuroendocrine cells in the lung. SCLC cells express several autocrine neurotransmitters/neuropeptides and their respective receptors. Expression of these neuronal markers is frequently regulated by RE1-silencing transcription factor (REST). In SCLC cells, an SCLC-specific isoform of REST (sREST) is highly expressed, whereas REST expression is undetectable, suggesting that the expression of sREST correlates with the pathogenesis of SCLC. Expression of sREST, which is derived through alternative splicing of REST, is abnormally regulated in SCLC cells, but the mechanism is unknown. Most recently, nSR100 (SRRM4) was described as an activator of REST alternative splicing. We now show that nSR100 is highly expressed in SCLC cells correlating with high sREST and low REST expression. Adhesion to the extracellular matrix (ECM) is thought to enhance tumorigenicity and confer resistance to apoptosis. Interestingly, nSR100 expression is enhanced in cells grown with ECM. Overexpression of REST caused repression of sREST and nSR100, the latter containing RE1 element controlled by REST. Culturing the SCLC cell line NCI-N417 cells with ECM also upregulated RE1-containing gene, the voltage-gated calcium channel subunit. Inhibition of the PI3K/Akt/mTOR pathway by LY294002 induced nSR100 expression, whereas the specific MEK/ERK inhibitor U0126 inhibited nSR100 expression. Repressing nSR100 by siRNA effectively repressed sREST, and conversely increased REST in NCI-N417 cells. Taken together, this report clarifies the ECM-dependent signaling pathway that impacts nSR100 expression and its regulation of alternative splicing in SCLC. IMPLICATIONS The splicing factor nSR100 may be novel SCLC-specific biomarker, as well as a therapeutic target.
Collapse
Affiliation(s)
- Masahito Shimojo
- Department of Medical Chemistry, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan.
| | | | | | | | | |
Collapse
|
333
|
Soshnev AA, Baxley RM, Manak JR, Tan K, Geyer PK. The insulator protein Suppressor of Hairy-wing is an essential transcriptional repressor in the Drosophila ovary. Development 2013; 140:3613-23. [PMID: 23884443 DOI: 10.1242/dev.094953] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Suppressor of Hairy-wing [Su(Hw)] is a DNA-binding factor required for gypsy insulator function and female germline development in Drosophila. The insulator function of the gypsy retrotransposon depends on Su(Hw) binding to clustered Su(Hw) binding sites (SBSs) and recruitment of the insulator proteins Centrosomal Protein 190 kD (CP190) and Modifier of mdg4 67.2 kD (Mod67.2). By contrast, the Su(Hw) germline function involves binding to non-clustered SBSs and does not require CP190 or Mod67.2. Here, we identify Su(Hw) target genes, using genome-wide analyses in the ovary to uncover genes with an ovary-bound SBS that are misregulated upon Su(Hw) loss. Most Su(Hw) target genes demonstrate enriched expression in the wild-type CNS. Loss of Su(Hw) leads to increased expression of these CNS-enriched target genes in the ovary and other tissues, suggesting that Su(Hw) is a repressor of neural genes in non-neural tissues. Among the Su(Hw) target genes is RNA-binding protein 9 (Rbp9), a member of the ELAV/Hu gene family. Su(Hw) regulation of Rbp9 appears to be insulator independent, as Rbp9 expression is unchanged in a genetic background that compromises the functions of the CP190 and Mod67.2 insulator proteins, even though both localize to Rbp9 SBSs. Rbp9 misregulation is central to su(Hw)(-/-) sterility, as Rbp9(+/-), su(Hw)(-/-) females are fertile. Eggs produced by Rbp9(+/-), su(Hw)(-/-) females show patterning defects, revealing a somatic requirement for Su(Hw) in the ovary. Our studies demonstrate that Su(Hw) is a versatile transcriptional regulatory protein with an essential developmental function involving transcriptional repression.
Collapse
Affiliation(s)
- Alexey A Soshnev
- Interdisciplinary Graduate Program in Molecular and Cellular Biology, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | | | |
Collapse
|
334
|
Lv J, Xin Y, Zhou W, Qiu Z. The epigenetic switches for neural development and psychiatric disorders. J Genet Genomics 2013; 40:339-346. [PMID: 23876774 DOI: 10.1016/j.jgg.2013.04.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2013] [Revised: 04/19/2013] [Accepted: 04/30/2013] [Indexed: 12/21/2022]
Abstract
The most remarkable feature of the nervous system is that the development and functions of the brain are largely reshaped by postnatal experiences, in joint with genetic landscapes. The nature vs. nurture argument reminds us that both genetic and epigenetic information is indispensable for the normal function of the brain. The epigenetic regulatory mechanisms in the central nervous system have been revealed over last a decade. Moreover, the mutations of epigenetic modulator genes have been shown to be implicated in neuropsychiatric disorders, such as autism spectrum disorders. The epigenetic study has initiated in the neuroscience field for a relative short period of time. In this review, we will summarize recent discoveries about epigenetic regulation on neural development, synaptic plasticity, learning and memory, as well as neuropsychiatric disorders. Although the comprehensive view of how epigenetic regulation contributes to the function of the brain is still not completed, the notion that brain, the most complicated organ of organisms, is profoundly shaped by epigenetic switches is widely accepted.
Collapse
Affiliation(s)
- Jingwen Lv
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai 201102, China
| | | | | | | |
Collapse
|
335
|
Pullen TJ, Rutter GA. When less is more: the forbidden fruits of gene repression in the adult β-cell. Diabetes Obes Metab 2013; 15:503-12. [PMID: 23121289 DOI: 10.1111/dom.12029] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 10/08/2012] [Accepted: 10/28/2012] [Indexed: 12/15/2022]
Abstract
Outside of the biological arena the term 'repression' often has a negative connotation. However, in the pancreatic β-cell a small group of genes, which are abundantly expressed in most if not all other mammalian tissues, are highly selectively repressed, with likely functional consequences. The two 'founder' members of this group, lactate dehydrogenase A (Ldha) and monocarboxylate transporter-1 (MCT-1/Slc16a1), are inactivated by multiple mechanisms including histone modifications and microRNA-mediated silencing. Their inactivation ensures that pyruvate and lactate, derived from muscle during exercise, do not stimulate insulin release inappropriately. Correspondingly, activating mutations in the MCT-1 promoter underlie 'exercise-induced hyperinsulinism' (EIHI) in man, a condition mimicked by forced over-expression of MCT-1 in the β-cell in mice. Furthermore, LDHA expression in the β-cell is upregulated in both human type 2 diabetes and in rodent models of the disease. Recent work by us and by others has identified a further ∼60 genes which are selectively inactivated in the β-cell, a list which we refine here up to seven by detailed comparison of the two studies. These genes include key regulators of cell proliferation and stimulus-secretion coupling. The present, and our earlier results, thus highlight the probable importance of shutting down a subset of 'disallowed' genes for the differentiated function of β-cells, and implicate previously unsuspected signalling pathways in the control of β-cell expansion and insulin secretion. Targeting of deregulated 'disallowed' genes in these cells may thus, in the future, provide new therapeutic avenues for type 2 diabetes.
Collapse
Affiliation(s)
- T J Pullen
- Section of Cell Biology, Department of Medicine, Imperial College London, London, UK
| | | |
Collapse
|
336
|
Faronato M, Patel V, Darling S, Dearden L, Clague MJ, Urbé S, Coulson JM. The deubiquitylase USP15 stabilizes newly synthesized REST and rescues its expression at mitotic exit. Cell Cycle 2013; 12:1964-77. [PMID: 23708518 PMCID: PMC3735711 DOI: 10.4161/cc.25035] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Reversible ubiquitylation of proteins contributes to their integrity, abundance and activity. The RE1-silencing transcription factor (REST) plays key physiological roles and is dysregulated in a spectrum of disease. It is rapidly turned over and is phosphorylated, polyubiquitylated and degraded en masse during neuronal differentiation and cell cycle progression. Through siRNA screening we identified the deubiquitylase USP15 as a key regulator of cellular REST. Both antagonism of REST polyubiquitylation and rescue of endogenous REST levels are dependent on the deubiquitylase activity of USP15. However, USP15 depletion does not destabilize pre-existing REST, but rather specifically impairs de novo REST synthesis. Indeed, we find that a small fraction of endogenous USP15 is associated with polysomes. In accordance with these findings, USP15 does not antagonize the degradation of phosphorylated REST at mitosis. Instead it is required for the rapid accumulation of newly synthesized REST on mitotic exit, thus playing a key role in its cell cycle oscillations. Importantly, this study reveals a novel role for a DUB in specifically promoting new protein synthesis.
Collapse
Affiliation(s)
- Monica Faronato
- Cellular & Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | | | | | | | | | | | | |
Collapse
|
337
|
Abstract
MicroRNAs (miRNAs) are important regulators of gene expression programs in the pancreas; however, little is known about the role of miRNA pathways during endocrine cell specification and maturation during neonatal life. In this study, we deleted Dicer1, an essential RNase for active miRNAs biogenesis, specifically from NGN3+ endocrine progenitor cells. We found that deletion of Dicer1 in endocrine progenitors did not affect the specification of hormone-expressing endocrine cells. However, the islets in the mutant mice in the neonatal period exhibited morphological defects in organization and loss of hormone expression, and the mutant mice subsequently developed diabetes. Dicer1-deficient β-cells lost insulin expression while maintaining the expression of β-cell transcription factors such as Pdx1 and Nkx6.1 early in the postnatal period. Surprisingly, transcriptional profiling showed that that the Dicer1-deficient endocrine cells expressed neuronal genes before the onset of diabetes. The derepression of neuronal genes was associated with a loss in binding of the neuronal transcriptional repressor RE-1-silencing transcription factor to its targets in Dicer1-deficient β-cells. These studies suggest that miRNAs play a critical role in suppressing neuronal genes during the maturation of endocrine cells.
Collapse
Affiliation(s)
- Murtaza S. Kanji
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
- Molecular Biology Interdepartmental Ph.D. Program (MBIDP), University of California, Los Angeles, Los Angeles, California
| | - Martin G. Martin
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Anil Bhushan
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
- Molecular Biology Interdepartmental Ph.D. Program (MBIDP), University of California, Los Angeles, Los Angeles, California
- Molecular, Cellular and Developmental Biology, University of California, Los Angeles, Los Angeles, California
- Corresponding author: Anil Bhushan,
| |
Collapse
|
338
|
Zhou G, Du T, Roizman B. The role of the CoREST/REST repressor complex in herpes simplex virus 1 productive infection and in latency. Viruses 2013; 5:1208-18. [PMID: 23628827 PMCID: PMC3712303 DOI: 10.3390/v5051208] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 04/21/2013] [Accepted: 04/23/2013] [Indexed: 12/12/2022] Open
Abstract
REST is a key component of the HDAC1 or 2, CoREST, LSD1, REST (HCLR) repressor complex. The primary function of the HCLR complex is to silence neuronal genes in non-neuronal cells. HCLR plays a role in regulating the expression of viral genes in productive infections as a donor of LDS1 for expression of α genes and as a repressor of genes expressed later in infection. In sensory neurons the HCLR complex is involved in the silencing of viral genome in the course of establishment of latency. The thesis of this article is that (a) sensory neurons evolved a mechanism to respond to the presence and suppress the transmission of infectious agents from the periphery to the CNS and (b) HSV evolved subservience to the HCLR with at least two objectives: to maintain a level of replication consistent with maximal person-to-person spread and to enable it to take advantage of neuronal innate immune responses to survive and be available for reactivation shielded from adaptive immune responses of the host.
Collapse
Affiliation(s)
| | | | - Bernard Roizman
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-773-702-1898; Fax: +1-773-702-1631
| |
Collapse
|
339
|
Hao N, Bhakti VLD, Peet DJ, Whitelaw ML. Reciprocal regulation of the basic helix-loop-helix/Per-Arnt-Sim partner proteins, Arnt and Arnt2, during neuronal differentiation. Nucleic Acids Res 2013; 41:5626-38. [PMID: 23599003 PMCID: PMC3675461 DOI: 10.1093/nar/gkt206] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Basic helix-loop-helix/Per-Arnt-Sim (bHLH/PAS) transcription factors function broadly in development, homeostasis and stress response. Active bHLH/PAS heterodimers consist of a ubiquitous signal-regulated subunit (e.g., hypoxia-inducible factors, HIF-1α/2α/3α; the aryl hydrocarbon receptor, AhR) or tissue-restricted subunit (e.g., NPAS1/3/4, Single Minded 1/2), paired with a general partner protein, aryl hydrocarbon receptor nuclear translocator (Arnt or Arnt2). We have investigated regulation of the neuron-enriched Arnt paralogue, Arnt2. We find high Arnt/Arnt2 ratios in P19 embryonic carcinoma cells and ES cells are dramatically reversed to high Arnt2/Arnt on neuronal differentiation. mRNA half-lives of Arnt and Arnt2 remain similar in both parent and neuronal differentiated cells. The GC-rich Arnt2 promoter, while heavily methylated in Arnt only expressing hepatoma cells, is methylation free in P19 and ES cells, where it is bivalent with respect to active H3K4me3 and repressive H3K27me3 histone marks. Typical of a 'transcription poised' developmental gene, H3K27me3 repressive marks are removed from Arnt2 during neuronal differentiation. Our data are consistent with a switch to predominant Arnt2 expression in neurons to allow specific functions of neuronal bHLH/PAS factors and/or to avoid neuronal bHLH/PAS factors from interfering with AhR/Arnt signalling.
Collapse
Affiliation(s)
- Nan Hao
- School of Molecular and Biomedical Science (Biochemistry), The University of Adelaide, Adelaide, South Australia 5005, Australia
| | | | | | | |
Collapse
|
340
|
Chen GL, Miller GM. Extensive alternative splicing of the repressor element silencing transcription factor linked to cancer. PLoS One 2013; 8:e62217. [PMID: 23614038 PMCID: PMC3628349 DOI: 10.1371/journal.pone.0062217] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 03/18/2013] [Indexed: 12/15/2022] Open
Abstract
The repressor element silencing transcription factor (REST) is a coordinate transcriptional and epigenetic regulator which functions as a tumor suppressor or an oncogene depending on cellular context, and a truncated splice variant REST4 has been linked to various types of cancer. We performed a comprehensive analysis of alternative splicing (AS) of REST by rapid amplification of cDNA ends and PCR amplification of cDNAs from various tissues and cell lines with specific primers. We identified 8 novel alternative exons including an alternate last exon which doubles the REST gene boundary, along with numerous 5'/3' splice sites and ends in the constitutive exons. With the combination of various splicing patterns (e.g. exon skipping and alternative usage of the first and last exons) that are predictive of altered REST activity, at least 45 alternatively spliced variants of coding and non-coding mRNA were expressed in a species- and cell-type/tissue-specific manner with individual differences. By examining the repertoire of REST pre-mRNA splicing in 27 patients with kidney, liver and lung cancer, we found that all patients without exception showed differential expression of various REST splice variants between paired tumor and adjacent normal tissues, with striking cell-type/tissue and individual differences. Moreover, we revealed that exon 3 skipping, which causes no frame shift but loss of a domain essential for nuclear translocation, was affected by pioglitazone, a highly selective activator of the peroxisome proliferator-activated receptor gamma (PPARγ) which contributes to cell differentiation and tumorigenesis besides its metabolic actions. Accordingly, this study demonstrates an extensive AS of REST pre-mRNA which redefines REST gene boundary and structure, along with a general but differential link between REST pre-mRNA splicing and various types of cancer. These findings advance our understanding of the complex, context-dependent regulation of REST gene expression and function, and provide potential biomarkers and therapeutic targets for cancer.
Collapse
Affiliation(s)
- Guo-Lin Chen
- Division of Neuroscience, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts, United States of America.
| | | |
Collapse
|
341
|
Das CM, Taylor P, Gireud M, Singh A, Lee D, Fuller G, Ji L, Fangusaro J, Rajaram V, Goldman S, Eberhart C, Gopalakrishnan V. The deubiquitylase USP37 links REST to the control of p27 stability and cell proliferation. Oncogene 2013; 32:1691-701. [PMID: 22665064 PMCID: PMC3435483 DOI: 10.1038/onc.2012.182] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 03/05/2012] [Accepted: 04/06/2012] [Indexed: 12/16/2022]
Abstract
The RE1 silencing transcription factor (REST) is a repressor of neuronal differentiation and its elevated expression in neural cells blocks neuronal differentiation. In this study, we demonstrate a role for REST in the control of proliferation of medulloblastoma cells. REST expression decreased the levels of cyclin-dependent kinase (CDK)NIB/p27, a CDK inhibitor and a brake of cell proliferation in these cells. The reciprocal relationship between REST and p27 was validated in human tumor samples. REST knockdown in medulloblastoma cells derepessed a novel REST target gene encoding the deubiquitylase ubiquitin (Ub)-specific peptidase 37 (USP37). Ectopically expressed wild-type USP37 formed a complex with p27, promoted its deubiquitination and stabilization and blocked cell proliferation. Knockdown of REST and USP37 prevented p27 stabilization and blocked the diminution in proliferative potential that normally accompanied REST loss. Unexpectedly, wild-type USP37 expression also induced the expression of REST-target neuronal differentiation genes even though REST levels were unaffected. In contrast, a mutant of USP37 carrying a site-directed change in a conserved cysteine failed to rescue REST-mediated p27 destabilization, maintenance of cell proliferation and blockade to neuronal differentiation. Consistent with these findings, a significant correlation between USP37 and p27 was observed in patient tumors. Collectively, these findings provide a novel connection between REST and the proteasomal machinery in the control of p27 and cell proliferation in medulloblastoma cells.
Collapse
Affiliation(s)
- Chandra M. Das
- Department of Pediatrics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Pete Taylor
- Department of Pediatrics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Monica Gireud
- Department of Pediatrics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Akanksha Singh
- Department of Pediatrics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Dean Lee
- Department of Pediatrics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Greg Fuller
- Department of Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Lingyun Ji
- Department of Biostatistics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Jason Fangusaro
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Veena Rajaram
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Stewart Goldman
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Charles Eberhart
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Vidya Gopalakrishnan
- Department of Pediatrics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
- Department of Brain Tumor Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
- Department of Centers for Cancer Epigenetics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
- Department of Stem Cells and Developmental Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
- Program in Neuroscience, The University of Texas Graduate School of Biomedical Sciences, Houston, Texas
| |
Collapse
|
342
|
Weiner JA, Jontes JD. Protocadherins, not prototypical: a complex tale of their interactions, expression, and functions. Front Mol Neurosci 2013; 6:4. [PMID: 23515683 PMCID: PMC3601302 DOI: 10.3389/fnmol.2013.00004] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 03/01/2013] [Indexed: 12/30/2022] Open
Abstract
The organization of functional neural circuits requires the precise and coordinated control of cell-cell interactions at nearly all stages of development, including neuronal differentiation, neuronal migration, axon outgrowth, dendrite arborization, and synapse formation and stabilization. This coordination is brought about by the concerted action of a large number of cell surface receptors, whose dynamic regulation enables neurons (and astrocytes) to adopt their proper roles within developing neural circuits. The protocadherins (Pcdhs) comprise a major family of cell surface receptors expressed in the developing vertebrate nervous system whose cellular and developmental roles are only beginning to be elucidated. In this review, we highlight selected recent results in several key areas of Pcdh biology and discuss their implications for our understanding of neural circuit formation and function.
Collapse
Affiliation(s)
- Joshua A Weiner
- Department of Biology, The University of Iowa Iowa City, IA, USA
| | | |
Collapse
|
343
|
González-Castañeda RE, Sánchez-González VJ, Flores-Soto M, Vázquez-Camacho G, Macías-Islas MA, Ortiz GG. Neural restrictive silencer factor and choline acetyltransferase expression in cerebral tissue of Alzheimer's Disease patients: A pilot study. Genet Mol Biol 2013; 36:28-36. [PMID: 23569405 PMCID: PMC3615522 DOI: 10.1590/s1415-47572013000100005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 10/15/2012] [Indexed: 02/05/2023] Open
Abstract
Decreased Choline Acetyltransferase (ChAT) brain level is one of the main biochemical disorders in Alzheimer’s Disease (AD). In rodents, recent data show that the CHAT gene can be regulated by a neural restrictive silencer factor (NRSF). The aim of the present work was to evaluate the gene and protein expression of CHAT and NRSF in frontal, temporal, entorhinal and parietal cortices of AD patient brains. Four brains from patients with AD and four brains from subjects without dementia were studied. Cerebral tissues were obtained and processed by the guanidine isothiocyanate method for RNA extraction. CHAT and NRSF gene and protein expression were determined by reverse transcription-polymerase chain reaction (RT-PCR) and Western blotting. CHAT gene expression levels were 39% lower in AD patients as compared to the control group (p < 0.05, U test). ChAT protein levels were reduced by 17% (p = 0.02, U test). NRSF gene expression levels were 86% higher in the AD group (p = 0.001, U test) as compared to the control group. In the AD subjects, the NRSF protein levels were 57% higher (p > 0.05, U test) than in the control subjects. These findings suggest for the first time that in the brain of AD patients high NRSF protein levels are related to low CHAT gene expression levels.
Collapse
Affiliation(s)
- Rocío E González-Castañeda
- Laboratorio de Desarrollo-Envejecimiento, Enfermedades Neurodegenerativas, División de Neurociencias, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, México. ; Instituto Tecnológico de Estudios Superiores de Monterrey, División de Ciencias de la Salud, Escuela de Medicina, Campus Guadalajara, Guadalajara, Jalisco, México
| | | | | | | | | | | |
Collapse
|
344
|
Wei C, Thatcher EJ, Olena AF, Cha DJ, Perdigoto AL, Marshall AF, Carter BD, Broadie K, Patton JG. miR-153 regulates SNAP-25, synaptic transmission, and neuronal development. PLoS One 2013; 8:e57080. [PMID: 23451149 PMCID: PMC3581580 DOI: 10.1371/journal.pone.0057080] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Accepted: 01/16/2013] [Indexed: 01/01/2023] Open
Abstract
SNAP-25 is a core component of the trimeric SNARE complex mediating vesicle exocytosis during membrane addition for neuronal growth, neuropeptide/growth factor secretion, and neurotransmitter release during synaptic transmission. Here, we report a novel microRNA mechanism of SNAP-25 regulation controlling motor neuron development, neurosecretion, synaptic activity, and movement in zebrafish. Loss of miR-153 causes overexpression of SNAP-25 and consequent hyperactive movement in early zebrafish embryos. Conversely, overexpression of miR-153 causes SNAP-25 down regulation resulting in near complete paralysis, mimicking the effects of treatment with Botulinum neurotoxin. miR-153-dependent changes in synaptic activity at the neuromuscular junction are consistent with the observed movement defects. Underlying the movement defects, perturbation of miR-153 function causes dramatic developmental changes in motor neuron patterning and branching. Together, our results indicate that precise control of SNAP-25 expression by miR-153 is critically important for proper neuronal patterning as well as neurotransmission.
Collapse
Affiliation(s)
- Chunyao Wei
- Department of Biological Sciences, Vanderbilt University and Medical School, Nashville, Tennessee, United States of America
| | - Elizabeth J. Thatcher
- Department of Biological Sciences, Vanderbilt University and Medical School, Nashville, Tennessee, United States of America
| | - Abigail F. Olena
- Department of Biological Sciences, Vanderbilt University and Medical School, Nashville, Tennessee, United States of America
| | - Diana J. Cha
- Department of Biological Sciences, Vanderbilt University and Medical School, Nashville, Tennessee, United States of America
| | - Ana L. Perdigoto
- Department of Biochemistry, Vanderbilt University and Medical School, Nashville, Tennessee, United States of America
| | - Andrew F. Marshall
- Department of Biological Sciences, Vanderbilt University and Medical School, Nashville, Tennessee, United States of America
| | - Bruce D. Carter
- Department of Biochemistry, Vanderbilt University and Medical School, Nashville, Tennessee, United States of America
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University and Medical School, Nashville, Tennessee, United States of America
| | - James G. Patton
- Department of Biological Sciences, Vanderbilt University and Medical School, Nashville, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
345
|
Mojtahedi S, Kordi MR, Hosseini SE, Omran SF, Soleimani M. Effect of treadmill running on the expression of genes that are involved in neuronal differentiation in the hippocampus of adult male rats. Cell Biol Int 2013; 37:276-83. [DOI: 10.1002/cbin.10022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 11/15/2012] [Indexed: 12/31/2022]
Affiliation(s)
- Shima Mojtahedi
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences; University of Tehran; Tehran; Iran
| | - Mohammad Reza Kordi
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences; University of Tehran; Tehran; Iran
| | | | - Simin Fallah Omran
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences; University of Tehran; Tehran; Iran
| | | |
Collapse
|
346
|
Low WC, Rujitanaroj PO, Lee DK, Messersmith PB, Stanton LW, Goh E, Chew SY. Nanofibrous scaffold-mediated REST knockdown to enhance neuronal differentiation of stem cells. Biomaterials 2013; 34:3581-90. [PMID: 23415645 DOI: 10.1016/j.biomaterials.2013.01.093] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 01/26/2013] [Indexed: 02/06/2023]
Abstract
At present, the recovery prospect for patients with chronic neurodegenerative diseases or acute trauma in the central nervous system is sub-optimal. The controlled differentiation of neural stem/progenitor cells (NPCs) to functional neurons is a possible treatment strategy. In contrast to the classical approach of biochemicals supplementation for guided stem cell commitment, this study explores the feasibility of directing neuronal differentiation through synergistic integration of three-dimensional nanofibrous topographical cues and scaffold-mediated knockdown of RE-1 silencing transcription factor (REST) in mouse NPCs. Taking advantage of the strong adhesive property and latent reactivity of mussel-inspired polydopamine (PD) coating, electrospun polycaprolactone (PCL) nanofibers were successfully functionalized with REST siRNAs (denoted as siREST PD-fiber). Sustained REST knockdown in NPCs was achieved for up to five days in vitro and the silencing efficiency was significantly higher than that mediated through siRNA adsorption onto non-PD coated sample controls. The silencing of REST, together with nanofiber topographical effect, significantly enhanced NPC neuronal commitment (57.5% Map2(+) cells in siREST PD-fiber vs. 43.5% in siREST PD-film vs. 50% in PD-fiber controls, p < 0.05) while reducing astrocytic and oligodendrocytic differentiation (10.7% O4(+) cells vs. ∼30% in siREST PD-film, p < 0.01). Taken together, the synergistic effects of scaffold-mediated REST knockdown and topographical cues from PD-modified nanofibers may be a useful strategy for generating functional neurons for therapeutic purposes.
Collapse
Affiliation(s)
- Wei Ching Low
- Division of Chemical and Biomolecular Engineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | | | | | | | | | | | | |
Collapse
|
347
|
Maze I, Noh KM, Allis CD. Histone regulation in the CNS: basic principles of epigenetic plasticity. Neuropsychopharmacology 2013; 38:3-22. [PMID: 22828751 PMCID: PMC3521967 DOI: 10.1038/npp.2012.124] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 05/07/2012] [Accepted: 05/08/2012] [Indexed: 12/25/2022]
Abstract
Postmitotic neurons are subject to a vast array of environmental influences that require the nuclear integration of intracellular signaling events to promote a wide variety of neuroplastic states associated with synaptic function, circuit formation, and behavioral memory. Over the last decade, much attention has been paid to the roles of transcription and chromatin regulation in guiding fundamental aspects of neuronal function. A great deal of this work has centered on neurodevelopmental and adulthood plasticity, with increased focus in the areas of neuropharmacology and molecular psychiatry. Here, we attempt to provide a broad overview of chromatin regulation, as it relates to central nervous system (CNS) function, with specific emphasis on the modes of histone posttranslational modifications, chromatin remodeling, and histone variant exchange. Understanding the functions of chromatin in the context of the CNS will aid in the future development of pharmacological therapeutics aimed at alleviating devastating neurological disorders.
Collapse
Affiliation(s)
- Ian Maze
- Laboratory of Chromatin Biology and Epigenetics, The Rockefeller University, New York, NY, USA
| | - Kyung-Min Noh
- Laboratory of Chromatin Biology and Epigenetics, The Rockefeller University, New York, NY, USA
| | - C David Allis
- Laboratory of Chromatin Biology and Epigenetics, The Rockefeller University, New York, NY, USA
| |
Collapse
|
348
|
Bergeron SA, Hannan MC, Codore H, Fero K, Li GH, Moak Z, Yokogawa T, Burgess HA. Brain selective transgene expression in zebrafish using an NRSE derived motif. Front Neural Circuits 2012; 6:110. [PMID: 23293587 PMCID: PMC3531662 DOI: 10.3389/fncir.2012.00110] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 12/07/2012] [Indexed: 11/13/2022] Open
Abstract
Transgenic technologies enable the manipulation and observation of circuits controlling behavior by permitting expression of genetically encoded reporter genes in neurons. Frequently though, neuronal expression is accompanied by transgene expression in non-neuronal tissues, which may preclude key experimental manipulations, including assessment of the contribution of neurons to behavior by ablation. To better restrict transgene expression to the nervous system in zebrafish larvae, we have used DNA sequences derived from the neuron-restrictive silencing element (NRSE). We find that one such sequence, REx2, when used in conjunction with several basal promoters, robustly suppresses transgene expression in non-neuronal tissues. Both in transient transgenic experiments and in stable enhancer trap lines, suppression is achieved without compromising expression within the nervous system. Furthermore, in REx2 enhancer trap lines non-neuronal expression can be de-repressed by knocking down expression of the NRSE binding protein RE1-silencing transcription factor (Rest). In one line, we show that the resulting pattern of reporter gene expression coincides with that of the adjacent endogenous gene, hapln3. We demonstrate that three common basal promoters are susceptible to the effects of the REx2 element, suggesting that this method may be useful for confining expression from many other promoters to the nervous system. This technique enables neural specific targeting of reporter genes and thus will facilitate the use of transgenic methods to manipulate circuit function in freely behaving larvae.
Collapse
Affiliation(s)
- Sadie A Bergeron
- Program in Genomics of Differentiation, Unit on Behavioral Neurogenetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development Bethesda, MD, USA
| | | | | | | | | | | | | | | |
Collapse
|
349
|
Abstract
During development, diverse cellular identities are established and maintained in the embryo. Although remarkably robust in vivo, cellular identities can be manipulated using experimental techniques. Lineage reprogramming is an emerging field at the intersection of developmental and stem cell biology in which a somatic cell is stably reprogrammed into a distinct cell type by forced expression of lineage-determining factors. Lineage reprogramming enables the direct conversion of readily available cells from patients (such as skin fibroblasts) into disease-relevant cell types (such as neurons and cardiomyocytes) or into induced pluripotent stem cells. Although remarkable progress has been made in developing novel reprogramming methods, the efficiency and fidelity of reprogramming need to be improved in order increase the experimental and translational utility of reprogrammed cells. Studying the mechanisms that prevent successful reprogramming should allow for improvements in reprogramming methods, which could have significant implications for regenerative medicine and the study of human disease. Furthermore, lineage reprogramming has the potential to become a powerful system for dissecting the mechanisms that underlie cell fate establishment and terminal differentiation processes. In this review, we will discuss how transcription factors interface with the genome and induce changes in cellular identity in the context of development and reprogramming.
Collapse
Affiliation(s)
- Thomas Vierbuchen
- Institute for Stem Cell Biology and Regenerative Medicine, Department of Pathology, and Cancer Biology Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | |
Collapse
|
350
|
Okamura D, Mochizuki K, Taniguchi H, Tokitake Y, Ikeda M, Yamada Y, Tournier C, Yamaguchi S, Tada T, Schöler HR, Matsui Y. REST and its downstream molecule Mek5 regulate survival of primordial germ cells. Dev Biol 2012; 372:190-202. [PMID: 23022299 DOI: 10.1016/j.ydbio.2012.09.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 09/12/2012] [Accepted: 09/15/2012] [Indexed: 01/09/2023]
Abstract
In mouse embryos, some primordial germ cells (PGCs) are eliminated by apoptosis, but the molecular pathways that lead to PGC survival versus apoptosis have not been fully characterized. Here, we found that REST (repressor element 1-silencing transcription factor), a transcription factor that binds a conserved regulatory element, NRSE/RE1, played a role in PGC survival. REST expression was higher in PGCs than in surrounding somatic cells. Moreover, in mouse embryos with a PGC-specific conditional REST mutation, the PGC population experienced more apoptosis and was significantly smaller than that in control embryos; these findings indicated that REST functioned in a cell-autonomous fashion that was critical for PGC survival. Several anti-apoptotic genes were among the previously identified REST-target gene candidates; moreover, some of these genes were downregulated in the REST-deficient PGCs. Mek5, which encodes a component in the a MAP kinase cascade, was one of these downregulated REST-target gene candidates, and a Mek5 mutation, like the REST mutation, caused an increase in PGC apoptosis; these finding suggested that REST promoted PGC survival via regulation of the Mek5 expression. Importantly, there were a normal number of PGCs in the REST mutants at birth, and both the male and female REST-mutant adults were fertile; these final observations revealed that the PGC population was very robust and could recover from a genetically induced reduction in cell number.
Collapse
Affiliation(s)
- Daiji Okamura
- Cell Resource Center for Biomedical Research, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|