201
|
Mao S, Li H, Sun Q, Zen K, Zhang CY, Li L. miR-17 regulates the proliferation and differentiation of the neural precursor cells during mouse corticogenesis. FEBS J 2014; 281:1144-58. [PMID: 24314167 DOI: 10.1111/febs.12680] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Revised: 11/08/2013] [Accepted: 12/02/2013] [Indexed: 01/28/2023]
Abstract
MicroRNAs (miRNAs) are endogenously expressed small, non-coding nucleotides that repress gene expression at the post-transcriptional level. In mammals, the developing brain contains a large, diverse group of miRNAs, which suggests that they play crucial roles in neural development. In the present study, we analyzed the miRNA expression patterns in the mouse cortex at various developmental stages. We found that miR-17 family miRNAs were highly expressed in the cortex during early developmental stages, and that their expression levels gradually decreased as the cortex developed. Further investigation revealed that the change in miR-17-5p expression occurred in the ventricular zone/sub-ventricular zone. In addition to promoting cell proliferation, miR-17-5p also influences the differentiation fate of neural precursor cells exposed to bone morphogenetic protein 2. Moreover, we show that these effects of miR-17-5p were mainly the result of regulating the bone morphogenetic protein signaling pathway by repressing expression of the bone morphogenetic protein type II receptor. Taken together, these findings suggest that miR-17 family members play a pivotal role in regulating cell activity during early development of the mouse cortex.
Collapse
Affiliation(s)
- Susu Mao
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University School of Life Sciences, China
| | | | | | | | | | | |
Collapse
|
202
|
The miR-17/106-p38 axis is a key regulator of the neurogenic-to-gliogenic transition in developing neural stem/progenitor cells. Proc Natl Acad Sci U S A 2014; 111:1604-9. [PMID: 24474786 DOI: 10.1073/pnas.1315567111] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Neural stem/progenitor cell (NSPC) multipotency is highly regulated so that specific neural networks form during development. NSPCs cannot respond to gliogenic signals without acquiring gliogenic competence and decreasing their neurogenic competence as development proceeds. Coup-tfI and Coup-tfII are triggers of these temporal NSPC competence changes. However, the downstream effectors of Coup-tfs that mediate the neurogenic-to-gliogenic competence transition remain unknown. Here, we identified the microRNA-17/106 (miR-17/106)-p38 axis as a critical regulator of this transition. Overexpression of miR-17 inhibited the acquisition of gliogenic competence and forced stage-progressed NSPCs to regain neurogenic competence without altering the methylation status of a glial gene promoter. We also identified Mapk14 (also known as p38) as a target of miR-17/106 and found that Mapk14 inhibition restored neurogenic competence after the neurogenic phase. These results demonstrate that the miR-17/106-p38 axis is a key regulator of the neurogenic-to-gliogenic NSPC competence transition and that manipulation of this axis permits bidirectional control of NSPC multipotency.
Collapse
|
203
|
Glycolipid and Glycoprotein Expression During Neural Development. ADVANCES IN NEUROBIOLOGY 2014; 9:185-222. [DOI: 10.1007/978-1-4939-1154-7_9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
204
|
Ding J, Hu H, Li X. SIOMICS: a novel approach for systematic identification of motifs in ChIP-seq data. Nucleic Acids Res 2013; 42:e35. [PMID: 24322294 PMCID: PMC3950686 DOI: 10.1093/nar/gkt1288] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The identification of transcription factor binding motifs is important for the study of gene transcriptional regulation. The chromatin immunoprecipitation (ChIP), followed by massive parallel sequencing (ChIP-seq) experiments, provides an unprecedented opportunity to discover binding motifs. Computational methods have been developed to identify motifs from ChIP-seq data, while at the same time encountering several problems. For example, existing methods are often not scalable to the large number of sequences obtained from ChIP-seq peak regions. Some methods heavily rely on well-annotated motifs even though the number of known motifs is limited. To simplify the problem, de novo motif discovery methods often neglect underrepresented motifs in ChIP-seq peak regions. To address these issues, we developed a novel approach called SIOMICS to de novo discover motifs from ChIP-seq data. Tested on 13 ChIP-seq data sets, SIOMICS identified motifs of many known and new cofactors. Tested on 13 simulated random data sets, SIOMICS discovered no motif in any data set. Compared with two recently developed methods for motif discovery, SIOMICS shows advantages in terms of speed, the number of known cofactor motifs predicted in experimental data sets and the number of false motifs predicted in random data sets. The SIOMICS software is freely available at http://eecs.ucf.edu/∼xiaoman/SIOMICS/SIOMICS.html.
Collapse
Affiliation(s)
- Jun Ding
- Department of Electric Engineering and Computer Science, University of Central Florida, Orlando, FL 32816, USA and Burnett School of Biomedical Science, University of Central Florida, Orlando, FL 32816, USA
| | | | | |
Collapse
|
205
|
Namihira M, Nakashima K. Mechanisms of astrocytogenesis in the mammalian brain. Curr Opin Neurobiol 2013; 23:921-7. [DOI: 10.1016/j.conb.2013.06.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 06/10/2013] [Indexed: 10/26/2022]
|
206
|
Physiological functions of endoplasmic reticulum stress transducer OASIS in central nervous system. Anat Sci Int 2013; 89:11-20. [PMID: 24242870 PMCID: PMC3889286 DOI: 10.1007/s12565-013-0214-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 10/27/2013] [Indexed: 02/02/2023]
Abstract
Eukaryotic cells can adapt to endoplasmic reticulum (ER) dysfunction by producing diverse signals from the ER to the cytosol or nucleus. These signaling pathways are collectively known as the unfolded protein response (UPR). The canonical branches of the UPR are mediated by three ER membrane-bound proteins: double-stranded RNA-dependent protein kinase (PKR)-like endoplasmic reticulum kinase (PERK), inositol-requiring enzyme-1 (IRE1) and activating transcription factor 6 (ATF6). These ER stress transducers basically play important roles in cell survival after ER stress. Recently, novel types of ER stress transducers that share a region of high sequence similarity with ATF6 have been identified. They have a transmembrane domain, which allows them to associate with the ER, and possess a transcription-activation domain and a basic leucine zipper (bZIP) domain. These membrane-bound bZIP transcription factors include OASIS, BBF2H7 CREBH, CREB4 and Luman, and are collectively referred to as OASIS family members. Despite their structural similarities with ATF6, differences in activating stimuli and tissue distribution indicate specialized functions of each member on regulating UPR signaling in specific organs and tissues. One of them, OASIS, is expressed preferentially in astrocytes in the central nervous system (CNS). OASIS temporally regulates the differentiation from neural precursor cells into astrocytes to promote the expression of Glial Cell Missing 1 through dynamic interactions among OASIS family members followed by accelerating demethylation of the Gfap promoter. This review is a summary of our current understanding of the physiological functions of OASIS in the CNS.
Collapse
|
207
|
Bagu ET, Layoun A, Calvé A, Santos MM. Friend of GATA and GATA-6 modulate the transcriptional up-regulation of hepcidin in hepatocytes during inflammation. Biometals 2013; 26:1051-65. [PMID: 24179092 DOI: 10.1007/s10534-013-9683-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 10/19/2013] [Indexed: 01/15/2023]
Abstract
Hepcidin is an antimicrobial peptide hormone that plays a central role in the metabolism of iron and its expression in the liver can be induced through two major pathways: the inflammatory pathway, mainly via IL-6; and the iron-sensing pathway, mediated by BMP-6. GATA-proteins are group of evolutionary conserved transcriptional regulators that bind to the consensus motif-WGATAR-in the promoter region. In hepatoma cells, GATA-proteins 4 and 6 in conjunction with the co-factor friend of GATA (FOG) were shown to modulate the transcription of HAMP. However, it is unclear as to which of the GATA-proteins drive the expression of HAMP in vivo. In this study, using in vitro and in vivo approaches, we investigated the relevance of GATA and FOG proteins in the expression of hepcidin following treatment with IL-6 and BMP-6. We found that treatment of Huh7 cells with either IL-6 or BMP-6 increased the HAMP promoter activity. The HAMP promoter activity following treatment with IL-6 or BMP-6 was further increased by co-transfection of the promoter with GATA proteins 4 and 6. However, co-transfection of the HAMP promoter with FOG proteins 1 or 2 repressed the promoter response to treatments with either IL-6 or BMP-6. The effects of both GATA and FOG proteins on the promoter activity in response to IL-6 or BMP-6 treatment were abrogated by mutation of the GATA response element-TTATCT-in the HAMP promoter region -103/-98. In vivo, treatment of mice with lipopolysaccharide led to a transient increase of Gata-6 expression in the liver that was positively correlated with the expression of hepcidin. Our results indicate that during inflammation GATA-6 is up-regulated in concert with hepcidin while GATA-4 and FOG (1 and 2) are repressed.
Collapse
Affiliation(s)
- Edward T Bagu
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Institut du Cancer de Montréal, (ICM), University of Montreal, Pavillon De Sève Porte Y-5625, 2099 rue Alexandre De Sève, Montreal, QC, H2L 4M1, Canada,
| | | | | | | |
Collapse
|
208
|
Le Dréau G, Martí E. The multiple activities of BMPs during spinal cord development. Cell Mol Life Sci 2013; 70:4293-305. [PMID: 23673983 PMCID: PMC11113619 DOI: 10.1007/s00018-013-1354-9] [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: 09/25/2012] [Revised: 04/25/2013] [Accepted: 04/29/2013] [Indexed: 12/19/2022]
Abstract
Bone morphogenetic proteins (BMPs) are one of the main classes of multi-faceted secreted factors that drive vertebrate development. A growing body of evidence indicates that BMPs contribute to the formation of the central nervous system throughout its development, from the initial shaping of the neural primordium to the generation and maturation of the different cell types that form the functional adult nervous tissue. In this review, we focus on the multiple activities of BMPs during spinal cord development, paying particular attention to recent results that highlight the complexity of BMP signaling during this process. These findings emphasize the unique capacity of these signals to mediate various functions in the same tissue throughout development, recruiting diverse effectors and strategies to instruct their target cells.
Collapse
Affiliation(s)
- Gwenvael Le Dréau
- Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/Baldiri i Reixac 10-15, 08028 Barcelona, Spain
| | - Elisa Martí
- Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/Baldiri i Reixac 10-15, 08028 Barcelona, Spain
| |
Collapse
|
209
|
Abstract
Spinal cord injury results from an insult inflicted on the spinal cord that usually encompasses its 4 major functions (motor, sensory, autonomic, and reflex). The type of deficits resulting from spinal cord injury arise from primary insult, but their long-term severity is due to a multitude of pathophysiological processes during the secondary phase of injury. The failure of the mammalian spinal cord to regenerate and repair is often attributed to the very feature that makes the central nervous system special-it becomes so highly specialized to perform higher functions that it cannot effectively reactivate developmental programs to re-build novel circuitry to restore function after injury. Added to this is an extensive gliotic and immune response that is essential for clearance of cellular debris, but also lays down many obstacles that are detrimental to regeneration. Here, we discuss how the mature chromatin state of different central nervous system cells (neural, glial, and immune) may contribute to secondary pathophysiology, and how restoring silenced developmental gene expression by altering histone acetylation could stall secondary damage and contribute to novel approaches to stimulate endogenous repair.
Collapse
Affiliation(s)
- Elisa M. York
- Department of Zoology (Life Sciences Institute), Brain Research Institute and International Collaboration on Repair Discoveries (iCORD), University of British Columbia, 2350 Health Sciences Blvd, V6T 1Z3 Vancouver, Canada
| | - Audrey Petit
- Department of Zoology (Life Sciences Institute), Brain Research Institute and International Collaboration on Repair Discoveries (iCORD), University of British Columbia, 2350 Health Sciences Blvd, V6T 1Z3 Vancouver, Canada
| | - A. Jane Roskams
- Department of Zoology (Life Sciences Institute), Brain Research Institute and International Collaboration on Repair Discoveries (iCORD), University of British Columbia, 2350 Health Sciences Blvd, V6T 1Z3 Vancouver, Canada
| |
Collapse
|
210
|
Schneider A, Chatterjee S, Bousiges O, Selvi BR, Swaminathan A, Cassel R, Blanc F, Kundu TK, Boutillier AL. Acetyltransferases (HATs) as targets for neurological therapeutics. Neurotherapeutics 2013; 10:568-88. [PMID: 24006237 PMCID: PMC3805875 DOI: 10.1007/s13311-013-0204-7] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The acetylation of histone and non-histone proteins controls a great deal of cellular functions, thereby affecting the entire organism, including the brain. Acetylation modifications are mediated through histone acetyltransferases (HAT) and deacetylases (HDAC), and the balance of these enzymes regulates neuronal homeostasis, maintaining the pre-existing acetyl marks responsible for the global chromatin structure, as well as regulating specific dynamic acetyl marks that respond to changes and facilitate neurons to encode and strengthen long-term events in the brain circuitry (e.g., memory formation). Unfortunately, the dysfunction of these finely-tuned regulations might lead to pathological conditions, and the deregulation of the HAT/HDAC balance has been implicated in neurological disorders. During the last decade, research has focused on HDAC inhibitors that induce a histone hyperacetylated state to compensate acetylation deficits. The use of these inhibitors as a therapeutic option was efficient in several animal models of neurological disorders. The elaboration of new cell-permeant HAT activators opens a new era of research on acetylation regulation. Although pathological animal models have not been tested yet, HAT activator molecules have already proven to be beneficial in ameliorating brain functions associated with learning and memory, and adult neurogenesis in wild-type animals. Thus, HAT activator molecules contribute to an exciting area of research.
Collapse
Affiliation(s)
- Anne Schneider
- />Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR7364, Université de Strasbourg-CNRS, GDR CNRS 2905, Faculté de Psychologie, 12 rue Goethe, 67000 Strasbourg, France
| | - Snehajyoti Chatterjee
- />Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR7364, Université de Strasbourg-CNRS, GDR CNRS 2905, Faculté de Psychologie, 12 rue Goethe, 67000 Strasbourg, France
| | - Olivier Bousiges
- />Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR7364, Université de Strasbourg-CNRS, GDR CNRS 2905, Faculté de Psychologie, 12 rue Goethe, 67000 Strasbourg, France
| | - B. Ruthrotha Selvi
- />Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore 560064 India
| | - Amrutha Swaminathan
- />Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore 560064 India
| | - Raphaelle Cassel
- />Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR7364, Université de Strasbourg-CNRS, GDR CNRS 2905, Faculté de Psychologie, 12 rue Goethe, 67000 Strasbourg, France
| | - Frédéric Blanc
- />Service de Neuropsychologie and CMRR (Centre Mémoire de Ressources et de recherche) Laboratoire ICube, Université de Strasbourg, CNRS, équipe IMIS-Neurocrypto, 1, place de l’Hôpital, 67000 Strasbourg, France
| | - Tapas K. Kundu
- />Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore 560064 India
| | - Anne-Laurence Boutillier
- />Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR7364, Université de Strasbourg-CNRS, GDR CNRS 2905, Faculté de Psychologie, 12 rue Goethe, 67000 Strasbourg, France
| |
Collapse
|
211
|
Gupta K, Chandran S, Hardingham GE. Human stem cell-derived astrocytes and their application to studying Nrf2-mediated neuroprotective pathways and therapeutics in neurodegeneration. Br J Clin Pharmacol 2013; 75:907-18. [PMID: 23126226 PMCID: PMC3612708 DOI: 10.1111/bcp.12022] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 10/28/2012] [Indexed: 02/07/2023] Open
Abstract
Glia, including astrocytes, are increasingly at the forefront of neurodegenerative research for their role in the modulation of neuronal function and survival. Improved understanding of underlying disease mechanisms, including the role of the cellular environment in neurodegeneration, is central to therapeutic development for these currently untreatable diseases. In these endeavours, experimental models that more closely reproduce the human condition have the potential to facilitate the transition between experimental studies in model organisms and patient trials. In this review we discuss the growing role of astrocytes in neurodegenerative diseases, and how astrocytes generated from human pluripotent stem cells represent a useful tool for analyzing astrocytic signalling and influence on neuronal function.
Collapse
Affiliation(s)
- Kunal Gupta
- Anne McLaren Laboratory for Regenerative Medicine & Cambridge Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0SZ, UK
| | | | | |
Collapse
|
212
|
Yamada T, Urano-Tashiro Y, Hashi Y, Sakumoto M, Akiyama H, Tashiro F. The U-box-type ubiquitin ligase PRP19β regulates astrocyte differentiation via ubiquitination of PTP1B. Brain Res 2013; 1524:12-25. [DOI: 10.1016/j.brainres.2013.06.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 06/01/2013] [Accepted: 06/05/2013] [Indexed: 11/15/2022]
|
213
|
Schneider L, Pellegatta S, Favaro R, Pisati F, Roncaglia P, Testa G, Nicolis SK, Finocchiaro G, d'Adda di Fagagna F. DNA damage in mammalian neural stem cells leads to astrocytic differentiation mediated by BMP2 signaling through JAK-STAT. Stem Cell Reports 2013; 1:123-38. [PMID: 24052948 PMCID: PMC3757751 DOI: 10.1016/j.stemcr.2013.06.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 06/11/2013] [Accepted: 06/12/2013] [Indexed: 01/17/2023] Open
Abstract
The consequences of DNA damage generation in mammalian somatic stem cells, including neural stem cells (NSCs), are poorly understood despite their potential relevance for tissue homeostasis. Here, we show that, following ionizing radiation-induced DNA damage, NSCs enter irreversible proliferative arrest with features of cellular senescence. This is characterized by increased cytokine secretion, loss of stem cell markers, and astrocytic differentiation. We demonstrate that BMP2 is necessary to induce expression of the astrocyte marker GFAP in irradiated NSCs via a noncanonical signaling pathway engaging JAK-STAT. This is promoted by ATM and antagonized by p53. Using a SOX2-Cre reporter mouse model for cell-lineage tracing, we demonstrate irradiation-induced NSC differentiation in vivo. Furthermore, glioblastoma assays reveal that irradiation therapy affects the tumorigenic potential of cancer stem cells by ablating self-renewal and inducing astroglial differentiation.
Collapse
Affiliation(s)
- Leonid Schneider
- IFOM Foundation-The FIRC Institute of Molecular Oncology Foundation, Via Adamello 16, 20139 Milan, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
214
|
Itoh T, Satou T, Dote K, Hashimoto S, Ito H. Effect of basic fibroblast growth factor on cultured rat neural stem cell in three-dimensional collagen gel. Neurol Res 2013; 27:429-32. [PMID: 15949242 DOI: 10.1179/016164105x18476] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES The effect of growth factors on the three-dimensional culture of neural stem cells has not been reported. We studied the effect of basic fibroblast growth factor (bFGF) on cultured rat neural stem cells in a three-dimensional culture. METHODS We cultured rat neural stem cells in collagen gel matrix for three-dimensional culture and examined the effect of bFGF under such culture conditions. RESULTS After 4 days culture, the cell density in the bFGF treatment group was 12 times that of the non-treatment group, reaching a significantly high value. In the bFGF treatment group, microtubule associate protein (MAP)-2-positive cell aggregation occurred, although in the bFGF non-treatment group there was no MAP-2-positive cell aggregation and few of the cells were sparsely distributed. Also, in the bFGF treatment group, MAP-2-positive cell aggregation had a luminal structure similar to neural rosettes. There was elongation of MAP-2-positive neurites from the cell aggregation to the circumference in the bFGF treatment group. DISCUSSION bFGF is known to induce the proliferation, but not the differentiation of neural stem cells in two-dimensional cultures. However, in the three-dimensional culture, bFGF induced both the proliferation and differentiation of neural stem cells. The three-dimensional culture is, therefore, considered a useful method for predicting the response of neural stem cells to cytokines or biologically active substances in vivo.
Collapse
Affiliation(s)
- Tatsuki Itoh
- Department of Pathology, Kinki University School of Medicine, Osaka, Japan.
| | | | | | | | | |
Collapse
|
215
|
Gámez B, Rodriguez-Carballo E, Ventura F. BMP signaling in telencephalic neural cell specification and maturation. Front Cell Neurosci 2013; 7:87. [PMID: 23761735 PMCID: PMC3671186 DOI: 10.3389/fncel.2013.00087] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 05/21/2013] [Indexed: 12/13/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) make up a family of morphogens that are critical for patterning, development, and function of the central and peripheral nervous system. Their effects on neural cells are pleiotropic and highly dynamic depending on the stage of development and the local niche. Neural cells display a broad expression profile of BMP ligands, receptors, and transducer molecules. Moreover, interactions of BMP signaling with other incoming morphogens and signaling pathways are crucial for most of these processes. The key role of BMP signaling suggests that it includes many regulatory mechanisms that restrict BMP activity both temporally and spatially. BMPs affect neural cell fate specification in a dynamic fashion. Initially they inhibit proliferation of neural precursors and promote the first steps in neuronal differentiation. Later on, BMP signaling effects switch from neuronal induction to promotion of astroglial identity and inhibition of neuronal or oligodendroglial lineage commitment. Furthermore, in postmitotic cells, BMPs regulate cell survival and death, to modulate neuronal subtype specification, promote dendritic and axonal growth and induce synapse formation and stabilization. In this review, we examine the canonical and non-canonical mechanisms of BMP signal transduction. Moreover, we focus on the specific role of BMPs in the nervous system including their ability to regulate neural stem cell proliferation, self-renewal, lineage specification, and neuronal function.
Collapse
Affiliation(s)
- Beatriz Gámez
- Departament de Ciències Fisiològiques II, Institut d'Investigació Biomèdica de Bellvitge, Universitat de Barcelona, L'Hospitalet de Llobregat Spain
| | | | | |
Collapse
|
216
|
Zouein FA, Kurdi M, Booz GW. Dancing rhinos in stilettos: The amazing saga of the genomic and nongenomic actions of STAT3 in the heart. JAKSTAT 2013; 2:e24352. [PMID: 24069556 PMCID: PMC3772108 DOI: 10.4161/jkst.24352] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 03/18/2013] [Accepted: 03/18/2013] [Indexed: 01/15/2023] Open
Abstract
A substantial body of evidence has shown that signal transducer and activator of transcription 3 (STAT3) has an important role in the heart in protecting the myocardium from ischemia and oxidative stress. These actions are attributed to STAT3 functioning as a transcription factor in upregulating cardioprotective genes. Loss of STAT3 has been implicated as well in the pathogenesis of heart failure and, in that context and in addition to the loss of a cardioprotective gene program, nuclear STAT3 has been identified as a transcriptional repressor important for the normal functioning of the ubiquitin-proteasome system for protein degradation. The later finding establishes a genomic role for STAT3 in controlling cellular homeostasis in cardiac myocytes independent of stress. Surprisingly, although a well-studied area, very few downstream gene targets of STAT3 in the heart have been definitively identified. In addition, STAT3 is now known to induce gene expression by noncanonical means that are not well characterized in the heart. On the other hand, recent evidence has shown that STAT3 has important nongenomic actions in cardiac myocytes that affect microtubule stability, mitochondrial respiration, and autophagy. These extranuclear actions of STAT3 involve protein–protein interactions that are incompletely understood, as is their regulation in both the healthy and injured heart. Moreover, how the diverse genomic and nongenomic actions of STAT3 crosstalk with each other is unchartered territory. Here we present an overview of what is and is not known about both the genomic and nongenomic actions of STAT3 in the heart from a structure-function perspective that focuses on the impact of posttranslational modifications and oxidative stress in regulating the actions and interactions of STAT3. Even though we have learnt a great deal about the role played by STAT3 in the heart, much more awaits to be discovered.
Collapse
Affiliation(s)
- Fouad A Zouein
- Department of Pharmacology and Toxicology; School of Medicine; and The Jackson Center for Heart Research at UMMC; The Cardiovascular-Renal Research Center; The University of Mississippi Medical Center; Jackson, MS USA
| | | | | |
Collapse
|
217
|
Senut MC, Cingolani P, Sen A, Kruger A, Shaik A, Hirsch H, Suhr ST, Ruden D. Epigenetics of early-life lead exposure and effects on brain development. Epigenomics 2013; 4:665-74. [PMID: 23244311 DOI: 10.2217/epi.12.58] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The epigenetic machinery plays a pivotal role in the control of many of the body's key cellular functions. It modulates an array of pliable mechanisms that are readily and durably modified by intracellular or extracellular factors. In the fast-moving field of neuroepigenetics, it is emerging that faulty epigenetic gene regulation can have dramatic consequences on the developing CNS that can last a lifetime and perhaps even affect future generations. Mounting evidence suggests that environmental factors can impact the developing brain through these epigenetic mechanisms and this report reviews and examines the epigenetic effects of one of the most common neurotoxic pollutants of our environment, which is believed to have no safe level of exposure during human development: lead.
Collapse
Affiliation(s)
- Marie-Claude Senut
- Institute of Environmental Health Sciences, CS Mott Center for Human Health & Development & Department of Obstetrics & Gynecology, Wayne State University, Detroit, MI 48201, USA
| | | | | | | | | | | | | | | |
Collapse
|
218
|
van Heesbeen HJ, Mesman S, Veenvliet JV, Smidt MP. Epigenetic mechanisms in the development and maintenance of dopaminergic neurons. Development 2013; 140:1159-69. [PMID: 23444349 DOI: 10.1242/dev.089359] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Mesodiencephalic dopaminergic (mdDA) neurons are located in the ventral mesodiencephalon and are involved in psychiatric disorders and severely affected in neurodegenerative diseases such as Parkinson's disease. mdDA neuronal development has received much attention in the last 15 years and many transcription factors involved in mdDA specification have been discovered. More recently however, the impact of epigenetic regulation has come into focus, and it's emerging that the processes of histone modification and DNA methylation form the basis of genetic switches that operate during mdDA development. Here, we review the epigenetic control of mdDA development, maturation and maintenance. As we highlight, epigenetic mechanisms play a pivotal role in all of these processes and the knowledge gathered from studying epigenetics in these contexts may aid our understanding of mdDA-related pathologies.
Collapse
Affiliation(s)
- Hendrikus J van Heesbeen
- Swammerdam Institute for Life Sciences, Science Park, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
219
|
Savary K, Caglayan D, Caja L, Tzavlaki K, Bin Nayeem S, Bergström T, Jiang Y, Uhrbom L, Forsberg-Nilsson K, Westermark B, Heldin CH, Ferletta M, Moustakas A. Snail depletes the tumorigenic potential of glioblastoma. Oncogene 2013; 32:5409-20. [PMID: 23524585 PMCID: PMC3898470 DOI: 10.1038/onc.2013.67] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 12/02/2012] [Accepted: 01/05/2013] [Indexed: 12/26/2022]
Abstract
Glioblastoma multiforme (GBM) is an aggressive brain malignancy characterized by high heterogeneity and invasiveness. It is increasingly accepted that the refractory feature of GBM to current therapies stems from the existence of few tumorigenic cells that sustain tumor growth and spreading, the so-called glioma-initiating cells (GICs). Previous studies showed that cytokines of the bone morphogenetic protein (BMP) family induce differentiation of the GICs, and thus act as tumor suppressors. Molecular pathways that explain this behavior of BMP cytokines remain largely elusive. Here, we show that BMP signaling induces Smad-dependent expression of the transcriptional regulator Snail in a rapid and sustained manner. Consistent with its already established promigratory function in other cell types, we report that Snail silencing decreases GBM cell migration. Consequently, overexpression of Snail increases GBM invasiveness in a mouse xenograft model. Surprisingly, we found that Snail depletes the GBM capacity to form gliomaspheres in vitro and to grow tumors in vivo, both of which are important features shared by GICs. Thus Snail, acting downstream of BMP signaling, dissociates the invasive capacity of GBM cells from their tumorigenic potential.
Collapse
Affiliation(s)
- K Savary
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Biomedical Center, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
220
|
Urayama S, Semi K, Sanosaka T, Hori Y, Namihira M, Kohyama J, Takizawa T, Nakashima K. Chromatin accessibility at a STAT3 target site is altered prior to astrocyte differentiation. Cell Struct Funct 2013; 38:55-66. [PMID: 23439558 DOI: 10.1247/csf.12034] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
DNA demethylation of astrocyte-specific gene promoters and STAT3 activation in neural precursor cells (NPCs) are essential for astrogliogenesis in the developing brain. To date, it remains unclear whether DNA methylation is the sole epigenetic determinant responsible for suppressing astrocyte-specific genes. Here, we used mouse embryonic stem cells (TKO ESCs) that lacked all 3 DNA methyltransferase genes, Dnmt1, Dnmt3a, and Dnmt3b, and thereby exhibit complete demethylation of the astrocyte-specific glial fibrillary acidic protein (Gfap) gene promoter. We found that although the Gfap promoter was demethylated, STAT3 failed to bind to its cognate element to induce Gfap transcription, whereas it induced transcription of a different target gene, Socs3. Moreover, although the Gfap promoter region containing the STAT3-binding site (GSBS) is enriched with transcription-repressive histone modifications, such as methylation of H3 at lysine 9 (H3K9me3) and H3K27me3, the reduction of these modifications in TKO ESCs was not sufficient for binding of STAT3 at GSBS. Furthermore, GSBS was digested by micrococcal nuclease in late-gestational NPCs that express GFAP upon LIF stimulation, but not in cells that show no expression of GFAP even in the presence of LIF, indicating that STAT3 can access GSBS in the former cells. We further showed that expression of NF-1A, which is known to potentiate differentiation of mid-gestational NPCs into astrocytes, increased its accessibility. Taken together, our results suggest that chromatin accessibility of GSBS plays a critical role in the regulation of Gfap expression.
Collapse
Affiliation(s)
- Satoshi Urayama
- Laboratory of Molecular Neuroscience, Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara, Japan
| | | | | | | | | | | | | | | |
Collapse
|
221
|
Gallicano GI. Modeling to optimize terminal stem cell differentiation. SCIENTIFICA 2013; 2013:574354. [PMID: 24278782 PMCID: PMC3820305 DOI: 10.1155/2013/574354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 12/18/2012] [Indexed: 06/02/2023]
Abstract
Embryonic stem cell (ESC), iPCs, and adult stem cells (ASCs) all are among the most promising potential treatments for heart failure, spinal cord injury, neurodegenerative diseases, and diabetes. However, considerable uncertainty in the production of ESC-derived terminally differentiated cell types has limited the efficiency of their development. To address this uncertainty, we and other investigators have begun to employ a comprehensive statistical model of ESC differentiation for determining the role of intracellular pathways (e.g., STAT3) in ESC differentiation and determination of germ layer fate. The approach discussed here applies the Baysian statistical model to cell/developmental biology combining traditional flow cytometry methodology and specific morphological observations with advanced statistical and probabilistic modeling and experimental design. The final result of this study is a unique tool and model that enhances the understanding of how and when specific cell fates are determined during differentiation. This model provides a guideline for increasing the production efficiency of therapeutically viable ESCs/iPSCs/ASC derived neurons or any other cell type and will eventually lead to advances in stem cell therapy.
Collapse
Affiliation(s)
- G. Ian Gallicano
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
222
|
Sakata H, Narasimhan P, Niizuma K, Maier CM, Wakai T, Chan PH. Interleukin 6-preconditioned neural stem cells reduce ischaemic injury in stroke mice. ACTA ACUST UNITED AC 2013; 135:3298-310. [PMID: 23169920 DOI: 10.1093/brain/aws259] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Transplantation of neural stem cells provides a promising therapy for stroke. Its efficacy, however, might be limited because of massive grafted-cell death after transplantation, and its insufficient capability for tissue repair. Interleukin 6 is a pro-inflammatory cytokine involved in the pathogenesis of various neurological disorders. Paradoxically, interleukin 6 promotes a pro-survival signalling pathway through activation of signal transducer and activator of transcription 3. In this study, we investigated whether cellular reprogramming of neural stem cells with interleukin 6 facilitates the effectiveness of cell transplantation therapy in ischaemic stroke. Neural stem cells harvested from the subventricular zone of foetal mice were preconditioned with interleukin 6 in vitro and transplanted into mouse brains 6 h or 7 days after transient middle cerebral artery occlusion. Interleukin 6 preconditioning protected the grafted neural stem cells from ischaemic reperfusion injury through signal transducer and activator of transcription 3-mediated upregulation of manganese superoxide dismutase, a primary mitochondrial antioxidant enzyme. In addition, interleukin 6 preconditioning induced secretion of vascular endothelial growth factor from the neural stem cells through activation of signal transducer and activator of transcription 3, resulting in promotion of angiogenesis in the ischaemic brain. Furthermore, transplantation of interleukin 6-preconditioned neural stem cells significantly attenuated infarct size and improved neurological performance compared with non-preconditioned neural stem cells. This interleukin 6-induced amelioration of ischaemic insults was abolished by transfecting the neural stem cells with signal transducer and activator of transcription 3 small interfering RNA before transplantation. These results indicate that preconditioning with interleukin 6, which reprograms neural stem cells to tolerate oxidative stress after ischaemic reperfusion injury and to induce angiogenesis through activation of signal transducer and activator of transcription 3, is a simple and beneficial approach for enhancing the effectiveness of cell transplantation therapy in ischaemic stroke.
Collapse
Affiliation(s)
- Hiroyuki Sakata
- Neurosurgical Laboratories, Stanford University, Stanford, CA 94305-5487, USA
| | | | | | | | | | | |
Collapse
|
223
|
Lilja T, Heldring N, Hermanson O. Like a rolling histone: Epigenetic regulation of neural stem cells and brain development by factors controlling histone acetylation and methylation. Biochim Biophys Acta Gen Subj 2013; 1830:2354-60. [DOI: 10.1016/j.bbagen.2012.08.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 07/12/2012] [Accepted: 08/07/2012] [Indexed: 01/24/2023]
|
224
|
Liu H, Jia D, Li A, Chau J, He D, Ruan X, Liu F, Li J, He L, Li B. p53 regulates neural stem cell proliferation and differentiation via BMP-Smad1 signaling and Id1. Stem Cells Dev 2013. [PMID: 23199293 DOI: 10.1089/scd.2012.0370] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Neural stem cells (NSCs) play essential roles in nervous system development and postnatal neuroregeneration and their deregulation underlies the development of neurodegenerative disorders. Yet how NSC proliferation and differentiation are controlled is not fully understood. Here we present evidence that tumor suppressor p53 regulates NSC proliferation and differentiation via the bone morphogenetic proteins (BMP)-Smad1 pathway and its target gene inhibitor of DNA binding 1 (Id1). p53 deficiency led to increased neurogenesis in vivo, and biased neuronal differentiation and augmented NSC proliferation of ex vivo NSCs. This is accompanied by elevated Smad1 expression/activation in the brain and NSC, which contributes to accelerated neuronal differentiation of p53(-/-) NSCs. p53 deficiency also leads to upregulation of Id1, whose expression is repressed by p53 in BMP-Smad1-dependent and -independent manners. Elevated Id1 expression contributes to augmented proliferation and, unexpectedly, accelerated neuronal differentiation of p53(-/-) NSCs as well. This study reveals a molecular mechanism by which tumor suppressor p53 controls NSC proliferation and differentiation and establishes a connection between p53 and Id1.
Collapse
Affiliation(s)
- Huijuan Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
225
|
Transcriptional Regulation and Specification of Neural Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 786:129-55. [DOI: 10.1007/978-94-007-6621-1_8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
226
|
Stine RR, Matunis EL. JAK-STAT signaling in stem cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 786:247-67. [PMID: 23696361 DOI: 10.1007/978-94-007-6621-1_14] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adult stem cells are essential for the regeneration and repair of tissues in an organism. Signals from many different pathways converge to regulate stem cell maintenance and differentiation while preventing overproliferation. Although each population of adult stem cells is unique, common themes arise by comparing the regulation of various stem cell types in an organism or by comparing similar stem cell types across species. The JAK-STAT signaling pathway, identified nearly two decades ago, is now known to be involved in many biological processes including the regulation of stem cells. Studies in Drosophila first implicated JAK-STAT signaling in the control of stem cell maintenance in the male germline stem cell microenvironment, or niche; subsequently it has been shown play a role in other niches in both Drosophila and mammals. In this chapter, we will address the role of JAK-STAT signaling in stem cells in the germline, intestinal, hematopoietic and neuronal niches in Drosophila as well as the hematopoietic and neuronal niches in mammals. We will comment on how the study of JAK-STAT signaling in invertebrate systems has helped to advance our understanding of signaling in vertebrates. In addition to the role of JAK- STAT signaling in stem cell niche homeostasis, we will also discuss the diseases, including cancers, that can arise when this pathway is misregulated.
Collapse
Affiliation(s)
- Rachel R Stine
- Department of Cell Biology, Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD, 21205 USA
| | | |
Collapse
|
227
|
Functions of BMP signaling in embryonic stem cell fate determination. Exp Cell Res 2013; 319:113-9. [DOI: 10.1016/j.yexcr.2012.09.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 09/30/2012] [Indexed: 01/08/2023]
|
228
|
Zhan R, Yamamoto M, Ueki T, Yoshioka N, Tanaka K, Morisaki H, Seiwa C, Yamamoto Y, Kawano H, Tsuruo Y, Watanabe K, Asou H, Aiso S. A DEAD-box RNA helicase Ddx54 protein in oligodendrocytes is indispensable for myelination in the central nervous system. J Neurosci Res 2012; 91:335-48. [PMID: 23239230 DOI: 10.1002/jnr.23162] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2012] [Revised: 09/22/2012] [Accepted: 10/02/2012] [Indexed: 02/05/2023]
Abstract
We recently reported that a new monoclonal antibody, 4F2, which labels oligodendroglial lineage cells, recognizes a DEAD-box RNA helicase Ddx54 and that Ddx54 binds to myelin basic protein (MBP) in brain and cultured oligodendrocytes. To elucidate the biological function of Ddx54, we generated a recombinant adenovirus, Ad-shRNA:Ddx54, expressing a short hairpin RNA to silence endogenous Ddx54 protein. The virus was intraventricularly injected into the brains of mice on postnatal day (PD) 2. The brains at PD 9 were then analyzed by immunohistochemistry. In untreated normal brain sections, as well as control brains that had been injected with Ad-β-Gal, myelination of axons occurred in the corpus callosum with filamentous patterns of immunosignals of myelin-associated glycoprotein (MAG) and MBP. In Ad-shRNA:Ddx54-injected brain, substantial amounts of MAG and MBP immunosignals were present, but MBP immunosignals accumulated in the subplate layer and did not intrude into the emerging white matter. Immunoblot analysis revealed that Ddx54 knockdown caused a significant decrease in the level of 21.5 kDa MBP isoform and Ddx54, but the amount of Olig2; 2',3'-cyclic nucleotide 3' phosphodiesterase; MAG; three MBP isoforms (14, 17.5, and 18 kDa); and QKI-5, QKI-6, and QKI-7 proteins remained unchanged. Transfection of the Ddx54 expression vector into luciferase reporter-introduced neuroepithelial cells resulted in upregulated MBP promoter activity. Immunoprecipitation of Ddx54 protein in MBP-transfected HEK293 cells indicated that Ddx54 may directly interact with MBP mRNA. These results suggest that Ddx54 protein play an important role in central nervous system myelination, presumably in myelin sheath formation after the differentiation of oligodendrocytes.
Collapse
Affiliation(s)
- Rui Zhan
- Center for Kampo Medicine, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
229
|
Abstract
We have defined functions of MEK in regulating gliogenesis in developing cerebral cortex using loss- and gain-of-function mouse genetics. Radial progenitors deficient in both Mek1 and Mek2 fail to transition to the gliogenic mode in late embryogenesis, and astrocyte and oligodendroglial precursors fail to appear. In exploring mechanisms, we found that the key cytokine-regulated gliogenic pathway is attenuated. Further, the Ets transcription family member Etv5/Erm is strongly regulated by MEK and Erm overexpression can rescue the gliogenic potential of Mek-deleted progenitors. Remarkably, Mek1/2-deleted mice surviving postnatally exhibit cortices almost devoid of astrocytes and oligodendroglia and exhibit neurodegeneration. Conversely, expression of constitutively active MEK1 leads to a major increase in numbers of astrocytes in the adult brain. We conclude that MEK is essential for acquisition of gliogenic competence by radial progenitors and that levels of MEK activity regulate gliogenesis in the developing cortex.
Collapse
|
230
|
Tan SL, Nishi M, Ohtsuka T, Matsui T, Takemoto K, Kamio-Miura A, Aburatani H, Shinkai Y, Kageyama R. Essential roles of the histone methyltransferase ESET in the epigenetic control of neural progenitor cells during development. Development 2012; 139:3806-16. [PMID: 22991445 DOI: 10.1242/dev.082198] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In the developing brain, neural progenitor cells switch differentiation competency by changing gene expression profiles that are governed partly by epigenetic control, such as histone modification, although the precise mechanism is unknown. Here we found that ESET (Setdb1), a histone H3 Lys9 (H3K9) methyltransferase, is highly expressed at early stages of mouse brain development but downregulated over time, and that ablation of ESET leads to decreased H3K9 trimethylation and the misregulation of genes, resulting in severe brain defects and early lethality. In the mutant brain, endogenous retrotransposons were derepressed and non-neural gene expression was activated. Furthermore, early neurogenesis was severely impaired, whereas astrocyte formation was enhanced. We conclude that there is an epigenetic role of ESET in the temporal and tissue-specific gene expression that results in proper control of brain development.
Collapse
Affiliation(s)
- Siok-Lay Tan
- Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
231
|
Selective STAT3-α or -β expression reveals spliceform-specific phosphorylation kinetics, nuclear retention and distinct gene expression outcomes. Biochem J 2012; 447:125-36. [PMID: 22799634 PMCID: PMC3441131 DOI: 10.1042/bj20120941] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Phosphorylation of STAT3 (signal transducer and activator of transcription 3) is critical for its nuclear import and transcriptional activity. Although a shorter STAT3β spliceform was initially described as a negative regulator of STAT3α, gene knockout studies have revealed that both forms play critical roles. We have expressed STAT3α and STAT3β at comparable levels to facilitate a direct comparison of their functional effects, and have shown their different cytokine-stimulated kinetics of phosphorylation and nuclear translocation. Notably, the sustained nuclear translocation and phosphorylation of STAT3β following cytokine exposure contrasted with a transient nuclear translocation and phosphorylation of STAT3α. Importantly, co-expression of the spliceforms revealed that STAT3β enhanced and prolonged the phosphorylation and nuclear retention of STAT3α, but a STAT3β R609L mutant, with a disrupted SH2 (Src homology 2) domain, was not tyrosine phosphorylated following cytokine stimulation and could not cross-regulate STAT3α. The physiological importance of prolonged phosphorylation and nuclear retention was indicated by transcriptome profiling of STAT3−/− cells expressing either STAT3α or STAT3β, revealing the complexity of genes that are up- and down-regulated by the STAT3 spliceforms, including a distinct set of STAT3β-specific genes regulated under basal conditions and after cytokine stimulation. These results highlight STAT3β as a significant transcriptional regulator in its own right, with additional actions to cross-regulate STAT3α phosphorylation and nuclear retention after cytokine stimulation.
Collapse
|
232
|
Yokogami K, Yamashita S, Takeshima H. Hypoxia-induced decreases in SOCS3 increase STAT3 activation and upregulate VEGF gene expression. Brain Tumor Pathol 2012; 30:135-43. [PMID: 23104276 DOI: 10.1007/s10014-012-0122-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 10/18/2012] [Indexed: 01/09/2023]
Abstract
Signal transducer and activator of transcription (STAT) 3 has been described as a master regulator of the signaling pathways that are involved in the mesenchymal transition of glioblastomas, which are the most aggressive type of tumors and which result in poor prognoses. Hypoxia, which is a strong inducer of vascular endothelial growth factor (VEGF), results in angiogenesis and the bulky growth of tumors. Here, we show that hypoxia induces VEGF gene expression through a STAT3 signaling cascade. Hypoxia increases the levels of aberrantly activated STAT3 by decreasing the levels of the suppressor of cytokine signaling (SOCS) 3, which is a negative regulator of the STAT3 signaling cascade. Activated STAT3 binds to the hypoxia-responsible element that is located -914 to -905 bp upstream of the transcription initiation site in the VEGF promoter and that transcriptionally regulates VEGF gene expression. This sequence closely resembled the previously defined sis-inducible element in the STAT3-binding sequences. The enforced overexpression of SOCS3 abolished the hypoxia-induced STAT3 activation and the STAT3-mediated transcriptional upregulation of the VEGF gene. In addition, activated STAT3 was found around necrotic foci in surgical specimens. These observations suggest that STAT3 is a molecular target of antiangiogenesis.
Collapse
Affiliation(s)
- Kiyotaka Yokogami
- Department of Neurosurgery, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan.
| | | | | |
Collapse
|
233
|
Meindl-Beinker NM, Matsuzaki K, Dooley S. TGF-β signaling in onset and progression of hepatocellular carcinoma. Dig Dis 2012; 30:514-23. [PMID: 23108308 DOI: 10.1159/000341704] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Transforming growth factor (TGF)-β is a central regulator in chronic liver disease, contributing to all stages of disease progression from initial liver injury through inflammation and fibrosis to cirrhosis and hepatocellular carcinoma. Liver damage-induced levels of active TGF-β enhance hepatocyte destruction and mediate hepatic stellate cell and fibroblast activation resulting in a wound-healing response, including myofibroblast generation and extracellular matrix deposition. Further evidence points to a decisive role of cytostatic and apoptotic functions mediated on hepatocytes, which is critical for the control of liver mass, with loss of TGF-β activities resulting in hyperproliferative disorders and cancer. This concept is based on studies that describe a bipartite role of TGF-β with tumor suppressor functions at early stages of liver damage and regeneration, whereas during cancer progression TGF-β may turn from a tumor suppressor into a tumor promoter that exacerbates invasive and metastatic behavior. We have delineated this molecular switch of the pathway from cytostatic to tumor promoting in further detail and identify activation of survival signaling pathways in hepatocytes as a most critical requirement. Targeting the TGF-β signaling pathway has been explored to inhibit liver disease progression. While interfering with TGF-β signaling in various short-term animal models has demonstrated promising results, liver disease progression in humans is a process of decades with different phases in which TGF-β or its targeting may have both beneficial and adverse outcomes. We emphasize that, in order to achieve therapeutic effects, targeting TGF-β signaling in the right cell type at the right time is required.
Collapse
Affiliation(s)
- Nadja M Meindl-Beinker
- Molecular Hepatology - Alcohol-Associated Diseases, Medical Clinic, Medical Faculty Mannheim of Heidelberg University, Germany
| | | | | |
Collapse
|
234
|
Bond AM, Bhalala OG, Kessler JA. The dynamic role of bone morphogenetic proteins in neural stem cell fate and maturation. Dev Neurobiol 2012; 72:1068-84. [PMID: 22489086 DOI: 10.1002/dneu.22022] [Citation(s) in RCA: 178] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The bone morphogenetic proteins (BMPs) are a group of powerful morphogens that are critical for development of the nervous system. The effects of BMP signaling on neural stem cells are myriad and dynamic, changing with each stage of development. During early development inhibition of BMP signaling differentiates neuroectoderm from ectoderm, and BMP signaling helps to specify neural crest. Thus modulation of BMP signaling underlies formation of both the central and peripheral nervous systems. BMPs secreted from dorsal structures then form a gradient which helps pattern the dorsal-ventral axis of the developing spinal cord and brain. During forebrain development BMPs sequentially induce neurogenesis and then astrogliogenesis and participate in neurite outgrowth from immature neurons. BMP signaling also plays a critical role in maintaining adult neural stem cell niches in the subventricular zone (SVZ) and subgranular zone (SGZ). BMPs are able to exert such diverse effects through closely regulated temporospatial expression and interaction with other signaling pathways.
Collapse
Affiliation(s)
- Allison M Bond
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | | | | |
Collapse
|
235
|
Martynoga B, Drechsel D, Guillemot F. Molecular control of neurogenesis: a view from the mammalian cerebral cortex. Cold Spring Harb Perspect Biol 2012; 4:4/10/a008359. [PMID: 23028117 DOI: 10.1101/cshperspect.a008359] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The mammalian nervous system is the most complex organ of any living organism. How this complexity is generated during neural development is just beginning to be elucidated. This article discusses the signaling, transcriptional, and epigenetic mechanisms that are involved in neural development. The first part focuses on molecules that control neuronal numbers through regulation of the timing of onset of neurogenesis, the timing of the neuronal-to-glial switch, and the rate of progenitor proliferation. The second part focuses on molecules that control neuronal diversity by generating spatially or temporally distinct populations of neuronal progenitors. Most of the studies discussed in this article are focused on the developing mammalian cerebral cortex, because this is one of the main model systems for neural developmental studies and many of the mechanisms identified in this tissue also operate elsewhere in the developing brain and spinal cord.
Collapse
Affiliation(s)
- Ben Martynoga
- Division of Molecular Neurobiology, National Institute for Medical Research, Mill Hill, London NW71AA, United Kingdom
| | | | | |
Collapse
|
236
|
Bench to bedside of neural stem cell in traumatic brain injury. Stem Cells Int 2012; 2012:141624. [PMID: 23028389 PMCID: PMC3458287 DOI: 10.1155/2012/141624] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 08/10/2012] [Accepted: 08/21/2012] [Indexed: 11/18/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the leading causes of major disability and death worldwide. Neural stem cells (NSCs) have recently been shown to contribute to the cellular remodelling that occurs following TBI and attention has been drawn to the area of neural stem cell as possible therapy for TBI. The NSCs may play an important role in the treatment of TBI by replacing the damaged cells and eventual remyelination. This paper summarized a critical assessment of recent data and developed a view comprising of six points to possible quality translation of NSCs in TBI.
Collapse
|
237
|
Role of Kruppel-like factor 4 in neurogenesis and radial neuronal migration in the developing cerebral cortex. Mol Cell Biol 2012; 32:4297-305. [PMID: 22907754 DOI: 10.1128/mcb.00838-12] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Transcription factors play key roles in the formation of a multilayered cerebral cortex consisting of neurons and glial cells. Krüppel-like factor 4 (KLF4) is expressed in neural stem cells and controls axonal regeneration. Its dysregulation leads to hydrocephalus in postnatal mouse brains. Here, we further show that KLF4 regulates neurogenesis and radial migration of neurons in the developing cerebral cortex. Neural progenitors with constitutive expression of KLF4 fail to migrate and develop into mature neurons but, rather, form cells with a glial identity. Notably, the JAK-STAT pathway is altered by KLF4, with increased phosphorylation of STAT3 at tyrosine 705. Blocking STAT3 activation with a dominant negative form can rescue the migration defect induced by constitutive KLF4 expression. Furthermore, downregulation of endogenous KLF4 significantly promotes radial migration and the transition of newly born migrating neurons from multipolar to bipolar morphology. Together, these results suggest that precise regulation of KLF4 expression is critical to neuronal differentiation and migration during the formation of a cerebral cortex.
Collapse
|
238
|
A critical cell-intrinsic role for serum response factor in glial specification in the CNS. J Neurosci 2012; 32:8012-23. [PMID: 22674276 DOI: 10.1523/jneurosci.5633-11.2012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Astrocytes and oligodendrocytes play crucial roles in nearly every facet of nervous system development and function, including neuronal migration, synaptogenesis, synaptic plasticity, and myelination. Previous studies have widely characterized the signaling pathways important for astrocyte differentiation and unveiled a number of transcription factors that guide oligodendrocyte differentiation in the CNS. However, the identities of the transcription factors critical for astrocyte specification in the brain remain unknown. Here we show that deletion of the stimulus-dependent transcription factor, serum response factor (SRF), in neural precursor cells (NPCs) (Srf-Nestin-cKO) results in nearly 60% loss in astrocytes and 50% loss in oligodendrocyte precursors at birth. Cultured SRF-deficient NPCs exhibited normal growth rate and capacity to self-renew. However, SRF-deficient NPCs generated fewer astrocytes and oligodendrocytes in response to several lineage-specific differentiation factors. These deficits in glial differentiation were rescued by ectopic expression of wild-type SRF in SRF-deficient NPCs. Interestingly, ectopic expression of a constitutively active SRF (SRF-VP16) in NPCs augmented astrocyte differentiation in the presence of pro-astrocytic factors. However, SRF-VP16 expression in NPCs had an inhibitory effect on oligodendrocyte differentiation. In contrast, mice carrying conditional deletion of SRF in developing forebrain neurons (Srf-NEX-cKO) did not exhibit any deficits in astrocytes in the brain. Together, our observations suggest that SRF plays a critical cell-autonomous role in NPCs to regulate astrocyte and oligodendrocyte specification in vivo and in vitro.
Collapse
|
239
|
Fish RJ, Neerman-Arbez M. Fibrinogen gene regulation. Thromb Haemost 2012; 108:419-26. [PMID: 22836683 DOI: 10.1160/th12-04-0273] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 07/11/2012] [Indexed: 01/08/2023]
Abstract
The Aα, Bβ and γ polypeptide chains of fibrinogen are encoded by a three gene cluster on human chromosome four. The fibrinogen genes (FGB-FGA-FGG) are expressed almost exclusively in hepatocytes where their output is coordinated to ensure a sufficient mRNA pool for each chain and maintain an abundant plasma fibrinogen protein level. Fibrinogen gene expression is controlled by the activity of proximal promoters which contain binding sites for hepatocyte transcription factors, including proteins which influence fibrinogen transcription in response to acute-phase inflammatory stimuli. The fibrinogen gene cluster also contains cis regulatory elements; enhancer sequences with liver activities identified by sequence conservation and functional genomics. While the transcriptional control of this gene cluster is fascinating biology, the medical impetus to understand fibrinogen gene regulation stems from the association of cardiovascular disease risk with high level circulating fibrinogen. In the general population this level varies from about 1.5 to 3.5 g/l. This variation between individuals is influenced by genotype, suggesting there are genetic variants contributing to fibrinogen levels which reside in fibrinogen regulatory loci. A complete picture of how fibrinogen genes are regulated will therefore point towards novel sources of regulatory variants. In this review we discuss regulation of the fibrinogen genes from proximal promoters and enhancers, the influence of acute-phase stimulation, post-transcriptional regulation by miRNAs and functional regulatory variants identified in genetic studies. Finally, we discuss the fibrinogen locus in light of recent advances in understanding chromosomal architecture and suggest future directions for researching the mechanisms that control fibrinogen expression.
Collapse
Affiliation(s)
- Richard J Fish
- Department of Genetic Medicine and Development, University of Geneva Medical Centre, Geneva, Switzerland.
| | | |
Collapse
|
240
|
Unfolded protein response, activated by OASIS family transcription factors, promotes astrocyte differentiation. Nat Commun 2012; 3:967. [PMID: 22828627 DOI: 10.1038/ncomms1971] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 06/22/2012] [Indexed: 01/27/2023] Open
Abstract
OASIS is a member of the CREB/ATF family of transcription factors and modulates cell- or tissue-specific unfolded protein response signalling. Here we show that this modulation has a critical role in the differentiation of neural precursor cells into astrocytes. Cerebral cortices of mice specifically deficient in OASIS (Oasis(-/-)) contain fewer astrocytes and more neural precursor cells than those of wild-type mice during embryonic development. Furthermore, astrocyte differentiation is delayed in primary cultured Oasis(-/-) neural precursor cells. The transcription factor Gcm1, which is necessary for astrocyte differentiation in Drosophila, is revealed to be a target of OASIS. Introduction of Gcm1 into Oasis(-/-) neural precursor cells improves the delayed differentiation of neural precursor cells into astrocytes by accelerating demethylation of the Gfap promoter. Gcm1 expression is temporally controlled by the unfolded protein response through interactions between OASIS family members during astrocyte differentiation. Taken together, our findings demonstrate a novel mechanism by which OASIS and its associated family members are modulated by the unfolded protein response to finely control astrocyte differentiation.
Collapse
|
241
|
Characterization of the expression and cell-surface localization of transmembrane protein 132A. Mol Cell Biochem 2012; 370:23-33. [PMID: 22821197 DOI: 10.1007/s11010-012-1394-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 07/07/2012] [Indexed: 10/28/2022]
Abstract
Transmembrane protein 132A (TMEM132A, KIAA1583) was first isolated as a novel gene that is enriched during the embryonic and postnatal stages of rat brain development and interacts with GRP78. However, the biological functions of TMEM132A are scarcely characterized because the protein does not contain any known structural domains. Using a cell-surface biotinylation assay and immunocytochemical staining, we found that TMEM132A is a transmembrane glycoprotein consisting of a large extracellular domain in the N-terminal region and a small cytosolic domain in the C-terminal region. Partial deletions of the intracellular domain of TMEM132A had little effect on its expression level and cell-surface localization in transfected HEK293 cells, whereas deletions of the extracellular domain hampered transport to the cell surface. The expression pattern of each N-terminal mutant was immunocytochemically confirmed in HeLa cells transfected with the same constructs. Treatment with tunicamycin, an inhibitor of protein glycosylation, led to the accumulation of the unglycosylated form of TMEM132A in inverse proportion to the glycosylated form; however, both forms were localized at the cell surface at almost equal rates. In contrast, GRP78 overexpression led to the accumulation of unglycosylated TMEM132A, which was not detected on the cell surface. Inhibition of ER-Golgi transport by treatment with brefeldin A or the overexpression of mutant Sar1 attenuated the amount of cell-surface localized TMEM132A in HEK293 cells. Treatment with reagents disrupting intracellular calcium rapidly down-regulated the amount of TMEM132A protein in Neuro2a cells without affecting the expression level of its mRNA. Taken together, our data show that the novel cell-surface localized glycoprotein, TMEM132A, is regulated by several factors, including GRP78, Sar1, and intracellular calcium, in a post-transcriptional manner.
Collapse
|
242
|
Scholl C, Weiβmüller K, Holenya P, Shaked-Rabi M, Tucker KL, Wölfl S. Distinct and overlapping gene regulatory networks in BMP- and HDAC-controlled cell fate determination in the embryonic forebrain. BMC Genomics 2012; 13:298. [PMID: 22748179 PMCID: PMC3460768 DOI: 10.1186/1471-2164-13-298] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 06/12/2012] [Indexed: 12/11/2022] Open
Abstract
Background Both bone morphogenetic proteins (BMPs) and histone deacetylases (HDACs) have previously been established to play a role in the development of the three major cell types of the central nervous system: neurons, astrocytes, and oligodendrocytes. We have previously established a connection between these two protein families, showing that HDACs suppress BMP-promoted astrogliogenesis in the embryonic striatum. Since HDACs act in the nucleus to effect changes in transcription, an unbiased analysis of their transcriptional targets could shed light on their downstream effects on BMP-signaling. Results Using neurospheres from the embryonic striatum as an in vitro system to analyze this phenomenon, we have performed microarray expression profiling on BMP2- and TSA-treated cultures, followed by validation of the findings with quantitative RT-PCR and protein analysis. In BMP-treated cultures we first observed an upregulation of genes involved in cell-cell communication and developmental processes such as members of BMP and canonical Wnt signaling pathways. In contrast, in TSA-treated cultures we first observed an upregulation of genes involved in chromatin modification and transcription. Interestingly, we could not record direct changes in the protein levels of canonical members of BMP2 signaling, but we did observe an upregulation of both the transcription factor STAT3 and its active isoform phospho-STAT3 at the protein level. Conclusions STAT3 and SMAD1/5/8 interact synergistically to promote astrogliogenesis, and thus we show for the first time that HDACs act to suppress BMP-promoted astrogliogenesis by suppression of the crucial partner STAT3.
Collapse
Affiliation(s)
- Catharina Scholl
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, 69120 Heidelberg, Germany.
| | | | | | | | | | | |
Collapse
|
243
|
Abstract
There are numerous examples of enduring effects of early experience on gene transcription and neural function. We review the emerging evidence for epigenetics as a candidate mechanism for such effects. There is now evidence that intracellular signals activated by environmental events can directly modify the epigenetic state of the genome, including CpG methylation, histone modifications and microRNAs. We suggest that this process reflects an activity-dependent epigenetic plasticity at the level of the genome, comparable with that observed at the synapse. This epigenetic plasticity mediates neuronal differentiation and phenotypic plasticity, including that associated with learning and memory. Altered epigenetic states are also associated with the risk for and expression of mental disorders. In a broader context, these studies define a biological basis for the interplay between environmental signals and the genome in the regulation of individual differences in behavior, cognition and physiology, as well as the risk for psychopathology.
Collapse
Affiliation(s)
- Judy Sng
- Integrative Neuroscience Program, Singapore Institute for Clinical Sciences, 30 Medial Drive, Singapore.
| | | |
Collapse
|
244
|
Ueki Y, Reh TA. Activation of BMP-Smad1/5/8 signaling promotes survival of retinal ganglion cells after damage in vivo. PLoS One 2012; 7:e38690. [PMID: 22701694 PMCID: PMC3368846 DOI: 10.1371/journal.pone.0038690] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 05/12/2012] [Indexed: 11/18/2022] Open
Abstract
While the essential role of bone morphogenetic protein (BMP) signaling in nervous system development is well established, its function in the adult CNS is poorly understood. We investigated the role of BMP signaling in the adult mouse retina following damage in vivo. Intravitreal injection of N-methyl-D-aspartic acid (NMDA) induced extensive retinal ganglion cell death by 2 days. During this period, BMP2, -4 and -7 were upregulated, leading to phosphorylation of the downstream effector, Smad1/5/8 in the inner retina, including in retinal ganglion cells. Expression of Inhibitor of differentiation 1 (Id1; a known BMP-Smad1/5/8 target) was also upregulated in the retina. This activation of BMP-Smad1/5/8 signaling was also observed following light damage, suggesting that it is a general response to retinal injuries. Co-injection of BMP inhibitors with NMDA effectively blocked the damage-induced BMP-Smad1/5/8 activation and led to further cell death of retinal ganglion cells, when compared with NMDA injection alone. Moreover, treatment of the retina with exogenous BMP4 along with NMDA damage led to a significant rescue of retinal ganglion cells. These data demonstrate that BMP-Smad1/5/8 signaling is neuroprotective for retinal ganglion cells after damage, and suggest that stimulation of this pathway can serve as a potential target for neuroprotective therapies in retinal ganglion cell diseases, such as glaucoma.
Collapse
Affiliation(s)
- Yumi Ueki
- Department of Biological Structure, University of Washington, Seattle, Washington, United States of America
| | - Thomas A. Reh
- Department of Biological Structure, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
245
|
Surmacz B, Noisa P, Risner-Janiczek JR, Hui K, Ungless M, Cui W, Li M. DLK1 promotes neurogenesis of human and mouse pluripotent stem cell-derived neural progenitors via modulating Notch and BMP signalling. Stem Cell Rev Rep 2012; 8:459-71. [PMID: 21761283 DOI: 10.1007/s12015-011-9298-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
A better understanding of the control of stem cell maintenance and differentiation fate choice is fundamental to effectively realising the potential of human pluripotent stem cells in disease modelling, drug screening and cell therapy. Dlk1 is a Notch related transmembrane protein that has been reportedly expressed in several neurogenic regions in the developing brain. In this study, we investigated the ability of Dlk1 in modulating the maintenance and differentiation of human and mouse ESC-derived neural progenitors. We found that DLK1, either employed as an extrinsic factor, or via transgene expression, consistently promoted the generation of neurons in both the mouse and human ESC-derived neural progenitors. DLK1 exerts this function by inducing cell cycle exit of the progenitors, as evidenced by an increase in the number of young neurons retaining BrdU labelling and cells expressing the cycling inhibitor P57Kip2. DLK1 antagonised the cell proliferation activity of Notch ligands Delta 1 and Jagged and inhibited Hes1-mediated Notch signaling as demonstrated by a luciferase reporter assay. Interestingly, we found that DLK1 promotes the neurogenic potential of human neural progenitor cells via suppression of Smad activation when they are challenged with BMP. Together, our data demonstrate for the first time a regulatory role for DLK1 in human and mouse neural progenitor differentiation and establish an interaction between DLK1 and Hes1-mediated Notch signaling in these cells. Furthermore, this study identifies DLK1 as a novel modulator of BMP/Smad signalling.
Collapse
Affiliation(s)
- Beata Surmacz
- Stem Cell Neurogenesis, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | | | | | | | | | | | | |
Collapse
|
246
|
Liu S, Yin F, Fan W, Wang S, Guo XR, Zhang JN, Tian ZM, Fan M. Over-expression of BMPR-IB reduces the malignancy of glioblastoma cells by upregulation of p21 and p27Kip1. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2012; 31:52. [PMID: 22650359 PMCID: PMC3408360 DOI: 10.1186/1756-9966-31-52] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 05/31/2012] [Indexed: 11/23/2022]
Abstract
Background In our previous study, we detected decreased expression of phospho-Smad1/5/8 and its upstream signaling molecule, bone morphogenetic protein receptor IB subunit (BMPR-IB), in certain glioblastoma tissues, unlike normal brain tissues. In order to clarify the functional roles and mechanism of BMPR-IB in the development of glioblastoma, we studied the effects of BMPR-IB overexpression on glioblastoma cell lines in vitro and in vivo. Methods We selected glioblastoma cell lines U251, U87, SF763, which have different expression of BMPR-IB to be the research subjects. Colony formation analysis and FACS were used to detect the effects of BMPR-IB on the growth and proliferation of glioblastoma cells in vivo. Immunofluresence was used to detect the differentiation changes after BMPR-IB overexpression or knocking-down. Then we used subcutaneous and intracranial tumor models to study the effect of BMPR-IB on the growth and differentiation of glioblastoma cells in vivo. The genetic alterations involved in this process were examined by real-time PCR and western blot analysis.ed. Results and conclusion Forced BMPR-IB expression in malignant human glioma cells, which exhibit lower expression of BMPR-IB, induced the phosphorylation and nuclear localization of smad1/5/8 and arrested the cell cycle in G1. Additionally, BMPR-IB overexpression could suppress anchorage-independent growth and promote differentiation of theses glioblastoma cells. Furthermore, overexpression of BMPR-IB inhibited the growth of subcutaneous and intracranial tumor xenografts and prolonged the survival of mice injected intracranially with BMPR-IB-overexpressing glioblastoma cells. Conversely, inhibition of BMPR-IB caused SF763 malignant glioma cells, a line known to exhibit high BMPR-IB expression that does not form tumors when used for xenografts, to show increased growth and regain tumorigenicity in a nude mouse model system, ultimately shortening the survival of these mice. We also observed significant accumulation of p21 and p27kip1 proteins in response to BMPR-IB overexpression. Our study suggests that overexpression of BMPR-IB may arrest and induce the differentiation of glioblastoma cells due to upregulation of p21 and p27kip1 in vitro and that in vivo and decreased expression of BMPR-IB in human glioblastoma cells contributes to glioma tumorigenicity. BMPR-IB could represent a new potential therapeutic target for malignant human gliomas.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Neurosurgery, Navy General Hospital, 100048 Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
247
|
Stipursky J, Francis D, Gomes FCA. Activation of MAPK/PI3K/SMAD Pathways by TGF-β 1 Controls Differentiation of Radial Glia into Astrocytes in vitro. Dev Neurosci 2012; 34:68-81. [DOI: 10.1159/000338108] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Accepted: 03/15/2012] [Indexed: 01/05/2023] Open
|
248
|
The Stat3/GR interaction code: predictive value of direct/indirect DNA recruitment for transcription outcome. Mol Cell 2012; 47:38-49. [PMID: 22633955 DOI: 10.1016/j.molcel.2012.04.021] [Citation(s) in RCA: 144] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 03/28/2012] [Accepted: 04/16/2012] [Indexed: 11/20/2022]
Abstract
Transcription factor recruitment to genomic sites of action is primarily due to direct protein:DNA interactions. The subsequent recruitment of coregulatory complexes leads to either transcriptional activation or repression. In contrast to this canonical scheme, some transcription factors, such as the glucocorticoid receptor (GR), behave as transcriptional repressors when recruited to target genes through protein tethering. We have investigated the genome-wide prevalence of tethering between GR and Stat3 and found nonreciprocal interactions, namely that GR tethering to DNA-bound Stat3 results in transcriptional repression, whereas Stat3 tethering to GR results in synergism. Further, other schemes of GR and Stat3 corecruitment to regulatory modules result in transcriptional synergism, including neighboring and composite binding sites. The results indicate extensive transcriptional interactions between Stat3 and GR; further, they provide a genome-wide assessment of transcriptional regulation by tethering and a molecular basis for integration of signals mediated by GR and Stats in health and disease.
Collapse
|
249
|
Neuron-astroglial interactions in cell-fate commitment and maturation in the central nervous system. Neurochem Res 2012; 37:2402-18. [PMID: 22614925 DOI: 10.1007/s11064-012-0798-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 04/18/2012] [Accepted: 05/07/2012] [Indexed: 01/24/2023]
Abstract
Neuron-astroglia interactions play a key role in several events of brain development, such as neuronal generation, migration, survival, and differentiation; axonal growth; and synapse formation and function. While there is compelling evidence of the effects of astrocyte factors on neurons, their effects on astrocytes have not been fully determined. In this review, we will focus on the role of neurons in astrocyte generation and maturation. Further, we highlight the great heterogeneity and diversity of astroglial and neural progenitors such as radial glia cells, and discuss the importance of the variety of cellular interactions in controlling the structural and functional organization of the brain. Finally, we present recent data on a new role of astrocytes in neuronal maturation, as mediators of the action of biolipids in the cerebral cortex. We will argue that the functional architecture of the brain depends on an intimate neuron-glia partnership, by briefly discussing the emerging view of how neuron-astrocyte dysfunctions might be associated with neurodegenerative diseases and neurological disorders.
Collapse
|
250
|
Olynik BM, Rastegar M. The genetic and epigenetic journey of embryonic stem cells into mature neural cells. Front Genet 2012; 3:81. [PMID: 22629283 PMCID: PMC3355330 DOI: 10.3389/fgene.2012.00081] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 04/25/2012] [Indexed: 12/14/2022] Open
Abstract
Epigenetic changes occur throughout life from embryonic development into adulthood. This results in the timely expression of developmentally important genes, determining the morphology and identity of different cell types and tissues within the body. Epigenetics regulate gene expression and cellular morphology through multiple mechanisms without alteration in the underlying DNA sequences. Different epigenetic mechanisms include chromatin condensation, post-translational modification of histone proteins, DNA cytosine marks, and the activity of non-coding RNA molecules. Epigenetics play key roles in development, stem cell differentiation, and have high impact in human disease. In this review, we will discuss our current knowledge about these epigenetic mechanisms, with a focus on histone and DNA marks. We will then talk about the genetics and epigenetics of embryonic stem cell self-renewal and differentiation into neural stem cells, and further into specific neuronal cell types.
Collapse
Affiliation(s)
- Brendan M. Olynik
- Regenerative Medicine Program, Faculty of Medicine, University of ManitobaWinnipeg, MB, Canada
- Department of Biochemistry and Medical Genetics, University of ManitobaWinnipeg, MB, Canada
| | - Mojgan Rastegar
- Regenerative Medicine Program, Faculty of Medicine, University of ManitobaWinnipeg, MB, Canada
- Department of Biochemistry and Medical Genetics, University of ManitobaWinnipeg, MB, Canada
| |
Collapse
|