1
|
Nalamalapu RR, Yue M, Stone AR, Murphy S, Saha MS. The tweety Gene Family: From Embryo to Disease. Front Mol Neurosci 2021; 14:672511. [PMID: 34262434 PMCID: PMC8273234 DOI: 10.3389/fnmol.2021.672511] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/18/2021] [Indexed: 12/31/2022] Open
Abstract
The tweety genes encode gated chloride channels that are found in animals, plants, and even simple eukaryotes, signifying their deep evolutionary origin. In vertebrates, the tweety gene family is highly conserved and consists of three members—ttyh1, ttyh2, and ttyh3—that are important for the regulation of cell volume. While research has elucidated potential physiological functions of ttyh1 in neural stem cell maintenance, proliferation, and filopodia formation during neural development, the roles of ttyh2 and ttyh3 are less characterized, though their expression patterns during embryonic and fetal development suggest potential roles in the development of a wide range of tissues including a role in the immune system in response to pathogen-associated molecules. Additionally, members of the tweety gene family have been implicated in various pathologies including cancers, particularly pediatric brain tumors, and neurodegenerative diseases such as Alzheimer’s and Parkinson’s disease. Here, we review the current state of research using information from published articles and open-source databases on the tweety gene family with regard to its structure, evolution, expression during development and adulthood, biochemical and cellular functions, and role in human disease. We also identify promising areas for further research to advance our understanding of this important, yet still understudied, family of genes.
Collapse
Affiliation(s)
- Rithvik R Nalamalapu
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| | - Michelle Yue
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| | - Aaron R Stone
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| | - Samantha Murphy
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| | - Margaret S Saha
- Department of Biology, College of William and Mary, Williamsburg, VA, United States
| |
Collapse
|
2
|
Radix Scutellariae Ameliorates Stress-Induced Depressive-Like Behaviors via Protecting Neurons through the TGF β3-Smad2/3-Nedd9 Signaling Pathway. Neural Plast 2020; 2020:8886715. [PMID: 33273910 PMCID: PMC7683137 DOI: 10.1155/2020/8886715] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/29/2020] [Accepted: 10/31/2020] [Indexed: 12/18/2022] Open
Abstract
Chronic stress can impair hippocampal neurogenesis, increase neuronal apoptosis, and cause depressive-like behaviors. Our previous studies found that Radix Scutellariae (RS) can rescue the stress-induced neuronal injury, but the mechanism is not clear. Here, we continued to investigate the underlying antidepressant mechanisms of the RS extract. A 7-week chronic unpredictable mild stress (CUMS) procedure was used to establish a murine depression model. 0.75 g/kg or 1.5 g/kg RS was administered daily to the mice during the last 4 weeks. Depressive-like behaviors were evaluated by the sucrose preference test (SPT), forced swimming test (FST), open field test (OFT), and tail suspension test (TST). The neuroprotective effect of RS was evaluated with the expression of hippocampal neuron-related markers and apoptosis-associated proteins by Nissl staining, immunohistochemistry, and western blot. Transforming growth factor-β3 (TGFβ3) pathway-related proteins were detected by western blot. Results showed that RS could ameliorate depressive-like behaviors, increase the expression of the antiapoptotic protein B-cell lymphoma 2 (BCL-2), reduce the expression of the proapoptotic protein BCL-2-associated X (BAX), and increase the number of doublecortin- (DCX-), microtubule-associated protein 2- (MAP2-), and neuronal nucleus- (NeuN-) positive cells in the hippocampus. Moreover, RS could reverse the CUMS-induced decrease of TGFβ3 protein, promote the phosphorylation of SMAD2/3, and increase the expression of downstream NEDD9 protein. These results suggest that RS could exert antidepressant effects via protecting neurons. And the molecular mechanism might be related to the regulation of the TGFβ3-SMAD2/3-NEDD9 pathway.
Collapse
|
3
|
Wu HN, Cao XL, Fang Z, Zhang YF, Han WJ, Yue KY, Cao Y, Zheng MH, Wang LL, Han H. Deficiency of Ttyh1 downstream to Notch signaling results in precocious differentiation of neural stem cells. Biochem Biophys Res Commun 2019; 514:842-847. [PMID: 31079925 DOI: 10.1016/j.bbrc.2019.04.181] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 04/27/2019] [Indexed: 12/18/2022]
Abstract
Mammalian neural stem cells (NSCs) are not only responsible for normal development of the central nervous system (CNS), but also participate in brain homeostasis and repair, thus hold promising clinical potentials in the treatment of neurodegenerative diseases and trauma. However the molecular networks regulating the stemness and differentiation of NSCs have not been fully understood. In this study, we show that Tweety-homolog 1 (Ttyh1), a five-pass transmembrane protein specifically expressed in mouse brain, is involved in maintaining stemness of murine NSCs. Blocking or activating Notch signal led to downregulation and upregulation of Ttyh1 in cultured NSCs, respectively, suggesting that Ttyh1 is under the control of Notch signaling. Knockdown of Ttyh1 in cultured NSCs resulted in a transient increase in the number and size of neurospheres, followed by a decrease of stemness as manifested by compromised neurosphere formation, downregulated stem cell markers, and increased neuronal differentiation. We generated Ttyh1 knockout mice by deleting its exon 4 using the CRISPR-Cas9 technology. Surprisingly, in contrast to a previous report, Ttyh1 knockout did not result in embryonic lethality. NSCs derived from Ttyh1 knockout mice phenocopied NSCs transfected with Ttyh1 siRNA. Immunofluorescence showed that loss of Ttyh1 leads to the increase of neurogenesis in adult mice. Taken together, these findings indicate that Ttyh1, which is likely downstream to Notch signaling, plays an important role in regulating NSCs.
Collapse
Affiliation(s)
- Hai-Ning Wu
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China; Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiu-Li Cao
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China; Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Zheng Fang
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China; Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Yu-Fei Zhang
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China; Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Wen-Juan Han
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China; Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Kang-Yi Yue
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China; Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Yuan Cao
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China; Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Min-Hua Zheng
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China; Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China.
| | - Li-Li Wang
- Key Laboratory of Synthetic and Natural Functional Molecular Chemistry of Ministry of Education, Institute of Modern Separation Science, Northwest University, Shaanxi Key Laboratory of Modern Separation Science, Xi'an, 710069, China
| | - Hua Han
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, China; Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
4
|
Kawaguchi A. Temporal patterning of neocortical progenitor cells: How do they know the right time? Neurosci Res 2018; 138:3-11. [PMID: 30227161 DOI: 10.1016/j.neures.2018.09.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 08/09/2018] [Accepted: 08/09/2018] [Indexed: 10/28/2022]
Abstract
During mammalian neocortical development, neural progenitor cells undergo sequential division to produce different types of progenies. Regulation of when and how many cells with a specific fate are produced from neural progenitor cells, i.e., 'temporal patterning' for cytogenesis, is crucial for the formation of the functional neocortex. Recently advanced techniques for transcriptome profiling at the single-cell level provide a solid basis to investigate the molecular nature underlying temporal patterning, including examining the necessity of cell-cycle progression. Evidence has indicated that cell-intrinsic programs and extrinsic cues coordinately regulate the timing of both the change in the division mode of neural progenitors from proliferative to neurogenic and their laminar fate transition from deep-layer to upper-layer neurons. Epigenetic modulation, transcriptional cascades, and post-transcriptional regulation are reported to function as cell-intrinsic programs, whereas extrinsic cues from the environment or surrounding cells supposedly function in a negative feedback or positive switching manner for temporal patterning. These findings suggest that neural progenitor cells have intrinsic temporal programs that can progress cell-autonomously and cell-cycle independently, while extrinsic cues play a critical role in tuning the temporal programs to let neural progenitor cells know the 'right' time to progress.
Collapse
Affiliation(s)
- Ayano Kawaguchi
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya, Aichi, 466-8550, Japan.
| |
Collapse
|
5
|
Kim J, Han D, Byun SH, Kwon M, Cho JY, Pleasure SJ, Yoon K. Ttyh1 regulates embryonic neural stem cell properties by enhancing the Notch signaling pathway. EMBO Rep 2018; 19:embr.201745472. [PMID: 30177553 DOI: 10.15252/embr.201745472] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 08/02/2018] [Accepted: 08/09/2018] [Indexed: 12/11/2022] Open
Abstract
Despite growing evidence linking Drosophila melanogaster tweety-homologue 1 (Ttyh1) to normal mammalian brain development and cell proliferation, its exact role has not yet been determined. Here, we show that Ttyh1 is required for the maintenance of neural stem cell (NSC) properties as assessed by neurosphere formation and in vivo analyses of cell localization after in utero electroporation. We find that enhanced Ttyh1-dependent stemness of NSCs is caused by enhanced γ-secretase activity resulting in increased levels of Notch intracellular domain (NICD) production and activation of Notch targets. This is a unique function of Ttyh1 among all other Ttyh family members. Molecular analyses revealed that Ttyh1 binds to the regulator of γ-secretase activity Rer1 in the endoplasmic reticulum and thereby destabilizes Rer1 protein levels. This is the key step for Ttyh1-dependent enhancement of γ-secretase activity, as Rer1 overexpression completely abolishes the effects of Ttyh1 on NSC maintenance. Taken together, these findings indicate that Ttyh1 plays an important role during mammalian brain development by positively regulating the Notch signaling pathway through the downregulation of Rer1.
Collapse
Affiliation(s)
- Juwan Kim
- College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, South Korea
| | - Dasol Han
- College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, South Korea
| | - Sung-Hyun Byun
- College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, South Korea
| | - Mookwang Kwon
- College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, South Korea
| | - Jae Youl Cho
- College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, South Korea
| | - Samuel J Pleasure
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Keejung Yoon
- College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, South Korea
| |
Collapse
|
6
|
Brütting C, Narasimhan H, Hoffmann F, Kornhuber ME, Staege MS, Emmer A. Investigation of Endogenous Retrovirus Sequences in the Neighborhood of Genes Up-regulated in a Neuroblastoma Model after Treatment with Hypoxia-Mimetic Cobalt Chloride. Front Microbiol 2018. [PMID: 29515560 PMCID: PMC5826361 DOI: 10.3389/fmicb.2018.00287] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human endogenous retroviruses (ERVs) have been found to be associated with different diseases, e.g., multiple sclerosis (MS). Most human ERVs integrated in our genome are not competent to replicate and these sequences are presumably silent. However, transcription of human ERVs can be reactivated, e.g., by hypoxia. Interestingly, MS has been linked to hypoxia since decades. As some patterns of demyelination are similar to white matter ischemia, hypoxic damage is discussed. Therefore, we are interested in the association between hypoxia and ERVs. As a model, we used human SH-SY5Y neuroblastoma cells after treatment with the hypoxia-mimetic cobalt chloride and analyzed differences in the gene expression profiles in comparison to untreated cells. The vicinity of up-regulated genes was scanned for endogenous retrovirus-derived sequences. Five genes were found to be strongly up-regulated in SH-SY5Y cells after treatment with cobalt chloride: clusterin, glutathione peroxidase 3, insulin-like growth factor 2, solute carrier family 7 member 11, and neural precursor cell expressed developmentally down-regulated protein 9. In the vicinity of these genes we identified large (>1,000 bp) open reading frames (ORFs). Most of these ORFs showed only low similarities to proteins from retro-transcribing viruses. However, we found very high similarity between retrovirus envelope sequences and a sequence in the vicinity of neural precursor cell expressed developmentally down-regulated protein 9. This sequence encodes the human endogenous retrovirus group FRD member 1, the encoded protein product is called syncytin 2. Transfection of syncytin 2 into the well-characterized Ewing sarcoma cell line A673 was not able to modulate the low immunostimulatory activity of this cell line. Future research is needed to determine whether the identified genes and the human endogenous retrovirus group FRD member 1 might play a role in the etiology of MS.
Collapse
Affiliation(s)
- Christine Brütting
- Department of Surgical and Conservative Paediatrics and Adolescent Medicine, Martin Luther University of Halle-Wittenberg, Halle, Germany.,Department of Neurology, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Harini Narasimhan
- Department of Surgical and Conservative Paediatrics and Adolescent Medicine, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Frank Hoffmann
- Department of Neurology, Hospital "Martha-Maria" Halle-Dölau, Halle, Germany
| | - Malte E Kornhuber
- Department of Neurology, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Martin S Staege
- Department of Surgical and Conservative Paediatrics and Adolescent Medicine, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Alexander Emmer
- Department of Neurology, Martin Luther University of Halle-Wittenberg, Halle, Germany
| |
Collapse
|
7
|
Chen L, Li J, Zhang YH, Feng K, Wang S, Zhang Y, Huang T, Kong X, Cai YD. Identification of gene expression signatures across different types of neural stem cells with the Monte-Carlo feature selection method. J Cell Biochem 2017; 119:3394-3403. [PMID: 29130544 DOI: 10.1002/jcb.26507] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 11/09/2017] [Indexed: 02/03/2023]
Abstract
Adult neural stem cells (NSCs) are a group of multi-potent, self-renewing progenitor cells that contribute to the generation of new neurons and oligodendrocytes. Three subtypes of NSCs can be isolated based on the stages of the NSC lineage, including quiescent neural stem cells (qNSCs), activated neural stem cells (aNSCs) and neural progenitor cells (NPCs). Although it is widely accepted that these three groups of NSCs play different roles in the development of the nervous system, their molecular signatures are poorly understood. In this study, we applied the Monte-Carlo Feature Selection (MCFS) method to identify the gene expression signatures, which can yield a Matthews correlation coefficient (MCC) value of 0.918 with a support vector machine evaluated by ten-fold cross-validation. In addition, some classification rules yielded by the MCFS program for distinguishing above three subtypes were reported. Our results not only demonstrate a high classification capacity and subtype-specific gene expression patterns but also quantitatively reflect the pattern of the gene expression levels across the NSC lineage, providing insight into deciphering the molecular basis of NSC differentiation.
Collapse
Affiliation(s)
- Lei Chen
- Schoolof Life Sciences, Shanghai University, Shanghai, P.R. China.,College of Information Engineering, Shanghai Maritime University, Shanghai, P.R. China
| | - JiaRui Li
- Schoolof Life Sciences, Shanghai University, Shanghai, P.R. China
| | - Yu-Hang Zhang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, P.R. China
| | - KaiYan Feng
- Department of Computer Science, Guangdong AIB Polytechnic, Guangzhou, Guangdong, P.R. China
| | - ShaoPeng Wang
- Schoolof Life Sciences, Shanghai University, Shanghai, P.R. China
| | - YunHua Zhang
- Anhui province key lab of Farmland Ecological Conversation and Pollution Prevention, School of Resources and Environment, Anhui Agricultural University, Hefei, P.R. China
| | - Tao Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Xiangyin Kong
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Yu-Dong Cai
- Schoolof Life Sciences, Shanghai University, Shanghai, P.R. China
| |
Collapse
|
8
|
Yang J, Menges S, Gu P, Tongbai R, Samuel M, Prather RS, Klassen H. Porcine Neural Progenitor Cells Derived from Tissue at Different Gestational Ages Can Be Distinguished by Global Transcriptome. Cell Transplant 2017; 26:1582-1595. [PMID: 29113465 PMCID: PMC5524599 DOI: 10.1177/0963689717723015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The impact of gestational age on mammalian neural progenitor cells is potentially important for both an understanding of neural development and the selection of donor cells for novel cell-based treatment strategies. In terms of the latter, it can be problematic to rely entirely on rodent models in which the gestational period is significantly shorter and the brain much smaller than is the case in humans. Here, we analyzed pig brain progenitor cells (pBPCs) harvested at 2 different gestational ages (E45 and E60) using gene expression profiles, obtained by microarray analysis and quantitative polymerase chain reaction (qPCR), across time in culture. Comparison of the global transcriptome of pBPCs from age-matched transgenic green flourescent protein (GFP)-expressing fetuses versus non-GFP-expressing fetuses did not reveal significant differences between the 2 cell types, whereas comparison between E45 and E60 pBPCs did show separation between the data sets by principle component analysis. Further examination by qPCR showed evidence of relative downregulation of proliferation markers and upregulation of glial markers in the gestationally older (E60) cells. Additional comparisons were made. This study provides evidence of age-related changes in the gene expression of cultured fetal porcine neural progenitors that are potentially relevant to the role of these cells during development and as donor cells for transplantation studies.
Collapse
Affiliation(s)
- Jing Yang
- 1 Stem Cell Research Center, University of California, Irvine, CA, USA.,2 Gavin Herbert Eye Institute, University of California, Irvine, CA, USA
| | - Steven Menges
- 1 Stem Cell Research Center, University of California, Irvine, CA, USA.,2 Gavin Herbert Eye Institute, University of California, Irvine, CA, USA
| | - Ping Gu
- 1 Stem Cell Research Center, University of California, Irvine, CA, USA.,2 Gavin Herbert Eye Institute, University of California, Irvine, CA, USA.,3 Present Address: Department of Ophthalmology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ronald Tongbai
- 1 Stem Cell Research Center, University of California, Irvine, CA, USA.,4 Present Address: Huntington Beach Eye Consultants, Huntington Beach, CA, USA
| | - Melissa Samuel
- 5 National Swine Resource and Research Center, University of Missouri, Columbia, MO, USA
| | - Randall S Prather
- 5 National Swine Resource and Research Center, University of Missouri, Columbia, MO, USA
| | - Henry Klassen
- 1 Stem Cell Research Center, University of California, Irvine, CA, USA.,2 Gavin Herbert Eye Institute, University of California, Irvine, CA, USA
| |
Collapse
|
9
|
Tweety-Homolog 1 Drives Brain Colonization of Gliomas. J Neurosci 2017; 37:6837-6850. [PMID: 28607172 DOI: 10.1523/jneurosci.3532-16.2017] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 03/16/2017] [Accepted: 04/19/2017] [Indexed: 01/25/2023] Open
Abstract
Early and progressive colonization of the healthy brain is one hallmark of diffuse gliomas, including glioblastomas. We recently discovered ultralong (>10 to hundreds of microns) membrane protrusions [tumor microtubes (TMs)] extended by glioma cells. TMs have been associated with the capacity of glioma cells to effectively invade the brain and proliferate. Moreover, TMs are also used by some tumor cells to interconnect to one large, resistant multicellular network. Here, we performed a correlative gene-expression microarray and in vivo imaging analysis, and identified novel molecular candidates for TM formation and function. Interestingly, these genes were previously linked to normal CNS development. One of the genes scoring highest in tests related to the outgrowth of TMs was tweety-homolog 1 (TTYH1), which was highly expressed in a fraction of TMs in mice and patients. Ttyh1 was confirmed to be a potent regulator of normal TM morphology and of TM-mediated tumor-cell invasion and proliferation. Glioma cells with one or two TMs were mainly responsible for effective brain colonization, and Ttyh1 downregulation particularly affected this cellular subtype, resulting in reduced tumor progression and prolonged survival of mice. The remaining Ttyh1-deficient tumor cells, however, had more interconnecting TMs, which were associated with increased radioresistance in those small tumors. These findings imply a cellular and molecular heterogeneity in gliomas regarding formation and function of distinct TM subtypes, with multiple parallels to neuronal development, and suggest that Ttyh1 might be a promising target to specifically reduce TM-associated brain colonization by glioma cells in patients.SIGNIFICANCE STATEMENT In this report, we identify tweety-homolog 1 (Ttyh1), a membrane protein linked to neuronal development, as a potent driver of tumor microtube (TM)-mediated brain colonization by glioma cells. Targeting of Ttyh1 effectively inhibited the formation of invasive TMs and glioma growth, but increased network formation by intercellular TMs, suggesting a functional and molecular heterogeneity of the recently discovered TMs with potential implications for future TM-targeting strategies.
Collapse
|
10
|
Cao XL, Zhang X, Zhang YF, Zhang YZ, Song CG, Liu F, Hu YY, Zheng MH, Han H. Expression and purification of mouse Ttyh1 fragments as antigens to generate Ttyh1-specific monoclonal antibodies. Protein Expr Purif 2016; 130:81-89. [PMID: 27678288 DOI: 10.1016/j.pep.2016.09.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 09/19/2016] [Accepted: 09/23/2016] [Indexed: 02/03/2023]
Abstract
Ttyh1 is a murine homolog of the Drosophila Tweety and is predicted as a five-pass transmembrane protein. The Ttyh1 mRNA is expressed in mouse brain tissues with a restricted pattern and in human glioma cells. Ttyh1 protein may function as a large-conductance chloride channel, however, the role of Ttyh1 in normal neural development and tumorigenesis has been largely unknown, at least partially due to the lack of effective antibodies. Here we report the expression in E. coli and purification of two recombinant Ttyh1 protein fragments corresponding to one of the predicted extracellular domains and the carboxyl terminus of the mouse Ttyh1. With these Ttyh1 protein products, a set of monoclonal antibodies (mAbs) against the mouse Ttyh1 protein was established by using conventional hybridoma techniques. The specificity of the anti-Ttyh1 mAbs was determined based on their activities in Western blotting and immunofluorescent analysis using embryonic brain tissues and cultured mouse neural stem cells (NSCs). We also show that the mouse Ttyh1 protein was expressed in cultured NSCs, most likely in membrane and cytoplasm. In mouse embryonic brains, it appeared that the Ttyh1 protein was specifically expressed in the apical edge of the ventricular zone as puncta-like structures, as determined by using immunofluorescence. Taken together, our study provided a useful tool for further exploration of the biological functions and pathological significance of Ttyh1 in mice.
Collapse
Affiliation(s)
- Xiu-Li Cao
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xing Zhang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yu-Fei Zhang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yi-Zhe Zhang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Chang-Geng Song
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Fan Liu
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yi-Yang Hu
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Min-Hua Zheng
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Hua Han
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, The Fourth Military Medical University, Xi'an, 710032, China; Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
11
|
Mitra SS, Feroze AH, Gholamin S, Richard C, Esparza R, Zhang M, Azad TD, Alrfaei B, Kahn SA, Hutter G, Guzman R, Creasey GH, Plant GW, Weissman IL, Edwards MSB, Cheshier S. Neural Placode Tissue Derived From Myelomeningocele Repair Serves as a Viable Source of Oligodendrocyte Progenitor Cells. Neurosurgery 2016. [PMID: 26225855 DOI: 10.1227/neu.0000000000000918] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The presence, characteristics, and potential clinical relevance of neural progenitor populations within the neural placodes of myelomeningocele patients remain to be studied. Neural stem cells are known to reside adjacent to ependyma-lined surfaces along the central nervous system axis. OBJECTIVE Given such neuroanatomic correlation and regenerative capacity in fetal development, we assessed myelomeningocele-derived neural placode tissue as a potentially novel source of neural stem and progenitor cells. METHODS Nonfunctional neural placode tissue was harvested from infants during the surgical repair of myelomeningocele and subsequently further analyzed by in vitro studies, flow cytometry, and immunofluorescence. To assess lineage potential, neural placode-derived neurospheres were subjected to differential media conditions. Through assessment of platelet-derived growth factor receptor α (PDGFRα) and CD15 cell marker expression, Sox2+Olig2+ putative oligodendrocyte progenitor cells were successfully isolated. RESULTS PDGFRαCD15 cell populations demonstrated the highest rate of self-renewal capacity and multipotency of cell progeny. Immunofluorescence of neural placode-derived neurospheres demonstrated preferential expression of the oligodendrocyte progenitor marker, CNPase, whereas differentiation to neurons and astrocytes was also noted, albeit to a limited degree. CONCLUSION Neural placode tissue contains multipotent progenitors that are preferentially biased toward oligodendrocyte progenitor cell differentiation and presents a novel source of such cells for use in the treatment of a variety of pediatric and adult neurological disease, including spinal cord injury, multiple sclerosis, and metabolic leukoencephalopathies.
Collapse
Affiliation(s)
- Siddhartha S Mitra
- ‡Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California; §Department of Neurosurgery, Lucile Packard Children's Hospital, Stanford University School of Medicine, Stanford, California; ¶Department of Neurosurgery, VA Palo Alto Health Care System, Stanford University School of Medicine, Palo Alto, California; ∥Department of Neurological Surgery, University of Washington, Seattle, Washington
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Hollins SL, Cairns MJ. MicroRNA: Small RNA mediators of the brains genomic response to environmental stress. Prog Neurobiol 2016; 143:61-81. [PMID: 27317386 DOI: 10.1016/j.pneurobio.2016.06.005] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 05/24/2016] [Accepted: 06/11/2016] [Indexed: 01/09/2023]
Abstract
The developmental processes that establish the synaptic architecture of the brain while retaining capacity for activity-dependent remodeling, are complex and involve a combination of genetic and epigenetic influences. Dysregulation of these processes can lead to problems with neural circuitry which manifest in humans as a range of neurodevelopmental syndromes, such as schizophrenia, bipolar disorder and fragile X mental retardation. Recent studies suggest that prenatal, postnatal and intergenerational environmental factors play an important role in the aetiology of stress-related psychopathology. A number of these disorders have been shown to display epigenetic changes in the postmortem brain that reflect early life experience. These changes affect the regulation of gene expression though chromatin remodeling (transcriptional) and post-transcriptional influences, especially small noncoding microRNA (miRNA). These dynamic and influential molecules appear to play an important function in both brain development and its adaption to stress. In this review, we examine the role of miRNA in mediating the brain's response to both prenatal and postnatal environmental perturbations and explore how stress- induced alterations in miRNA expression can regulate the stress response via modulation of the immune system. Given the close relationship between environmental stress, miRNA, and brain development/function, we assert that miRNA hold a significant position at the molecular crossroads between neural development and adaptations to environmental stress. A greater understanding of the dynamics that mediate an individual's predisposition to stress-induced neuropathology has major human health benefits and is an important area of research.
Collapse
Affiliation(s)
- Sharon L Hollins
- School of Biomedical Sciences and Pharmacy and the Hunter Medical Research Institute, the University of Newcastle, Callaghan, NSW 2308, Australia
| | - Murray J Cairns
- School of Biomedical Sciences and Pharmacy and the Hunter Medical Research Institute, the University of Newcastle, Callaghan, NSW 2308, Australia; Schizophrenia Research Institute, Sydney, NSW, Australia.
| |
Collapse
|
13
|
Cell-cycle-independent transitions in temporal identity of mammalian neural progenitor cells. Nat Commun 2016; 7:11349. [PMID: 27094546 PMCID: PMC4842982 DOI: 10.1038/ncomms11349] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 03/17/2016] [Indexed: 12/30/2022] Open
Abstract
During cerebral development, many types of neurons are sequentially generated by self-renewing progenitor cells called apical progenitors (APs). Temporal changes in AP identity are thought to be responsible for neuronal diversity; however, the mechanisms underlying such changes remain largely unknown. Here we perform single-cell transcriptome analysis of individual progenitors at different developmental stages, and identify a subset of genes whose expression changes over time but is independent of differentiation status. Surprisingly, the pattern of changes in the expression of such temporal-axis genes in APs is unaffected by cell-cycle arrest. Consistent with this, transient cell-cycle arrest of APs in vivo does not prevent descendant neurons from acquiring their correct laminar fates. Analysis of cultured APs reveals that transitions in AP gene expression are driven by both cell-intrinsic and -extrinsic mechanisms. These results suggest that the timing mechanisms controlling AP temporal identity function independently of cell-cycle progression and Notch activation mode.
Collapse
|
14
|
Wu W, Zhou J, Xu CT, Zhang J, Jin YJ, Sun GL. Derivation and growth characteristics of dental pulp stem cells from patients of different ages. Mol Med Rep 2015; 12:5127-34. [PMID: 26239849 DOI: 10.3892/mmr.2015.4106] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Accepted: 03/20/2015] [Indexed: 02/06/2023] Open
Abstract
The dental pulp contains a relatively low number of stem cells; however, it is considered to be a promising source of stem cells for use in regenerative therapy. To date, it has remained elusive whether there are certain differences in the dental pulp stem cells (DPSCs) from donors of different ages. In the present study, DPSC lines were derived using teeth from children, adolescents, adults and aged donors. The derivation efficiency, the proliferative and apoptotic rate, cell marker expression and the differentiation capacity were investigated and compared among these DPSC lines. The derivation efficacy was decreased with increasing donor age. Although a large part of cell surface markers was expressed in all DPSC lines, the expression of CD29 was downregulated in the DPSCs from aged teeth. In addition, the doubling time of DPSCs from aged teeth was prolonged and the number of apoptotic cells was increased with the propagation. These DPSCs were able to differentiate into a neuronal linage, which positively expressed the neuron-specific class III beta-tubulin and microtubule‑associated protein 2, as well as into an osteogenic lineage, which positively expressed CD45; however, these DPSCs from aged teeth were completely or partially deprived of differentiation capacity. By contrast, DPSCs from younger teeth displayed significantly higher vitality and a higher potential for use in dental regenerative medicine.
Collapse
Affiliation(s)
- Wei Wu
- Department of Stomatology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Jian Zhou
- Department of Stomatology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Chong-Tao Xu
- Department of Stomatology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Jie Zhang
- Department of Stomatology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yan-Jiao Jin
- Department of Stomatology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Geng-Lin Sun
- Department of Stomatology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
15
|
Greenman R, Gorelik A, Sapir T, Baumgart J, Zamor V, Segal-Salto M, Levin-Zaidman S, Aidinis V, Aoki J, Nitsch R, Vogt J, Reiner O. Non-cell autonomous and non-catalytic activities of ATX in the developing brain. Front Neurosci 2015; 9:53. [PMID: 25788872 PMCID: PMC4349085 DOI: 10.3389/fnins.2015.00053] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 02/06/2015] [Indexed: 12/20/2022] Open
Abstract
The intricate formation of the cerebral cortex requires a well-coordinated series of events, which are regulated at the level of cell-autonomous and non-cell autonomous mechanisms. Whereas cell-autonomous mechanisms that regulate cortical development are well-studied, the non-cell autonomous mechanisms remain poorly understood. A non-biased screen allowed us to identify Autotaxin (ATX) as a non-cell autonomous regulator of neural stem cells. ATX (also known as ENPP2) is best known to catalyze lysophosphatidic acid (LPA) production. Our results demonstrate that ATX affects the localization and adhesion of neuronal progenitors in a cell autonomous and non-cell autonomous manner, and strikingly, this activity is independent from its catalytic activity in producing LPA.
Collapse
Affiliation(s)
- Raanan Greenman
- Department of Molecular Genetics, Weizmann Institute of Science Rehovot, Israel
| | - Anna Gorelik
- Department of Molecular Genetics, Weizmann Institute of Science Rehovot, Israel
| | - Tamar Sapir
- Department of Molecular Genetics, Weizmann Institute of Science Rehovot, Israel
| | - Jan Baumgart
- University Medical Center, Institute for Microscopic Anatomy and Neurobiology, Johannes Gutenberg-University Mainz Mainz, Germany ; Central Laboratory Animal Facility, University Medical Center, Johannes Gutenberg-University Mainz Mainz, Germany
| | - Vanessa Zamor
- Department of Molecular Genetics, Weizmann Institute of Science Rehovot, Israel
| | - Michal Segal-Salto
- Department of Molecular Genetics, Weizmann Institute of Science Rehovot, Israel
| | - Smadar Levin-Zaidman
- Department of Chemical Research Support, Weizmann Institute of Science Rehovot, Israel
| | - Vassilis Aidinis
- Division of Immunology, Biomedical Sciences Research Center 'Alexander Fleming' Athens, Greece
| | - Junken Aoki
- Graduate School of Pharmaceutical Sciences, Tohoku University Miyagi, Japan
| | - Robert Nitsch
- University Medical Center, Institute for Microscopic Anatomy and Neurobiology, Johannes Gutenberg-University Mainz Mainz, Germany
| | - Johannes Vogt
- University Medical Center, Institute for Microscopic Anatomy and Neurobiology, Johannes Gutenberg-University Mainz Mainz, Germany
| | - Orly Reiner
- Department of Molecular Genetics, Weizmann Institute of Science Rehovot, Israel
| |
Collapse
|
16
|
Reinchisi G, Limaye PV, Singh MB, Antic SD, Zecevic N. Neurogenic potential of hESC-derived human radial glia is amplified by human fetal cells. Stem Cell Res 2013; 11:587-600. [PMID: 23651582 DOI: 10.1016/j.scr.2013.03.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 03/20/2013] [Accepted: 03/25/2013] [Indexed: 01/19/2023] Open
Abstract
The efficient production of human neocortical neurons from human embryonic stem cells (hESC) is the primary requirement for studying early stages of human cortical development. We used hESC to obtain radial glial cells (hESC-RG) and then compared them with RG cells isolated from human fetal forebrain. Fate of hESC-RG cells critically depends on intrinsic and extrinsic factors. The expression of Pax6 (intrinsic factor) has a similar neurogenic effect on hESC-RG differentiation as reported for human fetal RG cells. Factors from the microenvironment also play a significant role in determining hESC-RG cell fate. In contrast to control cultures, wherein hESC-RG generate mainly astroglia and far fewer neurons, in co-cultures with human fetal forebrain cells, the reverse was found to be true. This neurogenic effect was partly due to soluble factors from human fetal brain cultures. The detected shift towards neurogenesis has significance for developing future efficient neuro-differentiation protocols. Importantly, we established that hESC-RG cells are similar in many respects to human fetal RG cells, including their proliferative capacity, neurogenic potential, and ability to generate various cortical neuronal sub-types. Unlike fetal RG cells, the hESC-RG cells are readily available and can be standardized, features that have considerable practical advantages in research and clinics.
Collapse
Affiliation(s)
- Gisela Reinchisi
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | | | | | | |
Collapse
|
17
|
Chopp M, Zhang ZG. Enhancing Brain Reorganization and Recovery of Function after Stroke. Stroke 2011. [DOI: 10.1016/b978-1-4160-5478-8.10011-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
18
|
Kumada T, Yamanaka Y, Kitano A, Shibata M, Awaya T, Kato T, Okawa K, Abe T, Oshima N, Nakahata T, Heike T. Ttyh1, a Ca2+-binding protein localized to the endoplasmic reticulum, is required for early embryonic development. Dev Dyn 2010; 239:2233-45. [DOI: 10.1002/dvdy.22348] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
19
|
Shoemaker LD, Orozco NM, Geschwind DH, Whitelegge JP, Faull KF, Kornblum HI. Identification of differentially expressed proteins in murine embryonic and postnatal cortical neural progenitors. PLoS One 2010; 5:e9121. [PMID: 20161753 PMCID: PMC2817745 DOI: 10.1371/journal.pone.0009121] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Accepted: 01/18/2010] [Indexed: 12/11/2022] Open
Abstract
Background The central nervous system (CNS) develops from a heterogeneous pool of neural stem and progenitor cells (NSPC), the underlying differences among which are poorly understood. The study of NSPC would be greatly facilitated by the identification of additional proteins that mediate their function and that would distinguish amongst different progenitor populations. Methodology/Principal Findings To identify membrane and membrane-associated proteins expressed by NSPC, we used a proteomics approach to profile NSPC cultured as neurospheres (NS) isolated from the murine cortex during a period of neurogenesis (embryonic day 11.5, E11.5), as compared to NSPC isolated at a peak of gliogenesis (postnatal day 1, P0) and to differentiated E11.5 NS. 54 proteins were identified with high expression in E11.5 NS, including the TrkC receptor, several heterotrimeric G proteins, and the Neogenin receptor. 24 proteins were identified with similar expression in E11.5 and P0 NS over differentiated E11.5 NS, and 13 proteins were identified with high expression specifically in P0 NS compared to E11.5 NS. To illustrate the potential relevance of these identified proteins to neural stem cell biology, the function of Neogenin was further studied. Using Fluorescence Activated Cell Sorting (FACS) analysis, expression of Neogenin was associated with a self-renewing population present in both E11.5 and adult subventricular zone (SVZ) NS but not in P0 NS. E11.5 NS expressed a putative Neogenin ligand, RGMa, and underwent apoptosis when exposed to a ligand-blocking antibody. Conclusions/Significance There are fundamental differences between the continuously self-renewing and more limited progenitors of the developing cortex. We identified a subset of differentially expressed proteins that serve not only as a set of functionally important proteins, but as a useful set of markers for the subsequent analysis of NSPC. Neogenin is associated with the continuously self-renewing and neurogenic cells present in E11.5 cortical and adult SVZ NS, and the Neogenin/RGMa receptor/ligand pair may regulate cell survival during development.
Collapse
Affiliation(s)
- Lorelei D. Shoemaker
- Pasarow Mass Spectrometry Laboratory, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Neuropsychiatric Institute - Semel Institute for Neuroscience & Human Behavior and Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Brain Research Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Nicholas M. Orozco
- Neuropsychiatric Institute - Semel Institute for Neuroscience & Human Behavior and Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Brain Research Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Daniel H. Geschwind
- Neuropsychiatric Institute - Semel Institute for Neuroscience & Human Behavior and Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Brain Research Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Julian P. Whitelegge
- Pasarow Mass Spectrometry Laboratory, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Neuropsychiatric Institute - Semel Institute for Neuroscience & Human Behavior and Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Brain Research Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Kym F. Faull
- Pasarow Mass Spectrometry Laboratory, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Neuropsychiatric Institute - Semel Institute for Neuroscience & Human Behavior and Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Brain Research Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Harley I. Kornblum
- Neuropsychiatric Institute - Semel Institute for Neuroscience & Human Behavior and Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Brain Research Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
20
|
Phoenix TN, Temple S. Spred1, a negative regulator of Ras-MAPK-ERK, is enriched in CNS germinal zones, dampens NSC proliferation, and maintains ventricular zone structure. Genes Dev 2010; 24:45-56. [PMID: 20047999 DOI: 10.1101/gad.1839510] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Neural stem cells (NSCs) have great potential for self-renewal, which must be tightly regulated to generate appropriate cell numbers during development and to prevent tumor formation. The Ras-MAPK-ERK pathway affects mitogen-stimulated proliferation, and negative regulators are likely to be important for keeping self-renewal in check. Sprouty-related protein with an EVH1 domain (Spred1) is a recently discovered negative Ras-MAPK-ERK regulator linked to a neurofibromatosis 1 (NF-1)-like human syndrome; however, its role in CNS development has not been explored. We show that Spred1 is highly enriched in CNS germinal zones during neurogenesis. Spred1 knockdown increases NSC self-renewal and progenitor proliferation cell-autonomously, and overexpression causes premature differentiation. Surprisingly, Spred1 knockdown in vivo in the embryonic mouse forebrain frequently resulted in periventricular heterotopia, developmental abnormalities often associated with mutations in genes in the vesicular trafficking pathway that cause disruption of germinal zones and impair cell migration. In cortical progenitor cells, Spred1 localizes within distinct vesicles, indicating a potential role in transport. Spred1 knockdown gradually leads to disruption of the apical ventricular zone and loss of radial glia alignment. This impairs late neuronal migration, resulting in the formation of periventricular masses. Thus, Spred1 is critical for normal cortical development, as it modulates progenitor self-renewal/proliferation and helps maintain the integrity and organization of germinal zones.
Collapse
Affiliation(s)
- Timothy N Phoenix
- New York Neural Stem Cell Institute, Rensselaer, New York 12144, USA
| | | |
Collapse
|
21
|
Magold AI, Cacquevel M, Fraering PC. Gene expression profiling in cells with enhanced gamma-secretase activity. PLoS One 2009; 4:e6952. [PMID: 19763259 PMCID: PMC2739295 DOI: 10.1371/journal.pone.0006952] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Accepted: 07/27/2009] [Indexed: 01/10/2023] Open
Abstract
Background Processing by γ-secretase of many type-I membrane protein substrates triggers signaling cascades by releasing intracellular domains (ICDs) that, following nuclear translocation, modulate the transcription of different genes regulating a diverse array of cellular and biological processes. Because the list of γ-secretase substrates is growing quickly and this enzyme is a cancer and Alzheimer's disease therapeutic target, the mapping of γ-secretase activity susceptible gene transcription is important for sharpening our view of specific affected genes, molecular functions and biological pathways. Methodology/Principal Findings To identify genes and molecular functions transcriptionally affected by γ-secretase activity, the cellular transcriptomes of Chinese hamster ovary (CHO) cells with enhanced and inhibited γ-secretase activity were analyzed and compared by cDNA microarray. The functional clustering by FatiGO of the 1,981 identified genes revealed over- and under-represented groups with multiple activities and functions. Single genes with the most pronounced transcriptional susceptibility to γ-secretase activity were evaluated by real-time PCR. Among the 21 validated genes, the strikingly decreased transcription of PTPRG and AMN1 and increased transcription of UPP1 potentially support data on cell cycle disturbances relevant to cancer, stem cell and neurodegenerative diseases' research. The mapping of interactions of proteins encoded by the validated genes exclusively relied on evidence-based data and revealed broad effects on Wnt pathway members, including WNT3A and DVL3. Intriguingly, the transcription of TERA, a gene of unknown function, is affected by γ-secretase activity and was significantly altered in the analyzed human Alzheimer's disease brain cortices. Conclusions/Significance Investigating the effects of γ-secretase activity on gene transcription has revealed several affected clusters of molecular functions and, more specifically, 21 genes that hold significant potential for a better understanding of the biology of γ-secretase and its roles in cancer and Alzheimer's disease pathology.
Collapse
Affiliation(s)
- Alexandra I. Magold
- Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Matthias Cacquevel
- Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Patrick C. Fraering
- Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
22
|
Nielsen JA, Lau P, Maric D, Barker JL, Hudson LD. Integrating microRNA and mRNA expression profiles of neuronal progenitors to identify regulatory networks underlying the onset of cortical neurogenesis. BMC Neurosci 2009; 10:98. [PMID: 19689821 PMCID: PMC2736963 DOI: 10.1186/1471-2202-10-98] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Accepted: 08/19/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cortical development is a complex process that includes sequential generation of neuronal progenitors, which proliferate and migrate to form the stratified layers of the developing cortex. To identify the individual microRNAs (miRNAs) and mRNAs that may regulate the genetic network guiding the earliest phase of cortical development, the expression profiles of rat neuronal progenitors obtained at embryonic day 11 (E11), E12 and E13 were analyzed. RESULTS Neuronal progenitors were purified from telencephalic dissociates by a positive-selection strategy featuring surface labeling with tetanus-toxin and cholera-toxin followed by fluorescence-activated cell sorting. Microarray analyses revealed the fractions of miRNAs and mRNAs that were up-regulated or down-regulated in these neuronal progenitors at the beginning of cortical development. Nearly half of the dynamically expressed miRNAs were negatively correlated with the expression of their predicted target mRNAs. CONCLUSION These data support a regulatory role for miRNAs during the transition from neuronal progenitors into the earliest differentiating cortical neurons. In addition, by supplying a robust data set in which miRNA and mRNA profiles originate from the same purified cell type, this empirical study may facilitate the development of new algorithms to integrate various "-omics" data sets.
Collapse
Affiliation(s)
- Joseph A Nielsen
- Section of Developmental Genetics, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | | | | | |
Collapse
|
23
|
Vogel T, Ahrens S, Büttner N, Krieglstein K. Transforming growth factor beta promotes neuronal cell fate of mouse cortical and hippocampal progenitors in vitro and in vivo: identification of Nedd9 as an essential signaling component. ACTA ACUST UNITED AC 2009; 20:661-71. [PMID: 19587023 PMCID: PMC2820705 DOI: 10.1093/cercor/bhp134] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Transforming Growth Factor β (Tgfβ) and associated signaling effectors are expressed in the forebrain, but little is known about the role of this multifunctional cytokine during forebrain development. Using hippocampal and cortical primary cell cultures of developing mouse brains, this study identified Tgfβ-regulated genes not only associated with cell cycle exit of progenitors but also with adoption of neuronal cell fate. Accordingly, we observed not only an antimitotic effect of Tgfβ on progenitors but also an increased expression of neuronal markers in Tgfβ treated cultures. This effect was dependent upon Smad4. Furthermore, in vivo loss-of-function analyses using Tgfβ2−/−/Tgfβ3−/− double mutant mice showed the opposite effect of increased cell proliferation and fewer neurons in the cerebral cortex and hippocampus. Gata2, Runx1, and Nedd9 were candidate genes regulated by Tgfβ and known to be involved in developmental processes of neuronal progenitors. Using siRNA-mediated knockdown, we identified Nedd9 as an essential signaling component for the Tgfβ-dependent increase in neuronal cell fate. Expression of this scaffolding protein, which is mainly described as a signaling molecule of the β1-integrin pathway, was not only induced after Tgfβ treatment but was also associated with morphological changes of the Nestin-positive progenitor pool observed upon exposure to Tgfβ.
Collapse
Affiliation(s)
- Tanja Vogel
- Department of Neuroanatomy, Centre of Anatomy, Georg-August-University, 37075 Goettingen, Germany
| | | | | | | |
Collapse
|
24
|
Abstract
Restorative cell-based and pharmacological therapies for experimental stroke substantially improve functional outcome. These therapies target several types of parenchymal cells (including neural stem cells, cerebral endothelial cells, astrocytes, oligodendrocytes, and neurons), leading to enhancement of endogenous neurogenesis, angiogenesis, axonal sprouting, and synaptogenesis in the ischaemic brain. Interaction between these restorative events probably underpins the improvement in functional outcome. This Review provides examples of cell-based and pharmacological restorative treatments for stroke that stimulate brain plasticity and functional recovery. The molecular pathways activated by these therapies, which induce remodelling of the injured brain via angiogenesis, neurogenesis, and axonal and dendritic plasticity, are discussed. The ease of treating intact brain tissue to stimulate functional benefit in restorative therapy compared with treating injured brain tissue in neuroprotective therapy might more readily help with translation of restorative therapy from the laboratory to the clinic.
Collapse
Affiliation(s)
- Zheng Gang Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA
| | | |
Collapse
|
25
|
Jordan PM, Ojeda LD, Thonhoff JR, Gao J, Boehning D, Yu Y, Wu P. Generation of spinal motor neurons from human fetal brain-derived neural stem cells: role of basic fibroblast growth factor. J Neurosci Res 2009; 87:318-32. [PMID: 18803285 PMCID: PMC2738861 DOI: 10.1002/jnr.21856] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Neural stem cells (NSCs) have some specified properties but are generally uncommitted and so can change their fate after exposure to environmental cues. It is unclear to what extent this NSC plasticity can be modulated by extrinsic cues and what are the molecular mechanisms underlying neuronal fate determination. Basic fibroblast growth factor (bFGF) is a well-known mitogen for proliferating NSCs. However, its role in guiding stem cells for neuronal subtype specification is undefined. Here we report that in-vitro-expanded human fetal forebrain-derived NSCs can generate cholinergic neurons with spinal motor neuron properties when treated with bFGF within a specific time window. bFGF induces NSCs to express the motor neuron marker Hb9, which is blocked by specific FGF receptor inhibitors and bFGF neutralizing antibodies. This development of spinal motor neuron properties is independent of selective proliferation or survival and does not require high levels of MAPK activation. Thus our study indicates that bFGF can play an important role in modulating plasticity and neuronal fate of human NSCs and presumably has implications for exploring the full potential of brain NSCs for clinical applications, particularly in spinal motor neuron regeneration.
Collapse
Affiliation(s)
- Paivi M. Jordan
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555–0620, USA
| | - Luis D. Ojeda
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555–0620, USA
| | - Jason R. Thonhoff
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555–0620, USA
| | - Junling Gao
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555–0620, USA
| | - Darren Boehning
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555–0620, USA
| | - Yongjia Yu
- Department of Radiation Oncology, University of Texas Medical Branch, Galveston, Texas 77555–0620, USA
| | - Ping Wu
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555–0620, USA
| |
Collapse
|
26
|
Rahman R, Heath R, Grundy R. Cellular immortality in brain tumours: an integration of the cancer stem cell paradigm. Biochim Biophys Acta Mol Basis Dis 2009; 1792:280-8. [PMID: 19419702 DOI: 10.1016/j.bbadis.2009.01.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2008] [Revised: 01/21/2009] [Accepted: 01/21/2009] [Indexed: 02/01/2023]
Abstract
Brain tumours are a diverse group of neoplasms that continue to present a formidable challenge in our attempt to achieve curable intervention. Our conceptual framework of human brain cancer has been redrawn in the current decade. There is a gathering acceptance that brain tumour formation is a phenotypic outcome of dysregulated neurogenesis, with tumours viewed as abnormally differentiated neural tissue. In relation, there is accumulating evidence that brain tumours, similar to leukaemia and many solid tumours, are organized as a developmental hierarchy which is maintained by a small fraction of cells endowed with many shared properties of tissue stem cells. Proof that neurogenesis persists throughout adult life, compliments this concept. Although the cancer cell of origin is unclear, the proliferative zones that harbour stem cells in the embryonic, post-natal and adult brain are attractive candidates within which tumour-initiation may ensue. Dysregulated, unlimited proliferation and an ability to bypass senescence are acquired capabilities of cancerous cells. These abilities in part require the establishment of a telomere maintenance mechanism for counteracting the shortening of chromosomal termini. A strategy based upon the synthesis of telomeric repeat sequences by the ribonucleoprotein telomerase, is prevalent in approximately 90% of human tumours studied, including the majority of brain tumours. This review will provide a developmental perspective with respect to normal (neurogenesis) and aberrant (tumourigenesis) cellular turnover, differentiation and function. Within this context our current knowledge of brain tumour telomere/telomerase biology will be discussed with respect to both its developmental and therapeutic relevance to the hierarchical model of brain tumourigenesis presented by the cancer stem cell paradigm.
Collapse
Affiliation(s)
- Ruman Rahman
- School of Clinical Sciences, Queen's Medical Centre, Nottingham, NG7 2UH, UK.
| | | | | |
Collapse
|
27
|
MacDonald JL, Roskams AJ. Histone deacetylases 1 and 2 are expressed at distinct stages of neuro-glial development. Dev Dyn 2008; 237:2256-67. [PMID: 18651664 DOI: 10.1002/dvdy.21626] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The deacetylation of histone proteins, catalyzed by histone deacetylases (HDACs), is a common epigenetic modification of chromatin, associated with gene silencing. Although HDAC inhibitors are used clinically to treat nervous system disorders, such as epilepsy, very little is known about the expression pattern of the HDACs in the central nervous system. Identifying the cell types and developmental stages that express HDAC1 and HDAC2 within the brain is important for determining the therapeutic mode of action of HDAC inhibitors, and evaluating potential side effects. Here, we examined the expression of HDAC1 and HDAC2 in the murine brain at multiple developmental ages. HDAC1 is expressed in neural stem cells/progenitors and glia. In contrast, HDAC2 is initiated in neural progenitors and is up-regulated in post-mitotic neuroblasts and neurons, but not in fully differentiated glia. These results identify key developmental stages of HDAC expression and suggest transitions of neural development that may utilize HDAC1 and/or HDAC2.
Collapse
|
28
|
Howard BM, Zhicheng Mo, Filipovic R, Moore AR, Antic SD, Zecevic N. Radial glia cells in the developing human brain. Neuroscientist 2008; 14:459-73. [PMID: 18467668 PMCID: PMC2702478 DOI: 10.1177/1073858407313512] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human radial glia (RG) share many of the features described in rodents, but also have a number of characteristics unique to the human brain. Results obtained from different mammalian species including human and non-human primates reveal differences in the involvement of RG in neurogenesis and oligodendrogenesis and in the timing of the initial expression of typical RG immunomarkers. A common problem in studying the human brain is that experimental procedures using modern molecular and genetic methods, such as in vivo transduction with retroviruses or creation of knockout or transgenic mutants, are not possible. Nevertheless, abundant and valuable information about the development of the human brain has been revealed using postmortem human material. Additionally, a combination and spectrum of in vitro techniques are used to gain knowledge about normal developmental processes in the human brain, including better understanding of RG as progenitor cells. Molecular and functional characterization of multipotent progenitors, such as RG, is important for future cell replacement therapies in neurological and psychiatric disorders, which are often resistant to conventional treatments. The protracted time of development and larger size of the human brain could provide insight into processes that may go unnoticed in the much smaller rodent cortex, which develops over a much shorter period. With that in mind, we summarize results on the role of RG in the human fetal brain.
Collapse
|
29
|
Sanosaka T, Namihira M, Asano H, Kohyama J, Aisaki K, Igarashi K, Kanno J, Nakashima K. Identification of genes that restrict astrocyte differentiation of midgestational neural precursor cells. Neuroscience 2008; 155:780-8. [PMID: 18640244 DOI: 10.1016/j.neuroscience.2008.06.039] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Revised: 05/22/2008] [Accepted: 06/13/2008] [Indexed: 01/21/2023]
Abstract
During development of the mammalian CNS, neurons and glial cells (astrocytes and oligodendrocytes) are generated from common neural precursor cells (NPCs). However, neurogenesis precedes gliogenesis, which normally commences at later stages of fetal telencephalic development. Astrocyte differentiation of mouse NPCs at embryonic day (E) 14.5 (relatively late gestation) is induced by activation of the transcription factor signal transducer and activator of transcription (STAT) 3, whereas at E11.5 (mid-gestation) NPCs do not differentiate into astrocytes even when stimulated by STAT3-activating cytokines such as leukemia inhibitory factor (LIF). This can be explained in part by the fact that astrocyte-specific gene promoters are highly methylated in NPCs at E11.5, but other mechanisms are also likely to play a role. We therefore sought to identify genes involved in the inhibition of astrocyte differentiation of NPCs at midgestation. We first examined gene expression profiles in E11.5 and E14.5 NPCs, using Affymetrix GeneChip analysis, applying the Percellome method to normalize gene expression level. We then conducted in situ hybridization analysis for selected genes found to be highly expressed in NPCs at midgestation. Among these genes, we found that N-myc and high mobility group AT-hook 2 (Hmga2) were highly expressed in the E11.5 but not the E14.5 ventricular zone of mouse brain, where NPCs reside. Transduction of N-myc and Hmga2 by retroviruses into E14.5 NPCs, which normally differentiate into astrocytes in response to LIF, resulted in suppression of astrocyte differentiation. However, sustained expression of N-myc and Hmga2 in E11.5 NPCs failed to maintain the hypermethylated status of an astrocyte-specific gene promoter. Taken together, our data suggest that astrocyte differentiation of NPCs is regulated not only by DNA methylation but also by genes whose expression is controlled spatio-temporally during brain development.
Collapse
Affiliation(s)
- T Sanosaka
- Laboratory of Molecular Neuroscience, Graduate School of Biological Sciences, Nara Institute of Science and Technology, 8916-5, Takayama, Ikoma, Nara 630-0101, Japan
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Hartl D, Irmler M, Römer I, Mader MT, Mao L, Zabel C, de Angelis MH, Beckers J, Klose J. Transcriptome and proteome analysis of early embryonic mouse brain development. Proteomics 2008; 8:1257-65. [PMID: 18283662 DOI: 10.1002/pmic.200700724] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Mouse embryonic brain development involves sequential differentiation of multipotent progenitors into neurons and glia cells. Using microarrays and large 2-DE, we investigated the mouse brain transcriptome and proteome of embryonic days 9.5, 11.5, and 13.5. During this developmental period, neural progenitor cells shift from proliferation to neuronal differentiation. As expected, we detected numerous expression changes between all time points investigated, but interestingly, the rate of alteration remained in a similar range within 2 days of development. Furthermore, up- and down-regulation of gene products was balanced at each time point which was also seen at embryonic days 16-18. We hypothesize that during embryonic development, the rate of gene expression alteration is rather constant due to limited cellular resources such as energy, space, and free water. A similar complexity in terms of expressed genes and proteins suggests that changes in relative concentrations rather than an increase in the number of gene products dominate cellular differentiation. In general, expression of metabolism and cell cycle related gene products was down-regulated when precursor cells switched from proliferation to neuronal differentiation (days 9.5-11.5), whereas neuron specific gene products were up-regulated. A detailed functional analysis revealed their implication in differentiation related processes such as rearrangement of the actin cytoskeleton as well as Notch- and Wnt-signaling pathways.
Collapse
Affiliation(s)
- Daniela Hartl
- Institute for Human Genetics, Charité - University Medicine Berlin, Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Pinto L, Mader MT, Irmler M, Gentilini M, Santoni F, Drechsel D, Blum R, Stahl R, Bulfone A, Malatesta P, Beckers J, Götz M. Prospective isolation of functionally distinct radial glial subtypes--lineage and transcriptome analysis. Mol Cell Neurosci 2008; 38:15-42. [PMID: 18372191 DOI: 10.1016/j.mcn.2008.01.012] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Accepted: 01/07/2008] [Indexed: 12/18/2022] Open
Abstract
Since the discovery of radial glia as the source of neurons, their heterogeneity in regard to neurogenesis has been described by clonal and time-lapse analysis in vitro. However, the molecular determinants specifying neurogenic radial glia differently from radial glia that mostly self-renew remain ill-defined. Here, we isolated two radial glial subsets that co-exist at mid-neurogenesis in the developing cerebral cortex and their immediate progeny. While one subset generates neurons directly, the other is largely non-neurogenic but also gives rise to Tbr2-positive basal precursors, thereby contributing indirectly to neurogenesis. Isolation of these distinct radial glia subtypes allowed determining interesting differences in their transcriptome. These transcriptomes were also strikingly different from the transcriptome of radial glia isolated at the end of neurogenesis. This analysis therefore identifies, for the first time, the lineage origin of basal progenitors and the molecular differences of this lineage in comparison to directly neurogenic and gliogenic radial glia.
Collapse
Affiliation(s)
- Luisa Pinto
- Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Institute of Stem Cell Research, Ingolstädter Landstr. 1, 85764 Neuherberg/Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Mo Z, Moore AR, Filipovic R, Ogawa Y, Kazuhiro I, Antic SD, Zecevic N. Human cortical neurons originate from radial glia and neuron-restricted progenitors. J Neurosci 2007; 27:4132-45. [PMID: 17428991 PMCID: PMC6672534 DOI: 10.1523/jneurosci.0111-07.2007] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2006] [Revised: 03/08/2007] [Accepted: 03/09/2007] [Indexed: 12/13/2022] Open
Abstract
Understanding the molecular and physiological determinants of cortical neuronal progenitor cells is essential for understanding the development of the human brain in health and in disease. We used surface marker fucose N-acetyl lactosamine (LeX) (also known as CD15) to isolate progenitor cells from the cortical ventricular/subventricular zone of human fetal brain at the second trimester of gestation and to study their progeny in vitro. LeX+ cells had typical bipolar morphology, radial orientation, and antigen profiles, characterizing them as a subtype of radial glia (RG) cells. Four complementary experimental techniques (clonal analysis, immunofluorescence, transfection experiments, and patch-clamp recordings) indicated that this subtype of RG generates mainly astrocytes but also a small number of cortical neurons. The neurogenic capabilities of RGs were both region and stage dependent. Present results provide the first direct evidence that RGs in the human cerebral cortex serve as neuronal progenitors. Simultaneously, another progenitor subtype was identified as proliferating cells labeled with neuronal (beta-III-tubulin and doublecortin) but not RG markers [GFAP, vimentin, and BLBP (brain lipid-binding protein)]. Proliferative and antigenic characteristics of these cells suggested their neuron-restricted progenitor status. In summary, our in vitro study suggests that diverse populations of cortical progenitor cells, including multipotent RGs and neuron-restricted progenitors, contribute differentially to cortical neurogenesis at the second trimester of gestation in human cerebral cortex.
Collapse
Affiliation(s)
- Zhicheng Mo
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030, and
| | - Anna R. Moore
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030, and
| | - Radmila Filipovic
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030, and
| | - Yasuhiro Ogawa
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Myodaiji, Okazaki, Aichi 444-8787, Japan
| | - Ikenaka Kazuhiro
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Myodaiji, Okazaki, Aichi 444-8787, Japan
| | - Srdjan D. Antic
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030, and
| | - Nada Zecevic
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030, and
| |
Collapse
|
33
|
Bonnert TP, Bilsland JG, Guest PC, Heavens R, McLaren D, Dale C, Thakur M, McAllister G, Munoz-Sanjuan I. Molecular characterization of adult mouse subventricular zone progenitor cells during the onset of differentiation. Eur J Neurosci 2006; 24:661-75. [PMID: 16930398 DOI: 10.1111/j.1460-9568.2006.04912.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Adult mouse subventricular zone (SVZ) neural progenitor cells (NPCs) retain the capacity to generate multiple lineages in vitro and in vivo. Thus far, the mechanisms involved in the regulation of these cells have not been well elucidated. We have carried out RNA profiling of adult SVZ cell cultures undergoing differentiation, to identify pathways that regulate progenitor cell proliferation and to define a set of transcripts that can be used as molecular tools in the drug discovery process. We carried out a stepwise stratification of the results to identify transcripts specifically enriched in NPCs and validated some of these using comparative literature analysis, quantitative polymerase chain reaction and immunological techniques. The results show a set of transcription factors, secreted molecules and plasma membrane markers that are differentially regulated during differentiation. Pathway analysis highlights alterations in insulin growth factor, Wnt and transforming growth factor beta signalling cascades. Further characterization of these components could provide greater insight into the mechanisms involved in the regulation of neurogenesis in the adult brain.
Collapse
Affiliation(s)
- Timothy P Bonnert
- Molecular and Cellular Neuroscience Department, Merck Sharp & Dohme, The Neuroscience Research Centre, Terlings Park, Harlow, Essex CM20 2QR, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Merkle FT, Alvarez-Buylla A. Neural stem cells in mammalian development. Curr Opin Cell Biol 2006; 18:704-9. [PMID: 17046226 DOI: 10.1016/j.ceb.2006.09.008] [Citation(s) in RCA: 224] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2006] [Accepted: 09/29/2006] [Indexed: 11/22/2022]
Abstract
Neural stem cells (NSCs) are primary progenitors that give rise to neurons and glia in the embryonic, neonatal and adult brain. In recent years, we have learned three important things about these cells. First, NSCs correspond to cells previously thought to be committed glial cells. Second, embryonic and adult NSCs are lineally related: they transform from neuroepithelial cells into radial glia, then into cells with astroglial characteristics. Third, NSCs divide asymmetrically and often amplify the number of progeny they generate via symmetrically dividing intermediate progenitors. These advances challenge our traditional perceptions of glia and stem cells, and provide the foundation for understanding the molecular basis of mammalian NSC behavior.
Collapse
Affiliation(s)
- Florian T Merkle
- Department of Neurological Surgery and Program in Developmental and Stem Cell Biology, University of California, San Francisco, Box 0525, HSW 1201A, San Francisco, California 94143, USA
| | | |
Collapse
|
35
|
Cai J, Shin S, Wright L, Liu Y, Zhou D, Xue H, Khrebtukova I, Mattson MP, Svendsen CN, Rao MS. Massively parallel signature sequencing profiling of fetal human neural precursor cells. Stem Cells Dev 2006; 15:232-44. [PMID: 16646669 DOI: 10.1089/scd.2006.15.232] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
We have examined gene expression in multipotent neural precursor cells (NPCs) derived from human fetal (f) brain tissue and compared its expression profiles with embryonic stem (ESC) cells, embryoid body cell (EBC), and astrocyte precursors using the technique of massively parallel signature sequencing (MPSS). Gene expression profiles show that fNPCs express core neural stem cells markers and share expression profiles with astrocyte precursor cells (APCs) rather than ESC or EBC. Gene expression analysis shows that fNPCs differ from other adult stem and progenitor cells in their marker expression and activation of specific functional networks such as the transforming growth factorbeta (TGFbeta) and Notch signaling pathways. In addition, our results allow us to identify novel genes expressed in fNPCs and provide a detailed profile of fNPCs.
Collapse
Affiliation(s)
- Jingli Cai
- Stem Cell Biology Unit/Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Capela A, Temple S. LeX is expressed by principle progenitor cells in the embryonic nervous system, is secreted into their environment and binds Wnt-1. Dev Biol 2006; 291:300-13. [PMID: 16458284 DOI: 10.1016/j.ydbio.2005.12.030] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2005] [Revised: 12/12/2005] [Accepted: 12/14/2005] [Indexed: 11/22/2022]
Abstract
LeX/SSEA1/CD15 is an extracellular matrix-associated carbohydrate expressed by ES cells and by adult neural and bone marrow stem cells. It is important for cell adhesion, compaction and FGF2 responses of early embryonic stem cells; however, its function at later stages is not clear. We now show that LeX is expressed by primary mouse neural progenitor cells, including neural stem cells, neuroblasts and glioblasts, but not by their more differentiated products. LeX distinguishes highly proliferative cells even in the primitive neuroepithelium, demonstrating heterogeneity in cell potential before radial glia arise. At later stages, LeX expressing progenitors are frequently radial in morphology. Surface LeX expression can be used to enrich neural stem and progenitor cells from different CNS regions throughout development by FACS. We found that LeX expression is particularly strong in neural regions with prolonged neurogenesis, e.g., the olfactory epithelium, hippocampus, basal forebrain and cerebellum. These regions also express high levels of the growth factors FGF8 and/or Wnt-1. We show here that LeX-containing molecules in the developing nervous system bind Wnt-1. Our findings suggest that LeX, which is present on the surface of principle neural progenitors and secreted into their extracellular niche, may bind and present growth factors important for their proliferation and self-renewal.
Collapse
Affiliation(s)
- Alexandra Capela
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, NY 12208, USA
| | | |
Collapse
|
37
|
Barraud P, Thompson L, Kirik D, Björklund A, Parmar M. Isolation and characterization of neural precursor cells from theSox1-GFP reporter mouse. Eur J Neurosci 2005; 22:1555-69. [PMID: 16197496 DOI: 10.1111/j.1460-9568.2005.04352.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We have made use of a reporter mouse line in which enhanced green fluorescence protein (GFP) is inserted into the Sox1 locus. We show that the GFP reporter is coexpressed with the Sox1 protein as well as with other known markers for neural stem and progenitor cells, and can be used to identify and isolate these cells by fluorescence-activated cell sorting (FACS) from the developing or adult brain and from neurosphere cultures. All neurosphere-forming cells with the capacity for multipotency and self-renewal reside in the Sox1-GFP-expressing population. Thus, the Sox1-GFP reporter system is highly useful for identification, isolation and characterization of neural stem and progenitor cells, as well as for the validation of alternative means for isolating neural stem and progenitor cells. Further, transplantation experiments show that Sox1-GFP cells isolated from the foetal brain give rise to neurons and glia in vivo, and that many of the neurons display phenotypic characteristics appropriate for the developing brain region from which the Sox1-GFP precursors were derived. On the other hand, Sox1-GFP cells isolated from the adult subventricular zone or expanded neurosphere cultures gave rise almost exclusively to glial cells following transplantation. Thus, not all Sox1-GFP cells possess the same capacity for neuronal differentiation in vivo.
Collapse
Affiliation(s)
- Perrine Barraud
- Wallenberg Neuroscience Center, BMC A11, Section of Neurobiology, and Lund Strategic Research Center for Stem Cell Biology and Cell Therapy, Lund, University, SE-221 84 Lund, Sweden
| | | | | | | | | |
Collapse
|