1
|
Saade M, Martí E. Early spinal cord development: from neural tube formation to neurogenesis. Nat Rev Neurosci 2025; 26:195-213. [PMID: 39915695 DOI: 10.1038/s41583-025-00906-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2025] [Indexed: 03/26/2025]
Abstract
As one of the simplest and most evolutionarily conserved parts of the vertebrate nervous system, the spinal cord serves as a key model for understanding the principles of nervous system construction. During embryonic development, the spinal cord originates from a population of bipotent stem cells termed neuromesodermal progenitors, which are organized within a transient embryonic structure known as the neural tube. Neural tube morphogenesis differs along its anterior-to-posterior axis: most of the neural tube (including the regions that will develop into the brain and the anterior spinal cord) forms via the bending and dorsal fusion of the neural groove, but the establishment of the posterior region of the neural tube involves de novo formation of a lumen within a solid medullary cord. The early spinal cord primordium consists of highly polarized neural progenitor cells organized into a pseudostratified epithelium. Tight regulation of the cell division modes of these progenitors drives the embryonic growth of the neural tube and initiates primary neurogenesis. A rich history of observational and functional studies across various vertebrate models has advanced our understanding of the cellular events underlying spinal cord development, and these foundational studies are beginning to inform our knowledge of human spinal cord development.
Collapse
Affiliation(s)
- Murielle Saade
- Department of Cells and Tissues, Instituto de Biología Molecular de Barcelona CSIC, Barcelona, Spain.
| | - Elisa Martí
- Department of Cells and Tissues, Instituto de Biología Molecular de Barcelona CSIC, Barcelona, Spain.
| |
Collapse
|
2
|
Doucet D, Brubaker C, Turner D, Gregory CA. Factors affecting the role of canonical Wnt inhibitor Dickkopf-1 in cancer progression. Front Oncol 2023; 13:1114822. [PMID: 37007131 PMCID: PMC10050559 DOI: 10.3389/fonc.2023.1114822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 03/01/2023] [Indexed: 03/17/2023] Open
Abstract
BackgroundThe canonical Wnt inhibitor Dickkopf-1 (Dkk-1) has the capacity to modulate homeostasis between canonical and non-canonical Wnt pathways and also signal independently of Wnt. The specific effects of Dkk-1 activity on tumor physiology are therefore unpredictable with examples of Dkk-1 serving as either a driver or suppressor of malignancy. Given that Dkk-1 blockade may serve as a potential treatment for some types of cancer, we questioned whether it is possible to predict the role of Dkk-1 on tumor progression based on the tissue origin of the tumor.MethodsOriginal research articles that described Dkk-1 in terms a tumor suppressor or driver of cancer growth were identified. To determine the association between tumor developmental origin and the role of Dkk-1, a logistic regression was performed. The Cancer Genome Atlas database was interrogated for survival statistics based on tumor Dkk-1 expression.ResultsWe report that Dkk-1 is statistically more likely to serve as a suppressor in tumors arising from the ectoderm (p = 0.0198) or endoderm (p = 0.0334) but more likely to serve as a disease driver in tumors of mesodermal origin (p = 0.0155). Survival analyses indicated that in cases where Dkk-1 expression could be stratified, high Dkk-1 expression is usually associated with poor prognosis. This in part may be due to pro-tumorigenic role Dkk-1 plays on tumor cells but also through its influence on immunomodulatory and angiogenic processes in the tumor stroma.ConclusionDkk-1 has a context-specific dual role as a tumor suppressor or driver. Dkk-1 is significantly more likely to serve as a tumor suppressor in tumors arising from ectoderm and endoderm while the converse is true for mesodermal tumors. Patient survival data indicated high Dkk-1 expression is generally a poor prognostic indicator. These findings provide further support for the importance of Dkk-1 as a therapeutic cancer target in some cases.
Collapse
Affiliation(s)
- Dakota Doucet
- Medical Sciences Program, Texas A&M Health Science Center School of Medicine, Texas A&M University, Bryan, TX, United States
| | - Connor Brubaker
- Department of Statistics, Texas A&M University, College Station, TX, United States
| | - Donald Turner
- Department of Statistics, Texas A&M University, College Station, TX, United States
| | - Carl A. Gregory
- Department of Cell Biology and Genetics, Texas A&M Health Science Center School of Medicine, Texas A&M University, Bryan, TX, United States
- *Correspondence: Carl A. Gregory,
| |
Collapse
|
3
|
Geng S, Paul F, Kowalczyk I, Raimundo S, Sporbert A, Mamo TM, Hammes A. Balancing WNT signalling in early forebrain development: The role of LRP4 as a modulator of LRP6 function. Front Cell Dev Biol 2023; 11:1173688. [PMID: 37091972 PMCID: PMC10119419 DOI: 10.3389/fcell.2023.1173688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 03/28/2023] [Indexed: 04/25/2023] Open
Abstract
The specification of the forebrain relies on the precise regulation of WNT/ß-catenin signalling to support neuronal progenitor cell expansion, patterning, and morphogenesis. Imbalances in WNT signalling activity in the early neuroepithelium lead to congenital disorders, such as neural tube defects (NTDs). LDL receptor-related protein (LRP) family members, including the well-studied receptors LRP5 and LRP6, play critical roles in modulating WNT signalling capacity through tightly regulated interactions with their co-receptor Frizzled, WNT ligands, inhibitors and intracellular WNT pathway components. However, little is known about the function of LRP4 as a potential modulator of WNT signalling in the central nervous system. In this study, we investigated the role of LRP4 in the regulation of WNT signalling during early mouse forebrain development. Our results demonstrate that LRP4 can modulate LRP5- and LRP6-mediated WNT signalling in the developing forebrain prior to the onset of neurogenesis at embryonic stage 9.5 and is therefore essential for accurate neural tube morphogenesis. Specifically, LRP4 functions as a genetic modifier for impaired mitotic activity and forebrain hypoplasia, but not for NTDs in LRP6-deficient mutants. In vivo and in vitro data provide evidence that LRP4 is a key player in fine-tuning WNT signalling capacity and mitotic activity of mouse neuronal progenitors and of human retinal pigment epithelial (hTERT RPE-1) cells. Our data demonstrate the crucial roles of LRP4 and LRP6 in regulating WNT signalling and forebrain development and highlight the need to consider the interaction between different signalling pathways to understand the underlying mechanisms of disease. The findings have significant implications for our mechanistic understanding of how LRPs participate in controlling WNT signalling.
Collapse
Affiliation(s)
- Shuang Geng
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute for Biology, Free University of Berlin, Berlin, Germany
| | - Fabian Paul
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute for Biology, Free University of Berlin, Berlin, Germany
| | - Izabela Kowalczyk
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute for Biology, Free University of Berlin, Berlin, Germany
| | - Sandra Raimundo
- Advanced Light Microscopy Technology Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Anje Sporbert
- Advanced Light Microscopy Technology Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Tamrat Meshka Mamo
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- *Correspondence: Tamrat Meshka Mamo, ; Annette Hammes,
| | - Annette Hammes
- Neuroscience, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- *Correspondence: Tamrat Meshka Mamo, ; Annette Hammes,
| |
Collapse
|
4
|
Non-thermal plasma directly accelerates neuronal proliferation by stimulating axon formation. Sci Rep 2022; 12:15868. [PMID: 36151253 PMCID: PMC9508269 DOI: 10.1038/s41598-022-20063-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/08/2022] [Indexed: 11/08/2022] Open
Abstract
Among the various methods, Non Thermal Plasma (NTP) has been recently introduced and is being studied to recover the damaged nerve. In the recent years, several studies have suggested that NTP accelerates nerve cell regeneration, but the mechanism remains unknown. This study evaluated the effect of NTP on neuronal proliferation in SH-SY5Y (Human neuroblastoma cells) cells differentiated by retinoic acid (RA) and investigated the mechanism by which NTP promotes cell proliferation. We analyzed the morphology of differentiated SH-SY5Y cells, and performed western blot analysis and reverse transcription polymerase chain reaction (RT-PCR). Immunofluorescence analysis was performed in an in vivo study by categorizing Wistar A rats into three groups: non-nerve damage (Non-ND), nerve damage (ND), and nerve damage + NTP treatment (ND + NTP). The cell morphology analysis revealed that the number of cells increased and axonal elongation progressed after NTP treatment. In addition, western blots indicated that tau expression increased significantly after NTP treatment. The RT-PCR results revealed that the expression of tau, wnt3a, and β-catenin increased after NTP treatment. The in vivo immunofluorescence assay showed that NTP increased the markers for tau and S100B while regulating the over-expression of MAP2 and GAP43. NTP treatment accelerated cell proliferation and regeneration of damaged neurons in differentiated SH-SY5Y cells. These results establish the fact of NTP as a noninvasive and effective treatment for nerve injury.
Collapse
|
5
|
Gupta S, Kawaguchi R, Heinrichs E, Gallardo S, Castellanos S, Mandric I, Novitch BG, Butler SJ. In vitro atlas of dorsal spinal interneurons reveals Wnt signaling as a critical regulator of progenitor expansion. Cell Rep 2022; 40:111119. [PMID: 35858555 PMCID: PMC9414195 DOI: 10.1016/j.celrep.2022.111119] [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] [Received: 11/08/2021] [Revised: 04/12/2022] [Accepted: 06/28/2022] [Indexed: 11/03/2022] Open
Abstract
Restoring sensation after injury or disease requires a reproducible method for generating large quantities of bona fide somatosensory interneurons. Toward this goal, we assess the mechanisms by which dorsal spinal interneurons (dIs; dI1-dI6) can be derived from mouse embryonic stem cells (mESCs). Using two developmentally relevant growth factors, retinoic acid (RA) and bone morphogenetic protein (BMP) 4, we recapitulate the complete in vivo program of dI differentiation through a neuromesodermal intermediate. Transcriptional profiling reveals that mESC-derived dIs strikingly resemble endogenous dIs, with the correct molecular and functional signatures. We further demonstrate that RA specifies dI4-dI6 fates through a default multipotential state, while the addition of BMP4 induces dI1-dI3 fates and activates Wnt signaling to enhance progenitor proliferation. Constitutively activating Wnt signaling permits the dramatic expansion of neural progenitor cultures. These cultures retain the capacity to differentiate into diverse populations of dIs, thereby providing a method of increasing neuronal yield.
Collapse
Affiliation(s)
- Sandeep Gupta
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Riki Kawaguchi
- Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Eric Heinrichs
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Genetics and Genomics Graduate Program, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Salena Gallardo
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Interdepartmental Graduate Program, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Stephanie Castellanos
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; CIRM Bridges to Research Program, California State University, Northridge, Los Angeles, CA, USA
| | - Igor Mandric
- Department of Computer Science, Samueli School of Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Bennett G Novitch
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Intellectual & Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Samantha J Butler
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Intellectual & Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
6
|
Barzilai-Tutsch H, Morin V, Toulouse G, Chernyavskiy O, Firth S, Marcelle C, Serralbo O. Transgenic quails reveal dynamic TCF/β-catenin signaling during avian embryonic development. eLife 2022; 11:72098. [PMID: 35833630 PMCID: PMC9395189 DOI: 10.7554/elife.72098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 07/13/2022] [Indexed: 11/26/2022] Open
Abstract
The Wnt/β-catenin signaling pathway is highly conserved throughout evolution, playing crucial roles in several developmental and pathological processes. Wnt ligands can act at a considerable distance from their sources and it is therefore necessary to examine not only the Wnt-producing but also the Wnt-receiving cells and tissues to fully appreciate the many functions of this pathway. To monitor Wnt activity, multiple tools have been designed which consist of multimerized Wnt signaling response elements (TCF/LEF binding sites) driving the expression of fluorescent reporter proteins (e.g. GFP, RFP) or of LacZ. The high stability of those reporters leads to a considerable accumulation in cells activating the pathway, thereby making them easily detectable. However, this makes them unsuitable to follow temporal changes of the pathway’s activity during dynamic biological events. Even though fluorescent transcriptional reporters can be destabilized to shorten their half-lives, this dramatically reduces signal intensities, particularly when applied in vivo. To alleviate these issues, we developed two transgenic quail lines in which high copy number (12× or 16×) of the TCF/LEF binding sites drive the expression of destabilized GFP variants. Translational enhancer sequences derived from viral mRNAs were used to increase signal intensity and specificity. This resulted in transgenic lines efficient for the characterization of TCF/β-catenin transcriptional dynamic activities during embryogenesis, including using in vivo imaging. Our analyses demonstrate the use of this transcriptional reporter to unveil novel aspects of Wnt signaling, thus opening new routes of investigation into the role of this pathway during amniote embryonic development.
Collapse
Affiliation(s)
| | - Valerie Morin
- NeuroMyoGene Institute (INMG), Claude Bernard University Lyon 1, Lyon, France
| | - Gauthier Toulouse
- NeuroMyoGene Institute (INMG), Claude Bernard University Lyon 1, Lyon, France
| | | | | | - Christophe Marcelle
- NeuroMyoGene Institute (INMG), Claude Bernard University Lyon 1, LYON, France
| | | |
Collapse
|
7
|
Gupta S, Butler SJ. Getting in touch with your senses: Mechanisms specifying sensory interneurons in the dorsal spinal cord. WIREs Mech Dis 2021; 13:e1520. [PMID: 34730293 PMCID: PMC8459260 DOI: 10.1002/wsbm.1520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/10/2021] [Accepted: 01/11/2021] [Indexed: 11/18/2022]
Abstract
The spinal cord is functionally and anatomically divided into ventrally derived motor circuits and dorsally derived somatosensory circuits. Sensory stimuli originating either at the periphery of the body, or internally, are relayed to the dorsal spinal cord where they are processed by distinct classes of sensory dorsal interneurons (dIs). dIs convey sensory information, such as pain, heat or itch, either to the brain, and/or to the motor circuits to initiate the appropriate response. They also regulate the intensity of sensory information and are the major target for the opioid analgesics. While the developmental mechanisms directing ventral and dorsal cell fates have been hypothesized to be similar, more recent research has suggested that dI fates are specified by novel mechanisms. In this review, we will discuss the molecular events that specify dorsal neuronal patterning in the spinal cord, thereby generating diverse dI identities. We will then discuss how this molecular understanding has led to the development of robust stem cell methods to derive multiple spinal cell types, including the dIs, and the implication of these studies for treating spinal cord injuries and neurodegenerative diseases. This article is categorized under: Neurological Diseases > Stem Cells and Development.
Collapse
Affiliation(s)
- Sandeep Gupta
- Department of NeurobiologyUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Samantha J. Butler
- Department of NeurobiologyUniversity of California, Los AngelesLos AngelesCaliforniaUSA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell ResearchUniversity of California, Los AngelesLos AngelesCaliforniaUSA
- Intellectual and Developmental Disabilities Research CenterUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| |
Collapse
|
8
|
Shinozuka T, Takada S. Morphological and Functional Changes of Roof Plate Cells in Spinal Cord Development. J Dev Biol 2021; 9:jdb9030030. [PMID: 34449633 PMCID: PMC8395932 DOI: 10.3390/jdb9030030] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 01/09/2023] Open
Abstract
The most dorsal region, or roof plate, is the dorsal organizing center of developing spinal cord. This region is also involved in development of neural crest cells, which are the source of migratory neural crest cells. During early development of the spinal cord, roof plate cells secrete signaling molecules, such as Wnt and BMP family proteins, which regulate development of neural crest cells and dorsal spinal cord. After the dorso-ventral pattern is established, spinal cord dynamically changes its morphology. With this morphological transformation, the lumen of the spinal cord gradually shrinks to form the central canal, a cavity filled with cerebrospinal fluid that is connected to the ventricular system of the brain. The dorsal half of the spinal cord is separated by a glial structure called the dorsal (or posterior) median septum. However, underlying mechanisms of such morphological transformation are just beginning to be understood. Recent studies reveal that roof plate cells dramatically stretch along the dorso-ventral axis, accompanied by reduction of the spinal cord lumen. During this stretching process, the tips of roof plate cells maintain contact with cells surrounding the shrinking lumen, eventually exposed to the inner surface of the central canal. Interestingly, Wnt expression remains in stretched roof plate cells and activates Wnt/β-catenin signaling in ependymal cells surrounding the central canal. Wnt/β-catenin signaling in ependymal cells promotes proliferation of neural progenitor and stem cells in embryonic and adult spinal cord. In this review, we focus on the role of the roof plate, especially that of Wnt ligands secreted by roof plate cells, in morphological changes occurring in the spinal cord.
Collapse
Affiliation(s)
- Takuma Shinozuka
- Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Aichi, Okazaki 444-8787, Japan
- National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Aichi, Okazaki 444-8787, Japan
- Correspondence: (T.S.); (S.T.)
| | - Shinji Takada
- Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Aichi, Okazaki 444-8787, Japan
- National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Aichi, Okazaki 444-8787, Japan
- Department of Basic Biology, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), 5-1 Higashiyama, Myodaiji, Aichi, Okazaki 444-8787, Japan
- Correspondence: (T.S.); (S.T.)
| |
Collapse
|
9
|
Mutation in the Ciliary Protein C2CD3 Reveals Organ-Specific Mechanisms of Hedgehog Signal Transduction in Avian Embryos. J Dev Biol 2021; 9:jdb9020012. [PMID: 33805906 PMCID: PMC8103285 DOI: 10.3390/jdb9020012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 12/22/2022] Open
Abstract
Primary cilia are ubiquitous microtubule-based organelles that serve as signaling hubs for numerous developmental pathways, most notably the Hedgehog (Hh) pathway. Defects in the structure or function of primary cilia result in a class of diseases called ciliopathies. It is well known that primary cilia participate in transducing a Hh signal, and as such ciliopathies frequently present with phenotypes indicative of aberrant Hh function. Interestingly, the exact mechanisms of cilia-dependent Hh signaling transduction are unclear as some ciliopathic animal models simultaneously present with gain-of-Hh phenotypes in one organ system and loss-of-Hh phenotypes in another. To better understand how Hh signaling is perturbed across different tissues in ciliopathic conditions, we examined four distinct Hh-dependent signaling centers in the naturally occurring avian ciliopathic mutant talpid2 (ta2). In addition to the well-known and previously reported limb and craniofacial malformations, we observed dorsal-ventral patterning defects in the neural tube, and a shortened gastrointestinal tract. Molecular analyses for elements of the Hh pathway revealed that the loss of cilia impact transduction of an Hh signal in a tissue-specific manner at variable levels of the pathway. These studies will provide increased knowledge into how impaired ciliogenesis differentially regulates Hh signaling across tissues and will provide potential avenues for future targeted therapeutic treatments.
Collapse
|
10
|
Abstract
In the adult mouse spinal cord, the ependymal cell population that surrounds the central canal is thought to be a promising source of quiescent stem cells to treat spinal cord injury. Relatively little is known about the cellular origin of ependymal cells during spinal cord development, or the molecular mechanisms that regulate ependymal cells during adult homeostasis. Using genetic lineage tracing based on the Wnt target gene Axin2, we have characterized Wnt-responsive cells during spinal cord development. Our results revealed that Wnt-responsive progenitor cells are restricted to the dorsal midline throughout spinal cord development, which gives rise to dorsal ependymal cells in a spatially restricted pattern. This is contrary to previous reports that suggested an exclusively ventral origin of ependymal cells, suggesting that ependymal cells may retain positional identities in relation to their neural progenitors. Our results further demonstrated that in the postnatal and adult spinal cord, all ependymal cells express the Wnt/β-catenin signaling target gene Axin2, as well as Wnt ligands. Genetic elimination of β-catenin or inhibition of Wnt secretion in Axin2-expressing ependymal cells in vivo both resulted in impaired proliferation, indicating that Wnt/β-catenin signaling promotes ependymal cell proliferation. These results demonstrate the continued importance of Wnt/β-catenin signaling for both ependymal cell formation and regulation. By uncovering the molecular signals underlying the formation and regulation of spinal cord ependymal cells, our findings thus enable further targeting and manipulation of this promising source of quiescent stem cells for therapeutic interventions.
Collapse
|
11
|
Hibi M, Matsuda K, Takeuchi M, Shimizu T, Murakami Y. Evolutionary mechanisms that generate morphology and neural-circuit diversity of the cerebellum. Dev Growth Differ 2017; 59:228-243. [DOI: 10.1111/dgd.12349] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/10/2017] [Accepted: 03/13/2017] [Indexed: 01/10/2023]
Affiliation(s)
- Masahiko Hibi
- Bioscience and Biotechnology Center; Nagoya University; Nagoya 464-8601 Japan
- Graduate School of Science; Nagoya University; Nagoya Aichi 464-8602 Japan
| | - Koji Matsuda
- Bioscience and Biotechnology Center; Nagoya University; Nagoya 464-8601 Japan
- Graduate School of Science; Nagoya University; Nagoya Aichi 464-8602 Japan
| | - Miki Takeuchi
- Bioscience and Biotechnology Center; Nagoya University; Nagoya 464-8601 Japan
| | - Takashi Shimizu
- Bioscience and Biotechnology Center; Nagoya University; Nagoya 464-8601 Japan
- Graduate School of Science; Nagoya University; Nagoya Aichi 464-8602 Japan
| | - Yasunori Murakami
- Graduate School of Science and Engineering; Ehime University; Matsuyama 790-8577 Japan
| |
Collapse
|
12
|
Knoedler JR, Subramani A, Denver RJ. The Krüppel-like factor 9 cistrome in mouse hippocampal neurons reveals predominant transcriptional repression via proximal promoter binding. BMC Genomics 2017; 18:299. [PMID: 28407733 PMCID: PMC5390390 DOI: 10.1186/s12864-017-3640-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Accepted: 03/17/2017] [Indexed: 12/22/2022] Open
Abstract
Background Krüppel-like factor 9 (Klf9) is a zinc finger transcription factor that functions in neural cell differentiation, but little is known about its genomic targets or mechanism of action in neurons. Results We used the mouse hippocampus-derived neuronal cell line HT22 to identify genes regulated by Klf9, and we validated our findings in mouse hippocampus. We engineered HT22 cells to express a Klf9 transgene under control of the tetracycline repressor, and used RNA sequencing to identify genes modulated by Klf9. We found 217 genes repressed and 21 induced by Klf9. We also engineered HT22 cells to co-express biotin ligase and a Klf9 fusion protein containing an N-terminal biotin ligase recognition peptide. Using chromatin-streptavidin precipitation (ChSP) sequencing we identified 3,514 genomic regions where Klf9 associated. Seventy-five percent of these were within 1 kb of transcription start sites, and Klf9 associated in chromatin with 60% of the repressed genes. We analyzed the promoters of several repressed genes containing Klf9 ChSP peaks using transient transfection reporter assays and found that Klf9 repressed promoter activity, which was abolished after mutation of Sp/Klf-like motifs. Knockdown or knockout of Klf9 in HT22 cells caused dysregulation of Klf9 target genes. Chromatin immunoprecipitation assays showed that Klf9 associated in chromatin from mouse hippocampus with genes identified by ChSP sequencing on HT22 cells, and expression of Klf9 target genes was dysregulated in the hippocampus of neonatal Klf9-null mice. Gene ontology analysis revealed that Klf9 genomic targets include genes involved in cystokeletal remodeling, Wnt signaling and inflammation. Conclusions We have identified genomic targets of Klf9 in hippocampal neurons and created a foundation for future studies on how it functions in chromatin, and regulates neuronal morphology and survival across the lifespan. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3640-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Joseph R Knoedler
- Neuroscience Graduate Program, The University of Michigan, Ann Arbor, MI, 48109, USA.,Current address: Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Arasakumar Subramani
- Department of Molecular, Cellular and Developmental Biology, The University of Michigan, 3065C Kraus Natural Science Building, Ann Arbor, MI, 48109, USA
| | - Robert J Denver
- Neuroscience Graduate Program, The University of Michigan, Ann Arbor, MI, 48109, USA. .,Department of Molecular, Cellular and Developmental Biology, The University of Michigan, 3065C Kraus Natural Science Building, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
13
|
Kousa YA, Mansour TA, Seada H, Matoo S, Schutte BC. Shared molecular networks in orofacial and neural tube development. Birth Defects Res 2017; 109:169-179. [DOI: 10.1002/bdra.23598] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 10/17/2016] [Accepted: 10/21/2016] [Indexed: 11/06/2022]
Affiliation(s)
- Youssef A. Kousa
- Pediatric Residency Program; Children's National Health System; Washington DC
| | - Tamer A. Mansour
- Department of Population Health and Reproduction; University of California; Davis California
- Department of Clinical Pathology, College of Medicine; Mansoura University; Egypt
| | - Haitham Seada
- Department of Computer Science and Engineering, Computational Optimization and Innovation Laboratory; Michigan State University; East Lansing Michigan
| | - Samaneh Matoo
- Department of Modern Science; Islamic Azad University-Tehran Medical Branch; Tehran Iran
| | - Brian C. Schutte
- Department of Microbiology and Molecular Genetics and the Department of Pediatrics and Human Development; Michigan State University; East Lansing Michigan
| |
Collapse
|
14
|
AP-2α and AP-2β regulate dorsal interneuron specification in the spinal cord. Neuroscience 2017; 340:232-242. [DOI: 10.1016/j.neuroscience.2016.10.055] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 10/18/2016] [Accepted: 10/21/2016] [Indexed: 11/22/2022]
|
15
|
Molina A, Pituello F. Playing with the cell cycle to build the spinal cord. Dev Biol 2016; 432:14-23. [PMID: 28034699 DOI: 10.1016/j.ydbio.2016.12.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 12/14/2016] [Accepted: 12/20/2016] [Indexed: 12/31/2022]
Abstract
A fundamental issue in nervous system development and homeostasis is to understand the mechanisms governing the balance between the maintenance of proliferating progenitors versus their differentiation into post-mitotic neurons. Accumulating data suggest that the cell cycle and core regulators of the cell cycle machinery play a major role in regulating this fine balance. Here, we focus on the interplay between the cell cycle and cellular and molecular events governing spinal cord development. We describe the existing links between the cell cycle and interkinetic nuclear migration (INM). We show how the different morphogens patterning the neural tube also regulate the cell cycle machinery to coordinate proliferation and patterning. We give examples of how cell cycle core regulators regulate transcriptionally, or post-transcriptionally, genes involved in controlling the maintenance versus the differentiation of neural progenitors. Finally, we describe the changes in cell cycle kinetics occurring during neural tube patterning and at the time of neuronal differentiation, and we discuss future research directions to better understand the role of the cell cycle in cell fate decisions.
Collapse
Affiliation(s)
- Angie Molina
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, France.
| | - Fabienne Pituello
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, France.
| |
Collapse
|
16
|
Masuda T, Ishitani T. Context-dependent regulation of the β-catenin transcriptional complex supports diverse functions of Wnt/β-catenin signaling. J Biochem 2016; 161:9-17. [PMID: 28013224 DOI: 10.1093/jb/mvw072] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 09/14/2016] [Indexed: 12/15/2022] Open
Abstract
Wnt/β-catenin signaling is activated repeatedly during an animal's lifespan, and it controls gene expression through its essential nuclear effector, β-catenin, to regulate embryogenesis, organogenesis, and adult homeostasis. Although the β-catenin transcriptional complex has the ability to induce the expression of many genes to exert its diverse roles, it chooses and transactivates a specific gene set from among its numerous target genes depending on the context. For example, the β-catenin transcriptional complex stimulates the expression of cell cycle-related genes and consequent cell proliferation in neural progenitor cells, while it promotes the expression of neural differentiation-related genes in differentiating neurons. Recent studies using animal and cell culture models have gradually improved our understanding of the molecular basis underlying such context-dependent actions of the β-catenin transcriptional complex. Here, we describe eight mechanisms that support β-catenin-mediated context-dependent gene regulation, and their spatio-temporal regulation during vertebrate development. In addition, we discuss their contribution to the diverse functions of Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Takamasa Masuda
- Division of Cell Regulation Systems, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka 812-8582, Japan
| | - Tohru Ishitani
- Division of Cell Regulation Systems, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka 812-8582, Japan
| |
Collapse
|
17
|
Klhl31 attenuates β-catenin dependent Wnt signaling and regulates embryo myogenesis. Dev Biol 2015; 402:61-71. [DOI: 10.1016/j.ydbio.2015.02.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 02/16/2015] [Accepted: 02/17/2015] [Indexed: 11/19/2022]
|
18
|
Miranda M, Galli LM, Enriquez M, Szabo LA, Gao X, Hannoush RN, Burrus LW. Identification of the WNT1 residues required for palmitoylation by Porcupine. FEBS Lett 2014; 588:4815-24. [PMID: 25451226 DOI: 10.1016/j.febslet.2014.11.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 10/27/2014] [Accepted: 11/12/2014] [Indexed: 11/26/2022]
Abstract
The post-translational palmitoylation of WNT morphogens is critical for proper signaling during embryogenesis and adult homeostasis. The addition of palmitoyl groups to WNT proteins is catalyzed by Porcupine (PORCN). However, the Wnt amino acid residues required for recognition and palmitoylation by PORCN have not been fully characterized. We show that WNT1 residues 214-234 are sufficient for PORCN-dependent palmitoylation of Ser224. Substitution of Ser224 with Thr, but not Cys, is tolerated in palmitoylation and biological assays. Our data highlight the importance of palmitoylation for WNT1 activity and establish PORCN as an O-acyl transferase for WNT1.
Collapse
Affiliation(s)
- M Miranda
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA
| | - L M Galli
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA
| | - M Enriquez
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA
| | - L A Szabo
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA
| | - X Gao
- Department of Early Discovery Biochemistry, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - R N Hannoush
- Department of Early Discovery Biochemistry, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - L W Burrus
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA.
| |
Collapse
|
19
|
Tiwari SK, Agarwal S, Seth B, Yadav A, Ray RS, Mishra VN, Chaturvedi RK. Inhibitory Effects of Bisphenol-A on Neural Stem Cells Proliferation and Differentiation in the Rat Brain Are Dependent on Wnt/β-Catenin Pathway. Mol Neurobiol 2014; 52:1735-1757. [PMID: 25381574 DOI: 10.1007/s12035-014-8940-1] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 10/13/2014] [Indexed: 02/07/2023]
Abstract
Neurogenesis, a process of generation of new neurons, occurs throughout the life in the hippocampus and sub-ventricular zone (SVZ). Bisphenol-A (BPA), an endocrine disrupter used as surface coating for packaged food cans, injures the developing and adult brain. However, the effects of BPA on neurogenesis and underlying cellular and molecular mechanism(s) are still unknown. Herein, we studied the effect(s) of prenatal and early postnatal exposure of low dose BPA on Wnt/β-catenin signaling pathway that controls different steps of neurogenesis such as neural stem cell (NSC) proliferation and neuronal differentiation. Pregnant rats were treated with 4, 40, and 400 μg BPA/kg body weight orally daily from gestational day 6 to postnatal day 21. Both in vivo and in vitro studies showed that BPA alters NSC proliferation and differentiation. BPA impaired NSC proliferation (5'-bromo-2'-deoxyuridine (BrdU(+)) and nestin(+) cells) and neuronal differentiation (BrdU/doublecortin(+) and BrdU/neuronal nuclei (NeuN(+)) cells) in the hippocampus and SVZ as compared to control. It significantly altered expression/protein levels of neurogenic genes and the Wnt pathway genes in the hippocampus. BPA reduced cellular β-catenin and p-GSK-3β levels and decreased β-catenin nuclear translocation, and cyclin-D1 and TCF/LEF promoter luciferase activity. Specific activation and blockage of the Wnt pathway suggested involvement of this pathway in BPA-mediated inhibition of neurogenesis. Further, blockage of GSK-3β activity by SB415286 and GSK-3β small interfering RNA (siRNA) attenuated BPA-induced downregulation of neurogenesis. Overall, these results suggest significant inhibitory effects of BPA on NSC proliferation and differentiation in the rat via the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Shashi Kant Tiwari
- Developmental Toxicology Division, Systems Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 80-MG Marg, Lucknow, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Swati Agarwal
- Developmental Toxicology Division, Systems Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 80-MG Marg, Lucknow, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Brashket Seth
- Developmental Toxicology Division, Systems Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 80-MG Marg, Lucknow, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Anuradha Yadav
- Developmental Toxicology Division, Systems Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 80-MG Marg, Lucknow, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Ratan Singh Ray
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India.,Photobiology Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 80-MG Marg, Lucknow, Uttar Pradesh, India
| | - Vijay Nath Mishra
- Department of Neurology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Rajnish Kumar Chaturvedi
- Developmental Toxicology Division, Systems Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 80-MG Marg, Lucknow, Uttar Pradesh, India. .,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India.
| |
Collapse
|
20
|
Galli LM, Szabo LA, Li L, Htaik YM, Onguka O, Burrus LW. Concentration-dependent effects of WNTLESS on WNT1/3A signaling. Dev Dyn 2014; 243:1095-105. [PMID: 24866848 PMCID: PMC4140996 DOI: 10.1002/dvdy.24149] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 04/15/2014] [Accepted: 04/29/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND WNTLESS (WLS) is a multi-transmembrane protein that transports Wnt ligands from the Golgi to the cell surface. Although WLS loss-of-function experiments in the developing central nervous system reveal phenotypes consistent with defects in WNT1 and WNT3A signaling, data from complementary gain-of-function experiments have not yet been reported. Here, we report the phenotypic consequences of WLS overexpression in cultured cells and in the developing chick spinal cord. RESULTS Overexpression of small amounts of WLS along with either WNT1 or WNT3A promotes the Wnt/β-catenin pathway in HEK293T cells, while overexpression of higher levels of WLS inhibits the Wnt/β-catenin pathway in these cells. Similarly, overexpressed WLS inhibits the Wnt/β-catenin pathway in the developing spinal cord, as assessed by cell proliferation and specification. These effects appear to be Wnt-specific as overexpression of WLS inhibits the expression of FZD10, a target of β-catenin-dependent transcription. CONCLUSIONS Our results show that overexpression of WLS inhibits Wnt/β-catenin signaling in the spinal cord. As the activation of the Wnt/β-catenin pathway in the spinal cord requires WNT1 or WNT3A, our results are consistent with a model in which the relative concentration of WLS to Wnt regulates WNT1/3A signaling in the developing spinal cord.
Collapse
Affiliation(s)
- Lisa M. Galli
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA 94132
| | - Linda A. Szabo
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA 94132
| | - Lydia Li
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA 94132
| | - Yin Min Htaik
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA 94132
| | - Ouma Onguka
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA 94132
| | - Laura W. Burrus
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, CA 94132
| |
Collapse
|
21
|
Galli LM, Munji RN, Chapman SC, Easton A, Li L, Onguka O, Ramahi JS, Suriben R, Szabo LA, Teng C, Tran B, Hannoush RN, Burrus LW. Frizzled10 mediates WNT1 and WNT3A signaling in the dorsal spinal cord of the developing chick embryo. Dev Dyn 2014; 243:833-843. [PMID: 24599775 DOI: 10.1002/dvdy.24123] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 03/03/2014] [Accepted: 03/04/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND WNT1 and WNT3A drive a dorsal to ventral gradient of β-catenin-dependent Wnt signaling in the developing spinal cord. However, the identity of the receptors mediating downstream functions remains poorly understood. RESULTS In this report, we show that the spatiotemporal expression patterns of FZD10 and WNT1/WNT3A are highly correlated. We further show that in the presence of LRP6, FZD10 promotes WNT1 and WNT3A signaling using an 8xSuperTopFlash reporter assay. Consistent with a functional role for FZD10, we demonstrate that FZD10 is required for proliferation in the spinal cord. Finally, by using an in situ proximity ligation assay, we observe an interaction between FZD10 and WNT1 and WNT3A proteins. CONCLUSIONS Together, our results identify FZD10 as a receptor for WNT1 and WNT3A in the developing chick spinal cord.
Collapse
Affiliation(s)
- Lisa M Galli
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA
| | - Roeben N Munji
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA
| | - Susan C Chapman
- Clemson University, Biological Sciences, Long Hall, Clemson, SC, 29634, USA
| | - Ann Easton
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA
| | - Lydia Li
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA
| | - Ouma Onguka
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA
| | - Joseph S Ramahi
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA
| | - Rowena Suriben
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA
| | - Linda A Szabo
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA
| | - Camilla Teng
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA
| | - Baouyen Tran
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA
| | - Rami N Hannoush
- Department of Early Discovery Biochemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Laura W Burrus
- Department of Biology, San Francisco State University, 1600 Holloway Avenue, San Francisco, CA 94132, USA
| |
Collapse
|
22
|
Saritas-Yildirim B, Silva EM. The role of targeted protein degradation in early neural development. Genesis 2014; 52:287-99. [PMID: 24623518 DOI: 10.1002/dvg.22771] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 03/05/2014] [Accepted: 03/07/2014] [Indexed: 11/08/2022]
Abstract
As neural stem cells differentiate into neurons during neurogenesis, the proteome of the cells is restructured by de novo expression and selective removal of regulatory proteins. The control of neurogenesis at the level of gene regulation is well documented and the regulation of protein abundance through protein degradation via the Ubiquitin/26S proteasome pathway is a rapidly developing field. This review describes our current understanding of the role of the proteasome pathway in neurogenesis. Collectively, the studies show that targeted protein degradation is an important regulatory mechanism in the generation of new neurons.
Collapse
|
23
|
Nishino J, Kim S, Zhu Y, Zhu H, Morrison SJ. A network of heterochronic genes including Imp1 regulates temporal changes in stem cell properties. eLife 2013; 2:e00924. [PMID: 24192035 PMCID: PMC3817382 DOI: 10.7554/elife.00924] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Stem cell properties change over time to match the changing growth and regeneration demands of tissues. We showed previously that adult forebrain stem cell function declines during aging because of increased expression of let-7 microRNAs, evolutionarily conserved heterochronic genes that reduce HMGA2 expression. Here we asked whether let-7 targets also regulate changes between fetal and adult stem cells. We found a second let-7 target, the RNA binding protein IMP1, that is expressed by fetal, but not adult, neural stem cells. IMP1 expression was promoted by Wnt signaling and Lin28a expression and opposed by let-7 microRNAs. Imp1-deficient neural stem cells were prematurely depleted in the dorsal telencephalon due to accelerated differentiation, impairing pallial expansion. IMP1 post-transcriptionally inhibited the expression of differentiation-associated genes while promoting the expression of self-renewal genes, including Hmga2. A network of heterochronic gene products including Lin28a, let-7, IMP1, and HMGA2 thus regulates temporal changes in stem cell properties. DOI:http://dx.doi.org/10.7554/eLife.00924.001 Stem cells are found throughout the body, and play key roles in promoting tissue growth during fetal development, and in maintaining tissues in the adult. When stem cells divide, they can either give rise to more stem cells, or they can generate specialized cells required for tissue function. However, the properties of stem cells must change over time to match the changing growth and regeneration demands of tissues. A previous study by Nishino et al. has shown that expression of a micro RNA molecule called let-7 increases throughout adulthood, and this reduces the activity of stem cells in older animals. Now, Nishino et al. report that let-7, and other genes it regulates, also control the dramatic changes that occur in the properties of stem cells between fetal development and adulthood. Whereas stem cells in the fetal forebrain undergo rapid division and are capable of generating many different cell types, stem cells in the adult forebrain divide less often and can generate only a few specific types of cell. While Nishino et al. performed their study on stem cells in the brain, their results are likely to apply also to stem cells in other tissues. Nishino et al. show that let-7 regulates the production of an RNA binding protein called IMP1. Mice with stem cells that lack IMP1 have a smaller cerebral cortex than normal mice because their stem cells undergo fewer rounds of division before committing to become brain cells. Additional experiments revealed that IMP1 inhibits the expression of genes that trigger stem cells to commit to specific fates and promotes the expression of genes related to self-renewal. These results indicate that the gene that encodes IMP1 is expressed in fetal neural stem cells, but not in adult neural stem cells, and that the reduced production of this protein contributes to the developmental switch from highly proliferative neural stem cells in the fetus to the more quiescent stem cells found in adults. Further studies are likely to identify many more targets of let-7 that enable stem cells to adapt their properties to the changing needs of the organism over time. These results are interesting because let-7-regulated networks were first discovered based on their ability to regulate the timing of developmental transitions in worms. This suggests that the mechanisms employed by mammalian tissue stem cells to regulate changes in their properties over time, are at least partly evolutionarily conserved mechanisms inherited from invertebrates. DOI:http://dx.doi.org/10.7554/eLife.00924.002
Collapse
Affiliation(s)
- Jinsuke Nishino
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | | | | | | | | |
Collapse
|
24
|
Teslaa JJ, Keller AN, Nyholm MK, Grinblat Y. Zebrafish Zic2a and Zic2b regulate neural crest and craniofacial development. Dev Biol 2013; 380:73-86. [PMID: 23665173 DOI: 10.1016/j.ydbio.2013.04.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 04/25/2013] [Accepted: 04/29/2013] [Indexed: 11/25/2022]
Abstract
Holoprosencephaly (HPE), the most common malformation of the human forebrain, is associated with defects of the craniofacial skeleton. ZIC2, a zinc-finger transcription factor, is strongly linked to HPE and to a characteristic set of dysmorphic facial features in humans. We have previously identified important functions for zebrafish Zic2 in the developing forebrain. Here, we demonstrate that ZIC2 orthologs zic2a and zic2b also regulate the forming zebrafish craniofacial skeleton, including the jaw and neurocranial cartilages, and use the zebrafish to study Zic2-regulated processes that may contribute to the complex etiology of HPE. Using temporally controlled Zic2a overexpression, we show that the developing craniofacial cartilages are sensitive to Zic2 elevation prior to 24hpf. This window of sensitivity overlaps the critical expansion and migration of the neural crest (NC) cells, which migrate from the developing neural tube to populate vertebrate craniofacial structures. We demonstrate that zic2b influences the induction of NC at the neural plate border, while both zic2a and zic2b regulate NC migratory onset and strongly contribute to chromatophore development. Both Zic2 depletion and early ectopic Zic2 expression cause moderate, incompletely penetrant mispatterning of the NC-derived jaw precursors at 24hpf, yet by 2dpf these changes in Zic2 expression result in profoundly mispatterned chondrogenic condensations. We attribute this discrepancy to an additional role for Zic2a and Zic2b in patterning the forebrain primordium, an important signaling source during craniofacial development. This hypothesis is supported by evidence that transplanted Zic2-deficient cells can contribute to craniofacial cartilages in a wild-type background. Collectively, these data suggest that zebrafish Zic2 plays a dual role during craniofacial development, contributing to two disparate aspects of craniofacial morphogenesis: (1) neural crest induction and migration, and (2) early patterning of tissues adjacent to craniofacial chondrogenic condensations.
Collapse
Affiliation(s)
- Jessica J Teslaa
- Department of Zoology, University of Wisconsin, Madison, WI 53706, USA
| | | | | | | |
Collapse
|
25
|
Lewis AE, Vasudevan HN, O'Neill AK, Soriano P, Bush JO. The widely used Wnt1-Cre transgene causes developmental phenotypes by ectopic activation of Wnt signaling. Dev Biol 2013; 379:229-34. [PMID: 23648512 DOI: 10.1016/j.ydbio.2013.04.026] [Citation(s) in RCA: 207] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 04/08/2013] [Accepted: 04/19/2013] [Indexed: 10/26/2022]
Abstract
The Wnt1-Cre transgenic mouse line is extensively used in the study of the development of the neural crest and its derivatives and the midbrain. The Wnt1 gene has important developmental roles in formation of the midbrain-hindbrain boundary, regulation of midbrain size, and neurogenesis of ventral midbrain dopaminergic (mDA) neurons. Here, we report that Wnt1-Cre transgenic mice exhibit phenotypes in multiple aspects of midbrain development. Significant expansion of the midbrain and increased proliferation in the developing inferior colliculus is associated with ectopic expression of Wnt1. Marked elevation of Wnt1 expression in the ventral midbrain is correlated with disruption of the differentiation program of ventral mDA neurons. We find that these phenotypes can be attributed to ectopic expression of Wnt1 from the Wnt1-Cre transgene leading to the ectopic activation of canonical Wnt/β-catenin signaling. Since these caveats could complicate the utility of Wnt1-Cre in some developmental circumstances, we report a new Wnt1-Cre2 transgenic mouse line that can serve the same purposes as the original without the associated phenotypic complications. These studies reveal an important caveat to a widely-used reagent, provide an improved version of this reagent, and indicate that the original Wnt1-Cre transgenic mouse line may be useful as a gain of function model for interrogating Wnt signaling mechanisms in multiple aspects of midbrain development.
Collapse
Affiliation(s)
- Ace E Lewis
- Department of Cell and Tissue Biology, Program in Craniofacial and Mesenchymal Biology and Institute for Human Genetics, University of California at San Francisco, San Francisco, CA 94143, United States
| | | | | | | | | |
Collapse
|
26
|
Arai Y, Huttner WB, Calegari F. Neural Stem Cells. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
27
|
Tadesse T, Cheng Q, Xu M, Baro DJ, Young LJ, Pallas SL. Regulation of ephrin-A expression in compressed retinocollicular maps. Dev Neurobiol 2012; 73:274-96. [PMID: 23008269 DOI: 10.1002/dneu.22059] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 09/14/2012] [Accepted: 09/18/2012] [Indexed: 11/11/2022]
Abstract
Retinotopic maps can undergo compression and expansion in response to changes in target size, but the mechanism underlying this compensatory process has remained a mystery. The discovery of ephrins as molecular mediators of Sperry's chemoaffinity process allows a mechanistic approach to this important issue. In Syrian hamsters, neonatal, partial (PT) ablation of posterior superior colliculus (SC) leads to compression of the retinotopic map, independent of neural activity. Graded, repulsive EphA receptor/ephrin-A ligand interactions direct the formation of the retinocollicular map, but whether ephrins might also be involved in map compression is unknown. To examine whether map compression might be directed by changes in the ephrin expression pattern, we compared ephrin-A2 and ephrin-A5 mRNA expression between normal SC and PT SC using in situ hybridization and quantitative real-time PCR. We found that ephrin-A ligand expression in the compressed maps was low anteriorly and high posteriorly, as in normal animals. Consistent with our hypothesis, the steepness of the ephrin gradient increased in the lesioned colliculi. Interestingly, overall levels of ephrin-A2 and -A5 expression declined immediately after neonatal target damage, perhaps promoting axon outgrowth. These data establish a correlation between changes in ephrin-A gradients and map compression, and suggest that ephrin-A expression gradients may be regulated by target size. This in turn could lead to compression of the retinocollicular map onto the reduced target. These findings have important implications for mechanisms of recovery from traumatic brain injury.
Collapse
Affiliation(s)
- Tizeta Tadesse
- Neuroscience Institute, Department of Biology, Graduate Program in Neurobiology & Behavior, Georgia State University, Atlanta, Georgia, USA
| | | | | | | | | | | |
Collapse
|
28
|
Mulligan KA, Cheyette BNR. Wnt signaling in vertebrate neural development and function. J Neuroimmune Pharmacol 2012; 7:774-87. [PMID: 23015196 DOI: 10.1007/s11481-012-9404-x] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 09/10/2012] [Indexed: 02/03/2023]
Abstract
Members of the Wnt family of secreted signaling proteins influence many aspects of neural development and function. Wnts are required from neural induction and axis formation to axon guidance and synapse development, and even help modulate synapse activity. Wnt proteins activate a variety of downstream signaling pathways and can induce a similar variety of cellular responses, including gene transcription changes and cytoskeletal rearrangements. This review provides an introduction to Wnt signaling pathways and discusses current research on their roles in vertebrate neural development and function.
Collapse
Affiliation(s)
- Kimberly A Mulligan
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA 94158, USA
| | | |
Collapse
|
29
|
Abstract
The cerebellum controls smooth and skillful movements and it is also involved in higher cognitive and emotional functions. The cerebellum is derived from the dorsal part of the anterior hindbrain and contains two groups of cerebellar neurons: glutamatergic and gamma-aminobutyric acid (GABA)ergic neurons. Purkinje cells are GABAergic and granule cells are glutamatergic. Granule and Purkinje cells receive input from outside of the cerebellum from mossy and climbing fibers. Genetic analysis of mice and zebrafish has revealed genetic cascades that control the development of the cerebellum and cerebellar neural circuits. During early neurogenesis, rostrocaudal patterning by intrinsic and extrinsic factors, such as Otx2, Gbx2 and Fgf8, plays an important role in the positioning and formation of the cerebellar primordium. The cerebellar glutamatergic neurons are derived from progenitors in the cerebellar rhombic lip, which express the proneural gene Atoh1. The GABAergic neurons are derived from progenitors in the ventricular zone, which express the proneural gene Ptf1a. The mossy and climbing fiber neurons originate from progenitors in the hindbrain rhombic lip that express Atoh1 or Ptf1a. Purkinje cells exhibit mediolateral compartmentalization determined on the birthdate of Purkinje cells, and linked to the precise neural circuitry formation. Recent studies have shown that anatomy and development of the cerebellum is conserved between mammals and bony fish (teleost species). In this review, we describe the development of cerebellar neurons and neural circuitry, and discuss their evolution by comparing developmental processes of mammalian and teleost cerebellum.
Collapse
Affiliation(s)
- Mitsuhiro Hashimoto
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya, Aichi, 466-8550, Japan.
| | | |
Collapse
|
30
|
Le Dréau G, Martí E. Dorsal-ventral patterning of the neural tube: a tale of three signals. Dev Neurobiol 2012; 72:1471-81. [PMID: 22821665 DOI: 10.1002/dneu.22015] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 02/16/2012] [Accepted: 02/17/2012] [Indexed: 12/16/2022]
Abstract
Development of the vertebrate nervous system begins with the acquisition of neural identity from the midline dorsal-ectodermal cells of the gastrulating embryos. The subsequent progressive specification of the neural plate along its anterior-posterior and dorsal-ventral (DV) axes allows the generation of the tremendous variety of neuronal and glial cells that compose the vertebrate central nervous system (CNS). Studies on the development of the spinal cord, the anatomically simplest part of the CNS, have generated most of our current knowledge on the signaling events and the genetic networks that orchestrate the DV patterning of the neural plate. In this review, we discuss the recent advances in our understanding of these events and highlight unresolved questions. We focused our attention on the activity and the integration of the three main instructive cues: Sonic hedgehog, the Wnts and the Bone Morphogenetic Proteins, giving particular attention to the less well understood dorsal signaling events.
Collapse
Affiliation(s)
- Gwenvael Le Dréau
- Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, Spain
| | | |
Collapse
|
31
|
Hibi M, Shimizu T. Development of the cerebellum and cerebellar neural circuits. Dev Neurobiol 2012; 72:282-301. [DOI: 10.1002/dneu.20875] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
32
|
Precerebellar cell groups in the hindbrain of the mouse defined by retrograde tracing and correlated with cumulative Wnt1-cre genetic labeling. THE CEREBELLUM 2012; 10:570-84. [PMID: 21479970 DOI: 10.1007/s12311-011-0266-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The precerebellar nuclei are hindbrain and spinal cord centers that send fibers to the cerebellum. The neurons of the major hindbrain precerebellar nuclei are derived from the rhombic lip. Wnt1, a developmentally important gene involved in intercellular signaling, is expressed in the developing rhombic lip. We sought to investigate the relationship between the cell clusters expressing Wnt1 and the precerebellar nuclei in the hindbrain. We therefore defined the hindbrain precerebellar nuclei by retrograde tracing, following cerebellar injections of HRP, and compared these results with the cell clusters expressing Wnt1 in newborn mice. We found that 39 distinct hindbrain nuclei project to the cerebellum. Of these nuclei, all but three (namely the oral pontine reticular nucleus, the caudal pontine reticular nucleus, and the subcoeruleus nucleus) contain neurons expressing Wnt1. This shows a high degree of overlap between the precerebellar nuclei and the nuclei that express Wnt1. However, it should be noted that neurons expressing Wnt1 are also found in the superior olivary complex, which is a basal plate derivative lacking cerebellar projections.
Collapse
|
33
|
Lee HK, Deneen B. Daam2 is required for dorsal patterning via modulation of canonical Wnt signaling in the developing spinal cord. Dev Cell 2012; 22:183-96. [PMID: 22227309 DOI: 10.1016/j.devcel.2011.10.025] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Revised: 09/14/2011] [Accepted: 10/26/2011] [Indexed: 10/14/2022]
Abstract
The Daam family of proteins consists of Daam1 and Daam2. Although Daam1 participates in noncanonical Wnt signaling during gastrulation, Daam2 function remains completely uncharacterized. Here we describe the role of Daam2 in canonical Wnt signal transduction during spinal cord development. Loss-of-function studies revealed that Daam2 is required for dorsal progenitor identities and canonical Wnt signaling. These phenotypes are rescued by β-catenin, demonstrating that Daam2 functions in dorsal patterning through the canonical Wnt pathway. Complementary gain-of-function studies demonstrate that Daam2 amplifies Wnt signaling by potentiating ligand activation. Biochemical examination found that Daam2 association with Dvl3 is required for Wnt activity and dorsal patterning. Moreover, Daam2 stabilizes Dvl3/Axin2 binding, resulting in enhanced intracellular assembly of Dvl3/Axin2 complexes. These studies demonstrate that Daam2 modulates the formation of Wnt receptor complexes, revealing new insight into the functional diversity of Daam proteins and how canonical Wnt signaling contributes to pattern formation in the developing spinal cord.
Collapse
Affiliation(s)
- Hyun Kyoung Lee
- Center for Cell and Gene Therapy, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | | |
Collapse
|
34
|
Wilson NH, Stoeckli ET. Sonic Hedgehog regulates Wnt activity during neural circuit formation. VITAMINS AND HORMONES 2012; 88:173-209. [PMID: 22391304 DOI: 10.1016/b978-0-12-394622-5.00008-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Gradients of secreted morphogens, such as Sonic hedgehog (Shh), Wnt, and TGFβ/Bmp, have classically been shown to control many aspects of early development by regulating cell proliferation and determining cell fate. However, recent studies demonstrate that these molecules also play important and evolutionarily conserved roles in later aspects of neural development. Depending on the context, these molecules can elicit gene transcription in the nucleus, or alternatively can provide instructive signals at the growth cone that induce local and rapid changes in cytoskeletal organization. Shh can activate different cellular transduction pathways via its binding to alternative coreceptor complexes or simply by adaptation of its "classical" signaling pathway. However, in most of its activities during neural development, Shh does not act alone but rather in concert with other morphogens, particularly the Wnts. This review provides an overview of the mechanisms by which Shh signaling acts in concert with Wnts to mediate a myriad of cellular processes that are required for neural circuit formation.
Collapse
Affiliation(s)
- Nicole H Wilson
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
35
|
Sanchez-Simon F, Ledo A, Arevalo R, Rodriguez R. New insights into opioid regulatory pathways: influence of opioids on Wnt1 expression in zebrafish embryos. Neuroscience 2012; 200:237-47. [DOI: 10.1016/j.neuroscience.2011.10.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 10/14/2011] [Accepted: 10/15/2011] [Indexed: 01/24/2023]
|
36
|
Marinaro C, Pannese M, Weinandy F, Sessa A, Bergamaschi A, Taketo MM, Broccoli V, Comi G, Götz M, Martino G, Muzio L. Wnt signaling has opposing roles in the developing and the adult brain that are modulated by Hipk1. ACTA ACUST UNITED AC 2011; 22:2415-27. [PMID: 22095214 DOI: 10.1093/cercor/bhr320] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The canonical Wnt/Wingless pathway is implicated in regulating cell proliferation and cell differentiation of neural stem/progenitor cells. Depending on the context, β-Catenin, a key mediator of the Wnt signaling pathway, may regulate either cell proliferation or differentiation. Here, we show that β-Catenin signaling regulates the differentiation of neural stem/progenitor cells in the presence of the β-Catenin interactor Homeodomain interacting protein kinase-1 gene (Hipk1). On one hand, Hipk1 is expressed at low levels during the entire embryonic forebrain development, allowing β-Catenin to foster proliferation and to inhibit differentiation of neural stem/progenitor cells. On the other hand, Hipk1 expression dramatically increases in neural stem/progenitor cells, residing within the subventricular zone (SVZ), at the time when the canonical Wnt signaling induces cell differentiation. Analysis of mouse brains electroporated with Hipk1, and the active form of β-Catenin reveals that coexpression of both genes induces proliferating neural stem/progenitor cells to escape the cell cycle. Moreover, in SVZ derive neurospheres cultures, the overexpression of both genes increases the expression of the cell-cycle inhibitor P16Ink4. Therefore, our data confirm that the β-Catenin signaling plays a dual role in controlling cell proliferation/differentiation in the brain and indicate that Hipk1 is the crucial interactor able to revert the outcome of β-Catenin signaling in neural stem/progenitor cells of adult germinal niches.
Collapse
Affiliation(s)
- Cinzia Marinaro
- Neuroimmunology Unit, Institute of Experimental Neurology, Division of Neuroscience San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Taniguchi Y, Tanaka O, Sekiguchi M, Takekoshi S, Tsukamoto H, Kimura M, Imai K, Inoko H. Enforced expression of the transcription factor HOXD3 under the control of the Wnt1 regulatory element modulates cell adhesion properties in the developing mouse neural tube. J Anat 2011; 219:589-600. [PMID: 21929743 DOI: 10.1111/j.1469-7580.2011.01425.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
HOX genes expressed in a specific spatial and temporal manner play a crucial role in determining the body plan during the early development of vertebrates. In adult tissues, many HOX genes participate in normal hematopoiesis and carcinogenesis. We previously found that overexpression of the homeobox gene HOXD3 alters expression levels of cell adhesion molecules in human cancer cell lines. Here, we have investigated whether HOXD3 expression is related to the cell adhesion processes during mouse development focusing on dorsal midline cells or roof-plate cells of the neural tube and neural crest cells. We created transgenic mouse embryos, in which HOXD3 is expressed in the dorsal midline under the control of the Wnt1 regulatory element, and analyzed these embryos at embryonic day 10.5-13.5. In HOXD3-expressing transgenic embryos, although neural crest-derived structures in the trunk region appeared to be normal, striking abnormalities were found in the neural tube. In transgenic embryos expressing the lacZ gene under the control of the Wnt1 regulatory element, expression of lacZ was restricted to roof-plate cells within the neural tube. By contrast, in HOXD3-expressing transgenic embryos, expression of HOXD3 was not only located in the dorsal neural tube, but also had spread inside the ventricular zone in more ventral regions of the neural tube. These findings show that the HOXD3 transgene is expressed more broadly than the Wnt1 gene is normally expressed. Expression of both Wnt1 and Msx1, marker genes in the roof plate, was further extended ventrally in HOXD3-expressing embryos than in normal embryos, suggesting that expression of the HOXD3 transgene expands the roof plate ventrally within the neural tube. In the ventricular zone of HOXD3-expressing embryos at embryonic day 10.5, we observed an increase in the number of mitotic cells and failure of interkinetic nuclear migration of progenitor cells. Furthermore, in HOXD3-expressing embryos at embryonic day 12.5, the ventricular zone, in which progenitor cells became more loosely connected to each other, was composed of a large number of cells that did not express N-cadherin. Our results indicate that expression of HOXD3 is closely associated with modulation of cell-adhesive properties during embryonic development.
Collapse
Affiliation(s)
- Yasushi Taniguchi
- Division of Basic Molecular Science and Molecular Medicine, School of Medicine, Tokai University, Isehara, Kanagawa, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Ahrens MJ, Romereim S, Dudley AT. A re-evaluation of two key reagents for in vivo studies of Wnt signaling. Dev Dyn 2011; 240:2060-8. [PMID: 21793100 DOI: 10.1002/dvdy.22704] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2011] [Indexed: 11/10/2022] Open
Abstract
Conditional mutations and transcription-based reporters are important new tools for exploring the dynamic functions of biological pathways in vivo. While studying the role of the Wnt signaling pathway in cartilage, we observed that the β-catenin-dependent reporter TOPGAL was expressed in chondrocytes in which β-catenin was conditionally inactivated using a Col2a1::cre driver. Here we show that in these embryos recombination is complete and full-length β-catenin protein is absent in chondrocytes. Although a null allele in this context, the recombined β-catenin locus produces a stable transcript that encodes a truncated protein. The truncated protein alone fails to activate TOPFLASH, but strongly potentiates reporter activity in the presence of expressed β-catenin or Tcf4. Together, these data show that each mouse model exhibits specific undesirable properties, findings that strongly suggest the need for specific standards to ensure proper validation of this new generation of genetic tools.
Collapse
Affiliation(s)
- Molly J Ahrens
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois, USA
| | | | | |
Collapse
|
39
|
Horner VL, Caspary T. Disrupted dorsal neural tube BMP signaling in the cilia mutant Arl13b hnn stems from abnormal Shh signaling. Dev Biol 2011; 355:43-54. [PMID: 21539826 PMCID: PMC3119544 DOI: 10.1016/j.ydbio.2011.04.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Revised: 03/22/2011] [Accepted: 04/14/2011] [Indexed: 12/22/2022]
Abstract
In the embryonic neural tube, multiple signaling pathways work in concert to create functional neuronal circuits in the adult spinal cord. In the ventral neural tube, Sonic hedgehog (Shh) acts as a graded morphogen to specify neurons necessary for movement. In the dorsal neural tube, bone morphogenetic protein (BMP) and Wnt signals cooperate to specify neurons involved in sensation. Several signaling pathways, including Shh, rely on primary cilia in vertebrates. In this study, we used a mouse mutant with abnormal cilia, Arl13b(hnn), to study the relationship between cilia, cell signaling, and neural tube patterning. Arl13b(hnn) mutants have abnormal ventral neural tube patterning due to disrupted Shh signaling; in addition, dorsal patterning defects occur, but the cause of these is unknown. Here we show that the Arl13b(hnn) dorsal patterning defects result from abnormal BMP signaling. In addition, we find that Wnt ligands are abnormally expressed in Arl13b(hnn) mutants; surprisingly, however, downstream Wnt signaling is normal. We demonstrate that Arl13b is required non-autonomously for BMP signaling and Wnt ligand expression, indicating that the abnormal Shh signaling environment in Arl13b(hnn) embryos indirectly causes dorsal defects.
Collapse
Affiliation(s)
- Vanessa L. Horner
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Tamara Caspary
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
40
|
Miller-Delaney SFC, Lieberam I, Murphy P, Mitchell KJ. Plxdc2 is a mitogen for neural progenitors. PLoS One 2011; 6:e14565. [PMID: 21283688 PMCID: PMC3024984 DOI: 10.1371/journal.pone.0014565] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Accepted: 12/03/2010] [Indexed: 12/27/2022] Open
Abstract
The development of different brain regions involves the coordinated control of proliferation and cell fate specification along and across the neuraxis. Here, we identify Plxdc2 as a novel regulator of these processes, using in ovo electroporation and in vitro cultures of mammalian cells. Plxdc2 is a type I transmembrane protein with some homology to nidogen and to plexins. It is expressed in a highly discrete and dynamic pattern in the developing nervous system, with prominent expression in various patterning centres. In the chick neural tube, where Plxdc2 expression parallels that seen in the mouse, misexpression of Plxdc2 increases proliferation and alters patterns of neurogenesis, resulting in neural tube thickening at early stages. Expression of the Plxdc2 extracellular domain alone, which can be cleaved and shed in vivo, is sufficient for this activity, demonstrating a cell non-autonomous function. Induction of proliferation is also observed in cultured embryonic neuroepithelial cells (ENCs) derived from E9.5 mouse neural tube, which express a Plxdc2-binding activity. These experiments uncover a direct molecular activity of Plxdc2 in the control of proliferation, of relevance in understanding the role of this protein in various cancers, where its expression has been shown to be altered. They also implicate Plxdc2 as a novel component of the network of signalling molecules known to coordinate proliferation and differentiation in the developing nervous system.
Collapse
Affiliation(s)
| | - Ivo Lieberam
- Howard Hughes Medical Institute, Department of Biochemistry and Molecular Biophysics, Center for Neurobiology and Behavior, Columbia University, New York, New York, United States of America
| | - Paula Murphy
- Department of Zoology, Trinity College Dublin, Dublin, Ireland
| | - Kevin J. Mitchell
- Smurfit Institute of Genetics and Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
41
|
Neural Stem Cells. Regen Med 2011. [DOI: 10.1007/978-90-481-9075-1_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
42
|
Yoshinaga Y, Kagawa T, Shimizu T, Inoue T, Takada S, Kuratsu JI, Taga T. Wnt3a promotes hippocampal neurogenesis by shortening cell cycle duration of neural progenitor cells. Cell Mol Neurobiol 2010; 30:1049-58. [PMID: 20589426 PMCID: PMC11498854 DOI: 10.1007/s10571-010-9536-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Accepted: 06/10/2010] [Indexed: 02/03/2023]
Abstract
The effects of Wnt signaling on neural progenitor cells have been controversial. Activation of the canonical Wnt signaling pathway either promotes neural progenitor cell proliferation or accelerates their differentiation into postmitotic neurons. This study demonstrates that activation of the Wnt signaling pathway by itself induces neural progenitor cell proliferation but does not directly affect neuronal differentiation processes. To investigate whether Wnt signaling promotes expansion and/or differentiation of neural progenitor cells in the developing hippocampus, we prepared primary mouse hippocampal progenitors and treated them with Wnt3a in a chemically defined culture medium. Wnt3a increased the total number of cells, including the numbers of Ki67(+) proliferating cells and Tuj1(+) differentiated neurons. This result verified that Wnt3a promoted neural progenitor cell proliferation. Meanwhile, Wnt3a did not appear to actively enhance the neuronal differentiation process itself, because (1) the ratio of Tuj1(+) cells to the total cells, and (2) the ratio of BrdU(+) Tuj1(+) cells to the total BrdU(+) cells, were both comparable between cultures with or without Wnt3a. Indeed, Wnt3a caused no significant change in either cell survival or the proportion of symmetric and asymmetric cell divisions that directly affected neuron production. We finally demonstrated that the Wnt3a treatment simply shortened cell cycle duration of neural progenitor cells by 2.9 h. The accelerated cell cycle progression without affecting the ratio of symmetric/asymmetric cell divisions explains how Wnt signaling per se leads to the expansion of both proliferative cell population and differentiated neuronal cell population.
Collapse
Affiliation(s)
- Yutaka Yoshinaga
- Division of Cell Fate Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Kumamoto, 860-0811 Japan
- Department of Neurosurgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-0811 Japan
| | - Tetsushi Kagawa
- Division of Cell Fate Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Kumamoto, 860-0811 Japan
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510 Japan
| | - Takeshi Shimizu
- Division of Cell Fate Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Kumamoto, 860-0811 Japan
- Present Address: Vertebrate Axis Formation Team, Creative Research Promoting Program, RIKEN Center for Developmental Biology, 2-2-3 Minatozimaminami-machi, Cyuo-ku, Kobe, Hyogo 650-0047 Japan
| | - Toshihiro Inoue
- Division of Cell Fate Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Kumamoto, 860-0811 Japan
- Department of Ophthalmology and Visual Science, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-0811 Japan
| | - Shinji Takada
- Division of Molecular and Developmental Biology, Okazaki Institute for Integrative Biosciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787 Japan
| | - Jun-ichi Kuratsu
- Department of Neurosurgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-0811 Japan
| | - Tetsuya Taga
- Division of Cell Fate Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Kumamoto, 860-0811 Japan
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510 Japan
| |
Collapse
|
43
|
Ulloa F, Martí E. Wnt won the war: antagonistic role of Wnt over Shh controls dorso-ventral patterning of the vertebrate neural tube. Dev Dyn 2010; 239:69-76. [PMID: 19681160 DOI: 10.1002/dvdy.22058] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The spinal cord has been used as a model to dissect the mechanisms that govern the patterning of tissues during animal development, since the principles that rule the dorso-ventral patterning of the neural tube are applicable to other systems. Signals that determine the dorso-ventral axis of the spinal cord include Sonic hedgehog (Shh), acting as a bona fide morphogenetic signal to determine ventral progenitor identities, and members of the Bmp and the Wnt families, acting in the dorsal neural tube. Although Wnts have been initially recognized as important in proliferation of neural progenitor cells, their role in the dorso-ventral patterning has been controversial. In this review, we discuss recent reports that show an important contribution of the Wnt canonical pathway in dorso-ventral pattern formation. These data allow building a model by which the ventralizing activity of Shh is antagonized by Wnt activity through the expression of Gli3, a potent inhibitor of the Shh pathway. Therefore, antagonistic interactions between canonical Wnt, promoting dorsal identities, and Shh pathways, inducing ventral ones, would define the dorso-ventral patterning of the developing central nervous system.
Collapse
Affiliation(s)
- Fausto Ulloa
- Institute for Research in Biomedicine, Parc Cientific de Barcelona; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Department of Cell Biology, University of Barcelona, Barcelona, Spain
| | | |
Collapse
|
44
|
Abstract
Epithelial tubes are crucial to the function of organ systems including the cardiovascular system, pulmonary system, gastrointestinal tract, reproductive organ systems, excretory system, and auditory system. Using a variety of animal model systems, recent studies have substantiated the role of Wnt signaling via the canonical/beta-catenin-mediated trajectory, the non-canonical Wnt trajectories, or both, in forming epithelial tubular tissues. This review focuses on the involvement of the Wnt pathways in the induction, specification, proliferation, and morphogenesis involved in tubulogenesis within tissues including the lungs, kidneys, ears, mammary glands, gut, and heart. The ultimate goal is to describe the developmental processes forming the various tubulogenic organ systems to determine the relationships between these processes.
Collapse
Affiliation(s)
- Rachel K Miller
- Department of Biochemistry and Molecular Biology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA.
| | | |
Collapse
|
45
|
Garcia-Morales C, Liu CH, Abu-Elmagd M, Hajihosseini MK, Wheeler GN. Frizzled-10 promotes sensory neuron development in Xenopus embryos. Dev Biol 2009; 335:143-55. [PMID: 19716814 DOI: 10.1016/j.ydbio.2009.08.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 08/20/2009] [Accepted: 08/21/2009] [Indexed: 12/23/2022]
Abstract
Formation of the vertebrate nervous system requires coordinated cell-cell interactions, intracellular signalling events, gene transcription, and morphogenetic cell movements. Wnt signalling has been involved in regulating a wide variety of biological processes such as embryonic patterning, cell proliferation, cell polarity, motility, and the specification of cell fate. Wnt ligands associate with their receptors, members of the frizzled family (Fz). In Xenopus, five members of the frizzled family are expressed in the early nervous system. We have investigated the role of Xenopus frizzled-10 (Fz10) in neural development. We show that Fz10 is expressed in the dorsal neural ectoderm and neural folds in the region where primary sensory neurons develop. Fz10 mediates canonical Wnt signalling and interacts with Wnt1 and Wnt8 but not Wnt3a as shown in synergy assays. We find that Fz10 is required for the late stages of sensory neuron differentiation. Overexpression of Fz10 in Xenopus leads to an increase in the number of sensory neurons. Loss of Fz10 function using morpholinos inhibits the development of sensory neurons in Xenopus at later stages of neurogenesis and this can be rescued by co-injection of modified Fz10B and beta-catenin. In mouse P19 cells induced by retinoic acid to undergo neural differentiation, overexpression of Xenopus Fz10 leads to an increase in the number of neurons generated while siRNA knockdown of endogenous mouse Fz10 inhibits neurogenesis. Thus we propose Fz10 mediates Wnt1 signalling to determine sensory neural differentiation in Xenopus in vivo and in mouse cell culture.
Collapse
|
46
|
Effect of protocatechuic acid from Alpinia oxyphylla on proliferation of human adipose tissue-derived stromal cells in vitro. Mol Cell Biochem 2009; 330:47-53. [PMID: 19363594 DOI: 10.1007/s11010-009-0099-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2009] [Accepted: 03/30/2009] [Indexed: 12/31/2022]
Abstract
The effect of protocatechuic acid (PCA) from Alpinia oxyphylla and catapol from Rehmannia on the proliferation capacity of human adipose tissue-derived stromal cells (hADSCs) was investigated in vitro. Cell counts showed that treatment of hADSCs with PCA for 48 h increased the cell number in a dose-dependent manner, while no obvious effect of catapol on the proliferation of hADSCs was observed. In addition, the cell number of hADSCs treated by 1.5 mM PCA increased in a time-dependent manner. The flow cytometric analysis of DNA content demonstrated the cell cycle progress from the G0/G1 phase to the S phase. Western blot analysis revealed the elevated expression of cyclin D1 in hADSCs induced by PCA treatment. Cyclin D1-siRNA transfection significantly inhibit the promotion of cell proliferation by PCA. Furthermore, the flow cytometric analysis of the cell surface antigens and the multidifferential potential tests of PCA-treated hADSCs showed that the cells retained their functional characteristics of multipotential mesenchymal progenitors. It is concluded that PCA can effectively up-regulate the proliferation of hADSCs.
Collapse
|
47
|
Abstract
In all vertebrate brains, there is a period of widespread embryonic neurogenesis followed by specific regional neurogenesis that continues into adult stages. The Wnt signaling pathway, which is essential for numerous developmental processes, has also been suggested to be involved in neurogenesis. To help investigate the exact roles of canonical Wnt signaling in neurogenesis, here we examine the identity of Wnt-responsive cells in the zebrafish hypothalamus. This tissue is a useful diencephalic neurogenesis model containing evolutionarily conserved populations of neurons. We first performed in situ hybridization to show the expression patterns of Tcf family members and a canonical Wnt signaling reporter in the 50 hpf embryonic hypothalamus and larval/adult hypothalamus. We then used immunohistochemistry to identify the cell types of Wnt-responsive and Lef1-positive cells in both 50 hpf embryonic and adult hypothalamus. Our results indicate that Wnt-responsive cells in the hypothalamus are likely to be both mitotic progenitors and postmitotic precursors at embryonic stages, but only precursors at the adult stage. These data suggest that canonical Wnt signaling may be functionally required for maintenance of neural progenitor and precursor pools in the embryo, and for ongoing neurogenesis in the adult zebrafish.
Collapse
Affiliation(s)
- Xu Wang
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah
| | - Ji Eun Lee
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah
| | - Richard I. Dorsky
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah
- The Brain Institute, University of Utah, Salt Lake City, Utah
| |
Collapse
|
48
|
Yin ZS, Zhang H, Wang W, Hua XY, Hu Y, Zhang SQ, Li GW. Wnt-3a protein promote neuronal differentiation of neural stem cells derived from adult mouse spinal cord. Neurol Res 2008; 29:847-54. [PMID: 17609021 DOI: 10.1179/016164107x223539] [Citation(s) in RCA: 14] [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
BACKGROUND Wnt proteins as growth factor have multiple functions in neural development, and especially serve key roles in differentiation and development. Wnt-3a is an intercellular signaling molecule that is involved in a variety of morphogenetic events. The purpose of this study was to investigate the effects of Wnt-3a signal protein on proliferation and differentiation of neural stem cells derived from adult mouse spinal cord. METHODS Adult mouse neural stem cells were cultured with serum free incubation. The recombined plasmid pSecTag2/Hygro B-Wnt3a for eukaryotic expression transfected adult neural stem cell, then the expression protein was detected by Western blot. The differentiation of adult neural stem cells was identified by the immunocytochemical technique. RESULTS The inducing differentiated rates of neurons were improved greatly by Wnt-3a protein compared with control (p<0.05). CONCLUSION Wnt-3a has obvious influence on the neuronal differentiation of adult neural stem cell.
Collapse
Affiliation(s)
- Zong-Sheng Yin
- Department of Orthopaedics, First Affiliated Hospital of Anhui Medical University, Hefei 230032, China.
| | | | | | | | | | | | | |
Collapse
|
49
|
Nordin N, Li M, Mason JO. Expression profiles of Wnt genes during neural differentiation of mouse embryonic stem cells. CLONING AND STEM CELLS 2008; 10:37-48. [PMID: 18315501 DOI: 10.1089/clo.2007.0060] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The Wnt family of secreted signaling proteins regulates many aspects of animal development and the behavior of several types of stem cells, including embryonic stem (ES) cells. Activation of canonical Wnt signaling has been shown to either inhibit or promote the differentiation of ES cells into neurons, depending on the stage of differentiation. Here, we describe the expression of all 19 mouse Wnt genes during this process. Using the well-established retinoic acid induction protocol we found that all Wnt genes except Wnt8b are expressed as ES cells differentiate into neurons, many of them in dynamic patterns. The expression pattern of 12 Wnt genes was analyzed quantitatively at 2-day intervals throughout neural differentiation, showing that multiple Wnt genes are expressed at each stage. A large proportion of these, including both canonical and noncanonical Wnts, are expressed at highest levels during later stages of differentiation. The complexity of the patterns observed indicates that disentangling specific roles for individual Wnt genes in the differentiation process will be a significant challenge.
Collapse
Affiliation(s)
- Norshariza Nordin
- Centres for Integrative Physiology and Neuroscience Research, School of Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD United Kingdom
| | | | | |
Collapse
|
50
|
Lee K, Huh T, Lee CJ, Rhee M. Zic3zdefines the dorsal and vegetal neuroectoderm in the zebrafish embryonic development. Anim Cells Syst (Seoul) 2008. [DOI: 10.1080/19768354.2008.9647150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|