1
|
Decreased sensitivity in adolescent versus adult rats to the antidepressant-like effects of cannabidiol. Psychopharmacology (Berl) 2020; 237:1621-1631. [PMID: 32086540 DOI: 10.1007/s00213-020-05481-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 02/12/2020] [Indexed: 12/19/2022]
Abstract
RATIONALE Cannabidiol is a non-psychoactive phytocannabinoid with great therapeutic potential in diverse psychiatric disorders; however, its antidepressant potential has been mainly ascertained in adult rats. OBJECTIVES To compare the antidepressant-like response induced by cannabidiol in adolescent and adult rats and the possible parallel modulation of hippocampal neurogenesis. METHODS Male Sprague-Dawley rats were repeatedly treated with cannabidiol (3, 10, and 30 mg/kg) or vehicle (1 mL/kg) during adolescence (postnatal days, PND 27-33) or adulthood (PND 141-147) and exposed to 3 consecutive tests (forced swim, open field, two-bottle choice) that quantified behavioral despair, anxiety, and sucrose intake respectively. RESULTS Cannabidiol induced differential effects depending on the age and dose administered, with a decreased sensitivity observed in adolescent rats: (1) cannabidiol (30 mg/kg) decreased body weight only in adult rats; (2) cannabidiol ameliorated behavioral despair in adolescent and adult rats, but with a different dose sensitivity (10 vs. 30 mg/kg), and with a different extent (2 vs. 21 days post-treatment); (3) cannabidiol did not modulate anxiety-like behavior at any dose tested in adolescent or adult rats; and (4) cannabidiol increased sucrose intake in adult rats. CONCLUSIONS Our findings support the notion that cannabidiol exerts antidepressant- and anorexigenic-like effects in adult rats and demonstrate a decreased potential when administered in adolescent rats. Moreover, since cannabidiol did not modulate hippocampal neurogenesis (cell proliferation and early neuronal survival) in adolescent or adult rats, the results revealed potential antidepressant-like effects induced by cannabidiol without the need of regulating hippocampal neurogenesis.
Collapse
|
2
|
In Vitro Differentiation of Human Skin-Derived Cells into Functional Sensory Neurons-Like. Cells 2020; 9:cells9041000. [PMID: 32316463 PMCID: PMC7226083 DOI: 10.3390/cells9041000] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 02/07/2023] Open
Abstract
Skin-derived precursor cells (SKPs) are neural crest stem cells that persist in certain adult tissues, particularly in the skin. They can generate a large type of cell in vitro, including neurons. SKPs were induced to differentiate into sensory neurons (SNs) by molecules that were previously shown to be important for the generation of SNs: purmorphamine, CHIR99021, BMP4, GDNF, BDNF, and NGF. We showed that the differentiation of SKPs induced the upregulation of neurogenins. At the end of the differentiation protocol, transcriptional analysis was performed on BRN3A and a marker of pain-sensing nerve cell PRDM12 genes: 1000 times higher for PRDM12 and 2500 times higher for BRN3A in differentiated cells than they were in undifferentiated SKPs. Using immunostaining, we showed that 65% and 80% of cells expressed peripheral neuron markers BRN3A and PERIPHERIN, respectively. Furthermore, differentiated cells expressed TRPV1, PAR2, TRPA1, substance P, CGRP, HR1. Using calcium imaging, we observed that a proportion of cells responded to histamine, SLIGKV (a specific agonist of PAR2), polygodial (a specific agonist of TRPA1), and capsaicin (a specific agonist of TRPV1). In conclusion, SKPs are able to differentiate directly into functional SNs. These differentiated cells will be very useful for further in vitro studies.
Collapse
|
3
|
Curcumin Induces Neural Differentiation of Human Pluripotent Embryonal Carcinoma Cells through the Activation of Autophagy. BIOMED RESEARCH INTERNATIONAL 2019; 2019:4378710. [PMID: 30800669 PMCID: PMC6360631 DOI: 10.1155/2019/4378710] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 12/10/2018] [Accepted: 01/03/2019] [Indexed: 12/20/2022]
Abstract
Curcumin is a natural polyphenolic compound, isolated from Curcuma longa, and is an important ingredient of Asian foods. Curcumin has revealed its strong activities of anti-inflammatory, antioxidant, and anticancer. The efficient amount of curcumin could induce differentiation of stem cells and promoted the differentiation of glioma-initiating cells; however, the mechanisms underlying neural induction of curcumin have not yet been revealed. In this study, neural-inducing ability of curcumin was explored by using human pluripotent embryonal carcinoma cells, NTERA2 cells. The cells were induced toward neural lineage with curcumin and were compared with a standard neutralizing agent (retinoic acid). It was found that, after 14 days of the induction by curcumin, NTERA2 cells showed neuronal morphology and expressed neural-specific genes, including NeuroD, TUJ1, and PAX6. Importantly, curcumin activated neurogenesis of NTERA2 cells via the activation of autophagy, since autophagy-related genes, such as LC3, LAMP1, and ATG5, were upregulated along with the expression of neural genes. The inhibition of autophagy by chloroquine suppressed both autophagy and neural differentiation, highlighting the positive role of autophagy during neural differentiation. This autophagy-mediated neural differentiation of curcumin was found to be an ROS-dependent manner; curcumin induced ROS generation and suppressed antioxidant gene expression. Altogether, this study proposed the neural-inducing activity of curcumin via the regulation of autophagy within NTERA2 cells and underscored the health beneficial effects of curcumin for neurodegenerative disorders, such as Alzheimer's disease and Parkinson's disease.
Collapse
|
4
|
Lai F, Fagernes CE, Bernier NJ, Miller GM, Munday PL, Jutfelt F, Nilsson GE. Responses of neurogenesis and neuroplasticity related genes to elevated CO 2 levels in the brain of three teleost species. Biol Lett 2017; 13:rsbl.2017.0240. [PMID: 28855412 DOI: 10.1098/rsbl.2017.0240] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 08/08/2017] [Indexed: 12/20/2022] Open
Abstract
The continuous increase of anthropogenic CO2 in the atmosphere resulting in ocean acidification has been reported to affect brain function in some fishes. During adulthood, cell proliferation is fundamental for fish brain growth and for it to adapt in response to external stimuli, such as environmental changes. Here we report the first expression study of genes regulating neurogenesis and neuroplasticity in brains of three-spined stickleback (Gasterosteus aculeatus), cinnamon anemonefish (Amphiprion melanopus) and spiny damselfish (Acanthochromis polyacanthus) exposed to elevated CO2 The mRNA expression levels of the neurogenic differentiation factor (NeuroD) and doublecortin (DCX) were upregulated in three-spined stickleback exposed to high-CO2 compared with controls, while no changes were detected in the other species. The mRNA expression levels of the proliferating cell nuclear antigen (PCNA) and the brain-derived neurotrophic factor (BDNF) remained unaffected in the high-CO2 exposed groups compared to the control in all three species. These results indicate a species-specific regulation of genes involved in neurogenesis in response to elevated ambient CO2 levels. The higher expression of NeuroD and DCX mRNA transcripts in the brain of high-CO2-exposed three-spined stickleback, together with the lack of effects on mRNA levels in cinnamon anemonefish and spiny damselfish, indicate differences in coping mechanisms among fish in response to the predicted-future CO2 level.
Collapse
Affiliation(s)
- Floriana Lai
- Department of Biosciences, University of Oslo, Oslo, Norway
| | | | | | - Gabrielle M Miller
- Institute for Biology and Environmental Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Philip L Munday
- ARC Centre of Excellence for Coral Reef Studies, James Cook University, Townsville, Australia
| | - Fredrik Jutfelt
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | | |
Collapse
|
5
|
Moon M, Jeong HU, Choi JG, Jeon SG, Song EJ, Hong SP, Oh MS. Memory-enhancing effects of Cuscuta japonica Choisy via enhancement of adult hippocampal neurogenesis in mice. Behav Brain Res 2016; 311:173-182. [DOI: 10.1016/j.bbr.2016.05.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 05/08/2016] [Accepted: 05/12/2016] [Indexed: 10/21/2022]
|
6
|
Chen L, Liu Y, Lin QM, Xue L, Wang W, Xu JW. Electroacupuncture at Baihui (DU20) acupoint up-regulates mRNA expression of NeuroD molecules in the brains of newborn rats suffering in utero fetal distress. Neural Regen Res 2016; 11:604-9. [PMID: 27212921 PMCID: PMC4870917 DOI: 10.4103/1673-5374.180745] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
NeuroD plays a key regulatory effect on differentiation of neural stem cells into mature neurons in the brain. Thus, we assumed that electroacupuncture at Baihui (DU20) acupoint in newborn rats exposed to in utero fetal distress would influence expression of NeuroD. Electroacupuncture at Baihui was performed for 20 minutes on 3-day-old (Day 3) newborn Sprague-Dawley rats exposed to in utero fetal distress; electroacupuncture parameters consisted of sparse and dense waves at a frequency of 2–10 Hz. Real-time fluorescent quantitative PCR results demonstrated that mRNA expression of NeuroD, a molecule that indicates NeuroD, increased with prolonged time in brains of newborn rats, and peaked on Day 22. The level of mRNA expression was similar between Day 16 and Day 35. These findings suggest that electro acupuncture at Baihui acupoint could effectively increase mRNA expression of molecules involved in NeuroD in the brains of newborn rats exposed to in utero fetal distress.
Collapse
Affiliation(s)
- Lu Chen
- Neurobiology Research Center, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Yan Liu
- Neurobiology Research Center, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Qiao-Mei Lin
- Neurobiology Research Center, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Lan Xue
- Neurobiology Research Center, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Wei Wang
- Neurobiology Research Center, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Jian-Wen Xu
- Neurobiology Research Center, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
7
|
Zheng W, ZhuGe Q, Zhong M, Chen G, Shao B, Wang H, Mao X, Xie L, Jin K. Neurogenesis in adult human brain after traumatic brain injury. J Neurotrauma 2013; 30:1872-80. [PMID: 21275797 DOI: 10.1089/neu.2010.1579] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
While much work has been conducted regarding the neurogenesis response to traumatic brain injury (TBI) in rodents, it remains largely unknown whether neurogenesis in adult human brain also responds to TBI in a similar manner. Here, we performed immunocytochemistry on 11 brain specimens from patients with traumatic brain injury, who underwent surgical intervention. We found that expression of neural stem/progenitor cell (NSC) protein markers, including DCX, TUC4, PSA-NCAM, SOX2 and NeuroD, was increased in the perilesional cortex of human brain after TBI compared to that of normal brain. Confocal images showed that these NSC proteins were expressed in one single cell. We also found that proliferative markers were expressed in NSC protein-positive cells after TBI, and the number of proliferative NSCs was significantly increased after TBI. Our data suggest that TBI may also induce neurogenesis in human brain.
Collapse
Affiliation(s)
- WeiMing Zheng
- 1 Department of Neurosurgery, First Affiliated Hospital, Wenzhou Medical College , Wenzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Mc Cormack A, Taylor J, Gregersen N, George AM, Love DR. Delineation of 2q32q35 deletion phenotypes: two apparent "proximal" and "distal" syndromes. Case Rep Genet 2013; 2013:823451. [PMID: 23840981 PMCID: PMC3690635 DOI: 10.1155/2013/823451] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Accepted: 05/22/2013] [Indexed: 11/18/2022] Open
Abstract
We report on three patients with interstitial deletions of the long arm of chromosome 2 involving bands 2q32.1-q35. They presented with wide-ranging phenotypic variation including facial dysmorphisms, cleft palate, learning difficulties, behavioural issues and severe heart defects. Microarray analysis confirmed an 8.6 Mb deletion in patients 1 and 2 and a 24.7 Mb deletion in patient 3. We discuss the genes involved in the deleted regions including MYO1B, GLS, FRZB, SATB2, and CPS1 and compare the phenotype with those reported in the literature. Taken together, these data suggest that there is a spectrum of disease severity such that patients with deletions encompassing the region of 2q32.1q32.2, which includes the FRZB gene, show an apparently milder phenotype compared to those that lie further distal in 2q32.3q35 that encompasses the SATB2 gene.
Collapse
Affiliation(s)
- Adrian Mc Cormack
- Diagnostic Genetics, LabPlus, Auckland City Hospital, P.O. Box 110031, Auckland 1148, New Zealand
| | - Juliet Taylor
- Genetic Health Service New Zealand-Northern Hub, Auckland City Hospital, Private Bag 92024, Auckland 1142, New Zealand
| | - Nerine Gregersen
- Genetic Health Service New Zealand-Northern Hub, Auckland City Hospital, Private Bag 92024, Auckland 1142, New Zealand
| | - Alice M. George
- Diagnostic Genetics, LabPlus, Auckland City Hospital, P.O. Box 110031, Auckland 1148, New Zealand
| | - Donald R. Love
- Diagnostic Genetics, LabPlus, Auckland City Hospital, P.O. Box 110031, Auckland 1148, New Zealand
- School of Biological Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
9
|
Rashid MA, Katakura M, Kharebava G, Kevala K, Kim HY. N-Docosahexaenoylethanolamine is a potent neurogenic factor for neural stem cell differentiation. J Neurochem 2013; 125:869-84. [PMID: 23570577 DOI: 10.1111/jnc.12255] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 03/29/2013] [Accepted: 03/30/2013] [Indexed: 01/13/2023]
Abstract
Docosahexaenoic acid (DHA) has been shown to promote neuronal differentiation of neural stem cells (NSCs) in vivo and in vitro. Previously, we found that N-docosahexaenoylethanolamine (synaptamide), an endogenous DHA metabolite with an endocannabinoid-like structure, promotes neurite growth, synaptogenesis, and synaptic function. In this study, we demonstrate that synaptamide potently induces neuronal differentiation of NSCs. Differentiating NSCs were capable of synthesizing synaptamide from DHA. Treatment of NSCs with synaptamide at low nanomolar concentrations significantly increased the number of MAP2 and Tuj-1-positive neurons with concomitant induction of protein kinase A (PKA)/cAMP response element binding protein (CREB) phosphorylation. Conversely, PKA inhibitors or PKA knockdown abolished the synaptamide-induced neuronal differentiation of NSCs. URB597, a fatty acid amide hydrolase (FAAH) inhibitor, elevated the level of DHA-derived synaptamide and further potentiated the DHA- or synaptamide-induced neuronal differentiation of NSCs. Similarly, NSCs obtained from FAAH KO mice exhibited greater capacity to induce neuronal differentiation in response to DHA or synaptamide compared to the wild type NSCs. Neither synaptamide nor DHA affected NSC differentiation into GFAP-positive glia cells. These results suggest that endogenously produced synaptamide is a potent mediator for neurogenic differentiation of NSCs acting through PKA/CREB activation.
Collapse
Affiliation(s)
- Mohammad A Rashid
- Laboratory of Molecular Signaling, DICBR, NIAAA, NIH, Bethesda, Maryland 20892-9410, USA
| | | | | | | | | |
Collapse
|
10
|
Park S, Koh SE, Hur CY, Lee WD, Lim J, Lee YJ. Comparison of human first and third trimester placental mesenchymal stem cell. Cell Biol Int 2013; 37:242-9. [DOI: 10.1002/cbin.10032] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 11/23/2012] [Indexed: 01/28/2023]
Affiliation(s)
- Saeyoung Park
- Maria Biotech Co.; 102-9 Sinsuldong; 130-812 Seoul; Korea
| | - Seong-Eun Koh
- Department of Rehabilitation, School of Medicine; Konkuk University, 4-12 Hwayangdong; Seoul; Korea
| | | | - Won-Don Lee
- Maria Fertility Hospital; 103-10 Sinsuldong; Seoul; Korea
| | - Jinho Lim
- Maria Fertility Hospital; 103-10 Sinsuldong; Seoul; Korea
| | - Young-Jay Lee
- Maria Biotech Co.; 102-9 Sinsuldong; 130-812 Seoul; Korea
| |
Collapse
|
11
|
Nlend RN, Aït-Lounis A, Allagnat F, Cigliola V, Charollais A, Reith W, Haefliger JA, Meda P. Cx36 is a target of Beta2/NeuroD1, which associates with prenatal differentiation of insulin-producing β cells. J Membr Biol 2012; 245:263-73. [PMID: 22729650 DOI: 10.1007/s00232-012-9447-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 06/01/2012] [Indexed: 10/28/2022]
Abstract
The insulin-producing β cells of pancreatic islets are coupled by connexin36 (Cx36) channels. To investigate what controls the expression of this connexin, we have investigated its pattern during mouse pancreas development, and the influence of three transcription factors that are critical for β-cell development and differentiation. We show that (1) the Cx36 gene (Gjd2) is activated early in pancreas development and is markedly induced at the time of the surge of the transcription factors that determine β-cell differentiation; (2) the cognate protein is detected about a week later and is selectively expressed by β cells throughout the prenatal development of mouse pancreas; (3) a 2-kbp fragment of the Gjd2 promoter, which contains three E boxes for the binding of the bHLH factor Beta2/NeuroD1, ensures the expression of Cx36 by β cells; and (4) Beta2/NeuroD1 binds to these E boxes and, in the presence of the E47 ubiquitous cofactor, transactivates the Gjd2 promoter. The data identify Cx36 as a novel early marker of β cells and as a target of Beta2/NeuroD1, which is essential for β-cell development and differentiation.
Collapse
Affiliation(s)
- Rachel Nlend Nlend
- Department of Cell Physiology and Metabolism, University of Geneva, CMU, 1 Rue Michel Servet CH- 1211, Geneva 4, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Soares AR, Pereira PM, Ferreira V, Reverendo M, Simões J, Bezerra AR, Moura GR, Santos MAS. Ethanol exposure induces upregulation of specific microRNAs in zebrafish embryos. Toxicol Sci 2012; 127:18-28. [PMID: 22298809 DOI: 10.1093/toxsci/kfs068] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Prenatal exposure to ethanol leads to a myriad of developmental disorders known as fetal alcohol spectrum disorder, often characterized by growth and mental retardation, central nervous system damage, and specific craniofacial dysmorphic features. The mechanisms of ethanol toxicity are not fully understood, but exposure during development affects the expression of several genes involved in cell cycle control, apoptosis, and transcriptional regulation. MicroRNAs (miRNAs) are implicated in some of these processes, however, it is not yet clear if they are involved in ethanol-induced toxicity. In order to clarify this question, we have exposed zebrafish embryos to ethanol and evaluated whether a miRNA deregulation signature could be obtained. Zebrafish embryos were exposed to 1 and 1.5% of ethanol from 4 h postfertilization (hpf) to 24 hpf. The miRNA expression profiles obtained reveal significant miRNA deregulation and show that both ethanol concentrations upregulate miR-153a, miR-725, miR-30d, let-7k, miR-100, miR-738, and miR-732. Putative gene targets of deregulated miRNAs are involved in cell cycle control, apoptosis, and transcription, which are the main processes affected by ethanol toxicity. The conservation of affected mechanisms among vertebrates leads us to postulate that similar miRNA deregulation occurs in humans, highlighting a relevant role of miRNAs in ethanol toxicology.
Collapse
Affiliation(s)
- Ana Raquel Soares
- RNA Biology Laboratory, Department of Biology, CESAM, University of Aveiro, 3810-193 Aveiro, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Zhou FC, Zhao Q, Liu Y, Goodlett CR, Liang T, McClintick JN, Edenberg HJ, Li L. Alteration of gene expression by alcohol exposure at early neurulation. BMC Genomics 2011; 12:124. [PMID: 21338521 PMCID: PMC3056799 DOI: 10.1186/1471-2164-12-124] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Accepted: 02/21/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We have previously demonstrated that alcohol exposure at early neurulation induces growth retardation, neural tube abnormalities, and alteration of DNA methylation. To explore the global gene expression changes which may underline these developmental defects, microarray analyses were performed in a whole embryo mouse culture model that allows control over alcohol and embryonic variables. RESULT Alcohol caused teratogenesis in brain, heart, forelimb, and optic vesicle; a subset of the embryos also showed cranial neural tube defects. In microarray analysis (accession number GSM9545), adopting hypothesis-driven Gene Set Enrichment Analysis (GSEA) informatics and intersection analysis of two independent experiments, we found that there was a collective reduction in expression of neural specification genes (neurogenin, Sox5, Bhlhe22), neural growth factor genes [Igf1, Efemp1, Klf10 (Tieg), and Edil3], and alteration of genes involved in cell growth, apoptosis, histone variants, eye and heart development. There was also a reduction of retinol binding protein 1 (Rbp1), and de novo expression of aldehyde dehydrogenase 1B1 (Aldh1B1). Remarkably, four key hematopoiesis genes (glycophorin A, adducin 2, beta-2 microglobulin, and ceruloplasmin) were absent after alcohol treatment, and histone variant genes were reduced. The down-regulation of the neurospecification and the neurotrophic genes were further confirmed by quantitative RT-PCR. Furthermore, the gene expression profile demonstrated distinct subgroups which corresponded with two distinct alcohol-related neural tube phenotypes: an open (ALC-NTO) and a closed neural tube (ALC-NTC). Further, the epidermal growth factor signaling pathway and histone variants were specifically altered in ALC-NTO, and a greater number of neurotrophic/growth factor genes were down-regulated in the ALC-NTO than in the ALC-NTC embryos. CONCLUSION This study revealed a set of genes vulnerable to alcohol exposure and genes that were associated with neural tube defects during early neurulation.
Collapse
Affiliation(s)
- Feng C Zhou
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202, USA.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Lee JH, Zhou S, Smas CM. Identification of RANBP16 and RANBP17 as novel interaction partners for the bHLH transcription factor E12. J Cell Biochem 2011; 111:195-206. [PMID: 20503194 DOI: 10.1002/jcb.22689] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The ubiquitously expressed basic helix-loop-helix (bHLH) transcription factors E12 and E47, products of alternative splicing of the E2A/TCF3 gene, regulate diverse biological processes including cell growth, differentiation and development. To search for novel protein interactions for E12, we utilized the bHLH domain of E12 as a bait in yeast two-hybrid screening. Yeast two-hybrid, mammalian two-hybrid, and co-immunoprecipitation analyses demonstrate specific interaction of E12 with RANBP17, a novel member of the importin-beta superfamily; this interaction maps to the CRM1 homology region of RANBP17. Ectopic expression of RANBP17 leads to a approximately 3-fold increase in E2A/MyoD mediated transactivation of an E-box regulated luciferase reporter gene. Interaction and transactivation studies also revealed similar functions for RANBP16/XPO7. Furthermore, ectopic expression of either RANBP16 or RANBP17 resulted in increased level of endogenous transcript for the cyclin-dependent kinase inhibitor, p21(Waf1/Cip1), a well-characterized E2A target gene. Together, these biochemical and functional data reveal RANBP16 and RANBP17 as novel regulators of E2A protein action.
Collapse
Affiliation(s)
- Jun-Ho Lee
- Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine, Toledo, Ohio 43614, USA
| | | | | |
Collapse
|
15
|
The function and regulation of the bHLH gene, cato, in Drosophila neurogenesis. BMC DEVELOPMENTAL BIOLOGY 2010; 10:34. [PMID: 20346138 PMCID: PMC2851588 DOI: 10.1186/1471-213x-10-34] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Accepted: 03/26/2010] [Indexed: 01/17/2023]
Abstract
Background bHLH transcription factors play many roles in neural development. cousin of atonal (cato) encodes one such factor that is expressed widely in the developing sensory nervous system of Drosophila. However, nothing definitive was known of its function owing to the lack of specific mutations. Results We characterised the expression pattern of cato in detail using newly raised antibodies and GFP reporter gene constructs. Expression is predominantly in sensory lineages that depend on the atonal and amos proneural genes. In lineages that depend on the scute proneural gene, cato is expressed later and seems to be particularly associated with the type II neurons. Consistent with this, we find evidence that cato is a direct target gene of Atonal and Amos, but not of Scute. We generated two specific mutations of cato. Mutant embryos show several defects in chordotonal sensory lineages, most notably the duplication of the sensory neuron, which appears to be caused by an extra cell division. In addition, we show that cato is required to form the single chordotonal organ that persists in atonal mutant embryos. Conclusions We conclude that although widely expressed in the developing PNS, cato is expressed and regulated very differently in different sensory lineages. Mutant phenotypes correlate with cato's major expression in the chordotonal sensory lineage. In these cells, we propose that it plays roles in sense organ precursor maintenance and/or identity, and in controlling the number of cell divisions in the neuronal branch of the lineage arising from these precursors.
Collapse
|
16
|
Li H, Han YR, Bi C, Davila J, Goff LA, Thompson K, Swerdel M, Camarillo C, Ricupero CL, Hart RP, Plummer MR, Grumet M. Functional differentiation of a clone resembling embryonic cortical interneuron progenitors. Dev Neurobiol 2009; 68:1549-64. [PMID: 18814314 DOI: 10.1002/dneu.20679] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We have generated clones (L2.3 and RG3.6) of neural progenitors with radial glial properties from rat E14.5 cortex that differentiate into astrocytes, neurons, and oligodendrocytes. Here, we describe a different clone (L2.2) that gives rise exclusively to neurons, but not to glia. Neuronal differentiation of L2.2 cells was inhibited by bone morphogenic protein 2 (BMP2) and enhanced by Sonic Hedgehog (SHH) similar to cortical interneuron progenitors. Compared with L2.3, differentiating L2.2 cells expressed significantly higher levels of mRNAs for glutamate decarboxylases (GADs), DLX transcription factors, calretinin, calbindin, neuropeptide Y (NPY), and somatostatin. Increased levels of DLX-2, GADs, and calretinin proteins were confirmed upon differentiation. L2.2 cells differentiated into neurons that fired action potentials in vitro, and their electrophysiological differentiation was accelerated and more complete when cocultured with developing astroglial cells but not with conditioned medium from these cells. The combined results suggest that clone L2.2 resembles GABAergic interneuron progenitors in the developing forebrain.
Collapse
Affiliation(s)
- Hedong Li
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, Piscataway, New Jersey 08854-8082, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Siebzehnrubl FA, Blumcke I. Neurogenesis in the human hippocampus and its relevance to temporal lobe epilepsies. Epilepsia 2008; 49 Suppl 5:55-65. [PMID: 18522601 DOI: 10.1111/j.1528-1167.2008.01638.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Ample evidence points to the dentate gyrus as anatomical region for persistent neurogenesis in the adult mammalian brain. This has been confirmed in a variety of animal models under physiological as well as pathophysiological conditions. Notwithstanding, similar experiments are difficult to perform in humans. Postmortem studies demonstrated persisting neurogenesis in the elderly human brain. In addition, neural precursor cells can be isolated from surgical specimens obtained from patients with intractable temporal lobe epilepsy (TLE) and propagated or differentiated into neuronal and glial lineages. It remains a controversial issue, whether epileptic seizures have an effect on or even increase hippocampal neurogenesis in humans. Recent data support the notion that seizures induce neurogenesis in young patients, whereas the capacity of neuronal recruitment and proliferation decreases with age. Animal models of TLE further indicate that these newly generated neurons integrate into epileptogenic networks and contribute to increased seizure susceptibility. However, pathomorphological disturbances within the epileptic hippocampus, such as granule cell dispersion (GCD), may not directly result from compromised neurogenesis. Still, the majority of adult TLE patients present with significant dentate granule cell loss at an end stage of the disease, which relates to severe memory and learning disabilities. In conclusion, surgical specimens obtained from TLE patients represent an important tool to study mechanisms of stem cell recruitment, proliferation and differentiation in the human brain. In addition, increasing availability of surgical specimens opens new avenues to systematically explore disease pathomechanisms in chronic epilepsies.
Collapse
|
18
|
de Bont JM, Packer RJ, Michiels EM, den Boer ML, Pieters R. Biological background of pediatric medulloblastoma and ependymoma: a review from a translational research perspective. Neuro Oncol 2008; 10:1040-60. [PMID: 18676356 DOI: 10.1215/15228517-2008-059] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Survival rates of pediatric brain tumor patients have significantly improved over the years due to developments in diagnostic techniques, neurosurgery, chemotherapy, radiotherapy, and supportive care. However, brain tumors are still an important cause of cancer-related deaths in children. Prognosis is still highly dependent on clinical characteristics, such as the age of the patient, tumor type, stage, and localization, but increased knowledge about the genetic and biological features of these tumors is being obtained and might be useful to further improve outcome for these patients. It has become clear that the deregulation of signaling pathways essential in brain development, for example, sonic hedgehog (SHH), Wnt, and Notch pathways, plays an important role in pathogenesis and biological behavior, especially for medulloblastomas. More recently, data have become available about the cells of origin of brain tumors and the possible existence of brain tumor stem cells. Newly developed array-based techniques for studying gene expression, protein expression, copy number aberrations, and epigenetic events have led to the identification of other potentially important biological abnormalities in pediatric medulloblastomas and ependymomas.
Collapse
Affiliation(s)
- Judith M de Bont
- Department of Pediatric Oncology and Hematology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
19
|
Wullimann MF, Rink E, Vernier P, Schlosser G. Secondary neurogenesis in the brain of the African clawed frog, Xenopus laevis, as revealed by PCNA, Delta-1, Neurogenin-related-1, and NeuroD expression. J Comp Neurol 2008; 489:387-402. [PMID: 16025451 DOI: 10.1002/cne.20634] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
After primary neurogenesis in the Xenopus laevis embryo, a massive new surge of neurogenesis and related neurogenic and proneural gene expression occurs in the spinal cord at the beginning of the larval period (starting at Stage 46), which corresponds to well-documented secondary neurogenesis in larval zebrafish central nervous system development. Here, we document related neural proliferation and gene expression patterns in the brain of Xenopus, in various embryonic and larval stages, showing the distribution of proliferative cells (immunostaining of cells containing the proliferating cell nuclear antigen; the auxiliary protein of DNA polymerase delta; PCNA), and the activity of some critical genes expressed during neurogenesis (i.e., Delta-1, Neurogenin-related-1, NeuroD). This study reveals that the early larval stage in Xenopus (Stage 48) displays patterns of proliferation (PCNA), as well as of neurogenic (Delta-1) and proneural (Ngnr-1; NeuroD) gene expression that are qualitatively almost identical to those seen in the 3-day postembryonic zebrafish or the 12.5/13.5-day embryonic mouse. Furthermore, a comparable bauplan of early proliferation zones (including their neuromeric organization) as described in the postembryonic zebrafish apparently exists in tetrapods (Xenopus). Altogether, the data presented suggest a common brain bauplan on the level of early proliferation patterns and neurogenic/proneural gene activity in anamniotes, if not vertebrates.
Collapse
Affiliation(s)
- Mario F Wullimann
- Centre National de la Recherche Scientifique Institute of Neurobiology A. Fessard, F-91198 Gif-sur-Yvette, France.
| | | | | | | |
Collapse
|
20
|
Mencarelli MA, Caselli R, Pescucci C, Hayek G, Zappella M, Renieri A, Mari F. Clinical and molecular characterization of a patient with a 2q31.2-32.3 deletion identified by array-CGH. Am J Med Genet A 2007; 143A:858-65. [PMID: 17352388 DOI: 10.1002/ajmg.a.31602] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
We report on a patient with a de novo interstitial deletion of the long arm of chromosome 2 involving bands 2q31.2-2q32.3. The patient shows severe mental retardation, absence of speech, sleep disturbances, behavioral problems, and some dysmorphic features. In particular, he presents with macrocephaly, high forehead, thick and coarse hair, thick eyebrows, synophrys, increased inner and outer canthal distance, bifid nasal tip, high palate, micrognathia, dysmorphic right ear, and long and tapering fingers. Array-CGH analysis allowed us to identify and characterize a 2q interstitial deletion of about 13 Mb, involving the segment between cytogenetic bands 2q31.2 and 2q32.3. The deletion was confirmed by quantitative PCR. We compare the phenotype of our patient with those already reported in literature. In particular, we discuss the similarities shared with two recently reported patients, studied by array-CGH, who show an overlapping deletion. The common clinical features are: long face, high forehead, abnormal teeth and ears, midface hypoplasia, high palate, micrognathia, transparent and thin skin, high frequency of inguinal hernia, severe development impairment, and behavioral problems. Some genes located in the deleted region may be good candidates for the neurological phenotype such as ZNF533 and MYO1B, which are both involved in neuronal function. Furthermore, the GLS gene could be a good candidate in generating the behavioral phenotype in the patient. In fact, it encodes for the major enzyme yielding glutamate from glutamine and it can be implicated in behavioral disturbances in which glutamate acts as a neurotransmitter.
Collapse
|
21
|
Ben Abdallah NMB, Slomianka L, Lipp HP. Reversible effect of X-irradiation on proliferation, neurogenesis, and cell death in the dentate gyrus of adult mice. Hippocampus 2007; 17:1230-40. [PMID: 17764075 DOI: 10.1002/hipo.20358] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Therapeutic cranial X-irradiation causes cognitive deficits in adult and pediatric patients, in particular, when the exposed area includes the medial temporal lobes. Effects on adult neurogenesis within the hippocampus may be related to such deficits. To investigate this relation, we irradiated the brain of young adult C57Bl/6j mice with a single dose of 4 Gy at a dose-rate of 27.5 cGy/min. We observed an approximately 80% decrease in the number of cells immunoreactive for the proliferation marker Ki67, 16 and 48 h after exposure, which was restored to control values after 1 week. The number of doublecortin- and NeuroD-immunoreactive cells of neuronal lineage was reduced by 60-70% up to 1 week after irradiation, but not after 1 month. The number of pyknotic cells increased approximately 2.5 fold after 16 h, decreased to approximately 50% of control numbers after 48 h and 1 week, and was again at normal levels after 1 month. Granule cell number did not differ between different groups and time points. There was no apparent activation of microglia or astrocytes. Our findings consist of an acute and reversible effect of X-irradiation on proliferation, neurogenesis, and cell death. Transient changes of neurogenesis may play a role in transient impairments of cognitive performance of patients exposed to X-irradiation. We present an experimental approach to temporarily alter adult hippocampal neurogenesis (AhN), allowing mechanistic investigations of AhN and its relevance to cognitive performances. The work also represents a step toward optimized radiotherapy schedules.
Collapse
|
22
|
Abstract
Transcription factors (TFs) play pivotal roles in directing the formation of neurons and glia. Here I will review the recent genome-scale analysis of the expression of TFs in the developing mouse nervous system and discuss the logic by which TFs control the establishment of neuronal phenotype. Accumulating evidence suggests that while combinatorial action of TFs is able to define the basic framework of the nervous system, other control mechanisms, such as stochastic and epigenetic regulation of gene expression, also contribute to the generation of nerve cell diversity.
Collapse
Affiliation(s)
- Qiufu Ma
- Dana-Farber Cancer Institute and Department of Neurobiology, Harvard Medical School, 1 Jimmy Fund Way, Boston, MA 02115, USA.
| |
Collapse
|
23
|
Wang C, Bian W, Xia C, Zhang T, Guillemot F, Jing N. Visualization of bHLH transcription factor interactions in living mammalian cell nuclei and developing chicken neural tube by FRET. Cell Res 2006; 16:585-98. [PMID: 16775630 DOI: 10.1038/sj.cr.7310076] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Members of the basic helix-loop-helix (bHLH) gene family play important roles in vertebrate neurogenesis. In this study, confocal microscopy-based fluorescence resonance energy transfer (FRET) is used to monitor bHLH protein-protein interactions under various physiological conditions. Tissue-specific bHLH activators, NeuroD1, Mash1, Neurogenin1 (Ngn1), Neurogenin2 (Ngn2), and ubiquitous expressed E47 protein are tagged with enhanced yellow fluorescence protein (EYFP) and enhanced cyan fluorescence protein (ECFP), respectively. The subcellular localization and mobility of bHLH fusion proteins are examined in HEK293 cells. By transient transfection and in ovo electroporation, four pairs of tissue-specific bHLH activators and E47 protein are over-expressed in HEK293 cells and developing chick embryo neural tube. With the acceptor photobleaching method, FRET could be detected between these bHLH protein pairs in the nuclei of transfected cells and developing neural tubes. Mash1/E47 and Ngn2/E47 FRET pairs show higher FRET efficiencies in the medial and the lateral half of chick embryo neural tube, respectively. It suggests that these bHLH protein pairs formed functional DNA-protein complexes with regulatory elements of their downstream target genes in the specific regions. This work will help one understand the behaviours of bHLH factors in vivo.
Collapse
Affiliation(s)
- Chen Wang
- Key Laboratory of Stem Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | | | | | | | | | | |
Collapse
|
24
|
Schlosser G. Induction and specification of cranial placodes. Dev Biol 2006; 294:303-51. [PMID: 16677629 DOI: 10.1016/j.ydbio.2006.03.009] [Citation(s) in RCA: 282] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2005] [Revised: 12/22/2005] [Accepted: 12/23/2005] [Indexed: 12/17/2022]
Abstract
Cranial placodes are specialized regions of the ectoderm, which give rise to various sensory ganglia and contribute to the pituitary gland and sensory organs of the vertebrate head. They include the adenohypophyseal, olfactory, lens, trigeminal, and profundal placodes, a series of epibranchial placodes, an otic placode, and a series of lateral line placodes. After a long period of neglect, recent years have seen a resurgence of interest in placode induction and specification. There is increasing evidence that all placodes despite their different developmental fates originate from a common panplacodal primordium around the neural plate. This common primordium is defined by the expression of transcription factors of the Six1/2, Six4/5, and Eya families, which later continue to be expressed in all placodes and appear to promote generic placodal properties such as proliferation, the capacity for morphogenetic movements, and neuronal differentiation. A large number of other transcription factors are expressed in subdomains of the panplacodal primordium and appear to contribute to the specification of particular subsets of placodes. This review first provides a brief overview of different cranial placodes and then synthesizes evidence for the common origin of all placodes from a panplacodal primordium. The role of various transcription factors for the development of the different placodes is addressed next, and it is discussed how individual placodes may be specified and compartmentalized within the panplacodal primordium. Finally, tissues and signals involved in placode induction are summarized with a special focus on induction of the panplacodal primordium itself (generic placode induction) and its relation to neural induction and neural crest induction. Integrating current data, new models of generic placode induction and of combinatorial placode specification are presented.
Collapse
Affiliation(s)
- Gerhard Schlosser
- Brain Research Institute, AG Roth, University of Bremen, FB2, 28334 Bremen, Germany.
| |
Collapse
|
25
|
Aamodt E, Aamodt S. Neural specification and differentiation. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2006; 69:73-97. [PMID: 16492462 DOI: 10.1016/s0074-7742(05)69003-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Affiliation(s)
- Eric Aamodt
- Department of Biochemistry, Louisiana State University Health Sciences Center-Shreveport, Louisiana, USA
| | | |
Collapse
|
26
|
Pennesi ME, Bramblett DE, Cho JH, Tsai MJ, Wu SM. A role for bHLH transcription factors in retinal degeneration and dysfunction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2006; 572:155-61. [PMID: 17249569 DOI: 10.1007/0-387-32442-9_23] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Affiliation(s)
- Mark E Pennesi
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
27
|
Schlosser G. Evolutionary origins of vertebrate placodes: insights from developmental studies and from comparisons with other deuterostomes. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2005; 304:347-99. [PMID: 16003766 DOI: 10.1002/jez.b.21055] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Ectodermal placodes comprise the adenohypophyseal, olfactory, lens, profundal, trigeminal, otic, lateral line, and epibranchial placodes. The first part of this review presents a brief overview of placode development. Placodes give rise to a variety of cell types and contribute to many sensory organs and ganglia of the vertebrate head. While different placodes differ with respect to location and derivative cell types, all appear to originate from a common panplacodal primordium, induced at the anterior neural plate border by a combination of mesodermal and neural signals and defined by the expression of Six1, Six4, and Eya genes. Evidence from mouse and zebrafish mutants suggests that these genes promote generic placodal properties such as cell proliferation, cell shape changes, and specification of neurons. The common developmental origin of placodes suggests that all placodes may have evolved in several steps from a common precursor. The second part of this review summarizes our current knowledge of placode evolution. Although placodes (like neural crest cells) have been proposed to be evolutionary novelties of vertebrates, recent studies in ascidians and amphioxus have proposed that some placodes originated earlier in the chordate lineage. However, while the origin of several cellular and molecular components of placodes (e.g., regionalized expression domains of transcription factors and some neuronal or neurosecretory cell types) clearly predates the origin of vertebrates, there is presently little evidence that these components are integrated into placodes in protochordates. A scenario is presented according to which all placodes evolved from an adenohypophyseal-olfactory protoplacode, which may have originated in the vertebrate ancestor from the anlage of a rostral neurosecretory organ (surviving as Hatschek's pit in present-day amphioxus).
Collapse
|
28
|
Gaudillière B, Konishi Y, de la Iglesia N, Yao GL, Bonni A. A CaMKII-NeuroD signaling pathway specifies dendritic morphogenesis. Neuron 2005; 41:229-41. [PMID: 14741104 DOI: 10.1016/s0896-6273(03)00841-9] [Citation(s) in RCA: 201] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The elaboration of dendrites is fundamental to the establishment of neuronal polarity and connectivity, but the mechanisms that underlie dendritic morphogenesis are poorly understood. We found that the genetic knockdown of the transcription factor NeuroD in primary granule neurons including in organotypic cerebellar slices profoundly impaired the generation and maintenance of dendrites while sparing the development of axons. We also found that NeuroD mediated neuronal activity-dependent dendritogenesis. The activity-induced protein kinase CaMKII catalyzed the phosphorylation of NeuroD at distinct sites, including endogenous NeuroD at Ser336 in primary neurons, and thereby stimulated dendritic growth. These findings uncover an essential function for NeuroD in granule neuron dendritic morphogenesis. Our study also defines the CaMKII-NeuroD signaling pathway as a novel mechanism underlying activity-regulated dendritic growth that may play important roles in the developing and mature brain.
Collapse
Affiliation(s)
- Brice Gaudillière
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
29
|
Wong KK, Chang YM, Tsang YT, Perlaky L, Su J, Adesina A, Armstrong DL, Bhattacharjee M, Dauser R, Blaney SM, Chintagumpala M, Lau CC. Expression Analysis of Juvenile Pilocytic Astrocytomas by Oligonucleotide Microarray Reveals Two Potential Subgroups. Cancer Res 2005. [DOI: 10.1158/0008-5472.76.65.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Juvenile pilocytic astrocytoma (JPA) is one of the most common brain tumors in children. The expression profiles of 21 JPAs, determined using Affymetrix GeneChip U133A, were compared with subjects with normal cerebella. The genes involved in neurogenesis, cell adhesion, synaptic transmission, central nervous system development, potassium ion transport, protein dephosphorylation, and cell differentiation were found to be significantly deregulated in JPA. These 21 JPAs were further clustered into two major groups by unsupervised hierarchical clustering using a set of 848 genes with high covariance (0.5-10). Supervised analysis with Significance Analysis of Microarrays software between these two potential subgroups identified a list of significant differentially expressed genes involved in cell adhesion, regulation of cell growth, cell motility, nerve ensheathment, and angiogenesis. Immunostaining of myelin basic protein on paraffin sections derived from 18 incompletely resected JPAs suggests that JPA without myelin basic protein–positively stained tumor cells may have a higher tendency to progress.
Collapse
Affiliation(s)
| | - Yi-Mieng Chang
- 1Texas Children's Cancer Center,
- 2Departments of Pediatrics,
| | | | - Laszlo Perlaky
- 1Texas Children's Cancer Center,
- 2Departments of Pediatrics,
| | - Jack Su
- 1Texas Children's Cancer Center,
- 2Departments of Pediatrics,
| | | | | | | | | | | | | | - Ching C. Lau
- 1Texas Children's Cancer Center,
- 2Departments of Pediatrics,
| |
Collapse
|
30
|
Kamath SG, Chen N, Enkemann SA, Sanchez-Ramos J. Transcriptional profile of NeuroD expression in a human fetal astroglial cell line. Gene Expr 2005; 12:123-36. [PMID: 15892453 PMCID: PMC6009111 DOI: 10.3727/000000005783992133] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
NeuroD1, a member of the basic helix-loop-helix (bHLH) protein family, is a transcription factor that plays a pivotal role in terminal differentiation of neural progenitors. The primary objective was to generate an early transcriptional profile triggered by NeuroD1 to guide future studies on mechanisms of neuronal differentiation. The human NeuroD1 coding region was amplified from human fetal brain RNA using specific primers and cloned into a CMV expression vector (CT-GFP-TOPO/pcDNA3.1). Transfection of a fetal glial cell line with this construct resulted in expression of NeuroD1 in 13-15% of the cells. Markers typical of early neuronal development were observed by immunocytochemical staining in a small proportion of transfected cells. To enrich the population of NeuroD1-expressing cells, fluorescence-activated cell sorting (FACS) was used to purify and collect the NeuroD1/GFP+ cells. Total RNA was extracted from the pair of cultures (NeuroD1/GFP vs. control plasmid/GFP) and processed for gene expression studies. A final gene list was composed from those probe sets that were either increased or decreased in the NeuroD1-expressing cells in three independent experiments (p < 0.001). Each gene was investigated further for possible roles in neurogenesis and a subset of 177 genes was chosen based on the following characteristics: a) genes that are potential NeuroD1 dimerization partners, b) genes that modulate other bHLH transcription factors, c) genes related to development, and d) genes associated with neural induction, outgrowth, and terminal differentiation. DNA microarray analysis of NeuroD1 expression in an astroglial cell line produced a "snapshot" transcriptional profile that will be useful in deciphering the complex molecular code that specifies a neuronal fate.
Collapse
Affiliation(s)
- Siddharth G Kamath
- Department of Neurology, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | | | | | | |
Collapse
|
31
|
RADDE-GALLWITZ KRISTEN, PAN LING, GAN LIN, LIN XI, SEGIL NEIL, CHEN PING. Expression of Islet1 marks the sensory and neuronal lineages in the mammalian inner ear. J Comp Neurol 2004; 477:412-21. [PMID: 15329890 PMCID: PMC4158841 DOI: 10.1002/cne.20257] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Several basic helix-loop-helix (bHLH) genes have been shown to be essential for the generation of the auditory sensory hair cells or the spiral ganglion (SG) neurons that innervate the hair cells in the cochlea, as well as a variety of cell types in the other nervous systems. However, it remains elusive what cellular context-dependent mechanisms confer the inner ear-specific neuronal or sensory competency/identities. We explored the possibility that one of the mechanisms responsible for generating cellular diversity in the nervous system through cooperative action of bHLH and LIM-homeodomain (LIM-HD) transcriptional factors might also contribute to the inner ear-specific sensory and/or neuronal competency. Here, we show that Islet1 (Isl1), a LIM-HD protein, is expressed early in the otocyst in the region that gives rise to both the auditory sensory organ, the organ of Corti, and SG neurons. Subsequently, the expression of Isl1 is maintained in SG neurons but is transitory in the sensory lineage. At embryonic day 12 (E12) in mice, the expression of Isl1 marks distinctively the ventral portion of the nascent cochlear epithelium encompassing the primordial organ of Corti. At E13, Isl1 is maintained at relatively high levels in the sensory primordium while down-regulated in the other regions of the cochlear duct. As the sensory epithelium starts to differentiate, it is down-regulated in the entire cochlear epithelium. The expression of Isl1 in the developing inner ear reveals an early and likely a common step in the development of both sensory and neuronal lineages of the inner ear, and suggests its potential role in the inner ear-specific sensory and neuronal cell development.
Collapse
Affiliation(s)
| | - LING PAN
- Center for Aging and Developmental Biology, University of Rochester, Rochester, New York 14642
| | - LIN GAN
- Center for Aging and Developmental Biology, University of Rochester, Rochester, New York 14642
| | - XI LIN
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - NEIL SEGIL
- Department of Cell and Molecular Biology, House Ear Institute, Los Angeles, California 90057
| | - PING CHEN
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia 30322
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, Georgia 30322
- Department of Cell and Molecular Biology, House Ear Institute, Los Angeles, California 90057
- Correspondence to: Ping Chen, Department of Cell Biology, Emory University School of Medicine, 615 Michael Street, Atlanta, GA.
| |
Collapse
|
32
|
Cho JH, Tsai MJ. The role of BETA2/NeuroD1 in the development of the nervous system. Mol Neurobiol 2004; 30:35-47. [PMID: 15247487 DOI: 10.1385/mn:30:1:035] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2003] [Accepted: 12/19/2003] [Indexed: 11/11/2022]
Abstract
BETA2/NeuroD1 is a member of the basic helix-loop-helix (bHLH) transcription factor family, which has been shown to play a major role in development of the nervous system and formation of the endocrine system. Gain-of-function studies have indicated that BETA2/NeuroD1 is important for the neurogenesis of Xenopus embryos and several neurogenic cell lines. Disruption of the gene encoding BETA2/NeuroD1 leads to severe abnormalities of the developing mouse central nervous system as well as the peripheral nervous system. The focus of this article is on the recent progress in understanding the role of BETA2/NeuroD1 in the development of the nervous system.
Collapse
Affiliation(s)
- Jang-Hyeon Cho
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | | |
Collapse
|
33
|
Yasui R, Hasegawa M, Doi K, Shimizu KI, Ohtuski N, Ishida H, Nibu KI. Quantitative analysis of expression of NeuroD, GAP43 and receptor tyrosine kinase B in developing mouse olfactory neuroepithelium. Acta Otolaryngol 2004:90-4. [PMID: 15277044 DOI: 10.1080/03655230410017742] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVE The mammalian olfactory neuroepithelium (OE), which harbors olfactory receptor neurons (ORNs), has the unusual characteristic of continuous neurogenesis throughout its lifetime. This unique feature provides an excellent model for neuronal differentiation. Recently, we found dual-phase expression of NeuroD, a member of the basic helix-loop-helix transcription factor family, in the developing mouse OE, suggesting multiple roles of NeuroD during the development of mammalian ORNs. MATERIAL AND METHODS In order to better understand the molecular mechanism of the development of ORNs, we performed quantitative analysis of expression of NeuroD, GAP43 and receptor tyrosine kinase B (TrkB), as well as 5-bromo-2'-deoxyuridine (BrdU)-labeled cells, in the developing mouse OE from gestational Day 10 to postnatal Day 28. RESULTS During the embryonic period, NeuroD expression is mostly confined to the basal compartment. During the neonatal period, NeuroD expression is detected in two compartments: the middle compartment and the basal compartment. GAP43-expressing cells were located between these two NeuroD-positive layers. TrkB-expressing cells were located above the NeuroD-positive layer in the middle compartment. As the mice grew, the numbers of NeuroD-expressing cells and BrdU-labeled cells in the basal compartment significantly decreased, while the number of NeuroD-expressing cells in the middle compartment gradually increased. The number of TrkB-expressing cells dramatically increased. The number of GAP43-expressing cells also gradually increased. However, the relative proportion of GAP43 cells decreased as the OE developed. CONCLUSION NeuroD is a useful molecular marker for studying olfactory neurogenesis.
Collapse
Affiliation(s)
- Rie Yasui
- Department of Otolaryngology-Head and Neck Surgery, Kobe University Graduate School of Medicine, Japan
| | | | | | | | | | | | | |
Collapse
|
34
|
Affiliation(s)
- Hirotaka Watada
- Department of Medicine, Metabolism and Endocrinology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| |
Collapse
|
35
|
Kanda S, Tamada Y, Yoshidome A, Hayashi I, Nishiyama T. Over‐expression of bHLH genes facilitate neural formation of mouse embryonic stem (ES) cells in vitro. Int J Dev Neurosci 2004; 22:149-56. [PMID: 15140468 DOI: 10.1016/j.ijdevneu.2004.01.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2003] [Revised: 01/14/2004] [Accepted: 01/20/2004] [Indexed: 11/25/2022] Open
Abstract
Mouse embryonic stem (ES) cells are useful tools for investigating differentiation into neurons and glial cells in vitro. In order to induce ES cells to differentiate into neural cells, many researchers have investigated the efficiency of induction. Embryoid body (EB) formation and retinoic acid are potent differentiation inducers known to be a trigger at the early stage of development. Basic helix-loop-helix (bHLH) is one of the important transcription factors, which is essential for premature neural formation. In NeuroD2 and Mash1-transfected cells, neural formation was observed at day 6 after the plating of embryoid bodies in culture. Nestin was detected in NeuroD2- and Mash1-transfected cells at day 10, and strong signal was detected in Mash1 transfectants by RT-PCR analysis. Map2 and Nurr1 were also detected strongly at the early stage in transfected cells compared with the wild type control, especially in the Mash1 transfectant. In immunocytochemical analysis, Tuj1-positive neurons were detected at high frequency in Mash1 transfectants and some cells were stained by tyrosine hydrogenase (TH), a marker of dopaminergic neurons. These results demonstrate that bHLH has a potential activity at an early stage for ES cells and can induce effective and rapid neural differentiation in vitro.
Collapse
Affiliation(s)
- Seiji Kanda
- Regeneration Research Center for Intractable Diseases, Department of Public Health, Kansai Medical University, 10-15 Fumizono-cho, Moriguchi City, Osaka 570-8506, Japan.
| | | | | | | | | |
Collapse
|
36
|
Jin K, Sun Y, Xie L, Peel A, Mao XO, Batteur S, Greenberg DA. Directed migration of neuronal precursors into the ischemic cerebral cortex and striatum. Mol Cell Neurosci 2004; 24:171-89. [PMID: 14550778 DOI: 10.1016/s1044-7431(03)00159-3] [Citation(s) in RCA: 408] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Pathological processes, including cerebral ischemia, can enhance neurogenesis in the adult brain, but the fate of the newborn neurons that are produced and their role in brain repair are obscure. To determine if ischemia-induced neuronal proliferation is associated with migration of nascent neurons toward ischemic lesions, we mapped the migration of cells labeled by cell proliferation markers and antibodies against neuronal marker proteins, for up to 2 weeks after a 90-min episode of focal cerebral ischemia caused by occlusion of the middle cerebral artery. Doublecortin-immunoreactive cells in the rostral subventricular zone, but not the dentate gyrus, migrated into the ischemic penumbra of the adjacent striatum and, via the rostral migratory stream and lateral cortical stream, into the penumbra of ischemic cortex. These results indicate that after cerebral ischemia, new neurons are directed toward sites of brain injury, where they might be in a position to participate in brain repair and functional recovery.
Collapse
Affiliation(s)
- Kunlin Jin
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Montuenga LM, Guembe L, Burrell MA, Bodegas ME, Calvo A, Sola JJ, Sesma P, Villaro AC. The diffuse endocrine system: from embryogenesis to carcinogenesis. PROGRESS IN HISTOCHEMISTRY AND CYTOCHEMISTRY 2003; 38:155-272. [PMID: 12756892 DOI: 10.1016/s0079-6336(03)80004-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
In the present review we will summarise the current knowledge about the cells comprising the Diffuse Endocrine System (DES) in mammalian organs. We will describe the morphological, histochemical and functional traits of these cells in three major systems gastrointestinal, respiratory and prostatic. We will also focus on some aspects of their ontogeny and differentiation, as well as to their relevance in carcinogenesis, especially in neuroendocrine tumors. The first chapter describes the characteristics of DES cells and some of their specific biological and biochemical traits. The second chapter deals with DES in the gastrointestinal organs, with special reference to the new data on the differentiation mechanisms that leads to the appearance of endocrine cells from an undifferentiated stem cell. The third chapter is devoted to DES of the respiratory system and some aspects of its biological role, both, during development and adulthood. Neuroendocrine hyperplasia and neuroendocrine lung tumors are also addressed. Finally, the last chapter deals with the prostatic DES, discussing its probable functional role and its relevance in hormone-resistant prostatic carcinomas.
Collapse
Affiliation(s)
- Luis M Montuenga
- Department of Histology and Pathology, Schools of Science and Medicine, University of Navarra, 31080 Pamplona, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
1. Neural stem cells can be cultured from the CNS of different mammalian species at many stages of development. They have an extensive capacity for self-renewal and will proliferate ex vivo in response to mitogenic growth factors or following genetic modification with immortalising oncogenes. Neural stem cells are multipotent since their differentiating progeny will give rise to the principal cellular phenotypes comprising the mature CNS: neurons, astrocytes and oligodendrocytes. 2. Neural stem cells can also be derived from more primitive embryonic stem (ES) cells cultured from the blastocyst. ES cells are considered to be pluripotent since they can give rise to the full cellular spectrum and will, therefore, contribute to all three of the embryonic germ layers: endoderm, mesoderm and ectoderm. However, pluripotent cells have also been derived from germ cells and teratocarcinomas (embryonal carcinomas) and their progeny may also give rise to the multiple cellular phenotypes contributing to the CNS. In a recent development, ES cells have also been isolated and grown from human blastocysts, thus raising the possibility of growing autologous stem cells when combined with nuclear transfer technology. 3. There is now an emerging recognition that the adult mammalian brain, including that of primates and humans, harbours stem cell populations suggesting the existence of a previously unrecognised neural plasticity to the mature CNS, and thereby raising the possibility of promoting endogenous neural reconstruction. 4. Such reports have fuelled expectations for the clinical exploitation of neural stem cells in cell replacement or recruitment strategies for the treatment of a variety of human neurological conditions including Parkinson's disease (PD), Huntington's disease, multiple sclerosis and ischaemic brain injury. Owing to their migratory capacity within the CNS, neural stem cells may also find potential clinical application as cellular vectors for widespread gene delivery and the expression of therapeutic proteins. In this regard, they may be eminently suitable for the correction of genetically-determined CNS disorders and in the management of certain tumors responsive to cytokines. Since large numbers of stem cells can be generated efficiently in culture, they may obviate some of the technical and ethical limitations associated with the use of fresh (primary) embryonic neural tissue in current transplantation strategies. 5. While considerable recent progress has been made in terms of developing new techniques allowing for the long-term culture of human stem cells, the successful clinical application of these cells is presently limited by our understanding of both (i) the intrinsic and extrinsic regulators of stem cell proliferation and (ii) those factors controlling cell lineage determination and differentiation. Although such cells may also provide accessible model systems for studying neural development, progress in the field has been further limited by the lack of suitable markers needed for the identification and selection of cells within proliferating heterogeneous populations of precursor cells. There is a further need to distinguish between the committed fate (defined during normal development) and the potential specification (implying flexibility of fate through manipulation of its environment) of stem cells undergoing differentiation. 6. With these challenges lying ahead, it is the opinion of the authors that stem-cell therapy is likely to remain within the experimental arena for the foreseeable future. In this regard, few (if any) of the in vivo studies employing neural stem cell grafts have shown convincingly that behavioural recovery can be achieved in the various model paradigms. Moreover, issues relating to the quality control of cultured cells and their safety following transplantation have only begun to be addressed. 7. While on the one hand cell biotechnologists have been quick to realise the potential commercial value, human stem cell research and its clinical applications has been the subject of intense ethical and legislative considerations. The present chapter aims to review some recent aspects of stem cell research applicable to developmental neurobiology and the potential applications in clinical neuroscience.
Collapse
Affiliation(s)
- T Ostenfeld
- MRC Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
39
|
Fujita S. The Discovery of the Matrix Cell, the Identification of the Multipotent Neural Stem Cell and the Development of the Central Nervous System. Cell Struct Funct 2003; 28:205-28. [PMID: 14586132 DOI: 10.1247/csf.28.205] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
In the early 1960s I applied 3H-thymidine autoradiography to the study of the cells constituting the neural tube, and found that its wall was composed solely of one kind of single-layered epithelial cell, which perform an elevator movement between the mitotic and DNA-synthetic zones in the wall in accord with the cell cycle. They were identified as multipotent stem cells of the central nervous sytem (CNS) to which I gave the name of matrix cells. (3)H-thymidine autoradiography also revealed the chronology of development of these matrix cells: At first they proliferate only to expand the population (stage I), then switch to differentiate specific neuroblasts in given sequences (stage II), and finally change themselves into ependymoglioblasts, common progenitors of ependymal cells and neuroglia (stage III). Based on these findings, I proposed a monophyletic view of cytogenesis of the central nervous sytem. This matrix cell theory claiming the existence of multipotent stem cells has long been the target of severe criticism and not been accepted among neuro-embryologists for a long time. Recent findings by experimental and clinical neuroscientists on the importance of stem cells have renewed interest in the nature and biology of the multipotent neural stem cells. The present paper describes how the concept of the matrix cell (multipotent neural stem cells in vivo) emerged and what has come out from this view over the last 45 years, and how the basic concept of the matrix cell theory has recently been reconfirmed after a long period of controversy and neglect.
Collapse
Affiliation(s)
- Setsuya Fujita
- Louis Pasteur Center for Medical Research, Kyoto, Japan.
| |
Collapse
|
40
|
Abstract
The specialized endocrine and exocrine cells of the pancreas originally derive from a pool of apparently identical cells in the early gut endoderm. Serial changes in their gene expression program, controlled by a hierarchy of pancreatic transcription factors, direct this progression from multipotent progenitor cell to mature pancreatic cell. When the cells differentiate, this hierarchy of factors coalesces into a network of factors that maintain the differentiated phenotype of the cells. As we develop an understanding of the pancreatic transcription factors, we are also acquiring the tools with which we can ultimately control pancreatic cell differentiation.
Collapse
Affiliation(s)
- Maria E Wilson
- Department of Medicine, UCSF Diabetes Center, Hormone Research Institute, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143-0534, USA
| | | | | |
Collapse
|
41
|
Abstract
Helix-loop-helix (HLH) genes function as important regulators of neurogenesis in both the peripheral and central nervous systems. The olfactory system is an ideal tissue in which to study the role of these genes in regulating the acquisition of neuronal cell fate, particularly that of the olfactory receptor neuron (ORN). Here we describe the expression of several basic HLH (bHLH) and repeat HLH (rHLH) factors during olfactory placode development in Xenopus laevis. Our work reveals that a combination of both bHLH and rHLH genes are sequentially expressed within the nascent olfactory placode during normal development. Moreover, overexpression of the bHLH factor, Xenopus atonal homologue 5 (Xath5), promotes olfactory neural fate independent of cellular proliferation within a restricted domain at the anterior of the embryo. Collectively, our data argue that HLH genes are expressed in a cascade during olfactory placode development and that the activity of an atonal homologue, Xath5, can promote ORN fate but only in the appropriate developmental context.
Collapse
Affiliation(s)
- Carole J Burns
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah 84132-3401, USA
| | | |
Collapse
|
42
|
Firulli AB, Thattaliyath BD. Transcription factors in cardiogenesis: the combinations that unlock the mysteries of the heart. INTERNATIONAL REVIEW OF CYTOLOGY 2002; 214:1-62. [PMID: 11893163 DOI: 10.1016/s0074-7696(02)14002-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Heart formation is one of the first signs of organogenesis within the developing embryo and this process is conserved from flies to man. Completing the genetic roadmap of the molecular mechanisms that control the cell specification and differentiation of cells that form the developing heart has been an exciting and fast-moving area of research in the fields of molecular and developmental biology. At the core of these studies is an interest in the transcription factors that are responsible for initiation of a pluripotent cell to become programmed to the cardiac lineage and the subsequent transcription factors that implement the instructions set up by the cells commitment decision. To gain a better understanding of these pathways, cardiac-expressed transcription factors have been identified, cloned, overexpressed, and mutated to try to determine function. Although results vary depending on the gene in question, it is clear that there is a striking evolutionary conservation of the cardiogenic program among species. As we move up the evolutionary ladder toward man, we encounter cases of functional redundancy and combinatorial interactions that reflect the complex networks of gene expression that orchestrate heart development. This review focuses on what is known about the transcription factors implicated in heart formation and the role they play in this intricate genetic program.
Collapse
Affiliation(s)
- Anthony B Firulli
- Department of Physiology, University of Texas Health Science Center at San Antonio, San Antonio 78229, USA
| | | |
Collapse
|
43
|
Woodbury D, Reynolds K, Black IB. Adult bone marrow stromal stem cells express germline, ectodermal, endodermal, and mesodermal genes prior to neurogenesis. J Neurosci Res 2002; 69:908-17. [PMID: 12205683 DOI: 10.1002/jnr.10365] [Citation(s) in RCA: 340] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Bone marrow stromal stem cells (MSCs) normally differentiate into mesenchymal derivatives but recently have also been converted into neurons, classical ectodermal cells. To begin defining underlying mechanisms, we extended our characterization of MSCs and the differentiated neurons. In addition to expected mesodermal mRNAs, populations and clonal lines of MSCs expressed germinal, endodermal, and ectodermal genes. Thus, the MSCs are apparently "multidifferentiated" in addition to being multipotent. Conversely, the differentiating neurons derived from populations and clonal lines of MSCs expressed the specific markers beta-III tubulin, tau, neurofilament-M, TOAD-64, and synaptophysin de novo. The transmitter enzymes tyrosine hydroxylase and choline acetyltransferase were localized to neuronal subpopulations. Our observations suggest that MSCs are already multidifferentiated and that neural differentiation comprises quantitative modulation of gene expression rather than simple on-off switching of neural-specific genes.
Collapse
Affiliation(s)
- Dale Woodbury
- Department of Neuroscience and Cell Biology, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, USA.
| | | | | |
Collapse
|
44
|
Perry T, Lahiri DK, Chen D, Zhou J, Shaw KTY, Egan JM, Greig NH. A novel neurotrophic property of glucagon-like peptide 1: a promoter of nerve growth factor-mediated differentiation in PC12 cells. J Pharmacol Exp Ther 2002; 300:958-66. [PMID: 11861804 DOI: 10.1124/jpet.300.3.958] [Citation(s) in RCA: 232] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The insulinotropic hormone glucagon-like peptide-1 (7-36)-amide (GLP-1) has potent effects on glucose-dependent insulin secretion, insulin gene expression, and pancreatic islet cell formation and is presently in clinical trials as a therapy for type 2 diabetes mellitus. We report on the effects of GLP-1 and two of its long-acting analogs, exendin-4 and exendin-4 WOT, on neuronal proliferation and differentiation, and on the metabolism of two neuronal proteins in the rat pheochromocytoma (PC12) cell line, which has been shown to express the GLP-1 receptor. We observed that GLP-1 and exendin-4 induced neurite outgrowth in a manner similar to nerve growth factor (NGF), which was reversed by coincubation with the selective GLP-1 receptor antagonist exendin (9-39). Furthermore, exendin-4 could promote NGF-initiated differentiation and may rescue degenerating cells after NGF-mediated withdrawal. These effects were induced in the absence of cellular dysfunction and toxicity as quantitatively measured by 3-(4,5-cimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide and lactate dehydrogenase assays, respectively. Our findings suggest that such peptides may be used in reversing or halting the neurodegenerative process observed in neurodegenerative diseases, such as the peripheral neuropathy associated with type 2 diabetes mellitus and Alzheimer's and Parkinson's diseases. Due to its novel twin action, GLP-1 and exendin-4 have therapeutic potential for the treatment of diabetic peripheral neuropathy and these central nervous system disorders.
Collapse
Affiliation(s)
- TracyAnn Perry
- Section of Drug, Design, and Development, Laboratory of Neuroscience, Gerontology Research Center, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA.
| | | | | | | | | | | | | |
Collapse
|
45
|
Krüger M, Braun T. The neuronal basic helix-loop-helix transcription factor NSCL-1 is dispensable for normal neuronal development. Mol Cell Biol 2002; 22:792-800. [PMID: 11784856 PMCID: PMC133555 DOI: 10.1128/mcb.22.3.792-800.2002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The neuronal stem cell leukemia (NSCL) basic helix-loop-helix factors are neural cell-specific transcription factors. We have disrupted the NSCL-1 gene by homologous recombination and replaced the coding region with a beta-galactosidase reporter cassette to study the role of NSCL-1 in neuronal development and to follow the fate of NSCL-1 mutant cells. NSCL-1 mutant mice are viable and fertile on various genetic backgrounds and do not show any obvious signs of neurological malfunction. No differences in the distribution of NSCL-1 mutant or heterozygous neuronal cells were observed in the diencephalon, hippocampus, neocortex, and cerebellum at different stages of development. Likewise, no defects were found in the laminar organization of the cortex, and the distinct neuronal subpopulation appeared normal during development of the neocortex. Analysis of sensory neurons which strongly express NSCL-1 revealed that the spatiotemporal expression of neuronal differentiation factors, such as NeuroD and SCG-10, was not altered in developing distal and proximal cranial ganglia of mutant mice. In the cerebellum expression of NSCL-1 was confined to the proliferative and premigratory zone of the external granular layer and the internal granular layer. Interestingly, unlike cerebella of Math1(-/-) or NeuroD2(-/-) mice, NSCL-1-deficient mice have no obvious developmental defect, and neurons of the cerebellum appeared fully differentiated. Despite similar expression patterns of NSCL-1 and NSCL-2 in various areas of the diencephalon, including the arcuate nucleus and paraventricular nucleus, NSCL-1(-/-) mice are fertile and show no adult onset of obesity like NSCL-2 mutant mice. Double-mutant NSCL-1(-/-)-NSCL-2(-/-) mice do not show any additional obvious malformations of the central nervous system, although both genes are expressed in a largely overlapping pattern. Our results argue against a simple functional redundancy within the NSCL gene family.
Collapse
Affiliation(s)
- Markus Krüger
- Institute of Physiological Chemistry, University of Halle-Wittenberg, 06097 Halle, Germany
| | | |
Collapse
|
46
|
Fuhrmann S, Chow L, Reh TA. Molecular control of cell diversification in the vertebrate retina. Results Probl Cell Differ 2001; 31:69-91. [PMID: 10929402 DOI: 10.1007/978-3-540-46826-4_5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Affiliation(s)
- S Fuhrmann
- Department of Biological Structure, University of Washington, Seattle 98195, USA
| | | | | |
Collapse
|
47
|
Abstract
Traditional schemes of classifying nervous system malformations are based on descriptive morphogenesis of anatomic processes of ontogenesis, such as neurulation, neuroblast migration, and axonal pathfinding. This proposal is a first attempt to incorporate the recent molecular genetic data that explain programming of development etiologically. A scheme based purely on genetic mutations would not be practical, in part because only in a few dysgeneses are the specific defects known, but also because several genes might be involved sequentially and many genes inhibit or augment the expression of others. The same genes serve different functions at different stages and are involved in multiple organ systems. Some complex malformations, such as holoprosencephaly, result from several unrelated defective genes. Finally, a pure genetic classification would be too inflexible to incorporate some anatomic criteria. The basis for the proposed scheme is, therefore, disturbances in patterns of genetic expression; polarity gradients of the axes of the neural tube (eg, upregulation or downregulation of genetic influences); segmentation (eg, deletions of specific neuromeres, ectopic expression); mutations that cause change in cell lineage (eg, dysplastic gangliocytoma of cerebellum, myofiber differentiation within brain); and specific genes or molecules that mediate neuroblast migration in its early (eg, filamin-1), middle (eg, LIS1, double-cortin), or late course (eg, reelin, L1-CAM). The proposed scheme undoubtedly will undergo many future revisions, but it provides a starting point using currently available data.
Collapse
Affiliation(s)
- H B Sarnat
- Department of Neurology, University of Washington School of Medicine, Seattle, USA.
| |
Collapse
|
48
|
|
49
|
|
50
|
Bosse A, Stoykova A, Nieselt-Struwe K, Chowdhury K, Copeland NG, Jenkins NA, Gruss P. Identification of a novel mouse Iroquois homeobox gene, Irx5, and chromosomal localisation of all members of the mouse Iroquois gene family. Dev Dyn 2000; 218:160-74. [PMID: 10822268 DOI: 10.1002/(sici)1097-0177(200005)218:1<160::aid-dvdy14>3.0.co;2-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The Drosophila genes of the Iroquois-Complex encode homeodomain containing transcription factors that positively regulate the activity of certain proneural Achaete/Scute-C (AS-C) genes during the formation of external sensory organs (J. L. Gomez-Skarmeta and J. Modolell, EMBO J 17:181-190, 1996). Previously, we have identified three highly-related genes of the mouse Iroquois gene family that exert specific expression patterns in the central nervous system (A. Bosse et al., Mech Dev 69:169-181, 1997). In the present paper, we report the identification of a novel member of the Iroquois gene family, Irx5, that shows a restricted spatio/temporal expression during early mouse embryogenesis, distinct from the expression of Irx1-3. An extensive sequence analysis of 20 Iroquois-like genes from seven organisms reveals a high conservation of the homeodomain. Phylogenetic tree reconstruction showed a clustering of the members of the Iroquois gene family into groups of orthologous genes. Together, with the data obtained from the chromosomal mapping analysis, the results indicate that these genes have appeared in vertebrates during evolution as a result of gene duplication.
Collapse
Affiliation(s)
- A Bosse
- Department of Molecular Cell Biology, Max Planck Institute of Biophysical Chemistry, Göttingen, Germany
| | | | | | | | | | | | | |
Collapse
|