Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 22
1.
Dev Growth Differ ; 66(1): 21-34, 2024 Jan.
Article En | MEDLINE | ID: mdl-38239149

Inherited leukodystrophies are genetic disorders characterized by abnormal white matter in the central nervous system. Although individually rare, there are more than 400 distinct types of leukodystrophies with a cumulative incidence of 1 in 4500 live births. The pathophysiology of most leukodystrophies is poorly understood, there are treatments for only a few, and there is significant morbidity and mortality, suggesting a critical need for improvements in this field. A variety of animal, cell, and induced pluripotent stem cell-derived models have been developed for leukodystrophies, but with significant limitations in all models. Many leukodystrophies lack animal models, and extant models often show no or mixed recapitulation of key phenotypes. Zebrafish (Danio rerio) have become increasingly used as disease models for studying leukodystrophies due to their early onset of disease phenotypes and conservation of molecular and neurobiological mechanisms. Here, we focus on reviewing new zebrafish disease models for leukodystrophy or models with recent progress. This includes discussion of leukodystrophy with vanishing white matter disease, X-linked adrenoleukodystrophy, Zellweger spectrum disorders and peroxisomal disorders, PSAP deficiency, metachromatic leukodystrophy, Krabbe disease, hypomyelinating leukodystrophy-8/4H leukodystrophy, Aicardi-Goutières syndrome, RNASET2-deficient cystic leukoencephalopathy, hereditary diffuse leukoencephalopathy with spheroids-1 (CSF1R-related leukoencephalopathy), and ultra-rare leukodystrophies. Zebrafish models offer important potentials for the leukodystrophy field, including testing of new variants in known genes; establishing causation of newly discovered genes; and early lead compound identification for therapies. There are also unrealized opportunities to use humanized zebrafish models which have been sparsely explored.


Adrenoleukodystrophy , Leukodystrophy, Globoid Cell , Leukodystrophy, Metachromatic , Leukoencephalopathies , Animals , Zebrafish/genetics , Leukodystrophy, Metachromatic/genetics , Leukodystrophy, Metachromatic/therapy , Leukodystrophy, Globoid Cell/genetics , Leukodystrophy, Globoid Cell/therapy , Adrenoleukodystrophy/genetics , Leukoencephalopathies/therapy
2.
Stem Cells Dev ; 32(17-18): 524-538, 2023 09.
Article En | MEDLINE | ID: mdl-37358404

Neural progenitor cells are self-renewable, proliferative, and multipotent cell populations that generate diverse types of neurons and glia to build the nervous system. Transcription factors play critical roles in regulating various cellular processes; however, the transcription factors that regulate the development of neural progenitors are yet to be identified. In the present study, we demonstrated that zebrafish etv5a is expressed in the neural progenitor cells of the neuroectoderm. Downregulation of endogenous Etv5a function by etv5a morpholino or an etv5a dominant-negative variant increased the proliferation of sox2-positive neural progenitor cells, accompanied by inhibition of neurogenesis and gliogenesis. These phenotypes in Etv5a-depleted embryos could be rescued by a co-injection with etv5a cRNA. Etv5a overexpression reduced sox2 expression. Direct binding of Etv5a to the regulatory elements of sox2 was affirmed by chromatin immunoprecipitation. These data revealed that Etv5a directly suppressed sox2 expression to reduce the proliferation of neural progenitor cells. In addition, the expression of foxm1, a putative target gene of Etv5a and a direct upstream transcription factor of sox2, was upregulated in Etv5a-deficient embryos. Moreover, the suppression of Foxm1 function by the foxm1 dominant-negative construct nullified the phenotype of upregulated sox2 expression caused by Etv5a deficiency. Overall, our results indicated that Etv5a regulates the expression of sox2 via direct binding to the sox2 promoter and indirect regulation by inhibiting foxm1 expression. Hence, we revealed the role of Etv5a in the transcriptional hierarchy that regulates the proliferation of neural progenitor cells.


Transcription Factors , Zebrafish , Animals , Zebrafish/genetics , Zebrafish/metabolism , Cell Differentiation/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Neurons/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Cell Proliferation/genetics
3.
Int J Mol Sci ; 24(2)2023 Jan 15.
Article En | MEDLINE | ID: mdl-36675251

Bone morphogenetic protein (BMP) signaling regulates neural induction, neuronal specification, and neuronal differentiation. However, the role of BMP signaling in neural progenitors remains unclear. This is because interruption of BMP signaling before or during neural induction causes severe effects on subsequent neural developmental processes. To examine the role of BMP signaling in the development of neural progenitors in zebrafish, we bypassed the effect of BMP signaling on neural induction and suppressed BMP signaling at different time points during gastrulation using a temporally controlled transgenic line carrying a dominant-negative form of Bmp receptor type 1aa and a chemical inhibitor of BMP signaling, DMH1. Inhibiting BMP signaling from 8 hpf could bypass BMP regulation on neural induction, induce the number of proliferating neural progenitors, and reduce the number of neuronal precursors. Inhibiting BMP signaling upregulates the expression of the Notch downstream gene hairy/E(spl)-related 2 (her2). Inhibiting Notch signaling or knocking down the Her2 function reduced neural progenitor proliferation, whereas inactivating BMP signaling in Notch-Her2 deficient background restored the number of proliferating neural progenitors. These results reveal the time window for the proliferation of neural progenitors during zebrafish development and a fine balance between BMP and Notch signaling in regulating the proliferation of neural progenitor cells.


Neural Stem Cells , Zebrafish , Animals , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Neural Stem Cells/metabolism , Cell Proliferation , Gene Expression Regulation, Developmental
4.
Anticancer Res ; 41(12): 6135-6145, 2021 Dec.
Article En | MEDLINE | ID: mdl-34848468

BACKGROUND/AIM: This study aimed to explore RGS2 as a regulator of melanoma cell growth. MATERIALS AND METHODS: Effect of RGS2 over-expression was analyzed in three melanoma cell lines, and Rgs2 knockdown was performed in zebrafish. RESULTS: RGS2 was differentially expressed among the cell lines. In B16F10 cells, RGS2 over-expression inhibited MAPK and AKT activation, and prevented cell growth. A similar outcome was observed in A375 cells, but the MAPK signals were not suppressed. In A2058 cells, RGS2 repressed AKT activation, but without affecting cell growth. Moreover, MAPK and AKT constitutive activation abolished the RGS2 inhibitory effect on B16F10 cell growth. Rgs2 knockdown caused ectopic melanocyte differentiation, and promoted MAPK and AKT activation in zebrafish embryos. CONCLUSION: RGS2 prevents melanoma cell growth by inhibiting MAPK and AKT, but this effect depends on the overall cell genetic landscape. Further studies are warranted to investigate the anticancer therapeutic potential of RGS2 for melanoma.


Helix-Loop-Helix Motifs/physiology , Melanoma/drug therapy , Mitogen-Activated Protein Kinase Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RGS Proteins/therapeutic use , Animals , Humans , Melanoma/physiopathology , RGS Proteins/pharmacology , Signal Transduction , Zebrafish
5.
HGG Adv ; 2(3): 100034, 2021 Jul 08.
Article En | MEDLINE | ID: mdl-35047835

Leukodystrophies, genetic neurodevelopmental and/or neurodegenerative disorders of cerebral white matter, result from impaired myelin homeostasis and metabolism. Numerous genes have been implicated in these heterogeneous disorders; however, many individuals remain without a molecular diagnosis. Using whole-exome sequencing, biallelic variants in LSM7 were uncovered in two unrelated individuals, one with a leukodystrophy and the other who died in utero. LSM7 is part of the two principle LSM protein complexes in eukaryotes, namely LSM1-7 and LSM2-8. Here, we investigate the molecular and functional outcomes of these LSM7 biallelic variants in vitro and in vivo. Affinity purification-mass spectrometry of the LSM7 variants showed defects in the assembly of both LSM complexes. Lsm7 knockdown in zebrafish led to central nervous system defects, including impaired oligodendrocyte development and motor behavior. Our findings demonstrate that variants in LSM7 cause misassembly of the LSM complexes, impair neurodevelopment of the zebrafish, and may be implicated in human disease. The identification of more affected individuals is needed before the molecular mechanisms of mRNA decay and splicing regulation are added to the categories of biological dysfunctions implicated in leukodystrophies, neurodevelopmental and/or neurodegenerative diseases.

6.
Elife ; 92020 12 10.
Article En | MEDLINE | ID: mdl-33300869

Vanishing white matter disease (VWM) is a severe leukodystrophy of the central nervous system caused by mutations in subunits of the eukaryotic initiation factor 2B complex (eIF2B). Current models only partially recapitulate key disease features, and pathophysiology is poorly understood. Through development and validation of zebrafish (Danio rerio) models of VWM, we demonstrate that zebrafish eif2b mutants phenocopy VWM, including impaired somatic growth, early lethality, effects on myelination, loss of oligodendrocyte precursor cells, increased apoptosis in the CNS, and impaired motor swimming behavior. Expression of human EIF2B2 in the zebrafish eif2b2 mutant rescues lethality and CNS apoptosis, demonstrating conservation of function between zebrafish and human. In the mutants, intron 12 retention leads to expression of a truncated eif2b5 transcript. Expression of the truncated eif2b5 in wild-type larva impairs motor behavior and activates the ISR, suggesting that a feed-forward mechanism in VWM is a significant component of disease pathophysiology.


Disease Models, Animal , Eukaryotic Initiation Factor-2B/genetics , Eukaryotic Initiation Factor-2B/metabolism , Leukoencephalopathies/genetics , Leukoencephalopathies/metabolism , Animals , Humans , Leukoencephalopathies/physiopathology , Mutation , Stress, Physiological/physiology , Zebrafish
7.
Dev Dyn ; 247(12): 1264-1275, 2018 12.
Article En | MEDLINE | ID: mdl-30358936

BACKGROUND: Many molecules and signaling pathways involved in neural development play a role in neurodegenerative diseases and brain tumor progression. Peroxisome proliferator-activated receptor (PPAR) proteins regulate the differentiation of tissues and the progression of many diseases. However, the role of these proteins in neural development is unclear. RESULTS: We examined the function of Pparα in the neural development of zebrafish. Two duplicate paralogs for mammalian PPARA/Ppara, namely pparaa and pparab, are present in the zebrafish genome. Both pparaa and pparab are expressed in the developing central nervous system in zebrafish embryos. Inhibiting the function of Pparα by using either the PPARα/Pparα antagonist GW6471 or pparaa or pparab truncated constructs produced identical phenotypes, which were sufficient to reduce the proliferation of neuronal and glial precursor cells without affecting the formation of neural progenitors. CONCLUSIONS: We demonstrated that both Pparαa and Pparαb proteins are essential regulators of the proliferation of neuronal and glial precursors. This study provides a better understanding of the functions of PPARα/Pparα in neural development and further expands our knowledge of the potential role of PPARα/Pparα in neurological disorders and brain tumors. Developmental Dynamics 247:1264-1275, 2018. © 2018 Wiley Periodicals, Inc.


Cell Proliferation/drug effects , Central Nervous System/cytology , Neuroglia/cytology , Neurons/cytology , PPAR alpha/physiology , Stem Cells/cytology , Animals , Central Nervous System/embryology , Neurogenesis , PPAR alpha/deficiency , Zebrafish/embryology
8.
Exp Neurol ; 304: 114-124, 2018 06.
Article En | MEDLINE | ID: mdl-29522758

Hexanucleotide repeat expansions in the C9orf72 gene are a common genetic cause of familial and sporadic amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). However, the function of C9orf72 in neural development and the pathogenic mechanism underlying neurodegeneration are unknown. We found that disrupting C9orf72 expression by using C9orf72 constructs that lack the complete DENN domain result in reduced GTPase activity in zebrafish embryos, demonstrating the indispensability of the complete DENN domain. This effect was phenocopied by knocking down endogenous C9orf72 expression by using morpholinos. C9orf72-deficient zebrafish embryos exhibited impaired axonogenesis and motility defects. The C9orf72 deficiency upregulated the expression of tp53 and caused neuronal apoptosis. Knockdown Tp53 in the C9orf72-deficient embryos rescued only the apoptotic phenotype but not the phenotype with axonal and motility defects. The C9orf72 deficiency also induced ccng1 (encodes Cyclin G1) mRNA expression, and injection of a dominant-negative Cyclin G1 construct rescued the axonal impairment, apoptosis, and motility defects in the C9orf72-deficient embryos. Our results revealed the GTPase activity of C9orf72 and demonstrated that Cyclin G1 is an essential downstream mediator for C9orf72 in neural development and motility. Furthermore, downregulating Cyclin G1 was sufficient to rescue all the defects caused by C9orf72 deficiency. In summary, we revealed a novel regulatory mechanism underlying the role of C9orf72 in neurological and motility defects. This result facilitates understanding the function of the C9orf72 gene in the developing nervous system and provides a potential mechanism underlying the pathogenesis of ALS-FTD.


C9orf72 Protein/metabolism , Cyclin G1/metabolism , Motor Activity/physiology , Neurogenesis/physiology , Animals , Apoptosis/physiology , Zebrafish
9.
Cell Death Dis ; 9(1): 10, 2018 01 09.
Article En | MEDLINE | ID: mdl-29317613

Glucose-6-phosphate dehydrogenase (G6PD) is a housekeeping enzyme involved in the pentose phosphate shunt for producing nicotinamide adenine dinucleotide phosphate (NADPH). Severe G6PD deficiency leads to embryonic lethality, but the underlying mechanism is unclear. In the current study, the effects of G6PD on epithelial-mesenchymal transition (EMT), especially during embryonic development, were investigated. The knockdown of G6PD induced morphological changes, accompanied by the suppression of epithelial markers, E-cadherin and ß-catenin, in A549 and MDCK cells. Such modulation of EMT was corroborated by the enhancement of migration ability in G6PD-knockdown A549 cells. Zebrafish embryos with g6pd knockdown exhibited downregulation of the E-cadherin/ß-catenin adhesion molecules and impaired embryonic development through reduction in epiboly rate and increase in cell shedding at the embryo surface. The dysregulation in zebrafish embryonic development caused by g6pd knockdown could be rescued through human G6PD or CDH1 (E-cadherin gene) cRNA coinjection. The Smad3/miR-200b axis was dysregulated upon G6PD knockdown, and the reconstitution of SMAD3 in G6PD-knockdown A549 cells restored the expression of E-cadherin/ß-catenin. The inhibition of NADPH oxidase (NOX) activation through the loss of p22phox signaling was involved in the dysregulation of the Smad3/miR-200b axis upon G6PD knockdown. The reconstitution of G6PD led to the recovery of the regulation of NOX/Smad3/miR-200b signaling and increased the expression of E-cadherin/ß-catenin in G6PD-knockdown cells. Thus, these results suggest that in the EMT process, G6PD plays an important regulatory role as an integral component of the NOX/Smad3/miR-200b axis.


Epithelial-Mesenchymal Transition , Glucosephosphate Dehydrogenase/metabolism , MicroRNAs/metabolism , NADPH Oxidases/metabolism , Smad3 Protein/metabolism , A549 Cells , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion Molecules/metabolism , Dogs , Embryo, Nonmammalian/metabolism , Embryonic Development , Glucosephosphate Dehydrogenase/antagonists & inhibitors , Glucosephosphate Dehydrogenase/genetics , Humans , Madin Darby Canine Kidney Cells , NADPH Oxidases/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , Zebrafish/growth & development , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
10.
Oncotarget ; 8(38): 63110-63120, 2017 Sep 08.
Article En | MEDLINE | ID: mdl-28968975

Glioblastomas are among the most fatal brain tumors; however, the molecular determinants of their tumorigenic behavior are not adequately defined. In this study, we analyzed the role of KMT2A in the glioblastoma cell line U-87 MG. KMT2A knockdown promoted cell proliferation. Moreover, it increased the DNA methylation of NOTCH1 and NOTCH3 and reduced the expression of NOTCH1 and NOTCH3. NOTCH1 or NOTCH3 activation inhibited U-87 MG cell proliferation, whereas NOTCH1 and NOTCH3 inhibition by shRNAs induced cell proliferation, thus demonstrating the tumor-suppressive ability of NOTCH1 and NOTCH3 in U-87 MG cells. The induced cell proliferation caused by KMT2A knockdown could be nullified by using either constitutively active NOTCH1 or constitutively active NOTCH3. This result demonstrates that KMT2A positively regulates NOTCH1 and NOTCH3 and that this mechanism is essential for inhibiting the U-87 MG cell proliferation. The role of KMT2A knockdown in promoting tumor growth was further confirmed in vivo by transplanting U-87 MG cells into the brains of zebrafish larvae. In conclusion, we identified KMT2A-NOTCH as a negative regulatory cascade for glioblastoma cell proliferation, and this result provides important information for KMT2A- or NOTCH-targeted therapeutic strategies for brain tumors.

11.
Dev Genes Evol ; 227(3): 219-230, 2017 06.
Article En | MEDLINE | ID: mdl-28154937

The forkhead box subclass O (FoxO) family of proteins is a group of highly evolutionary conserved transcription factors that regulate various cellular processes and embryonic development. Dysregulated expressions of FOXO genes have been identified in numerous tumors and genetic disorders. The expression of FOXO/Foxo, particularly FOXO4/Foxo4 and FOXO6/Foxo6, in the developing nervous system has not been fully characterized. Here, we identified zebrafish foxo4, foxo6a, and foxo6b homologs and demonstrated that all three genes were expressed in the developing nervous system. foxo4, foxo6a, and foxo6b displayed ubiquitous expression in the brain and later in distinct brain tissues. In addition, these three genes were expressed in different retinal layers in a time-dependent manner. Furthermore, the mRNA expression of all three genes was significantly downregulated after treatment with a selective PI3-kinase (PI3K) inhibitor, LY294002. Our results suggest that foxo4, foxo6a, and foxo6b play important roles in the developing brain and retina and that the transcriptional levels of these genes are regulated by PI3-kinase signaling.


Brain/metabolism , Forkhead Transcription Factors/genetics , Retina/metabolism , Zebrafish Proteins/genetics , Zebrafish/growth & development , Zebrafish/genetics , Amino Acid Sequence , Animals , Embryo, Nonmammalian/metabolism , Gene Expression Regulation , Phosphoinositide-3 Kinase Inhibitors , Phylogeny , Sequence Alignment , Signal Transduction , Transcription, Genetic , Zebrafish/metabolism
12.
Stem Cells ; 35(4): 1003-1014, 2017 04.
Article En | MEDLINE | ID: mdl-27790787

Neural crest progenitor cells, which give rise to many ectodermal and mesodermal derivatives, must maintain a delicate balance of apoptosis and proliferation for their final tissue contributions. Here we show that zebrafish bmp5 is expressed in neural crest progenitor cells and that it activates the Smad and Erk signaling pathways to regulate cell survival and proliferation, respectively. Loss-of-function analysis showed that Bmp5 was required for cell survival and this response is mediated by the Smad-Msxb signaling cascade. However, the Bmp5-Smad-Msxb signaling pathway had no effect on cell proliferation. In contrast, Bmp5 was sufficient to induce cell proliferation through the Mek-Erk-Id3 signaling cascade, whereas disruption of this signaling cascade had no effect on cell survival. Taken together, our results demonstrate an important regulatory mechanism for bone morphogenic protein-initiated signal transduction underlying the formation of neural crest progenitors. Stem Cells 2017;35:1003-1014.


Neural Crest/cytology , Neural Crest/metabolism , Signal Transduction , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Apoptosis , Bone Morphogenetic Protein 5 , Cell Proliferation , Cell Survival , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Gene Knockdown Techniques , Models, Biological , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Smad Proteins/metabolism , Zebrafish/embryology
13.
Biochim Biophys Acta Mol Cell Res ; 1864(3): 463-474, 2017 Mar.
Article En | MEDLINE | ID: mdl-27979767

Neural crest cells are multipotent progenitors that migrate extensively and differentiate into numerous derivatives. The developmental plasticity and migratory ability of neural crest cells render them an attractive model for studying numerous aspects of cell progression. We observed that zebrafish rgs2 was expressed in neural crest cells. Disrupting Rgs2 expression by using a dominant negative rgs2 construct or rgs2 morpholinos reduced GTPase-activating protein activity, induced the formation of neural crest progenitors, increased the proliferation of nonectomesenchymal neural crest cells, and inhibited the formation of ectomesenchymal neural crest derivatives. The transcription of pparda (which encodes Pparδ, a Wnt-activated transcription factor) was upregulated in Rgs2-deficient embryos, and Pparδ inhibition using a selective antagonist in the Rgs2-deficient embryos repaired neural crest defects. Our results clarify the mechanism through which the Rgs2-Pparδ cascade regulates neural crest development; specifically, Pparδ directly binds to the promoter and upregulates the transcription of the neural crest specifier sox10. This study reveals a unique regulatory mechanism, the Rgs2-Pparδ-Sox10 signaling cascade, and defines a key molecular regulator, Rgs2, in neural crest development.


Neural Crest/metabolism , Neurogenesis/genetics , PPAR delta/genetics , RGS Proteins/genetics , SOXE Transcription Factors/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Amino Acid Sequence , Animals , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Neural Crest/growth & development , PPAR delta/metabolism , Promoter Regions, Genetic , RGS Proteins/metabolism , SOXE Transcription Factors/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transcriptional Activation , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt Signaling Pathway , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/metabolism
14.
Neural Dev ; 10: 28, 2015 Dec 30.
Article En | MEDLINE | ID: mdl-26714454

BACKGROUND: Notch signaling has been conserved throughout evolution and plays a fundamental role in various neural developmental processes and the pathogenesis of several human cancers and genetic disorders. However, how Notch signaling regulates various cellular processes remains unclear. Although Deltex proteins have been identified as cytoplasmic downstream elements of the Notch signaling pathway, few studies have been reported on their physiological role. RESULTS: We isolated zebrafish deltex1 (dtx1) and showed that this gene is primarily transcribed in the developing nervous system, and its spatiotemporal expression pattern suggests a role in neural differentiation. The transcription of dtx1 was suppressed by the direct binding of the Notch downstream transcription factors Her2 and Her8a. Overexpressing the complete coding sequence of Dtx1 was necessary for inducing neuronal and glial differentiation. By contrast, disrupting Dtx1 expression by using a Dtx1 construct without the RING finger domain reduced neuronal and glial differentiation. This effect was phenocopied by the knockdown of endogenous Dtx1 expression by using morpholinos, demonstrating the essential function of the RING finger domain and confirming the knockdown specificity. Cell proliferation and apoptosis were unaltered in Dtx1-overexpressed and -deficient zebrafish embryos. Examination of the expression of her2 and her8a in embryos with altered Dtx1 expression showed that Dxt1-induced neuronal differentiation did not require a regulatory effect on the Notch-Hairy/E(Spl) pathway. However, both Dtx1 and Notch activation induced glial differentiation, and Dtx1 and Notch activation negatively inhibited each other in a reciprocal manner, which achieves a proper balance for the expression of Dtx1 and Notch to facilitate glial differentiation. We further confirmed that the Dtx1-Notch-Hairy/E(Spl) cascade was sufficient to induce neuronal and glial differentiation by concomitant injection of an active form of Notch with dtx1, which rescued the neuronogenic and gliogenic defects caused by the activation of Notch signaling. CONCLUSIONS: Our results demonstrated that Dtx1 is regulated by Notch-Hairy/E(Spl) signaling and is a major factor specifically regulating neural differentiation. Thus, our results provide new insights into the mediation of neural development by the Notch signaling pathway.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , DNA-Binding Proteins/metabolism , Neuroglia/metabolism , Neurons/metabolism , Receptors, Notch/metabolism , Zebrafish Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Neurogenesis/physiology , Neuroglia/cytology , Neurons/cytology , Receptors, Notch/genetics , Signal Transduction/physiology , Zebrafish , Zebrafish Proteins/genetics
15.
Stem Cells Dev ; 24(24): 2943-51, 2015 Dec 15.
Article En | MEDLINE | ID: mdl-26414806

Growth-arrest-specific 7 (Gas7) is preferentially expressed in the nervous system and plays an important role during neuritogenesis in vertebrates. We recently demonstrated that gas7 is highly expressed in zebrafish neurons, where it regulates neural development. The possibility that gas7 may also regulate the development of other tissues remains to be examined. In this study, we investigate the role of Gas7 in the development of craniofacial tissues. Knockdown of gas7 using morpholino oligomers produced abnormal phenotypes in neural crest (NC) cells and their derivatives. NC-derived cartilage maturation was altered in Gas7 morphants as revealed by aberrant sox9b and dlx2 expression, a phenotype that could be rescued by coinjection of gas7 mRNA. While rhombomere morphology remained normal in Gas7 morphants, we observed reduced expression of the prechondrogenic genes sox9b and dlx2 in cells populating the posterior pharyngeal arches, but the fundamental structure of pharyngeal arches was preserved. In addition, NC cell sublineages that migrate to form neurons, glial cells, and melanocytes were altered in Gas7 morphants as revealed by aberrant expression of neurod, foxd3, and mitfa, respectively. Development of NC progenitors was also examined in Gas7 morphants at 12 hpf, and we observed that the reduction of cell precursors in Gas7 morphants was due to increased apoptosis level. These results indicate that the formation of NC progenitors and derivatives depends on Gas7 expression. Our observations also suggest that Gas7 regulates the formation of NC derivatives constituting the internal tissues of pharyngeal arches, without affecting the fundamental structure of mesodermal-derived pharyngeal arches.


Nerve Tissue Proteins/metabolism , Neural Crest/metabolism , Zebrafish Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Morphogenesis , Nerve Tissue Proteins/genetics , Neural Crest/embryology , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish , Zebrafish Proteins/genetics
16.
Dev Neurobiol ; 75(5): 452-62, 2015 May.
Article En | MEDLINE | ID: mdl-25284327

Multiple epigenetic factors play a critical role in cell proliferation and differentiation. However, their function in embryogenesis, especially in neural development, is currently unclear. The Trithorax group (TrxG) homolog KMT2A (MLL1) is an important epigenetic regulator during development and has an especially well-defined role in hematopoiesis. Translocation and aberrant expression of KMT2A is often observed in many tumors, indicating its proto-oncogenic character. Here, we show that Kmt2a was essential for neural development in zebrafish embryos. Disrupting the expression of Kmt2a using morpholino antisense oligonucleotides and a dominant-negative variant resulted in neurogenic phenotypes, including downregulated proliferation of neural progenitors, premature differentiation of neurons, and impaired gliogenesis. This study therefore revealed a novel function of Kmt2a in cell proliferation and differentiation, providing further insight into the function of TrxG proteins in neural development and brain tumors.


Cell Differentiation/physiology , Cell Proliferation , Gene Expression Regulation, Developmental/physiology , Histone-Lysine N-Methyltransferase/metabolism , Neural Stem Cells/cytology , Neurogenesis/physiology , Neuroglia/cytology , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Neurons/physiology
17.
Dev Biol ; 397(1): 116-28, 2015 Jan 01.
Article En | MEDLINE | ID: mdl-25446033

The study of molecular regulation in neural development provides information to understand how diverse neural cells are generated. It also helps to establish therapeutic strategies for the treatment of neural degenerative disorders and brain tumors. The Hairy/E(spl) family members are potential targets of Notch signaling, which is fundamental to neural cell maintenance, cell fate decisions, and compartment boundary formation. In this study, we isolated a zebrafish homolog of Hairy/E(spl), her2, and showed that this gene is expressed in neural progenitor cells and in the developing nervous system. The expression of her2 required Notch activation, as revealed by a Notch-defective mutant and a chemical inhibitor, N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT). The endogenous expression of Her2 was altered by both overexpression and morpholino-knockdown approaches, and the results demonstrated that Her2 was both necessary and sufficient to promote the proliferation of neural progenitors by inhibiting the transcription of the cell cycle inhibitors cdkn1a, cdkn1ba, and cdkn1bb. Her2 knockdown caused premature neuronal differentiation, which indicates that Her2 is essential for inhibiting neuronal differentiation. At a later stage of neural development, Her2 could induce glial differentiation. The overexpression of Her2 constructs lacking the bHLH or WRPW domain phenocopied the effect of the morpholino knockdown, demonstrating the essential function of these two domains and further confirming the knockdown specificity. In conclusion, our data reveal that Her2 promotes progenitor proliferation and maintains progenitor characteristics by inhibiting neuronal differentiation. Together, these two mechanisms ensure the proper development of the neural progenitor cell pool.


Gene Expression Regulation, Developmental , Neurogenesis/physiology , Neuroglia/metabolism , Neurons/metabolism , Receptor, ErbB-2/physiology , Zebrafish Proteins/physiology , Animals , Cell Cycle , Cell Differentiation , Cell Proliferation , Dipeptides/chemistry , Gene Expression Profiling , Genes, Dominant , Receptor, ErbB-2/genetics , Signal Transduction , Time Factors , Zebrafish , Zebrafish Proteins/genetics
18.
J Cell Sci ; 126(Pt 24): 5626-34, 2013 Dec 15.
Article En | MEDLINE | ID: mdl-24101720

Hematopoietic and vascular endothelial cells constitute the circulatory system and are both generated from the ventral mesoderm. However, the molecules and signaling pathways involved in ventral mesoderm formation and specification remain unclear. We found that zebrafish etv5a was expressed in the ventral mesoderm during gastrulation. Knockdown of Etv5a using morpholinos increased the proliferation of ventral mesoderm cells and caused defects in hematopoietic derivatives and in vascular formation. By contrast, the formation of other mesodermal derivatives, such as pronephros, somites and the gut wall, was not affected. Knockdown specificity was further confirmed by overexpression of an etv5a construct lacking its acidic domain. In conclusion, our data reveal that etv5a is essential for the inhibition of ventral mesoderm cell proliferation and for the formation of the hemato-vascular lineage.


Cell Proliferation , Mesoderm/cytology , Proto-Oncogene Proteins c-ets/physiology , Zebrafish Proteins/physiology , Zebrafish/embryology , Animals , Apoptosis , Cell Differentiation , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Gastrulation , Gene Expression , Gene Knockdown Techniques , Morpholinos/genetics , Neovascularization, Physiologic
19.
PLoS One ; 8(1): e54262, 2013.
Article En | MEDLINE | ID: mdl-23342113

Akt1 is well known for its role in regulating cell proliferation, differentiation, and apoptosis and is implicated in tumors and several neurological disorders. However, the role of Akt1 in neural development has not been well defined. We have isolated zebrafish akt1 and shown that this gene is primarily transcribed in the developing nervous system, and its spatiotemporal expression pattern suggests a role in neural differentiation. Injection of akt1 morpholinos resulted in loss of neuronal precursors with a concomitant increase in post-mitotic neurons, indicating that knockdown of Akt1 is sufficient to cause premature differentiation of neurons. A similar phenotype was observed in embryos deficient for Notch signaling. Both the ligand (deltaA) and the downstream target of Notch (her8a) were downregulated in akt1 morphants, indicating that Akt1 is required for Delta-Notch signaling. Furthermore, akt1 expression was downregulated in Delta-Notch signaling-deficient embryos and could be induced by constitutive activation of Notch signaling. In addition, knockdown of Akt1 was able to nullify the inhibition of neuronal differentiation caused by constitutive activation of Notch signaling. Taken together, these results provide in vivo evidence that Akt1 interacts with Notch signaling reciprocally and provide an explanation of why Akt1 is essential for the inhibition of neuronal differentiation.


Neurons/cytology , Neurons/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Notch/metabolism , Zebrafish Proteins/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Proto-Oncogene Proteins c-akt/genetics , Receptors, Notch/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Zebrafish , Zebrafish Proteins/genetics
20.
Dev Biol ; 375(1): 1-12, 2013 Mar 01.
Article En | MEDLINE | ID: mdl-23328254

Delta/notch-like epidermal growth factor (EGF)-related receptor (DNER) is a single-pass transmembrane protein found to be a novel ligand in the Notch signaling pathway. Its function was previously characterized in the developing cerebellum and inner ear hair cells. In this study, we isolated a zebrafish homolog of DNER and showed that this gene is expressed in the developing nervous system. Overexpression of dner or the intracellular domain of dner was sufficient to inhibit the proliferation of neural progenitors and induce neuronal and glial differentiation. In contrast, the knockdown of endogenous Dner expression using antisense morpholino oligonucleotides increased the proliferation of neural progenitors and maintained neural cells in a progenitor status through inhibition of neuronal and glial differentiation. Through analysis of the antagonistic effect on the Delta ligand and the role of the potential downstream mediator Deltex1, we showed that Dner acts in Notch-dependent and Notch-independent manner. This is the first study to demonstrate a role for Dner in neural progenitors and neuronal differentiation and provides new insights into mediation of neuronal development and differentiation by the Notch signaling pathway.


DNA-Binding Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neurogenesis , Neuroglia/cytology , Neurons/cytology , Receptors, Cell Surface/metabolism , Receptors, Notch/metabolism , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Cell Proliferation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Knockdown Techniques , Morpholinos , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nervous System/embryology , Nervous System/metabolism , Neural Stem Cells/physiology , Neuroglia/physiology , Neurons/physiology , Oligodeoxyribonucleotides, Antisense , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Sequence Alignment , Signal Transduction , Zebrafish , Zebrafish Proteins/biosynthesis , Zebrafish Proteins/genetics
...