Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Genet ; 18(5): e1010207, 2022 05.
Article in English | MEDLINE | ID: mdl-35580127

ABSTRACT

In developing melanocytes and in melanoma cells, multiple paralogs of the Activating-enhancer-binding Protein 2 family of transcription factors (TFAP2) contribute to expression of genes encoding pigmentation regulators, but their interaction with Microphthalmia transcription factor (MITF), a master regulator of these cells, is unclear. Supporting the model that TFAP2 facilitates MITF's ability to activate expression of pigmentation genes, single-cell seq analysis of zebrafish embryos revealed that pigmentation genes are only expressed in the subset of mitfa-expressing cells that also express tfap2 paralogs. To test this model in SK-MEL-28 melanoma cells we deleted the two TFAP2 paralogs with highest expression, TFAP2A and TFAP2C, creating TFAP2 knockout (TFAP2-KO) cells. We then assessed gene expression, chromatin accessibility, binding of TFAP2A and of MITF, and the chromatin marks H3K27Ac and H3K27Me3 which are characteristic of active enhancers and silenced chromatin, respectively. Integrated analyses of these datasets indicate TFAP2 paralogs directly activate enhancers near genes enriched for roles in pigmentation and proliferation, and directly repress enhancers near genes enriched for roles in cell adhesion. Consistently, compared to WT cells, TFAP2-KO cells proliferate less and adhere to one another more. TFAP2 paralogs and MITF co-operatively activate a subset of enhancers, with the former necessary for MITF binding and chromatin accessibility. By contrast, TFAP2 paralogs and MITF do not appear to co-operatively inhibit enhancers. These studies reveal a mechanism by which TFAP2 profoundly influences the set of genes activated by MITF, and thereby the phenotype of pigment cells and melanoma cells.


Subject(s)
Melanoma , Microphthalmos , Animals , Cell Proliferation/genetics , Chromatin/genetics , Chromatin/metabolism , Gene Expression Regulation, Developmental , Melanocytes/metabolism , Melanoma/genetics , Melanoma/metabolism , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Microphthalmos/genetics , Pigmentation/genetics , Zebrafish/genetics , Zebrafish/metabolism
2.
Methods Mol Biol ; 2403: 91-106, 2022.
Article in English | MEDLINE | ID: mdl-34913119

ABSTRACT

Danio rerio (zebrafish), traditionally used in forward genetic screens, has in the last decade become a popular model for reverse genetic studies with the introduction of TALENS, zinc finger nucleases, and CRISPR/Cas9. Unexpectedly, homozygous frameshift mutations generated by these tools frequently result in phenotypes that are less penetrant than those seen in embryos injected with antisense morpholino oligonucleotides targeting the same gene. One explanation for the difference is that some frameshift mutations result in nonsense-mediated decay of the gene transcript, a process which can induce expression of homologous genes. This form of genetic compensation, called transcriptional adaptation, does not occur when the mutant allele results in no RNA transcripts being produced from the targeted gene. Such RNA-less mutants can be generated by deleting a gene's promoter using a pair of guide RNAs and Cas9 protein. Here, we present a protocol and use it to generate alleles of arhgap29b and slc41a1 that lack detectable zygotic transcription. In the case of the arhgap29b mutant, an emerging phenotype did not segregate with the promoter deletion mutation, highlighting the potential for off-target mutagenesis with these tools. In summary, this chapter describes a method to generate zebrafish mutants that avoid a form of genetic compensation that occurs in many frameshift mutants.


Subject(s)
Zebrafish , Alleles , Animals , CRISPR-Cas Systems/genetics , Cation Transport Proteins , RNA , RNA, Guide, Kinetoplastida/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
3.
J Invest Dermatol ; 133(1): 68-77, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22931925

ABSTRACT

IFN regulatory factor 6 (IRF6) is a transcription factor that, in mammals, is required for the differentiation of skin, breast epithelium, and oral epithelium. However, the transcriptional targets that mediate these effects are currently unknown. In zebrafish and frog embryos, Irf6 is necessary for differentiation of the embryonic superficial epithelium, or periderm. Here we use microarrays to identify genes that are expressed in the zebrafish periderm and whose expression is inhibited by a dominant-negative variant of Irf6 (dnIrf6). These methods identify Grainyhead-like 3 (Grhl3), an ancient regulator of the epidermal permeability barrier, as acting downstream of Irf6. In human keratinocytes, IRF6 binds conserved elements near the GRHL3 [corrected] promoter. We show that one of these elements has enhancer activity in human keratinocytes and zebrafish periderm, suggesting that Irf6 directly stimulates Grhl3 expression in these tissues. Simultaneous inhibition of grhl1 and grhl3 disrupts periderm differentiation in zebrafish, and, intriguingly, forced grhl3 expression restores periderm markers in both zebrafish injected with dnIrf6 and frog embryos depleted of Irf6. Finally, in Irf6-deficient mouse embryos, Grhl3 expression in the periderm and oral epithelium is virtually absent. These results indicate that Grhl3 is a key effector of Irf6 in periderm differentiation.


Subject(s)
DNA-Binding Proteins/biosynthesis , Germ Layers/growth & development , Germ Layers/metabolism , Interferon Regulatory Factors/metabolism , Transcription Factors/biosynthesis , Zebrafish Proteins/biosynthesis , Zebrafish Proteins/metabolism , Animals , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental/genetics , Gene Silencing , Germ Layers/embryology , Humans , Keratinocytes/metabolism , Mice , Promoter Regions, Genetic , Transcription Factors/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
4.
PLoS Genet ; 8(10): e1002966, 2012.
Article in English | MEDLINE | ID: mdl-23055939

ABSTRACT

Sensory hair cells are essential for hearing and balance. Their development from epithelial precursors has been extensively characterized with respect to transcriptional regulation, but not in terms of posttranscriptional influences. Here we report on the identification and functional characterization of an alternative-splicing regulator whose inactivation is responsible for defective hair-cell development, deafness, and impaired balance in the spontaneous mutant Bronx waltzer (bv) mouse. We used positional cloning and transgenic rescue to locate the bv mutation to the splicing factor-encoding gene Ser/Arg repetitive matrix 4 (Srrm4). Transcriptome-wide analysis of pre-mRNA splicing in the sensory patches of embryonic inner ears revealed that specific alternative exons were skipped at abnormally high rates in the bv mice. Minigene experiments in a heterologous expression system confirmed that these skipped exons require Srrm4 for inclusion into the mature mRNA. Sequence analysis and mutagenesis experiments showed that the affected transcripts share a novel motif that is necessary for the Srrm4-dependent alternative splicing. Functional annotations and protein-protein interaction data indicated that the encoded proteins cluster in the secretion and neurotransmission pathways. In addition, the splicing of a few transcriptional regulators was found to be Srrm4 dependent, and several of the genes known to be targeted by these regulators were expressed at reduced levels in the bv mice. Although Srrm4 expression was detected in neural tissues as well as hair cells, analyses of the bv mouse cerebellum and neocortex failed to detect splicing defects. Our data suggest that Srrm4 function is critical in the hearing and balance organs, but not in all neural tissues. Srrm4 is the first alternative-splicing regulator to be associated with hearing, and the analysis of bv mice provides exon-level insights into hair-cell development.


Subject(s)
Alternative Splicing , Deafness/genetics , Mutation , Nerve Tissue Proteins/genetics , Animals , Base Sequence , Cell Line , Cerebellum/metabolism , Cluster Analysis , Disease Models, Animal , Gene Order , Hair Cells, Auditory, Inner/metabolism , Humans , Mice , Molecular Sequence Data , Nucleotide Motifs , Phenotype , RNA Precursors/chemistry , RNA Precursors/genetics , RNA Precursors/metabolism , Transcriptome , Transgenes
5.
PLoS Genet ; 6(9): e1001122, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20862309

ABSTRACT

A model of the gene-regulatory-network (GRN), governing growth, survival, and differentiation of melanocytes, has emerged from studies of mouse coat color mutants and melanoma cell lines. In this model, Transcription Factor Activator Protein 2 alpha (TFAP2A) contributes to melanocyte development by activating expression of the gene encoding the receptor tyrosine kinase Kit. Next, ligand-bound Kit stimulates a pathway activating transcription factor Microphthalmia (Mitf), which promotes differentiation and survival of melanocytes by activating expression of Tyrosinase family members, Bcl2, and other genes. The model predicts that in both Tfap2a and Kit null mutants there will be a phenotype of reduced melanocytes and that, because Tfap2a acts upstream of Kit, this phenotype will be more severe, or at least as severe as, in Tfap2a null mutants in comparison to Kit null mutants. Unexpectedly, this is not the case in zebrafish or mouse. Because many Tfap2 family members have identical DNA-binding specificity, we reasoned that another Tfap2 family member may work redundantly with Tfap2a in promoting Kit expression. We report that tfap2e is expressed in melanoblasts and melanophores in zebrafish embryos and that its orthologue, TFAP2E, is expressed in human melanocytes. We provide evidence that Tfap2e functions redundantly with Tfap2a to maintain kita expression in zebrafish embryonic melanophores. Further, we show that, in contrast to in kita mutants where embryonic melanophores appear to differentiate normally, in tfap2a/e doubly-deficient embryonic melanophores are small and under-melanized, although they retain expression of mitfa. Interestingly, forcing expression of mitfa in tfap2a/e doubly-deficient embryos partially restores melanophore differentiation. These findings reveal that Tfap2 activity, mediated redundantly by Tfap2a and Tfap2e, promotes melanophore differentiation in parallel with Mitf by an effector other than Kit. This work illustrates how analysis of single-gene mutants may fail to identify steps in a GRN that are affected by the redundant activity of related proteins.


Subject(s)
Cell Differentiation , Melanophores/cytology , Melanophores/metabolism , Transcription Factor AP-2/metabolism , Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cell Death , Cell Lineage , Cells, Cultured , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Humans , Melanocytes/cytology , Melanocytes/metabolism , Mice , Microphthalmia-Associated Transcription Factor/genetics , Models, Biological , Mutation/genetics , Organ Specificity/genetics , Proto-Oncogene Proteins c-kit/metabolism , Transcription Factor AP-2/genetics , Transcription Factors/genetics , Transcriptional Activation/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
6.
J Invest Dermatol ; 127(8): 2020-30, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17290233

ABSTRACT

Transient receptor potential melastatin 7 (TRPM7) is a broadly expressed, non-selective cation channel. Studies in cultured cells implicate TRPM7 in regulation of cell growth, spreading, and survival. However, zebrafish trpm7 homozygous mutants display death of melanophores and temporary paralysis, but no gross morphological defects during embryonic stages. This phenotype implies that melanophores are unusually sensitive to decreases in Trpm7 levels, a hypothesis we investigate here. We find that pharmacological inhibition of caspases does not rescue melanophore viability in trpm7 mutants, implying that melanophores die by a mechanism other than apoptosis. Consistent with this possibility, ultrastructural analysis of dying melanophores in trpm7 mutants reveals abnormal melanosomes and evidence of a ruptured plasma membrane, indicating that cell death occurs by necrosis. Interestingly, inhibition of melanin synthesis largely prevents melanophore cell death in trpm7 mutants. These results suggest that melanophores require Trpm7 in order to detoxify intermediates of melanin synthesis. We find that unlike TRPM1, TRPM7 is expressed in human melanoma cell lines, indicating that these cells may also be sensitized to reduction of TRPM7 levels.


Subject(s)
Melanins/biosynthesis , Melanophores/cytology , TRPM Cation Channels/physiology , Zebrafish Proteins/physiology , Animals , Apoptosis , Caspase Inhibitors , Cell Line, Tumor , Cell Survival , Embryo, Nonmammalian/cytology , Magnesium/pharmacology , Melanoma/chemistry , Mutation , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins c-kit/physiology , TRPM Cation Channels/analysis , TRPM Cation Channels/genetics , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/physiology , Zebrafish , Zebrafish Proteins/analysis , Zebrafish Proteins/genetics
7.
Dev Dyn ; 233(4): 1560-70, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15968639

ABSTRACT

Certain aggressive melanoma cell lines exhibit a dedifferentiated phenotype, expressing genes that are characteristic of various cell types including endothelial, neural, and stem cells. Moreover, we have shown that aggressive melanoma cells can participate in neovascularization in vivo and vasculogenic mimicry in vitro, demonstrating that these cells respond to microenvironmental cues and manifest developmental plasticity. To explore this plasticity further, we transplanted human metastatic melanoma cells into zebrafish blastula-stage embryos and monitored their behavior post-transplantation. The data show that human metastatic melanoma cells placed in the zebrafish embryo survive, exhibit motility, and divide. The melanoma cells do not form tumors nor integrate into host organs, but instead become scattered throughout the embryo in interstitial spaces, reflecting the dedifferentiated state of the cancer cells. In contrast to the fate of melanoma cells, human melanocytes transplanted into zebrafish embryos most frequently become distributed to their normal microenvironment of the skin, revealing that the zebrafish embryo contains possible homing cues that can be interpreted by normal human cells. Finally, we show that within the zebrafish embryo, metastatic melanoma cells retain their dedifferentiated phenotype. These results demonstrate the utility of the zebrafish embryonic model for the study of tumor cell plasticity and suggest that this experimental paradigm can be a powerful one in which to investigate tumor-microenvironment interactions.


Subject(s)
Cell Division/physiology , Cell Movement/physiology , Melanoma/pathology , Zebrafish/physiology , Animals , Cell Line, Tumor , Fibroblasts/physiology , Fibroblasts/transplantation , Humans , Melanocytes/physiology , Melanocytes/transplantation , Microscopy, Confocal , Neoplasm Metastasis , Neoplasm Transplantation , Transplantation, Heterologous
8.
Mech Dev ; 121(11): 1365-76, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15454266

ABSTRACT

An outstanding problem in the study of vertebrate development is the identification of the genes that direct neural crest precursor cells to adopt and maintain specific differentiated cell fates. In an effort to identify such genes, we have carried out a mutagenesis screen in zebrafish and isolated mutants that lack neural crest-derived melanophores. In this manuscript we describe the phenotype of one such mutant, touchtone(b722) (tct), and the map position of the gene it defines. Analysis of expression of dopachrome tautomerase (dct) and microphthalmia (mitfa) suggests that melanophore precursors are specified normally in homozygous tct mutants. However, differentiated melanophores are pale, small, and about half of them have disappeared by 48 h of development, apparently by cell death. We show that melanophores require Tct function cell autonomously. Signals from the receptor tyrosine kinase receptor C-kit are essential for survival of melanophores in zebrafish and mammals. However, differences in the phenotypes of tct and c-kit homozygous mutants, and an absence of interaction between c-kit and tct heterozygotes, suggest that Tct functions independently of the C-kit pathway. Other neural crest derivatives, including other pigment cell types, appear normal in tct mutants. Interestingly, tct mutant embryos undergo a temporary period of near complete paralyzis during the second day of development, although markers of axons of motor and sensory neurons look normal in this period. A fraction of tct(b722) mutants survive to adulthood, but mutant adults are small, indicating a role for Tct in post-larval growth. The tct gene maps to a small interval near a telomere of chromosome 18. Thus, we have identified a zebrafish gene that when mutated produces semi-viable offspring and that may serve as a model of human diseases that have both pigmentation and neurological symptoms.


Subject(s)
Melanophores/cytology , Zebrafish Proteins/physiology , Zebrafish/embryology , Animals , Cell Death/physiology , Cell Differentiation/physiology , Cell Survival , Chromosome Mapping , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/physiology , Gene Expression Regulation, Developmental , Melanophores/chemistry , Melanophores/physiology , Mutation/genetics , Phenotype , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins c-kit/physiology , TRPM Cation Channels , Zebrafish/genetics , Zebrafish Proteins/analysis , Zebrafish Proteins/genetics
9.
Dev Biol ; 265(1): 246-61, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14697367

ABSTRACT

The genes that control development of embryonic melanocytes are poorly defined. Although transcription factor Ap-2alpha is expressed in neural crest (NC) cells, its role in development of embryonic melanocytes and other neural crest derivatives is unclear because mouse Ap-2alpha mutants die before melanogenesis. We show that zebrafish embryos injected with morpholino antisense oligonucleotides complementary to ap-2alpha (ap-2alpha MO) complete early morphogenesis normally and have neural crest cells. Expression of c-kit, which encodes the receptor for the Steel ligand, is reduced in these embryos, and, similar to zebrafish c-kit mutant embryos, embryonic melanophores are reduced in number and migration. The effects of ap-2alpha MO injected into heterozygous and homozygous c-kit mutants support the notion that Ap-2alpha works through C-kit and additional target genes to mediate melanophore cell number and migration. In contrast to c-kit mutant embryos, in ap-2alpha MO-injected embryos, melanophores are small and under-pigmented, and unexpectedly, analysis of mosaic embryos suggests Ap-2alpha regulates melanophore differentiation through cell non-autonomous targets. In addition to melanophore phenotypes, we document reduction of other neural crest derivatives in ap-2alpha MO-injected embryos, including jaw cartilage, enteric neurons, and sympathetic neurons. These results reveal that Ap-2alpha regulates multiple steps of melanophore development, and is required for development of other neuronal and non-neuronal neural crest derivatives.


Subject(s)
Autonomic Pathways/embryology , Branchial Region/embryology , DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental , Melanophores/physiology , Transcription Factors/physiology , Zebrafish/embryology , Amino Acid Sequence , Animals , Antisense Elements (Genetics) , Gene Expression Profiling , Molecular Sequence Data , Neural Crest/embryology , Proto-Oncogene Proteins c-kit , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Transcription Factor AP-2 , Zebrafish Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...