Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Genesis ; 62(1): e23580, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37974491

RESUMO

Bop1 can promote cell proliferation and is a component of the Pes1-Bop1-WDR12 (PeBoW) complex that regulates ribosomal RNA processing and biogenesis. In embryos, however, bop1 mRNA is highly enriched in the neural plate, cranial neural crest and placodes, and potentially may interact with Six1, which also is expressed in these tissues. Recent work demonstrated that during development, Bop1 is required for establishing the size of the tadpole brain, retina and cranial cartilages, as well as controlling neural tissue gene expression levels. Herein, we extend this work by assessing the effects of Bop1 knockdown at neural plate and larval stages. Loss of Bop1 expanded neural plate gene expression domains (sox2, sox11, irx1) and reduced neural crest (foxd3, sox9), placode (six1, sox11, irx1, sox9) and epidermal (dlx5) expression domains. At larval stages, Bop1 knockdown reduced the expression of several otic vesicle genes (six1, pax2, irx1, sox9, dlx5, otx2, tbx1) and branchial arch genes that are required for chondrogenesis (sox9, tbx1, dlx5). The latter was not the result of impaired neural crest migration. Together these observations indicate that Bop1 is a multifunctional protein that in addition to its well-known role in ribosomal biogenesis functions during early development to establish the craniofacial precursor domains.


Assuntos
Crista Neural , Fatores de Transcrição , Crista Neural/metabolismo , Fatores de Transcrição/metabolismo , Cabeça , Crânio/metabolismo , Ribossomos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento
2.
Dev Biol ; 494: 71-84, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36521641

RESUMO

The morphogenesis of the otic vesicle (OV) to form inner ear organs serves as an excellent model system to understand cell fate acquisition on a single cell level. Tbx2 and Tbx3 (Tbx2/3) encode closely related T-box transcription factors that are expressed widely in the mammalian OV. Inactivation of both genes in the OV (Tbx2/3cKO) results in failed morphogenesis into inner ear organs. To understand the basis of these defects, single cell RNA-sequencing (scRNA-seq) was performed on the OV lineage, in controls versus Tbx2/3cKO embryos. We identified a multipotent population termed otic progenitors in controls that are marked by expression of the known otic placode markers Eya1, Sox2, and Sox3 as well as new markers Fgf18, Cxcl12, and Pou3f3. The otic progenitor population was increased three-fold in Tbx2/3cKO embryos, concomitant with dysregulation of genes in these cells as well as reduced progression to more differentiated states of prosensory and nonsensory cells. An ectopic neural population of cells was detected in the posterior OV of Tbx2/3cKO embryos but had reduced maturation to delaminated neural cells. As all three cell fates were affected in Tbx2/3cKO embryos, we suggest that Tbx2/3 promotes progression of multipotent otic progenitors to more differentiated cell types in the OV.


Assuntos
Orelha Interna , Animais , Diferenciação Celular/genética , Orelha Interna/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Mamíferos/metabolismo , Morfogênese , Sistema Nervoso/metabolismo , Fatores de Transcrição/metabolismo , Proteínas com Domínio T
3.
Development ; 148(8)2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33795231

RESUMO

All epithelial components of the inner ear, including sensory hair cells and innervating afferent neurons, arise by patterning and differentiation of epithelial progenitors residing in a simple sphere, the otocyst. Here, we identify the transcriptional repressors TBX2 and TBX3 as novel regulators of these processes in the mouse. Ablation of Tbx2 from the otocyst led to cochlear hypoplasia, whereas loss of Tbx3 was associated with vestibular malformations. The loss of function of both genes (Tbx2/3cDKO) prevented inner ear morphogenesis at midgestation, resulting in indiscernible cochlear and vestibular structures at birth. Morphogenetic impairment occurred concomitantly with increased apoptosis in ventral and lateral regions of Tbx2/3cDKO otocysts around E10.5. Expression analyses revealed partly disturbed regionalisation, and a posterior-ventral expansion of the neurogenic domain in Tbx2/3cDKO otocysts at this stage. We provide evidence that repression of FGF signalling by TBX2 is important to restrict neurogenesis to the anterior-ventral otocyst and implicate another T-box factor, TBX1, as a crucial mediator in this regulatory network.


Assuntos
Apoptose , Orelha Interna/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Organogênese , Transdução de Sinais , Proteínas com Domínio T/biossíntese , Animais , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Camundongos , Camundongos Knockout , Proteínas com Domínio T/genética
4.
Development ; 148(17)2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34414417

RESUMO

Branchio-oto-renal syndrome (BOR) is a disorder characterized by hearing loss, and craniofacial and/or renal defects. Variants in the transcription factor Six1 and its co-factor Eya1, both of which are required for otic development, are linked to BOR. We previously identified Sobp as a potential Six1 co-factor, and SOBP variants in mouse and humans cause otic phenotypes; therefore, we asked whether Sobp interacts with Six1 and thereby may contribute to BOR. Co-immunoprecipitation and immunofluorescence experiments demonstrate that Sobp binds to and colocalizes with Six1 in the cell nucleus. Luciferase assays show that Sobp interferes with the transcriptional activation of Six1+Eya1 target genes. Experiments in Xenopus embryos that either knock down or increase expression of Sobp show that it is required for formation of ectodermal domains at neural plate stages. In addition, altering Sobp levels disrupts otic vesicle development and causes craniofacial cartilage defects. Expression of Xenopus Sobp containing the human variant disrupts the pre-placodal ectoderm similar to full-length Sobp, but other changes are distinct. These results indicate that Sobp modifies Six1 function and is required for vertebrate craniofacial development, and identify Sobp as a potential candidate gene for BOR.


Assuntos
Desenvolvimento Ósseo , Proteínas de Homeodomínio/metabolismo , Metaloproteínas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Xenopus/metabolismo , Animais , Síndrome Brânquio-Otorrenal/embriologia , Síndrome Brânquio-Otorrenal/genética , Núcleo Celular/metabolismo , Orelha Interna/embriologia , Orelha Interna/metabolismo , Ectoderma/embriologia , Ectoderma/metabolismo , Expressão Gênica , Proteínas de Homeodomínio/genética , Larva/crescimento & desenvolvimento , Metaloproteínas/genética , Crista Neural/embriologia , Crista Neural/metabolismo , Proteínas Nucleares/genética , Ligação Proteica , Proteínas Tirosina Fosfatases/metabolismo , Ativação Transcricional , Proteínas de Xenopus/genética , Xenopus laevis
5.
Dev Dyn ; 252(12): 1407-1427, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37597164

RESUMO

BACKGROUND: Members of the sulfotransferase superfamily (SULT) influence the activity of a wide range of hormones, neurotransmitters, metabolites and xenobiotics. However, their roles in developmental processes are not well characterized even though they are expressed during embryogenesis. We previously found in a microarray screen that Six1 up-regulates LOC100037047, which encodes XB5850668.L, an uncharacterized sulfotransferase. RESULTS: Since Six1 is required for patterning the embryonic ectoderm into its neural plate, neural crest, preplacodal and epidermal domains, we used loss- and gain-of function assays to characterize the role of XB5850668.L during this process. Knockdown of endogenous XB5850668.L resulted in the reduction of epidermal, neural crest, cranial placode and otic vesicle gene expression domains, concomitant with neural plate expansion. Increased levels had minimal effects, but infrequently expanded neural plate and neural crest gene domains, and infrequently reduced cranial placode and otic vesicle gene domains. Mutation of two key amino acids in the sulfotransferase catalytic domain required for PAPS binding and enzymatic activity tended to reduce the effects of overexpressing the wild-type protein. CONCLUSIONS: Our analyses indicates that XB5850668.L is a member of the SULT2 family that plays important roles in patterning the embryonic ectoderm. Some aspects of its influence likely depend on sulfotransferase activity.


Assuntos
Ectoderma , Crista Neural , Crista Neural/metabolismo , Crânio/metabolismo , Desenvolvimento Embrionário/genética , Sulfotransferases/genética , Sulfotransferases/metabolismo , Regulação da Expressão Gênica no Desenvolvimento
6.
Dev Genes Evol ; 2023 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-37815616

RESUMO

As proposed by Wilhelm Roux in 1885, the key goal of experimental embryology ("Entwicklungsmechanik") was to elucidate whether organisms or their parts develop autonomously ("self-differentiation") or require interactions with other parts or the environment. However, experimental embryologists soon realized that concepts like "self-differentiation" only make sense when applied to particular parts or units of the developing embryo as defined both in time and space. Whereas the formation of tissues or organs may initially depend on interactions with surrounding tissues, they later become independent of such interactions or "determined." Moreover, the determination of a particular tissue or organ primordium has to be distinguished from the spatially coordinated determination of its parts-what we now refer to as "patterning." While some primordia depend on extrinsic influences (e.g., signals from adjacent tissues) for proper patterning, others rely on intrinsic mechanisms. Such intrinsically patterned units may behave as "morphogenetic fields" that can compensate for lost parts and regulate their size and proper patterning. While these insights were won by experimental embryologists more than 100 years ago, they retain their relevance today. To enable the generation of more life-like organoids in vitro for studying developmental processes and diseases in a dish, questions about the spatiotemporal units of development (when and how tissues and organs are determined and patterned) need to be increasingly considered. This review briefly sketches this conceptual history and its continued relevance by focusing on the determination and patterning of the inner ear with a specific emphasis on some studies published in this journal.

7.
J Anat ; 243(1): 78-89, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36748120

RESUMO

Live imaging in the zebrafish embryo using tissue-specific expression of fluorescent proteins can yield important insights into the mechanisms that drive sensory organ morphogenesis and cell differentiation. Morphogenesis of the semicircular canal ducts of the vertebrate inner ear requires a complex rearrangement of epithelial cells, including outgrowth, adhesion, fusion and perforation of epithelial projections to generate pillars of tissue that form the hubs of each canal. We report the insertion sites and expression patterns of two enhancer trap lines in the developing zebrafish embryo, each of which highlight different aspects of epithelial cell morphogenesis in the inner ear. A membrane-linked EGFP driven by smad6b regulatory sequences is expressed throughout the otic epithelium, most strongly on the lateral side of the ear and in the sensory cristae. A second enhancer trap line, with cytoplasmic EGFP driven by frizzled1 (fzd1) regulatory sequences, specifically marks cells of the ventral projection and pillar in the developing ear, and marginal cells in the sensory cristae, together with variable expression in the retina and epiphysis, and neurons elsewhere in the developing central nervous system. We have used a combination of methods to identify the insertion sites of these two transgenes, which were generated through random insertion, and show that Targeted Locus Amplification is a rapid and reliable method for the identification of insertion sites of randomly inserted transgenes.


Assuntos
Canais Semicirculares , Peixe-Zebra , Animais , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Epitélio/metabolismo , Morfogênese/fisiologia , Regulação da Expressão Gênica no Desenvolvimento
8.
Dev Biol ; 469: 160-171, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33131705

RESUMO

The inner ear comprises four epithelial domains: the cochlea, vestibule, semicircular canals, and endolymphatic duct/sac. These structures are segregated at embryonic day 13.5 (E13.5). However, these four anatomical structures remain undefined at E10.5. Here, we aimed to identify lineage-specific genes in the early developing inner ear using published data obtained from single-cell RNA-sequencing (scRNA-seq) of embryonic mice. We downloaded 5000 single-cell transcriptome data, named 'auditory epithelial trajectory', from the Mouse Organogenesis Cell Atlas. The dataset was supposed to include otic epithelial cells at E9.5-13.5. We projected the 5000 â€‹cells onto a two-dimensional space encoding the transcriptional state and visualised the pattern of otic epithelial cell differentiation. We identified 15 clusters, which were annotated as one of the four components of the inner ear epithelium using known genes that characterise the four different tissues. Additionally, we classified 15 clusters into sub-regions of the four inner ear components. By comparing transcriptomes between these 15 clusters, we identified several candidates of lineage-specific genes. Characterising these new candidate genes will help future studies about inner ear development.


Assuntos
Orelha Interna/embriologia , Orelha Interna/metabolismo , Animais , Diferenciação Celular/genética , Cóclea/metabolismo , Simulação por Computador , Orelha Interna/citologia , Embrião de Mamíferos/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Hibridização In Situ , Camundongos , Camundongos Endogâmicos ICR , RNA Mensageiro/metabolismo , RNA-Seq , Análise de Célula Única , Vestíbulo do Labirinto/metabolismo
9.
Biochem Biophys Res Commun ; 623: 96-103, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-35878429

RESUMO

The transcription factor FOXG1 plays an important role in inner ear development; however, the cis-regulatory mechanisms controlling the inner-ear-specific expression of FOXG1 are poorly understood. In this study, we aimed to identify the element that specifically regulates FoxG1 expression in the otic vesicle, which develops into the inner ear, through comparative genome analysis between vertebrate species and chromatin immunoprecipitation. The cis-regulatory element (E2) identified showed high evolutionary conservation among vertebrates in the genomic DNA of FoxG1 spanning approximately 3 Mbp. We identified core sequences important for the activity of the otic-vesicle-specific enhancer through in vitro and in vivo reporter assays for various E2 enhancer mutants and determined the consensus sequence for SOX DNA binding. In addition, SoxE, a subfamily of the Sox family, was simultaneously expressed in the otic vesicles of developing embryos and showed a similar protein expression pattern as that of FoxG1. Furthermore, SOXE transcription factors induced specific transcriptional activity through the FoxG1 Otic enhancer (E2b). These findings suggest that the interaction between the otic enhancer of FoxG1 and SOXE transcription factor, in which the otic expression of FoxG1 is evolutionarily well-conserved, is important during early development of the inner ear, a sensory organ important for survival in nature.


Assuntos
Orelha Interna , Fatores de Transcrição SOXE , Animais , DNA/metabolismo , Orelha Interna/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição SOXE/genética , Fatores de Transcrição/metabolismo
10.
Development ; 146(18)2019 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-31488567

RESUMO

The mammalian cochlea develops from a ventral outgrowth of the otic vesicle in response to Shh signaling. Mouse embryos lacking Shh or its essential signal transduction components display cochlear agenesis; however, a detailed understanding of the transcriptional network mediating this process is unclear. Here, we describe an integrated genomic approach to identify Shh-dependent genes and associated regulatory sequences that promote cochlear duct morphogenesis. A comparative transcriptome analysis of otic vesicles from mouse mutants exhibiting loss (Smoecko ) and gain (Shh-P1) of Shh signaling reveal a set of Shh-responsive genes partitioned into four expression categories in the ventral half of the otic vesicle. This target gene classification scheme provides novel insight into several unanticipated roles for Shh, including priming the cochlear epithelium for subsequent sensory development. We also mapped regions of open chromatin in the inner ear by ATAC-seq that, in combination with Gli2 ChIP-seq, identified inner ear enhancers in the vicinity of Shh-responsive genes. These datasets are useful entry points for deciphering Shh-dependent regulatory mechanisms involved in cochlear duct morphogenesis and establishment of its constituent cell types.


Assuntos
Cóclea/embriologia , Cóclea/metabolismo , Genoma , Proteínas Hedgehog/metabolismo , Morfogênese/genética , Animais , Sequência de Bases , Embrião de Mamíferos/metabolismo , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica no Desenvolvimento , Camundongos Transgênicos , Reprodutibilidade dos Testes
11.
Dev Dyn ; 250(11): 1524-1551, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33830554

RESUMO

Progress in understanding mechanisms of inner ear development has been remarkably rapid in recent years. The research community has benefited from the availability of several diverse model organisms, including zebrafish, chick, and mouse. The complexity of the inner ear has proven to be a challenge, and the complexity of the mammalian cochlea in particular has been the subject of intense scrutiny. Zebrafish lack a cochlea and exhibit a number of other differences from amniote species, hence they are sometimes seen as less relevant for inner ear studies. However, accumulating evidence shows that underlying cellular and molecular mechanisms are often highly conserved. As a case in point, consideration of the diverse functions of Fgf and its downstream effectors reveals many similarities between vertebrate species, allowing meaningful comparisons the can benefit the entire research community. In this review, I will discuss mechanisms by which Fgf controls key events in early otic development in zebrafish and provide direct comparisons with chick and mouse.


Assuntos
Orelha Interna , Modelos Animais , Peixe-Zebra , Animais , Orelha Interna/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Mamíferos/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
12.
Genesis ; 59(12): e23453, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34664392

RESUMO

The vertebrate Six (Sine oculis homeobox) family of homeodomain transcription factors plays critical roles in the development of several organs. Six1 plays a central role in cranial placode development, including the precursor tissues of the inner ear, as well as other cranial sensory organs and the kidney. In humans, mutations in SIX1 underlie some cases of Branchio-oto-renal (BOR) syndrome, which is characterized by moderate-to-severe hearing loss. We utilized CRISPR/Cas9 technology to establish a six1 mutant line in Xenopus tropicalis that is available to the research community. We demonstrate that at larval stages, the six1-null animals show severe disruptions in gene expression of putative Six1 target genes in the otic vesicle, cranial ganglia, branchial arch, and neural tube. At tadpole stages, six1-null animals display dysmorphic Meckel's, ceratohyal, and otic capsule cartilage morphology. This mutant line will be of value for the study of the development of several organs as well as congenital syndromes that involve these tissues.


Assuntos
Síndrome Brânquio-Otorrenal/genética , Anormalidades Congênitas/genética , Perda Auditiva/genética , Proteínas de Homeodomínio/genética , Proteínas de Xenopus/genética , Animais , Região Branquial/crescimento & desenvolvimento , Região Branquial/patologia , Síndrome Brânquio-Otorrenal/fisiopatologia , Sistemas CRISPR-Cas/genética , Anormalidades Congênitas/patologia , Desenvolvimento Embrionário/genética , Gânglios Parassimpáticos/crescimento & desenvolvimento , Gânglios Parassimpáticos/patologia , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/genética , Perda Auditiva/fisiopatologia , Humanos , Tubo Neural/crescimento & desenvolvimento , Tubo Neural/patologia , Crânio/crescimento & desenvolvimento , Crânio/patologia , Fatores de Transcrição/genética , Xenopus/genética , Xenopus/crescimento & desenvolvimento
13.
FASEB J ; 32(7): 3984-3992, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29475374

RESUMO

Radial spokes are structurally conserved, macromolecular complexes that are essential for the motility of 9 + 2 motile cilia. In Chlamydomonas species, mutations in radial spoke proteins result in ciliary motility defects. However, little is known about the function of radial spoke proteins during embryonic development. Here, we investigated the role of a novel radial spoke protein, leucine-rich repeat containing protein 23 (Lrrc23), during zebrafish embryonic development. Mutations in lrrc23 resulted in a selective otolith formation defect during early ear development. Similar otolith defects were also present in the radial spoke head 3 homolog ( rsph3) and radial spoke head 4 homolog A ( rsph4a) radial spoke mutants. Notably, the radial spoke protein mutations specifically affected ciliary motility in the otic vesicle (OV), whereas motile cilia in other organs functioned normally. Via high-speed video microscopy, we found that motile cilia formation was stochastic and transient in the OV. Importantly, all the motile cilia in the OV beat circularly, in contrast to the planar beating pattern of typical 9 + 2 motile cilia. We identified the key time frame for motile cilia formation during OV development. Finally, we showed that the functions of radial spoke proteins were conserved between zebrafish and Tetrahymena. Together, our results suggest that radial spoke proteins are essential for ciliary motility in the OV and that radial spoke-regulated OV motile cilia represent a unique type of cilia during early zebrafish embryonic development.-Han, X., Xie, H., Wang, Y., Zhao, C. Radial spoke proteins regulate otolith formation during early zebrafish development.


Assuntos
Cílios/metabolismo , Proteínas do Citoesqueleto/metabolismo , Membrana dos Otólitos/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Proteínas do Citoesqueleto/genética , Mutação , Membrana dos Otólitos/citologia , Membrana dos Otólitos/crescimento & desenvolvimento , Proteínas de Protozoários/metabolismo , Tetrahymena , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
14.
Dev Dyn ; 246(6): 451-465, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28295855

RESUMO

BACKGROUND: Paired organs in animals are largely bilaterally symmetric despite inherent noise in most biological processes. How is precise organ shape and size achieved during development despite this noise? Examining paired organ development is a challenge because it requires repeated quantification of two structures in parallel within living embryos. Here we combine bilateral quantification of morphology through time with asymmetric perturbations to study regulation of organ shape, size, and symmetry in developing organ pairs. RESULTS: We present quantitative live imaging tools to measure the shape and size of the developing inner ears on both the left and right side simultaneously over time. By quantifying variation between the left and right inner ear (intrinsic noise) and between different individuals (extrinsic noise), we find that initial variability decreases over time in normal development to achieve symmetry. Early asymmetry is increased by environmental stress, but symmetry is still recovered over subsequent developmental time. Using multiple unilateral perturbations including Fgf signaling and ultraviolet light, we find that growth can be adjusted to compensate for a range of initial size and shape differences. CONCLUSIONS: We propose that symmetry in developmental systems does not emerge through precise deterministic bilateral development, but rather through feedback mechanisms that adjust morphogenesis rates to account for variation. Developmental Dynamics 246:451-465, 2016. © 2017 Wiley Periodicals, Inc.


Assuntos
Orelha Interna/crescimento & desenvolvimento , Morfogênese , Organogênese/fisiologia , Animais , Orelha Interna/anatomia & histologia , Orelha Interna/embriologia , Retroalimentação Fisiológica , Microscopia Confocal , Tempo , Peixe-Zebra
15.
J Neurosci Res ; 95(5): 1194-1203, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28105691

RESUMO

Fetal alcohol exposure can cause Fetal Alcohol Spectrum Disorders (FASD), completely preventable developmental disabilities characterized by permanent birth defects. However, specific gestational timing when developing organs are most sensitive to alcohol exposure is unclear. In this study, we examined the temporal effects of embryonic alcohol exposure on octavolateral organs in zebrafish (Danio rerio), including inner ears and lateral line neuromasts that function in hearing, balance, and hydrodynamic detection, respectively. To determine an alcohol-sensitive period in the first 24 hours post fertilization (hpf), Et(krt4:EGFP)sqet4 zebrafish that express green fluorescent protein in sensory hair cells were treated in 2% alcohol for 2, 3, and 5-hours. Octavolateral organs of control and alcohol-exposed larvae were examined at 3, 5, and 7 days post fertilization (dpf). Using confocal and light microscopy, we found that alcohol-exposed larvae had significantly smaller otic vesicles and saccular otoliths than control larvae at 3 dpf. Only alcohol-exposed larvae from 12-17 hpf had smaller otic vesicles at 5 dpf, smaller saccular otoliths at 7 dpf and fewer saccular hair cells, neuromasts and hair cells per neuromast at 3 dpf. In addition, auditory function was assessed by microphonic potential recordings from inner ear hair cells in response to 200-Hz stimulation. Hearing sensitivity was reduced for alcohol-exposed larvae from 7-12 and 12-17 hpf. Our results show that 12-17 hpf is an alcohol-sensitive time window when morphology and function of zebrafish octavolateral organs are most vulnerable to alcohol exposure. This study implies that embryonic alcohol exposure timing during early development can influence severity of hearing deficits. © 2017 Wiley Periodicals, Inc.


Assuntos
Depressores do Sistema Nervoso Central/toxicidade , Orelha Interna/efeitos dos fármacos , Etanol/toxicidade , Células Ciliadas Auditivas/efeitos dos fármacos , Audição/efeitos dos fármacos , Fatores Etários , Análise de Variância , Animais , Animais Geneticamente Modificados , Orelha Interna/embriologia , Embrião não Mamífero , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Audição/fisiologia , Perda Auditiva/induzido quimicamente , Queratina-4/genética , Queratina-4/metabolismo , Larva/efeitos dos fármacos , Sistema da Linha Lateral/efeitos dos fármacos , Sistema da Linha Lateral/embriologia , Peixe-Zebra
16.
Biochim Biophys Acta ; 1839(6): 425-37, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24768923

RESUMO

AGR2 is a member of the protein disulfide isomerase (PDI) family, which is implicated in cancer cell growth and metastasis, asthma, and inflammatory bowel disease. Despite the contributions of this protein to several biological processes, the regulatory mechanisms controlling expression of the AGR2 gene in different organs remain unclear. Zebrafish anterior gradient 2 (agr2) is expressed in several organs, including the otic vesicles that contain mucus-secreting cells. To elucidate the regulatory mechanisms controlling agr2 expression in otic vesicles, we generated a Tg(-6.0 k agr2:EGFP) transgenic fish line that expressed EGFP in a pattern recapitulating that of agr2. Double immunofluorescence studies were used to demonstrate that Agr2 and GFP colocalize in the semicircular canals and supporting cells of all sensory patches in the otic vesicles of Tg(-6.0 k agr2:EGFP) embryos. Transient/stable transgenic analyses coupled with 5'-end deletion revealed that a 100 bp sequence within the -2.6 to -2.5 kbp region upstream of agr2 directs EGFP expression specifically in the otic vesicles. Two HMG-binding motifs were detected in this region. Mutation of these motifs prevented EGFP expression. Furthermore, EGFP expression in the otic vesicles was prevented by knockdown of the sox10 gene. This corresponded with decreased agr2 expression in the otic vesicles of sox10 morphants during different developmental stages. Electrophoretic mobility shift assays were used to show that Sox10 binds to HMG-binding motifs located within the -2.6 to -2.5 kbp region upstream of agr2. These results demonstrate that agr2 expression in the otic vesicles of zebrafish embryos is regulated by Sox10.


Assuntos
Orelha/fisiologia , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição SOXE/metabolismo , Canais Semicirculares/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Ensaio de Desvio de Mobilidade Eletroforética , Embrião não Mamífero/citologia , Imunofluorescência , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hibridização In Situ , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição SOXE/genética , Canais Semicirculares/citologia , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
17.
Dev Dyn ; 243(10): 1275-85, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24677486

RESUMO

BACKGROUND: Vertebrate otic and epibranchial placodes develop in close proximity in response to localized fibroblast growth factor (Fgf) signaling. Although less is known about epibranchial induction, the process of otic induction in highly conserved, with important roles for Fgf3 and Fgf8 reported in all species examined. Fgf10 is also critical for otic induction in mouse, but the only zebrafish ortholog examined to date, fgf10a, is not expressed early enough to play such a role. A second zebrafish ortholog, fgf10b, has not been previously examined. RESULTS: We find that zebrafish fgf10b is expressed at tailbud stage in paraxial cephalic mesoderm beneath prospective epibranchial tissue, lateral to the developing otic placode. Knockdown of fgf10b does not affect initial otic induction but impairs subsequent accumulation of otic cells. Formation of epibranchial placodes and ganglia are also moderately impaired. Combinatorial disruption of fgf10b and fgf3 exacerbates the deficiency of otic cells and eliminates epibranchial induction entirely. Disruption of fgf10b and fgf24 also strongly reduces, but does not eliminate, epibranchial induction. CONCLUSIONS: fgf10b participates in a late phase of otic induction and, in combination with fgf3, is especially critical for epibranchial induction.


Assuntos
Região Branquial/embriologia , Orelha/embriologia , Indução Embrionária/genética , Fator 3 de Crescimento de Fibroblastos/fisiologia , Fatores de Crescimento de Fibroblastos/fisiologia , Mesoderma/metabolismo , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra , Animais , Animais Geneticamente Modificados , Padronização Corporal/genética , Região Branquial/metabolismo , Embrião não Mamífero , Fator 10 de Crescimento de Fibroblastos/fisiologia , Peixe-Zebra/embriologia , Peixe-Zebra/genética
18.
Aquat Toxicol ; 259: 106539, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37086653

RESUMO

Dioxins are a class of highly toxic and persistent environmental pollutants that have been shown through epidemiological and laboratory-based studies to act as developmental teratogens. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), the most potent dioxin congener, has a high affinity for the aryl hydrocarbon receptor (AHR), a ligand activated transcription factor. TCDD-induced AHR activation during development impairs nervous system, cardiac, and craniofacial development. Despite the robust phenotypes previously reported, the characterization of developmental malformations and our understanding of the molecular targets mediating TCDD-induced developmental toxicity remains limited. In zebrafish, TCDD-induced craniofacial malformations are produced, in part, by the downregulation of SRY-box transcription factor 9b (sox9b), a member of the SoxE gene family. sox9b, along with fellow SoxE gene family members sox9a and sox10, have important functions in the development of the otic placode, the otic vesicle, and, ultimately, the inner ear. Given that sox9b is a known target of TCDD and that transcriptional interactions exist among SoxE genes, we asked whether TCDD exposure impaired the development of the zebrafish auditory system, specifically the otic vesicle, which gives rise to the sensory components of the inner ear. Using immunohistochemistry, in vivo confocal imaging, and time-lapse microscopy, we assessed the impact of TCDD exposure on zebrafish otic vesicle development. We found exposure resulted in structural deficits, including incomplete pillar fusion and altered pillar topography, leading to defective semicircular canal development. The observed structural deficits were accompanied by reduced collagen type II expression in the ear. Together, our findings reveal the otic vesicle as a novel target of TCDD-induced toxicity, suggest that the function of multiple SoxE genes may be affected by TCDD exposure, and provide insight into how environmental contaminants contribute to congenital malformations.


Assuntos
Dioxinas , Orelha Interna , Dibenzodioxinas Policloradas , Poluentes Químicos da Água , Animais , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Dibenzodioxinas Policloradas/toxicidade , Dioxinas/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Poluentes Orgânicos Persistentes/metabolismo , Poluentes Químicos da Água/toxicidade , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/metabolismo , Orelha Interna/metabolismo
19.
Cell Rep ; 42(6): 112545, 2023 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-37227818

RESUMO

An abundance of research has recently highlighted the susceptibility of cochleovestibular ganglion (CVG) neurons to noise damage and aging in the adult cochlea, resulting in hearing deficits. Furthering our understanding of the transcriptional cascades that contribute to CVG development may provide insight into how these cells can be regenerated to treat inner ear dysfunction. Here we perform a high-depth single-cell RNA sequencing analysis of the E10.5 otic vesicle and its surrounding tissues, including CVG precursor neuroblasts and emerging CVG neurons. Clustering and trajectory analysis of otic-lineage cells reveals otic markers and the changes in gene expression that occur from neuroblast delamination toward the development of the CVG. This dataset provides a valuable resource for further identifying the mechanisms associated with CVG development from neurosensory competent cells within the otic vesicle.


Assuntos
Cóclea , Células-Tronco Neurais , Camundongos , Animais , Células-Tronco Neurais/metabolismo , Neurônios , Regulação da Expressão Gênica no Desenvolvimento
20.
Front Cell Dev Biol ; 11: 1274788, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37854072

RESUMO

Introduction: The Six1 transcription factor plays important roles in the development of cranial sensory organs, and point mutations underlie craniofacial birth defects. Because Six1's transcriptional activity can be modulated by interacting proteins, we previously screened for candidate interactors and identified zinc-finger MYM-containing protein 4 (Zmym4) by its inclusion of a few domains with a bona fide cofactor, Sine oculis binding protein (Sobp). Although Zmym4 has been implicated in regulating early brain development and certain cancers, its role in craniofacial development has not previously been described. Methods: We used co-immunoprecipitation and luciferase-reporter assays in cultured cells to test interactions between Zmym4 and Six1. We used knock-down and overexpression of Zmym4 in embryos to test for its effects on early ectodermal gene expression, neural crest migration and craniofacial cartilage formation. Results: We found no evidence that Zmym4 physically or transcriptionally interacts with Six1 in cultured cells. Nonetheless, knockdown of endogenous Zmym4 in embryos resulted in altered early cranial gene expression, including those expressed in the neural border, neural plate, neural crest and preplacodal ectoderm. Experimentally increasing Zmym4 levels had minor effects on neural border or neural plate genes, but altered the expression of neural crest and preplacodal genes. At larval stages, genes expressed in the otic vesicle and branchial arches showed reduced expression in Zmym4 morphants. Although we did not detect defects in neural crest migration into the branchial arches, loss of Zmym4 resulted in aberrant morphology of several craniofacial cartilages. Discussion: Although Zmym4 does not appear to function as a Six1 transcriptional cofactor, it plays an important role in regulating the expression of embryonic cranial genes in tissues critical for normal craniofacial development.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa